首页 > 最新文献

Checkpoints and Immunomodulation最新文献

英文 中文
Abstract A23: Anti-PD-1 antibody scFv producing recombinant Bifidobacterium exerts antitumor effect in a larger fraction of the treated mice compared to full-length anti-PD-1 antibody A23:与全长抗pd -1抗体相比,产生抗pd -1抗体的重组双歧杆菌在更大比例的治疗小鼠中发挥抗肿瘤作用
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A23
Koichiro Shioya, Li Wang, T. Matsumura, Hitomi Shimizu, Y. Kanari, Y. Seki, Yuko Shimatani, S. Taniguchi, S. Kataoka
Anti-PD-1 therapy has improved therapeutic outcomes of patients in multiple cancer types. However, the therapy has demonstrated clinical benefits in only a small fraction of patients. The reason of the limited response in clinical practice is not fully understood. In an aim to improve anti-cancer drug delivery and potency, we have been developing in situ delivery and production system (i-DPS) by modifying a non-pathogenic anaerobic bacterium, Bifidobacterium, which localizes and proliferates only in the hypoxic environment like solid tumors after intravenous administration, produces anticancer proteins, enzymes or other pharmacologically active molecules selectively at the tumor site. Here we present anti-human PD-1 antibody scFv producing i-DPS in cancer immunotherapy, which could be specifically delivered to and amplified only at the hypoxic sites of solid tumors. A series of in vitro assays has been performed to confirm the stable expression and secretion of anti-human PD-1 scFv by recombinant Bifidobacterium, the binding inhibition of PD-1/PD-L1 interaction and elevated IFN gamma production in mixed lymphocyte culture by anti-human PD-1 scFv secreted from recombinant Bifidobacterium. Anti-murine PD-1 scFv producing Bifidobacterium as surrogate systemically administered to the syngeneic mice model demonstrated significant tumor growth inhibition. Of particular interest, the suppression of tumor growth was observed in a larger fraction of the treated mice while the control anti-PD-1 antibody showed the effect on only a few mice. The analysis of tumor infiltrating lymphocytes and myeloid cells will be presented as well. Taken together, i-DPS for anti-PD-1 antibody provides a new promising immune-therapeutic modality to target hypoxic solid tumors and also provides a unique insight for antibody drug delivery in cancer immunotherapy. Citation Format: Koichiro Shioya, Li Wang, Tomio Matsumura, Hitomi Shimizu, Yasuyoshi Kanari, Yuji Seki, Yuko Shimatani, Shun’ichiro Taniguchi, Shiro Kataoka. Anti-PD-1 antibody scFv producing recombinant Bifidobacterium exerts antitumor effect in a larger fraction of the treated mice compared to full-length anti-PD-1 antibody [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A23.
抗pd -1治疗改善了多种癌症类型患者的治疗效果。然而,该疗法仅在一小部分患者中显示出临床益处。临床实践中反应有限的原因尚不完全清楚。为了提高抗癌药物的递送和药效,我们一直在开发原位递送和生产系统(i-DPS),通过修饰一种非致病性厌氧菌双歧杆菌,该细菌在静脉给药后仅在实体瘤等缺氧环境中定位和增殖,在肿瘤部位选择性地产生抗癌蛋白、酶或其他药理活性分子。在这里,我们提出了在癌症免疫治疗中产生i-DPS的抗人PD-1抗体scFv,它可以特异性地递送到实体肿瘤的缺氧部位并在其上扩增。一系列体外实验证实重组双歧杆菌稳定表达和分泌抗人PD-1 scFv,重组双歧杆菌分泌的抗人PD-1 scFv结合抑制PD-1/PD-L1相互作用,提高混合淋巴细胞中IFN γ的产生。产生抗小鼠PD-1 scFv的双歧杆菌作为替代物系统给药于同基因小鼠模型,显示出明显的肿瘤生长抑制作用。特别有趣的是,肿瘤生长的抑制在大部分治疗小鼠中被观察到,而对照抗pd -1抗体仅对少数小鼠起作用。肿瘤浸润淋巴细胞和骨髓细胞的分析也将被提出。综上所述,抗pd -1抗体i-DPS提供了一种新的有前景的针对缺氧实体瘤的免疫治疗方式,也为癌症免疫治疗中的抗体药物递送提供了独特的见解。引文格式:盐谷光一郎,王丽,松村富雄,清水仁美,金利康吉,关祐二,岛谷优子,谷口顺一郎,片冈四郎。与全长抗pd -1抗体相比,产生抗pd -1抗体的重组双歧杆菌在更大比例的治疗小鼠中发挥抗肿瘤作用[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫,2018;6(9增刊):摘要nr A23。
{"title":"Abstract A23: Anti-PD-1 antibody scFv producing recombinant Bifidobacterium exerts antitumor effect in a larger fraction of the treated mice compared to full-length anti-PD-1 antibody","authors":"Koichiro Shioya, Li Wang, T. Matsumura, Hitomi Shimizu, Y. Kanari, Y. Seki, Yuko Shimatani, S. Taniguchi, S. Kataoka","doi":"10.1158/2326-6074.TUMIMM17-A23","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A23","url":null,"abstract":"Anti-PD-1 therapy has improved therapeutic outcomes of patients in multiple cancer types. However, the therapy has demonstrated clinical benefits in only a small fraction of patients. The reason of the limited response in clinical practice is not fully understood. In an aim to improve anti-cancer drug delivery and potency, we have been developing in situ delivery