首页 > 最新文献

Cancer immunology research最新文献

英文 中文
CD4+ Regulatory T Cells in Human Cancer: Subsets, Origin, and Molecular Regulation. 人类癌症中的 CD4+ 调节性 T 细胞:亚群、起源和分子调控
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-02 DOI: 10.1158/2326-6066.CIR-23-0517
Julian Swatler, Marco De Luca, Ivano Rotella, Veronica Lise, Emilia Maria Cristina Mazza, Enrico Lugli

CD4+CD25hiFOXP3+ regulatory T cells (Treg) play major roles in the maintenance of immune tolerance, prevention of inflammation, and tissue homeostasis and repair. In contrast with these beneficial roles, Tregs are abundant in virtually all tumors and have been mechanistically linked to disease progression, metastases development, and therapy resistance. Tregs are thus recognized as a major target for cancer immunotherapy. Compared with other sites in the body, tumors harbor hyperactivated Treg subsets whose molecular characteristics are only beginning to be elucidated. Here, we describe current knowledge of intratumoral Tregs and discuss their potential cellular and tissue origin. Furthermore, we describe currently recognized molecular regulators that drive differentiation and maintenance of Tregs in cancer, with a special focus on those signals regulating their chronic immune activation, with relevant implications for cancer progression and therapy.

CD4+CD25hiFOXP3+ 调节性 T 细胞(Treg)在维持免疫耐受、预防炎症、组织稳态和修复方面发挥着重要作用。与这些有益作用形成鲜明对比的是,调节性 Tregs 在几乎所有肿瘤中都很丰富,而且在机理上与疾病进展、转移发展和耐药性有关。因此,Tregs 被认为是癌症免疫疗法的主要靶点。与体内其他部位相比,肿瘤蕴藏着过度激活的 Treg 亚群,其分子特征才刚刚开始被阐明。在此,我们将介绍目前对肿瘤内 Tregs 的了解,并讨论其潜在的细胞和组织来源。此外,我们还描述了目前公认的驱动肿瘤内 Tregs 分化和维持的分子调控因子,并特别关注调控其慢性免疫激活的信号,这些信号对癌症的进展和治疗具有重要意义。
{"title":"CD4+ Regulatory T Cells in Human Cancer: Subsets, Origin, and Molecular Regulation.","authors":"Julian Swatler, Marco De Luca, Ivano Rotella, Veronica Lise, Emilia Maria Cristina Mazza, Enrico Lugli","doi":"10.1158/2326-6066.CIR-23-0517","DOIUrl":"10.1158/2326-6066.CIR-23-0517","url":null,"abstract":"<p><p>CD4+CD25hiFOXP3+ regulatory T cells (Treg) play major roles in the maintenance of immune tolerance, prevention of inflammation, and tissue homeostasis and repair. In contrast with these beneficial roles, Tregs are abundant in virtually all tumors and have been mechanistically linked to disease progression, metastases development, and therapy resistance. Tregs are thus recognized as a major target for cancer immunotherapy. Compared with other sites in the body, tumors harbor hyperactivated Treg subsets whose molecular characteristics are only beginning to be elucidated. Here, we describe current knowledge of intratumoral Tregs and discuss their potential cellular and tissue origin. Furthermore, we describe currently recognized molecular regulators that drive differentiation and maintenance of Tregs in cancer, with a special focus on those signals regulating their chronic immune activation, with relevant implications for cancer progression and therapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140334758","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Immune Suppressor IGSF1 as a Potential Target for Cancer Immunotherapy. 作为癌症免疫疗法潜在靶点的免疫抑制因子 IGSF1。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-02 DOI: 10.1158/2326-6066.CIR-23-0817
Dong-In Koh, Minki Lee, Yoon Sun Park, Jae-Sik Shin, Joseph Kim, Yea Seong Ryu, Jun Hyung Lee, Seunggeon Bae, Mi So Lee, Jun Ki Hong, Hong-Rae Jeong, Mingee Choi, Seung-Woo Hong, Dong Kwan Kim, Hyun-Kyung Lee, Bomi Kim, Yoo Sang Yoon, Dong-Hoon Jin

The development of first-generation immune-checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 ushered in a new era in anticancer therapy. Although immune-checkpoint blockade therapies have shown clinical success, a substantial number of patients yet fail to benefit. Many studies are under way to discover next-generation immunotherapeutic targets. Immunoglobulin superfamily member 1 (IGSF1) is a membrane glycoprotein proposed to regulate thyroid function. Despite containing 12 immunoglobin domains, a possible role for IGSF1, in immune response, remains unknown. Here, our studies revealed that IGSF1 is predominantly expressed in tumors but not normal tissues, and increased expression is observed in PD-L1low non-small cell lung cancer (NSCLC) cells as compared with PD-L1high cells. Subsequently, we developed and characterized an IGSF1-specific human monoclonal antibody, WM-A1, that effectively promoted antitumor immunity and overcame the limitations of first-generation immune-checkpoint inhibitors, likely via a distinct mechanism of action. We further demonstrated high WM-A1 efficacy in humanized peripheral blood mononuclear cells (PBMC), and syngeneic mouse models, finding additive efficacy in combination with an anti-PD-1 (a well-characterized checkpoint inhibitor). These findings support IGSF1 as an immune target that might complement existing cancer immunotherapeutics.