and production system (i-DPS) by modifying a non-pathogenic anaerobic bacterium, Bifidobacterium, which localizes and proliferates only in the hypoxic environment like solid tumors after intravenous administration, produces anticancer proteins, enzymes or other pharmacologically active molecules selectively at the tumor site. Here we present anti-human PD-1 antibody scFv producing i-DPS in cancer immunotherapy, which could be specifically delivered to and amplified only at the hypoxic sites of solid tumors. A series of in vitro assays has been performed to confirm the stable expression and secretion of anti-human PD-1 scFv by recombinant Bifidobacterium, the binding inhibition of PD-1/PD-L1 interaction and elevated IFN gamma production in mixed lymphocyte culture by anti-human PD-1 scFv secreted from recombinant Bifidobacterium. Anti-murine PD-1 scFv producing Bifidobacterium as surrogate systemically administered to the syngeneic mice model demonstrated significant tumor growth inhibition. Of particular interest, the suppression of tumor growth was observed in a larger fraction of the treated mice while the control anti-PD-1 antibody showed the effect on only a few mice. The analysis of tumor infiltrating lymphocytes and myeloid cells will be presented as well. Taken together, i-DPS for anti-PD-1 antibody provides a new promising immune-therapeutic modality to target hypoxic solid tumors and also provides a unique insight for antibody drug delivery in cancer immunotherapy. Citation Format: Koichiro Shioya, Li Wang, Tomio Matsumura, Hitomi Shimizu, Yasuyoshi Kanari, Yuji Seki, Yuko Shimatani, Shun’ichiro Taniguchi, Shiro Kataoka. Anti-PD-1 antibody scFv producing recombinant Bifidobacterium exerts antitumor effect in a larger fraction of the treated mice compared to full-length anti-PD-1 antibody [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A23.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80785409","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract A14: Surgical removal of metastatic lesions increases T-cell reactivity to tumor-associated antigens in stage III melanoma patients 摘要:手术切除转移性病变可增加III期黑色素瘤患者t细胞对肿瘤相关抗原的反应性
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A14
Y. Coaña, Fríða Björk Gunnarsdóttir, Maria Wolodarski, S. E. Brage, G. Masucci, J. Hansson, R. Kiessling
Currently, there are hardly any peripheral blood biomarkers that allow for identification of patients’ prognosis or survival in advanced melanoma patients. Our main objective was to compare the reactivity of peripheral blood T cells of patients with stage III or IV metastatic melanoma to Tumor-Associated Antigens, before and after surgery where metastatic lesions were removed. Peripheral blood mononuclear cells were isolated from blood samples taken before and after surgery and were stimulated over the course of two weeks with overlapping peptide pools of three known melanoma antigens: MelanA, NY-ESO-1, and Cripto-1. After 12 days, cells were re-stimulated and analyzed using multicolor flow cytometry. CD4 and CD8 positive cells were analyzed for cytokine production. McNemar’s test was used to analyze changes before and after surgery, and correlation between cell reactivity and cytokine production with progression free survival was determined by Kaplan Meier analysis. Surgical removal of metastatic lesions changed reactivity of T cells to MelanA, NY-ESO-1 and Cripto-1. The presence of CD4 T cells that produced IL-17 and/or TNF-a after stimulation was correlated with a worse progression-free survival (PFS). We show here that surgical removal of metastases increases T-cell reactivity in melanoma patients and that certain cytokine profiles may be associated to PFS. This could provide insight into the complexity of the correlation between a specific T-cell response and a favorable immune response to metastatic melanoma. Citation Format: Yago Pico de Coana, Friða Bjork Gunnarsdottir, Maria Wolodarski, Suzanne Egyhazi Brage, Giuseppe Masucci, Johan Hansson, Rolf Kiessling. Surgical removal of metastatic lesions increases T-cell reactivity to tumor-associated antigens in stage III melanoma patients [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A14.
目前,几乎没有任何外周血生物标志物可以识别晚期黑色素瘤患者的预后或生存。我们的主要目的是比较III期或IV期转移性黑色素瘤患者在转移病灶切除手术前后外周血T细胞对肿瘤相关抗原的反应性。从手术前后采集的血液样本中分离外周血单个核细胞,并在两周内用三种已知黑色素瘤抗原(MelanA, NY-ESO-1和Cripto-1)重叠的肽池刺激。12天后,再次刺激细胞,用多色流式细胞术分析。分析CD4和CD8阳性细胞的细胞因子产生情况。McNemar试验分析手术前后的变化,Kaplan Meier分析细胞反应性和细胞因子产生与无进展生存期的相关性。手术切除转移灶改变了T细胞对MelanA、NY-ESO-1和Cripto-1的反应性。刺激后产生IL-17和/或TNF-a的CD4 T细胞的存在与较差的无进展生存期(PFS)相关。我们在这里表明,手术切除转移瘤增加了黑色素瘤患者的t细胞反应性,某些细胞因子谱可能与PFS有关。这可以深入了解特异性t细胞反应和对转移性黑色素瘤有利的免疫反应之间的复杂关系。引文格式:Yago Pico de Coana, frizha Bjork Gunnarsdottir, Maria Wolodarski, Suzanne Egyhazi Brage, Giuseppe Masucci, Johan Hansson, Rolf Kiessling。在III期黑色素瘤患者中,手术切除转移性病变可增加t细胞对肿瘤相关抗原的反应性[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A14。
{"title":"Abstract A14: Surgical removal of metastatic lesions increases T-cell reactivity to tumor-associated antigens in stage III melanoma patients","authors":"Y. Coaña, Fríða Björk Gunnarsdóttir, Maria Wolodarski, S. E. Brage, G. Masucci, J. Hansson, R. Kiessling","doi":"10.1158/2326-6074.TUMIMM17-A14","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A14","url":null,"abstract":"Currently, there are hardly any peripheral blood biomarkers that allow for identification of patients’ prognosis or survival in advanced melanoma patients. Our main objective was to compare the reactivity of peripheral blood T cells of patients with stage III or IV metastatic melanoma to Tumor-Associated Antigens, before and after surgery where metastatic lesions were removed. Peripheral blood mononuclear cells were isolated from blood samples taken before and after surgery and were stimulated over the course of two weeks with overlapping peptide pools of three known melanoma antigens: MelanA, NY-ESO-1, and Cripto-1. After 12 days, cells were re-stimulated and analyzed using multicolor flow cytometry. CD4 and CD8 positive cells were analyzed for cytokine production. McNemar’s test was used to analyze changes before and after surgery, and correlation between cell reactivity and cytokine production with progression free survival was determined by Kaplan Meier analysis. Surgical removal of metastatic lesions changed reactivity of T cells to MelanA, NY-ESO-1 and Cripto-1. The presence of CD4 T cells that produced IL-17 and/or TNF-a after stimulation was correlated with a worse progression-free survival (PFS). We show here that surgical removal of metastases increases T-cell reactivity in melanoma patients and that certain cytokine profiles may be associated to PFS. This could provide insight into the complexity of the correlation between a specific T-cell response and a favorable immune response to metastatic melanoma. Citation Format: Yago Pico de Coana, Friða Bjork Gunnarsdottir, Maria Wolodarski, Suzanne Egyhazi Brage, Giuseppe Masucci, Johan Hansson, Rolf Kiessling. Surgical removal of metastatic lesions increases T-cell reactivity to tumor-associated antigens in stage III melanoma patients [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A14.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83085461","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A15: Immune-related changes in breast cancer tumor evolution A15:乳腺癌肿瘤演变中的免疫相关变化
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A15
C. R. G. D. Alcazar, S. Huh, Muhammad B Ekram, A. Trinh, Lin L. Liu, F. Beca, Zi Xiaoyuan, M. Kwak, Helga Bergholtz, Ying Su, L. Ding, H. Russnes, A. Richardson, Kirsten Babski, E. M. Kim, C. H. McDonnell, Jon Wagner, Ron Rowberry, G. Freeman, D. Dillon, T. Sørlie, L. Coussens, J. Garber, R. Fan, Kristie A. Bobolis, Joon Jeong, S. Y. Park, F. Michor, K. Polyak
Immunotherapy is a highly promising therapeutic option in metastatic disease albeit only in a subset of patients possibly due to heterogeneity in the mechanisms by which tumors escape immune surveillance. Immune cells shape tumor evolution directly (e.g., anti-tumor immune response) and indirectly (e.g., changing the microenvironment) by selecting for cancer cells with certain properties. We hypothesized that the in situ (DCIS) to invasive ductal carcinoma (IDC) transition is a critical tumor progression step for immune escape in breast cancer that defines subsequent tumor evolution. In DCIS, cancer cells are physically separated from the stroma by the basement membrane and myoepithelial cell layer, and tumor-infiltrating leukocytes are rarely detected in direct contact with cancer cells. In contrast, in IDC, cancer cells and leukocytes are intermingled, thus, only cancer cells that can survive in this