以 PD-1/PD-L1 和 CTLA-4 为靶点的第一代免疫检查点抑制剂的开发开创了抗癌疗法的新纪元。虽然免疫检查点阻断疗法在临床上取得了成功,但仍有大量患者未能从中获益。根据最近的报道,许多发现下一代免疫治疗靶点的研究正在进行中。在这里,我们通过蛋白质组分析发现了一个新的免疫治疗靶点--IGSF1(免疫球蛋白超家族成员1),它是一种膜糖蛋白,被认为能调节甲状腺功能。尽管IGSF1含有12个免疫球蛋白结构域,但它在免疫反应中可能扮演的角色仍然未知。我们的研究发现,IGSF1主要在肿瘤中表达,而不在正常组织中表达,与PD-L1高表达的细胞相比,IGSF1在PD-L1低表达的NSCLC细胞中表达增加。随后,我们开发并鉴定了一种IGSF1特异性人单克隆抗体WM-A1,它能有效促进抗肿瘤免疫,克服第一代免疫检查点抑制剂的局限性,可能是通过一种独特的作用机制。我们进一步证明了 WM-A1 在人源化外周血单核细胞(PBMCs)和合成小鼠模型中的高效力,发现它与抗 PD-1 抗体(一种特征明显的检查点抑制剂)联合使用具有协同增效作用。这些研究结果支持将 IGSF1 作为一种新型免疫靶点,以补充现有的癌症免疫疗法。
{"title":"The Immune Suppressor IGSF1 as a Potential Target for Cancer Immunotherapy.","authors":"Dong-In Koh, Minki Lee, Yoon Sun Park, Jae-Sik Shin, Joseph Kim, Yea Seong Ryu, Jun Hyung Lee, Seunggeon Bae, Mi So Lee, Jun Ki Hong, Hong-Rae Jeong, Mingee Choi, Seung-Woo Hong, Dong Kwan Kim, Hyun-Kyung Lee, Bomi Kim, Yoo Sang Yoon, Dong-Hoon Jin","doi":"10.1158/2326-6066.CIR-23-0817","DOIUrl":"10.1158/2326-6066.CIR-23-0817","url":null,"abstract":"<p><p>The development of first-generation immune-checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 ushered in a new era in anticancer therapy. Although immune-checkpoint blockade therapies have shown clinical success, a substantial number of patients yet fail to benefit. Many studies are under way to discover next-generation immunotherapeutic targets. Immunoglobulin superfamily member 1 (IGSF1) is a membrane glycoprotein proposed to regulate thyroid function. Despite containing 12 immunoglobin domains, a possible role for IGSF1, in immune response, remains unknown. Here, our studies revealed that IGSF1 is predominantly expressed in tumors but not normal tissues, and increased expression is observed in PD-L1low non-small cell lung cancer (NSCLC) cells as compared with PD-L1high cells. Subsequently, we developed and characterized an IGSF1-specific human monoclonal antibody, WM-A1, that effectively promoted antitumor immunity and overcame the limitations of first-generation immune-checkpoint inhibitors, likely via a distinct mechanism of action. We further demonstrated high WM-A1 efficacy in humanized peripheral blood mononuclear cells (PBMC), and syngeneic mouse models, finding additive efficacy in combination with an anti-PD-1 (a well-characterized checkpoint inhibitor). These findings support IGSF1 as an immune target that might complement existing cancer immunotherapeutics.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139575187","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
Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer. 通过对癌症患者进行地诺单抗治疗,用 RANKL 阻断剂进行免疫调节。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-02 DOI: 10.1158/2326-6066.CIR-23-0184
Hewitt Chang, Jaqueline Marquez, Brandon K Chen, Daniel M Kim, Michael L Cheng, Eric V Liu, Hai Yang, Li Zhang, Meenal Sinha, Alexander Cheung, Serena S Kwek, Eric D Chow, Mark Bridge, Rahul R Aggarwal, Terence W Friedlander, Eric J Small, Mark Anderson, Lawrence Fong