environment will play a role in disease progression. To dissect mechanisms of immune escape in breast cancer, we first analyzed the composition of leukocytes in normal breast tissues, DCIS, and IDC by polychromatic FACS. We found that DCIS and IDC contained significantly higher numbers of leukocytes, compared to normal breast, whereas in normal tissues more leukocytes were in the stromal than in the epithelial fraction. We also observed significant differences in the relative frequencies of several CD45+ cell types including increased neutrophils and decreased CD8+/CD4+ T cell ratios in tumors compared to normal stroma. Next, we analyzed the gene expression profiles of CD45+CD3+ T cells and found gene set enrichment of cytotoxic cells in DCIS including CD8+ T cells and NKT cells when compared to IDC. Conversely, we found enrichment for gene sets corresponding to regulatory T cells in IDC when compared to DCIS. Overall this suggested that DCIS had a more activated immune environment and IDC a more suppressed immune environment. We further explored this result by immunofluorescence (IF) and found fewer activated GZMB+CD8+ T cells in IDC than in DCIS, including a set of matched DCIS and locally recurrent IDC tissues. We also found that the TCR clonotype was more diverse in DCIS than in normal breast and IDCs. Interestingly we detected a few relatively frequent clones that were shared among different DCIS, one of which was previously shown to recognize a protein from the Epstein-Bar virus. To elucidate mechanisms of immune evasion in IDC, we performed IF analysis of immune checkpoint proteins PD-L1 and TIGIT and found significant differences between DCIS and IDC. TIGIT-expressing T cells were more slightly frequent in DCIS than in IDC. PD-L1 expression was higher in the epithelial cancer cells in triple negative IDC compared to DCIS, and amplification of CD274 (encoding PD-L1) was only detected in triple negative IDCs. Given the close proximity of ERBB2 (encoding HER2) to a cluster of genes encoding several chemokines, we analyzed the HER2+ samples fro
免疫治疗是一种非常有前途的转移性疾病的治疗选择,尽管仅适用于一小部分患者,可能是由于肿瘤逃避免疫监视的机制存在异质性。免疫细胞通过选择具有某些特性的癌细胞,直接(例如,抗肿瘤免疫反应)或间接(例如,改变微环境)影响肿瘤的进化。我们假设原位癌(DCIS)向浸润性导管癌(IDC)的转变是乳腺癌免疫逃逸的关键肿瘤进展步骤,这决定了随后的肿瘤演变。在DCIS中,癌细胞被基底膜和肌上皮细胞层物理地与间质分离,肿瘤浸润的白细胞很少与癌细胞直接接触。相反,在IDC中,癌细胞和白细胞是混杂的,因此只有能够在这种环境中生存的癌细胞才会在疾病进展中发挥作用。为了剖析乳腺癌中免疫逃逸的机制,我们首先通过多色FACS分析了正常乳腺组织、DCIS和IDC中白细胞的组成。我们发现,与正常乳腺相比,DCIS和IDC的白细胞数量明显增加,而在正常组织中,基质中的白细胞多于上皮部分。我们还观察到,与正常基质相比,肿瘤中几种CD45+细胞类型的相对频率存在显著差异,包括中性粒细胞增加和CD8+/CD4+ T细胞比例降低。接下来,我们分析了CD45+CD3+ T细胞的基因表达谱,发现与IDC相比,DCIS细胞毒性细胞包括CD8+ T细胞和NKT细胞的基因集富集。相反,与DCIS相比,我们发现IDC中与调节性T细胞相关的基因组富集。总体而言,这表明DCIS具有更激活的免疫环境,而IDC具有更抑制的免疫环境。我们通过免疫荧光(IF)进一步探讨了这一结果,发现IDC中活化的GZMB+CD8+ T细胞少于DCIS,包括一组匹配的DCIS和局部复发的IDC组织。我们还发现TCR克隆型在DCIS中比在正常乳腺和肝癌中更多样化。有趣的是,我们发现了一些在不同DCIS之间共享的相对频繁的克隆,其中一个先前被证明可以识别来自爱泼斯坦-巴尔病毒的蛋白质。为了阐明IDC中免疫逃避的机制,我们对免疫检查点蛋白PD-L1和TIGIT进行了IF分析,发现DCIS和IDC之间存在显著差异。表达tigit的T细胞在DCIS中的频率略高于IDC。与DCIS相比,三阴性IDC上皮癌细胞中PD-L1的表达更高,CD274(编码PD-L1)的扩增仅在三阴性IDC中检测到。鉴于ERBB2(编码HER2)与编码几种趋化因子的基因簇非常接近,我们分析了来自TCGA的HER2+样本。我们发现17q12趋化因子簇(CC)与ERBB2的共扩增在HER+ER+发光样肿瘤中富集,而在HER2+ER乳腺肿瘤中不富集。我们还发现,在缺乏CC增益的肿瘤中,T细胞激活和抑制相关基因的表达更高。此外,通过多色FISH和IF评估肿瘤样本,我们确定CC扩增与CD8+ T细胞活化之间存在负相关。总的来说,我们的结果显示在肿瘤进展过程中癌细胞和免疫微环境的共同进化。引用形式:Carlos R. Gil del Alcazar, SungJin Huh, Muhammad B. Ekram, Anne Trinh, Lin L. Liu, Francisco Beca, Zi Xiaoyuan, Misuk Kwak, Helga Bergholtz, Ying Su, Lina Ding, Lina Ding, Hege G. Russnes, Andrea L. Richardson, Kirsten Babski, Elizabeth Min Hui Kim, Charles H. McDonnell, III, Jon Wagner, Ron Rowberry, Gordon Freeman, Deborah Dillon, Therese Sorlie, Lisa M. Coussens, Judy E. Garber, Rong Fan, Kristie Bobolis, Joon Jeong, So Yeon Park, Franziska Michor, Kornelia Polyak。乳腺癌肿瘤演变中的免疫相关变化[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A15。
{"title":"Abstract A15: Immune-related changes in breast cancer tumor evolution","authors":"C. R. G. D. Alcazar, S. Huh, Muhammad B Ekram, A. Trinh, Lin L. Liu, F. Beca, Zi Xiaoyuan, M. Kwak, Helga Bergholtz, Ying Su, L. Ding, H. Russnes, A. Richardson, Kirsten Babski, E. M. Kim, C. H. McDonnell, Jon Wagner, Ron Rowberry, G. Freeman, D. Dillon, T. Sørlie, L. Coussens, J. Garber, R. Fan, Kristie A. Bobolis, Joon Jeong, S. Y. Park, F. Michor, K. Polyak","doi":"10.1158/2326-6074.TUMIMM17-A15","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A15","url":null,"abstract":"Immunotherapy is a highly promising therapeutic option in metastatic disease albeit only in a subset of patients possibly due to heterogeneity in the mechanisms by which tumors escape immune surveillance. Immune cells shape tumor evolution directly (e.g., anti-tumor immune response) and indirectly (e.g., changing the microenvironment) by selecting for cancer cells with certain properties. We hypothesized that the in situ (DCIS) to invasive ductal carcinoma (IDC) transition is a critical tumor progression step for immune escape in breast cancer that defines subsequent tumor evolution. In DCIS, cancer cells are physically separated from the stroma by the basement membrane and myoepithelial cell layer, and tumor-infiltrating leukocytes are rarely detected in direct contact with cancer cells. In contrast, in IDC, cancer cells and leukocytes are intermingled, thus, only cancer cells that can survive in this environment will play a role in disease progression. To dissect mechanisms of immune escape in breast cancer, we first analyzed the composition of leukocytes in normal breast tissues, DCIS, and IDC by polychromatic FACS. We found that DCIS and IDC contained significantly higher numbers of leukocytes, compared to normal breast, whereas in normal tissues more leukocytes were in the stromal than in the epithelial fraction. We also observed significant differences in the relative frequencies of several CD45+ cell types including increased neutrophils and decreased CD8+/CD4+ T cell ratios in tumors compared to normal stroma. Next, we analyzed the gene expression profiles of CD45+CD3+ T cells and found gene set enrichment of cytotoxic cells in DCIS including CD8+ T cells and NKT cells when compared to IDC. Conversely, we found enrichment for gene sets corresponding to regulatory T cells in IDC when compared to DCIS. Overall this suggested that DCIS had a more activated immune environment and IDC a more suppressed immune environment. We further explored this result by immunofluorescence (IF) and found fewer activated GZMB+CD8+ T cells in IDC than in DCIS, including a set of matched DCIS and locally recurrent IDC tissues. We also found that the TCR clonotype was more diverse in DCIS than in normal breast and IDCs. Interestingly we detected a few relatively frequent clones that were shared among different DCIS, one of which was previously shown to recognize a protein from the Epstein-Bar virus. To elucidate mechanisms of immune evasion in IDC, we performed IF analysis of immune checkpoint proteins PD-L1 and TIGIT and found significant differences between DCIS and IDC. TIGIT-expressing T cells were more slightly frequent in DCIS than in IDC. PD-L1 expression was higher in the epithelial cancer cells in triple negative IDC compared to DCIS, and amplification of CD274 (encoding PD-L1) was only detected in triple negative IDCs. Given the close proximity of ERBB2 (encoding HER2) to a cluster of genes encoding several chemokines, we analyzed the HER2+ samples fro","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88926361","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A38: Blood-based T cell biomarkers and soluble PD-L1 predict responses and immune-related toxicity to PD-1 blockade in melanoma and lung cancer 血液T细胞生物标志物和可溶性PD-L1可预测黑色素瘤和肺癌患者对PD-1阻断的反应和免疫相关毒性
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A38
R. Dronca, A. Mansfield, Xin Liu, S. Harrington, E. Enninga, S. Markovic, L. Kottschade, R. McWilliams, M. Block, W. Nevala, M. Thompson, Haidong Dong
Despite unprecedented successes with immune checkpoint blockade in advanced cancers, these agents provide durable clinical benefit in only a subset of patients. In addition, the clinical management of patients receiving immune checkpoint blockade remains quite challenging due to the unpredictable and kinetically heterogeneous responses which can manifest as late responses, pseudoprogression, or hyperprogression in subsets of patients. The dynamics and heterogeneity of programmed cell death protein ligand 1 (PD-L1, aka B7-H1) expression confound its use as a predictive biomarker in cancer immunotherapy, but blood-based biomarkers have the potential to predict responders and detect mechanisms of resistance to immunotherapy. We previously reported that Bim (BH3-only protein) is a downstream signaling molecule of the PD-1 pathway, and that Bim levels in circulating tumor-reactive T-cells reflected patient responses to anti-PD-1 therapy in melanoma. We also discovered the existence of soluble PD-L1 (sPD-L1) in the plasma of cancer patients and showed a correlation of sPD-L1 with cancer stages in renal