Denosumab is a fully human mAb that binds receptor activator of NFκB ligand (RANKL). It is routinely administered to patients with cancer to reduce the incidence of new bone metastasis. RANK-RANKL interactions regulate bone turnover by controlling osteoclast recruitment, development, and activity. However, these interactions also can regulate immune cells including dendritic cells and medullary thymic epithelial cells. Inhibition of the latter results in reduced thymic negative selection of T cells and could enhance the generation of tumor-specific T cells. We examined whether administering denosumab could modify modulate circulating immune cells in patients with cancer. Blood was collected from 23 patients with prostate cancer and 3 patients with renal cell carcinoma, all of whom had advanced disease and were receiving denosumab, prior to and during denosumab treatment. Using high-dimensional mass cytometry, we found that denosumab treatment by itself induced modest effects on circulating immune cell frequency and activation. We also found minimal changes in the circulating T-cell repertoire and the frequency of new thymic emigrants with denosumab treatment. However, when we stratified patients by whether they were receiving chemotherapy and/or steroids, patients receiving these concomitant treatments showed significantly greater immune modulation, including an increase in the frequency of natural killer cells early and classical monocytes later. We also saw broad induction of CTLA-4 and TIM3 expression in circulating lymphocytes and some monocyte populations. These findings suggest that denosumab treatment by itself has modest immunomodulatory effects, but when combined with conventional cancer treatments, can lead to the induction of immunologic checkpoints. See related Spotlight by Nasrollahi and Davar, p. 383.

地诺单抗是一种全人源单克隆抗体,能与核因子κB受体激活剂配体(RANKL)结合。它是癌症患者的常规用药,可降低新的骨转移发生率。RANK-RANKL 的相互作用通过控制破骨细胞的招募、发育和活性来调节骨转换。不过,这些相互作用也能调节免疫细胞,包括树突状细胞和髓质胸腺上皮细胞(mTECs)。抑制后者会导致胸腺负性选择 T 细胞减少,并可能促进肿瘤特异性 T 细胞的生成。我们研究了使用地诺单抗是否能改变癌症患者循环免疫细胞的变化。我们采集了 23 名前列腺癌患者和 3 名肾细胞癌患者的血液,这些患者均为晚期患者,正在接受地诺单抗治疗。通过使用高维质谱仪,我们发现地诺单抗治疗本身对循环免疫细胞的频率和活化影响不大。我们还发现,在使用地诺单抗治疗时,循环 T 细胞群和新胸腺移植物的频率变化极小。然而,当我们根据患者是否接受化疗和/或类固醇治疗对其进行分层时,同时接受这些治疗的患者表现出的免疫调节明显更强,包括早期 NK 细胞频率的增加和后期经典单核细胞频率的增加。我们还发现循环淋巴细胞和一些单核细胞群中 CTLA-4 和 TIM3 的表达受到广泛诱导。这些研究结果表明,地诺单抗治疗本身具有适度的免疫调节作用,但如果与常规癌症治疗相结合,则可诱导免疫检查点。
{"title":"Immune Modulation with RANKL Blockade through Denosumab Treatment in Patients with Cancer.","authors":"Hewitt Chang, Jaqueline Marquez, Brandon K Chen, Daniel M Kim, Michael L Cheng, Eric V Liu, Hai Yang, Li Zhang, Meenal Sinha, Alexander Cheung, Serena S Kwek, Eric D Chow, Mark Bridge, Rahul R Aggarwal, Terence W Friedlander, Eric J Small, Mark Anderson, Lawrence Fong","doi":"10.1158/2326-6066.CIR-23-0184","DOIUrl":"10.1158/2326-6066.CIR-23-0184","url":null,"abstract":"<p><p>Denosumab is a fully human mAb that binds receptor activator of NFκB ligand (RANKL). It is routinely administered to patients with cancer to reduce the incidence of new bone metastasis. RANK-RANKL interactions regulate bone turnover by controlling osteoclast recruitment, development, and activity. However, these interactions also can regulate immune cells including dendritic cells and medullary thymic epithelial cells. Inhibition of the latter results in reduced thymic negative selection of T cells and could enhance the generation of tumor-specific T cells. We examined whether administering denosumab could modify modulate circulating immune cells in patients with cancer. Blood was collected from 23 patients with prostate cancer and 3 patients with renal cell carcinoma, all of whom had advanced disease and were receiving denosumab, prior to and during denosumab treatment. Using high-dimensional mass cytometry, we found that denosumab treatment by itself induced modest effects on circulating immune cell frequency and activation. We also found minimal changes in the circulating T-cell repertoire and the frequency of new thymic emigrants with denosumab treatment. However, when we stratified patients by whether they were receiving chemotherapy and/or steroids, patients receiving these concomitant treatments showed significantly greater immune modulation, including an increase in the frequency of natural killer cells early and classical monocytes later. We also saw broad induction of CTLA-4 and TIM3 expression in circulating lymphocytes and some monocyte populations. These findings suggest that denosumab treatment by itself has modest immunomodulatory effects, but when combined with conventional cancer treatments, can lead to the induction of immunologic checkpoints. See related Spotlight by Nasrollahi and Davar, p. 383.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10993769/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139563555","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
Immunomodulatory Effects of RANK/RANKL Blockade in Patients with Cancer. RANK/RANKL 阻断剂对癌症患者的免疫调节作用
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-02 DOI: 10.1158/2326-6066.CIR-23-1091
Elham Nasrollahi, Diwakar Davar

In cancer, multiple factors converge upon receptor activator of nuclear factor κB (RANK) and its ligand (RANKL) signaling to promote the development of bone metastases; agents that inhibit RANKL signaling reduce skeletal-related events (SRE) in patients with cancer. In addition, RANKL signaling is important in augmenting the ability of dendritic cells (DC) to stimulate both naïve T-cell proliferation and the survival of RANK+ T cells. In this issue, Chang and colleagues using high-dimensional cytometry to evaluate immunomodulatory effects of denosumab in patients with advanced solid, observe early on treatment changes in multiple compartments, and greater effects in patients receiving concurrent chemotherapy or steroids. See related article by Chang et al., p. 453 (4).