cell carcinoma and with prognosis in metastatic melanoma. Here we evaluated the frequency of tumor-reactive T cells with Bim expression along with effector memory profile and sPD-L1 as biomarkers of response in a new cohort of patients with metastatic melanoma and lung cancer undergoing anti-PD-1 therapy. We recruited 100 cancer patients treated with anti-PD-1 therapy who had peripheral blood collected at baseline and at each subsequent radiographic tumor evaluation. Frequencies of Bim+ and effector memory CD8+ T cells were measured by flow cytometry in gated tumor-reactive CD11a high PD1+ CD8+ T cells. We also measured levels of sPD-L1 in plasma at baseline and serially during treatment with ELISA. Baseline T cell markers and sPD-L1 levels and their percent changes in patients who had a radiographic response were compared to those who had progressive disease (PD). We found that patients with objective response after 4 cycles of anti-PD-1 therapy had higher frequency of Bim+ CD8+ T cells at baseline compared to patients with PD (mean 43% vs. 30%, P=0.0484). The frequencies of Bim+ T cells decreased significantly after the first 3 months of treatment in responders compared with progressors (mean -16% vs. + 40% P=0.0111). The frequency of effector memory CD8+ T cells also dramatically increased in responders in 3-6 months after anti-PD-1 therapy (mean 89% vs. 9.56%, p=0.002). High baseline sPD-L1 was associated with progression on anti-PD-1 therapy (mean 2.8 ng/mL vs. 0.7 ng/mL, p=0.07) or development of immune-related toxicities and the levels increased by 12 weeks in patients progressing on therapy. Conclusion: Measurements of T cell biomarkers (Bim and effector memory) and sPD-L1 levels provide a new noninvasive way to predict and monitor patient responses to anti-PD-1 monotherapy in melanoma and lung cancers. Note: This abstract was not presented at th
尽管免疫检查点阻断在晚期癌症中取得了前所未有的成功,但这些药物仅在一小部分患者中提供持久的临床益处。此外,接受免疫检查点阻断的患者的临床管理仍然相当具有挑战性,因为不可预测和动力学异质性的反应可能表现为晚期反应、假进展或亚群患者的超进展。程序性细胞死亡蛋白配体1 (PD-L1,又名B7-H1)表达的动态和异质性使其在癌症免疫治疗中作为预测性生物标志物的用途混淆,但基于血液的生物标志物具有预测应答和检测免疫治疗耐药机制的潜力。我们之前报道过Bim (BH3-only蛋白)是PD-1通路的下游信号分子,循环肿瘤反应性t细胞中的Bim水平反映了黑色素瘤患者对抗PD-1治疗的反应。我们还发现癌症患者血浆中存在可溶性PD-L1 (sPD-L1),并显示sPD-L1与肾细胞癌的癌症分期和转移性黑色素瘤的预后相关。在一组接受抗pd -1治疗的转移性黑色素瘤和肺癌患者中,我们评估了带有Bim表达的肿瘤反应性T细胞的频率,以及效应记忆谱和sPD-L1作为反应的生物标志物。我们招募了100名接受抗pd -1治疗的癌症患者,他们在基线和随后的每次放射学肿瘤评估时采集外周血。在门控肿瘤反应性CD11a高PD1+ CD8+ T细胞中,用流式细胞术检测Bim+和效应记忆CD8+ T细胞的频率。我们还测量了基线和治疗期间血浆中sPD-L1的水平。基线T细胞标志物和sPD-L1水平及其在有放射学反应的患者中变化的百分比与患有进展性疾病(PD)的患者进行比较。我们发现,在4个周期的抗PD-1治疗后客观缓解的患者在基线时的Bim+ CD8+ T细胞频率高于PD患者(平均43%对30%,P=0.0484)。与进展者相比,缓解者在治疗的前3个月后,Bim+ T细胞的频率显著下降(平均-16% vs. + 40% P=0.0111)。在抗pd -1治疗后3-6个月,应答者中效应记忆CD8+ T细胞的频率也显著增加(平均89%比9.56%,p=0.002)。高基线sPD-L1与抗pd -1治疗的进展(平均2.8 ng/mL vs 0.7 ng/mL, p=0.07)或免疫相关毒性的发生相关,并且在治疗进展的患者中,sPD-L1水平在12周内升高。结论:T细胞生物标志物(Bim和效应记忆)和sPD-L1水平的测量为预测和监测黑色素瘤和肺癌患者对抗pd -1单药治疗的反应提供了一种新的无创方法。注:本摘要未在会议上发表。引文格式:Roxana S. Dronca, Aaron Mansfield, Liu Xin, Susan Harrington, Elizabeth Enninga, Svetomir Markovic, Lisa Kottschade, Rob Mcwilliams, Matthew Block, Wendy Nevala, Michael Thompson,董海东基于血液的T细胞生物标志物和可溶性PD-L1预测黑色素瘤和肺癌对PD-1阻断的反应和免疫相关毒性[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫,2018;6(9增刊):摘要nr A38。
{"title":"Abstract A38: Blood-based T cell biomarkers and soluble PD-L1 predict responses and immune-related toxicity to PD-1 blockade in melanoma and lung cancer","authors":"R. Dronca, A. Mansfield, Xin Liu, S. Harrington, E. Enninga, S. Markovic, L. Kottschade, R. McWilliams, M. Block, W. Nevala, M. Thompson, Haidong Dong","doi":"10.1158/2326-6074.TUMIMM17-A38","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A38","url":null,"abstract":"Despite unprecedented successes with immune checkpoint blockade in advanced cancers, these agents provide durable clinical benefit in only a subset of patients. In addition, the clinical management of patients receiving immune checkpoint blockade remains quite challenging due to the unpredictable and kinetically heterogeneous responses which can manifest as late responses, pseudoprogression, or hyperprogression in subsets of patients. The dynamics and heterogeneity of programmed cell death protein ligand 1 (PD-L1, aka B7-H1) expression confound its use as a predictive biomarker in cancer immunotherapy, but blood-based biomarkers have the potential to predict responders and detect mechanisms of resistance to immunotherapy. We previously reported that Bim (BH3-only protein) is a downstream signaling molecule of the PD-1 pathway, and