在癌症中,核因子κB受体活化因子(RANK)及其配体(RANKL)信号传导会汇聚多种因素,促进骨转移的发生;抑制RANKL信号传导的药物可减少癌症患者骨骼相关事件(SRE)的发生。此外,RANKL信号在增强树突状细胞(DC)刺激幼稚T细胞增殖和RANK+ T细胞存活的能力方面也很重要。在本期的文章中,Chang及其同事使用高维细胞测定法评估了地诺单抗对晚期实体瘤患者的免疫调节作用,观察到治疗早期多个分区的变化,以及对同时接受化疗或类固醇治疗的患者的更大作用。参见Chang等人的相关文章(4)。
{"title":"Immunomodulatory Effects of RANK/RANKL Blockade in Patients with Cancer.","authors":"Elham Nasrollahi, Diwakar Davar","doi":"10.1158/2326-6066.CIR-23-1091","DOIUrl":"10.1158/2326-6066.CIR-23-1091","url":null,"abstract":"<p><p>In cancer, multiple factors converge upon receptor activator of nuclear factor κB (RANK) and its ligand (RANKL) signaling to promote the development of bone metastases; agents that inhibit RANKL signaling reduce skeletal-related events (SRE) in patients with cancer. In addition, RANKL signaling is important in augmenting the ability of dendritic cells (DC) to stimulate both naïve T-cell proliferation and the survival of RANK+ T cells. In this issue, Chang and colleagues using high-dimensional cytometry to evaluate immunomodulatory effects of denosumab in patients with advanced solid, observe early on treatment changes in multiple compartments, and greater effects in patients receiving concurrent chemotherapy or steroids. See related article by Chang et al., p. 453 (4).</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139905139","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
Dual Chimeric Antigen Receptor T Cells Targeting CD38 and SLAMF7 with Independent Signaling Demonstrate Preclinical Efficacy and Safety in Multiple Myeloma. 靶向 CD38 和 SLAMF7 的双嵌合抗原受体 T 细胞具有独立信号传导功能,在多发性骨髓瘤中显示出临床前疗效和安全性。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-02 DOI: 10.1158/2326-6066.CIR-23-0839
Nathalie Roders, Cecilia Nakid-Cordero, Fabio Raineri, Maxime Fayon, Audrey Abecassis, Caroline Choisy, Elisabeth Nelson, Claire Maillard, David Garrick, Alexis Talbot, Jean-Paul Fermand, Bertrand Arnulf, Jean-Christophe Bories

Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma targeting B-cell maturation antigen (BCMA) induces high overall response rates. However, relapse still occurs and novel strategies for targeting multiple myeloma cells using CAR T-cell therapy are needed. SLAMF7 (also known as CS1) and CD38 on tumor plasma cells represent potential alternative targets for CAR T-cell therapy in multiple myeloma, but their expression on activated T cells and other hematopoietic cells raises concerns about the efficacy and safety of such treatments. Here, we used CRISPR/Cas9 deletion of the CD38 gene in T cells and developed DCAR, a double CAR system targeting CD38 and CS1 through activation and costimulation receptors, respectively. Inactivation of CD38 enhanced the anti-multiple myeloma activity of DCAR T in vitro. Edited DCAR T cells showed strong in vitro and in vivo responses specifically against target cells expressing both CD38 and CS1. Furthermore, we provide evidence that, unlike anti-CD38 CAR T-cell therapy, which elicited a rapid immune reaction against hematopoietic cells in a humanized mouse model, DCAR T cells showed no signs of toxicity. Thus, DCAR T cells could provide a safe and efficient alternative to anti-BCMA CAR T-cell therapy to treat patients with multiple myeloma.

以B细胞成熟抗原(BCMA)为靶点的嵌合抗原受体T细胞(CAR-T)疗法治疗多发性骨髓瘤(MM)的总体反应率很高。然而,复发仍时有发生,因此需要使用 CAR-T 靶向 MM 细胞的新策略。肿瘤浆细胞上的SLAMF7(又称CS1)和CD38是CAR-T治疗MM的潜在替代靶点,但它们在活化T细胞和其他造血细胞上的表达引起了人们对此类治疗的有效性和安全性的担忧。在这里,我们利用 CRISPR/Cas9 技术删除了 T 细胞中的 CD38 基因,并开发出了 DCAR,一种分别通过活化受体和共刺激受体靶向 CD38 和 CS1 的双 CAR 系统。CD38失活增强了DCAR-T的体外抗MM活性。编辑后的 DCAR-T 在体外和体内都表现出了针对同时表达 CD38 和 CS1 的靶细胞的强烈特异性反应。此外,我们还提供证据表明,在人源化小鼠模型中,抗 CD38 CAR-T 会引起针对造血细胞的快速免疫反应,而 DCAR-T 则不同,它没有毒性迹象。因此,DCAR-T 可以安全有效地替代抗 BCMA CAR-T 治疗 MM 患者。
{"title":"Dual Chimeric Antigen Receptor T Cells Targeting CD38 and SLAMF7 with Independent Signaling Demonstrate Preclinical Efficacy and Safety in Multiple Myeloma.","authors":"Nathalie Roders, Cecilia Nakid-Cordero, Fabio Raineri, Maxime Fayon, Audrey Abecassis, Caroline Choisy, Elisabeth Nelson, Claire Maillard, David Garrick, Alexis Talbot, Jean-Paul Fermand, Bertrand Arnulf, Jean-Christophe Bories","doi":"10.1158/2326-6066.CIR-23-0839","DOIUrl":"10.1158/2326-6066.CIR-23-0839","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma targeting B-cell maturation antigen (BCMA) induces high overall response rates. However, relapse still occurs and novel strategies for targeting multiple myeloma cells using CAR T-cell therapy are needed. SLAMF7 (also known as CS1) and CD38 on tumor plasma cells represent potential alternative targets for CAR T-cell therapy in multiple myeloma, but their expression on activated T cells and other hematopoietic cells raises concerns about the efficacy and safety of such treatments. Here, we used CRISPR/Cas9 deletion of the CD38 gene in T cells and developed DCAR, a double CAR system targeting CD38 and CS1 through activation and costimulation receptors, respectively. Inactivation of CD38 enhanced the anti-multiple myeloma activity of DCAR T in vitro. Edited DCAR T cells showed strong in vitro and in vivo responses specifically against target cells expressing both CD38 and CS1. Furthermore, we provide evidence that, unlike anti-CD38 CAR T-cell therapy, which elicited a rapid immune reaction against hematopoietic cells in a humanized mouse model, DCAR T cells showed no signs of toxicity. Thus, DCAR T cells could provide a safe and efficient alternative to anti-BCMA CAR T-cell therapy to treat patients with multiple myeloma.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139575186","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
Allogeneic CAR T Cells: Complex Cellular Therapy Designs Test the Limits of Our Preclinical Models. 异体 CAR T 细胞:复杂的细胞疗法设计考验我们临床前模型的极限。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-02 DOI: 10.1158/2326-6066.CIR-24-0204
Paolo F Caimi, Jan Joseph Melenhorst

All chimeric antigen receptor (CAR) T-cell products currently approved by the FDA are autologous, which poses several challenges for widespread use. In this issue, Degagné and colleagues present their preclinical research on creating off-the-shelf CAR T cells for multiple myeloma. They utilized the CRISPR/Cas12a genome editing platform and gene knock-in techniques to eliminate alloreactivity and decrease susceptibility to natural killer (NK)-cell elimination. This work has led to an ongoing phase I trial of off-the-shelf CAR T cells for multiple myeloma treatment. See related article by Degagné et al., p. 462 (2).