that Bim levels in circulating tumor-reactive T-cells reflected patient responses to anti-PD-1 therapy in melanoma. We also discovered the existence of soluble PD-L1 (sPD-L1) in the plasma of cancer patients and showed a correlation of sPD-L1 with cancer stages in renal cell carcinoma and with prognosis in metastatic melanoma. Here we evaluated the frequency of tumor-reactive T cells with Bim expression along with effector memory profile and sPD-L1 as biomarkers of response in a new cohort of patients with metastatic melanoma and lung cancer undergoing anti-PD-1 therapy. We recruited 100 cancer patients treated with anti-PD-1 therapy who had peripheral blood collected at baseline and at each subsequent radiographic tumor evaluation. Frequencies of Bim+ and effector memory CD8+ T cells were measured by flow cytometry in gated tumor-reactive CD11a high PD1+ CD8+ T cells. We also measured levels of sPD-L1 in plasma at baseline and serially during treatment with ELISA. Baseline T cell markers and sPD-L1 levels and their percent changes in patients who had a radiographic response were compared to those who had progressive disease (PD). We found that patients with objective response after 4 cycles of anti-PD-1 therapy had higher frequency of Bim+ CD8+ T cells at baseline compared to patients with PD (mean 43% vs. 30%, P=0.0484). The frequencies of Bim+ T cells decreased significantly after the first 3 months of treatment in responders compared with progressors (mean -16% vs. + 40% P=0.0111). The frequency of effector memory CD8+ T cells also dramatically increased in responders in 3-6 months after anti-PD-1 therapy (mean 89% vs. 9.56%, p=0.002). High baseline sPD-L1 was associated with progression on anti-PD-1 therapy (mean 2.8 ng/mL vs. 0.7 ng/mL, p=0.07) or development of immune-related toxicities and the levels increased by 12 weeks in patients progressing on therapy. Conclusion: Measurements of T cell biomarkers (Bim and effector memory) and sPD-L1 levels provide a new noninvasive way to predict and monitor patient responses to anti-PD-1 monotherapy in melanoma and lung cancers. Note: This abstract was not presented at th","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90353204","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract A33: Tumor derived T cell clones for evaluation of check point inhibitor therapeutics A33:肿瘤来源的T细胞克隆用于检查点抑制剂治疗的评估
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A33
R. Shields, S. Saux, Sophie Li, M. Sathe, T. Mcclanahan, R. D. Malefyt
Improving clinical outcomes using immunotherapeutics in cancer treatments will depend on a variety of factors including the composition and phenotype of effector T cell in tumors, the inhibitory factors from myeloid, tumor and stromal compartments of the tumor and selection of efficacious combination of therapeutic immunomodulatory agents that either neutralize immune checkpoint inhibitors or activate T cell costimulatory pathways. To address the issue of selecting optimal combinations of immune modulating agents we developed an in-vitro culture system for allo-stimulating and expanding tumor infiltrating lymphocytes (TILS) from tumor digests with an MHC Class II alloantigen expressing cell line. We selected allo-specific CD4 and CD8 clones from expanded TILS with an exhausted phenotype confirmed by cell surface expression of PD-1, LAG3 and TIGIT. T cell clones were incubated with the alloantigen expressing cell lines transduced with different combinations of immodulatory ligands and compared the effect of single agent versus combinations of checkpoint inhibitory monoclonal antibodies on T cell proliferation and cytokine production. Taqman and flow cytometry analysis was performed on several clones following allo-stimulation. A decrease in TIL clonality with repeated allo-stimulation was confirmed by TcR beta chain sequencing. In-vitro IFN gamma and Granzyme B production was enhanced with anti-PD-1 antibodies in combination with either anti-LAG3 or anti-TIGIT antibodies. This in-vitro system has the potential of screening combinations of immodulatory antibodies in a variety of TILS from different tumor types and can improve our understanding of new checkpoint inhibitor combinations. Citation Format: Robert L. Shields, Sabine Le Saux, Sophie Li, Manjiri Sathe, Terri McClanahan, Rene De Waal Malefyt. Tumor derived T cell clones for evaluation of check point inhibitor therapeutics [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A33.