目前美国食品及药物管理局批准的所有嵌合抗原受体(CAR)T细胞产品都是自体细胞,这给广泛应用带来了一些挑战。在本期杂志中,Degagné及其同事介绍了他们针对多发性骨髓瘤创建现成CAR T细胞的临床前研究。他们利用CRISPR/Cas12a基因组编辑平台和基因敲入技术消除异体活性,降低对自然杀伤(NK)细胞的易感性。这项工作促使现成的 CAR T 细胞用于多发性骨髓瘤治疗的 I 期试验正在进行中。参见 Degagné 等人的相关文章,第 462 页(2)。
{"title":"Allogeneic CAR T Cells: Complex Cellular Therapy Designs Test the Limits of Our Preclinical Models.","authors":"Paolo F Caimi, Jan Joseph Melenhorst","doi":"10.1158/2326-6066.CIR-24-0204","DOIUrl":"10.1158/2326-6066.CIR-24-0204","url":null,"abstract":"<p><p>All chimeric antigen receptor (CAR) T-cell products currently approved by the FDA are autologous, which poses several challenges for widespread use. In this issue, Degagné and colleagues present their preclinical research on creating off-the-shelf CAR T cells for multiple myeloma. They utilized the CRISPR/Cas12a genome editing platform and gene knock-in techniques to eliminate alloreactivity and decrease susceptibility to natural killer (NK)-cell elimination. This work has led to an ongoing phase I trial of off-the-shelf CAR T cells for multiple myeloma treatment. See related article by Degagné et al., p. 462 (2).</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140334757","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
Engineering CD3/CD137 Dual Specificity into a DLL3-Targeted T-Cell Engager Enhances T-Cell Infiltration and Efficacy against Small-Cell Lung Cancer. 将 CD3/CD137 双特异性工程化为 DLL3 靶向 T 细胞吸引器,增强 T 细胞浸润和对小细胞肺癌的疗效
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-04-01 DOI: 10.1158/2326-6066.CIR-23-0638
Hirofumi Mikami, Shu Feng, Yutaka Matsuda, Shinya Ishii, Sotaro Naoi, Yumiko Azuma, Hiroaki Nagano, Kentaro Asanuma, Yoko Kayukawa, Toshiaki Tsunenari, Shogo Kamikawaji, Ryutaro Iwabuchi, Junko Shinozuka, Masaki Yamazaki, Haruka Kuroi, Samantha Shu Wen Ho, Siok Wan Gan, Priyanka Chichili, Chai Ling Pang, Chiew Ying Yeo, Shun Shimizu, Naoka Hironiwa, Yasuko Kinoshita, Yuichiro Shimizu, Akihisa Sakamoto, Masaru Muraoka, Noriyuki Takahashi, Tatsuya Kawa, Hirotake Shiraiwa, Futa Mimoto, Kenji Kashima, Mika Kamata-Sakurai, Shumpei Ishikawa, Hiroyuki Aburatani, Takehisa Kitazawa, Tomoyuki Igawa

Small-cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICI) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all patients with SCLC are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared with a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.

小细胞肺癌(SCLC)是一种侵袭性癌症,免疫检查点抑制剂(ICI)对其治疗效果有限。双特异性 T 细胞捕获剂是治疗对 ICI 耐药的肿瘤的有前途的替代疗法,但并非所有 SCLC 患者都有反应。在此,为了将 CD137 的调控功能整合到 T 细胞吸引剂中,从而提高疗效,我们生成了 CD3/CD137 双特异性 Fab,并设计了 DLL3 靶向三特异性抗体(DLL3 三特异性)。生成的 CD3/CD137 双特异性 Fab 能竞争性地与 CD3 和 CD137 结合,以防止 CD3 和 CD137 在不依赖 DLL3 的情况下发生交联,从而导致全身性 T 细胞活化。我们证实,与传统的 DLL3 靶向双特异性 T 细胞吞噬剂相比,DLL3 三特异性能更好地控制肿瘤生长,并显著增加瘤内 T 细胞的数量。这些研究结果表明,DLL3三特异性可通过同时诱导CD137成本刺激发挥强大的疗效,为SCLC的治疗提供了一种前景广阔的选择。
{"title":"Engineering CD3/CD137 Dual Specificity into a DLL3-Targeted T-Cell Engager Enhances T-Cell Infiltration and Efficacy against Small-Cell Lung Cancer.","authors":"Hirofumi Mikami, Shu Feng, Yutaka Matsuda, Shinya Ishii, Sotaro Naoi, Yumiko Azuma, Hiroaki Nagano, Kentaro Asanuma, Yoko Kayukawa, Toshiaki Tsunenari, Shogo Kamikawaji, Ryutaro Iwabuchi, Junko Shinozuka, Masaki Yamazaki, Haruka Kuroi, Samantha Shu Wen Ho, Siok Wan Gan, Priyanka Chichili, Chai Ling Pang, Chiew Ying Yeo, Shun Shimizu, Naoka Hironiwa, Yasuko Kinoshita, Yuichiro Shimizu, Akihisa Sakamoto, Masaru Muraoka, Noriyuki Takahashi, Tatsuya Kawa, Hirotake Shiraiwa, Futa Mimoto, Kenji Kashima, Mika Kamata-Sakurai, Shumpei Ishikawa, Hiroyuki Aburatani, Takehisa Kitazawa, Tomoyuki Igawa","doi":"10.1158/2326-6066.CIR-23-0638","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0638","url":null,"abstract":"<p><p>Small-cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICI) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all patients with SCLC are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared with a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140334754","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
An NFAT1-C3a-C3aR Positive Feedback Loop in Tumor-Associated Macrophages Promotes a Glioma Stem Cell Malignant Phenotype. 肿瘤相关巨噬细胞中的 NFAT1-C3a-C3aR 正反馈环促进胶质瘤干细胞恶性表型的形成
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2326-6066.CIR-23-0418
Yaochuan Zhang, Yifu Song, Xiaoliang Wang, Mengwu Shi, Yibin Lin, Dongxia Tao, Sheng Han

Extensive infiltration by tumor-associated macrophages (TAM) in combination with myeloid-derived suppressor cells constitute the immunosuppressive microenvironment and promote the malignant phenotype of gliomas. The aggressive mesenchymal (MES)-subtype glioma stem cells (GSC) are prominent in the immunosuppressive microenvironment of gliomas. However, the underlying immune-suppressive mechanisms are still unknown. The current study showed that the antitumor immune microenvironment was activated in glioma in Nfat1-/- mice, suggesting induction of the immune-suppressive microenvironment by nuclear factor of activated T cells-1 (NFAT1). In TAMs, NFAT1 could upregulate the transcriptional activity of complement 3 (C3) and increase the secretion of C3a, which could then bind to C3aR and promote M2-like macrophage polarization by activating TIM-3. Simultaneously, C3a/C3aR activated the Ca2+-NFAT1 pathway, forming a positive feedback loop for the M2-like polarization of TAMs, which further promoted the MES transition of GSCs. Finally, disruption of this feedback loop using a C3aR inhibitor significantly inhibited glioma growth both in vitro and in vivo. The current study demonstrated that a NFAT1-C3a-C3aR positive feedback loop induces M2-like TAMs and further promotes the malignant phenotype of GSCs, which might be the potential therapeutic target for glioma.

肿瘤相关巨噬细胞(TAM)与髓源抑制细胞的广泛浸润构成了免疫抑制微环境,并促进了胶质瘤的恶性表型。侵袭性间充质(MES)亚型胶质瘤干细胞(GSC)在胶质瘤的免疫抑制微环境中表现突出。然而,其背后的免疫抑制机制仍然未知。目前的研究表明,Nfat1-/-小鼠胶质瘤的抗肿瘤免疫微环境被激活,这表明活化T细胞核因子-1(NFAT1)诱导了免疫抑制微环境。在TAMs中,NFAT1能上调补体3(C3)的转录活性,增加C3a的分泌,然后C3a能与C3aR结合,通过激活TIM-3促进M2样巨噬细胞极化。同时,C3a/C3aR激活了Ca2+-NFAT1通路,形成了TAMs M2样极化的正反馈回路,进一步促进了GSCs的MES转变。最后,使用 C3aR 抑制剂破坏这一反馈环路可显著抑制胶质瘤在体外和体内的生长。目前的研究表明,NFAT1-C3a-C3aR正反馈环诱导M2样TAMs,并进一步促进GSCs的恶性表型,这可能是胶质瘤的潜在治疗靶点。
{"title":"An NFAT1-C3a-C3aR Positive Feedback Loop in Tumor-Associated Macrophages Promotes a Glioma Stem Cell Malignant Phenotype.","authors":"Yaochuan Zhang, Yifu Song, Xiaoliang Wang, Mengwu Shi, Yibin Lin, Dongxia Tao, Sheng Han","doi":"10.1158/2326-6066.CIR-23-0418","DOIUrl":"10.1158/2326-6066.CIR-23-0418","url":null,"abstract":"<p><p>Extensive infiltration by tumor-associated macrophages (TAM) in combination with myeloid-derived suppressor cells constitute the immunosuppressive microenvironment and promote the malignant phenotype of gliomas. The aggressive mesenchymal (MES)-subtype glioma stem cells (GSC) are prominent in the immunosuppressive microenvironment of gliomas. However, the underlying immune-suppressive mechanisms are still unknown. The current study showed that the antitumor immune microenvironment was activated in glioma in Nfat1-/- mice, suggesting induction of the immune-suppressive microenvironment by nuclear factor of activated T cells-1 (NFAT1). In TAMs, NFAT1 could upregulate the transcriptional activity of complement 3 (C3) and increase the secretion of C3a, which could then bind to C3aR and promote M2-like macrophage polarization by activating TIM-3. Simultaneously, C3a/C3aR activated the Ca2+-NFAT1 pathway, forming a positive feedback loop for the M2-like polarization of TAMs, which further promoted the MES transition of GSCs. Finally, disruption of this feedback loop using a C3aR inhibitor significantly inhibited glioma growth both in vitro and in vivo. The current study demonstrated that a NFAT1-C3a-C3aR positive feedback loop induces M2-like TAMs and further promotes the malignant phenotype of GSCs, which might be the potential therapeutic target for glioma.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139575184","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
A Sampling of Highlights from the Literature. 文献精华选集》。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2326-6066.CIR-12-3-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-12-3-WWR","DOIUrl":"10.1158/2326-6066.CIR-12-3-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140027437","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
FcγRIIB Is an Immune Checkpoint Limiting the Activity of Treg-Targeting Antibodies in the Tumor Microenvironment. FcγRIIB是一种免疫检查点,它限制了肿瘤微环境中Treg靶向抗体的活性。
IF 8.1 1区 医学 Q1 Medicine Pub Date : 2024-03-04 DOI: 10.1158/2326-6066.CIR-23-0389
David A Knorr, Lucas Blanchard, Rom S Leidner, Shawn M Jensen, Ryan Meng, Andrew Jones, Carmen Ballesteros-Merino, Richard B Bell, Maria Baez, Alessandra Marino, David Sprott, Carlo B Bifulco, Brian Piening, Rony Dahan, Juan C Osorio, Bernard A Fox, Jeffrey V Ravetch

Preclinical murine data indicate that fragment crystallizable (Fc)-dependent depletion of intratumoral regulatory T cells (Treg) is a major mechanism of action of anti-CTLA-4. However, the two main antibodies administered to patients (ipilimumab and tremelimumab) do not recapitulate these effects. Here, we investigate the underlying mechanisms responsible for the limited Treg depletion observed with these therapies. Using an immunocompetent murine model humanized for CTLA-4 and Fcγ receptors (FcγR), we show that ipilimumab and tremelimumab exhibit limited Treg depletion in tumors. Immune profiling of the tumor microenvironment (TME) in both humanized mice and humans revealed high expression of the inhibitory Fc receptor, FcγRIIB, which limits antibody-dependent cellular cytotoxicity/phagocytosis. Blocking FcγRIIB in humanized mice rescued the Treg-depleting capacity and antitumor activity of ipilimumab. Furthermore, Fc engineering of antibodies targeting Treg-associated targets (CTLA-4 or CCR8) to minimize FcγRIIB binding significantly enhanced Treg depletion, resulting in increased antitumor activity across various tumor models. Our results define the inhibitory FcγRIIB as an immune checkpoint limiting antibody-mediated Treg depletion in the TME, and demonstrate Fc engineering as an effective strategy to overcome this limitation and improve the efficacy of Treg-targeting antibodies.

临床前小鼠数据表明,Fc依赖性瘤内调节性T细胞(Tregs)耗竭是抗CTLA-4的主要作用机制。然而,患者使用的两种主要抗体(伊匹单抗(Ipilimumab)和特雷莫单抗(Tremelimumab))并不能再现这些效应。在此,我们研究了这些疗法导致Treg消耗有限的内在机制。我们利用一种CTLA-4和Fcγ受体(FcγRs)人源化的免疫功能健全的小鼠模型,证明伊匹单抗和特瑞莫单抗在肿瘤中表现出有限的Treg消耗。人源化小鼠和人类肿瘤微环境(TME)的免疫分析表明,抑制性Fc受体FcγRIIB的高表达限制了抗体依赖性细胞毒性/吞噬作用(ADCC/ADCP)。在人源化小鼠体内阻断FcγRIIB可以挽救伊匹单抗的Treg清除能力和抗肿瘤活性。此外,对针对Treg相关靶点(CTLA-4或CCR8)的抗体进行Fc工程设计,使FcγRIIB结合最小化,也能显著增强Treg消耗能力,从而提高各种肿瘤模型的抗肿瘤活性。我们的研究结果确定了抑制性 FcγRIIB 是限制抗体介导的 TME 中 Treg 消耗的免疫检查点,并证明 Fc 工程是克服这一限制和提高 Treg 靶向抗体疗效的有效策略。
{"title":"FcγRIIB Is an Immune Checkpoint Limiting the Activity of Treg-Targeting Antibodies in the Tumor Microenvironment.","authors":"David A Knorr, Lucas Blanchard, Rom S Leidner, Shawn M Jensen, Ryan Meng, Andrew Jones, Carmen Ballesteros-Merino, Richard B Bell, Maria Baez, Alessandra Marino, David Sprott, Carlo B Bifulco, Brian Piening, Rony Dahan, Juan C Osorio, Bernard A Fox, Jeffrey V Ravetch","doi":"10.1158/2326-6066.CIR-23-0389","DOIUrl":"10.1158/2326-6066.CIR-23-0389","url":null,"abstract":"<p><p>Preclinical murine data indicate that fragment crystallizable (Fc)-dependent depletion of intratumoral regulatory T cells (Treg) is a major mechanism of action of anti-CTLA-4. However, the two main antibodies administered to patients (ipilimumab and tremelimumab) do not recapitulate these effects. Here, we investigate the underlying mechanisms responsible for the limited Treg depletion observed with these therapies. Using an immunocompetent murine model humanized for CTLA-4 and Fcγ receptors (FcγR), we show that ipilimumab and tremelimumab exhibit limited Treg depletion in tumors. Immune profiling of the tumor microenvironment (TME) in both humanized mice and humans revealed high expression of the inhibitory Fc receptor, FcγRIIB, which limits antibody-dependent cellular cytotoxicity/phagocytosis. Blocking FcγRIIB in humanized mice rescued the Treg-depleting capacity and antitumor activity of ipilimumab. Furthermore, Fc engineering of antibodies targeting Treg-associated targets (CTLA-4 or CCR8) to minimize FcγRIIB binding significantly enhanced Treg depletion, resulting in increased antitumor activity across various tumor models. Our results define the inhibitory FcγRIIB as an immune checkpoint limiting antibody-mediated Treg depletion in the TME, and demonstrate Fc engineering as an effective strategy to overcome this limitation and improve the efficacy of Treg-targeting antibodies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10911703/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139037362","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
Cancer immunology research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号: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