使用免疫疗法改善癌症治疗的临床结果将取决于多种因素,包括肿瘤中效应T细胞的组成和表型,来自骨髓、肿瘤和肿瘤间质室的抑制因子,以及选择有效的治疗性免疫调节剂组合,这些组合可以中和免疫检查点抑制剂或激活T细胞共刺激途径。为了解决选择最佳免疫调节剂组合的问题,我们开发了一种体外培养系统,用于从肿瘤消化系统中刺激和扩大肿瘤浸润淋巴细胞(TILS),使用MHC II类同种异体抗原表达细胞系。我们从扩增的TILS中选择了同种异体特异性CD4和CD8克隆,这些克隆具有耗尽表型,通过细胞表面PD-1、LAG3和TIGIT的表达证实。将T细胞克隆与用不同组合的不调节配体转导的表达同种抗原的细胞系一起培养,比较单药与检查点抑制性单克隆抗体联合使用对T细胞增殖和细胞因子产生的影响。在allo刺激后对几个克隆进行Taqman和流式细胞术分析。TcR β链测序证实了重复同种异体刺激后TIL克隆性的降低。抗pd -1抗体联合抗lag3或抗tigit抗体可增强体外IFN γ和颗粒酶B的产生。该体外系统具有筛选来自不同肿瘤类型的各种TILS中的不调节抗体组合的潜力,并且可以提高我们对新的检查点抑制剂组合的理解。引文格式:Robert L. Shields, Sabine Le Saux, Sophie Li, Manjiri Sathe, Terri McClanahan, Rene De Waal Malefyt。肿瘤来源的T细胞克隆用于检查点抑制剂治疗的评估[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫,2018;6(9增刊):摘要nr A33。
{"title":"Abstract A33: Tumor derived T cell clones for evaluation of check point inhibitor therapeutics","authors":"R. Shields, S. Saux, Sophie Li, M. Sathe, T. Mcclanahan, R. D. Malefyt","doi":"10.1158/2326-6074.TUMIMM17-A33","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A33","url":null,"abstract":"Improving clinical outcomes using immunotherapeutics in cancer treatments will depend on a variety of factors including the composition and phenotype of effector T cell in tumors, the inhibitory factors from myeloid, tumor and stromal compartments of the tumor and selection of efficacious combination of therapeutic immunomodulatory agents that either neutralize immune checkpoint inhibitors or activate T cell costimulatory pathways. To address the issue of selecting optimal combinations of immune modulating agents we developed an in-vitro culture system for allo-stimulating and expanding tumor infiltrating lymphocytes (TILS) from tumor digests with an MHC Class II alloantigen expressing cell line. We selected allo-specific CD4 and CD8 clones from expanded TILS with an exhausted phenotype confirmed by cell surface expression of PD-1, LAG3 and TIGIT. T cell clones were incubated with the alloantigen expressing cell lines transduced with different combinations of immodulatory ligands and compared the effect of single agent versus combinations of checkpoint inhibitory monoclonal antibodies on T cell proliferation and cytokine production. Taqman and flow cytometry analysis was performed on several clones following allo-stimulation. A decrease in TIL clonality with repeated allo-stimulation was confirmed by TcR beta chain sequencing. In-vitro IFN gamma and Granzyme B production was enhanced with anti-PD-1 antibodies in combination with either anti-LAG3 or anti-TIGIT antibodies. This in-vitro system has the potential of screening combinations of immodulatory antibodies in a variety of TILS from different tumor types and can improve our understanding of new checkpoint inhibitor combinations. Citation Format: Robert L. Shields, Sabine Le Saux, Sophie Li, Manjiri Sathe, Terri McClanahan, Rene De Waal Malefyt. Tumor derived T cell clones for evaluation of check point inhibitor therapeutics [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A33.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75656517","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A34: Understanding mechanisms of checkpoint blockade in EGFR-driven glioblastoma 摘要:了解egfr驱动的胶质母细胞瘤的检查点阻断机制
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A34
Alan T Yeo, A. Charest
Background and Rationale: Glioblastoma multiforme (GBM) is the most common type of malignant glioma and has a poor prognosis, which calls for the exploration of alternative therapeutic approaches. Checkpoint blockade immunotherapies have produced significant clinical responses among a spectrum of malignancies. There are ongoing clinical trials for treatment of GBM with checkpoints inhibitors; however, it is anticipated that responses will be varied. With a preclinical GBM mouse model, we systematically evaluated the antitumor efficacy of antibodies targeting PD-1 and CTLA-4 administered as single agent monotherapy or combination in an EGFR genetically driven immunocompetent mouse model of GBM. In addition, we performed comprehensive immunophenotyping in EGFR-driven GBM. Methods: Transgenic EGFR, CDKN2A null, PTEN floxed mice with a floxed luciferase reporter were stereotactically injected intracranially with a TGFa- iCre lentivirus. Cohorts of mice were imaged with bioluminescence to detect growing tumors and were treated with murine monoclonal antibodies against PD-1, CTLA-4 and combination every 3 days for 3 doses beginning post tumor detection. Treated mice and controls were followed for overall survival and analysis of tumor infiltrating immune cells. Another cohort of mice was sacrificed 3 days post completion of treatment and immunophenotyping was performed by flow cytometry. Results: We observe that only mice treated with combination blockade of PD-1 and CTLA-4 had improved survival compared to untreated controls. Single agent monotherapies were ineffective as measured in no improvement in survival compared to controls. A small subset of mice treated with combination blockade of PD-1 and CTLA-4 displayed long term survival up to 100 days post tumor detection. Bioluminescence imaging revealed cytostatic effects in mice treated with combination blockade of PD-1 and CTLA-4 and in a smaller subset, sustained regression. Monotherapy treatment displayed continuing tumor growth similar to untreated controls. Characterization of tumor infiltrating immune cells displayed evidence that only combination treatment of PD-1 and CTLA-4 correlates with an increase in the number of CD8 T cell infiltrate and a decrease in the number of granulocytic MDSC (G-MDSC) infiltrate while monotherapy does not affect the ratio of CD8 to G-MDSC. Conclusions: Immune checkpoint blockade of both PD-1 and CTLA-4 provides survival benefit whereas monotherapies are ineffective in our genetically engineered mouse model of GBM. Studies aimed at revealing the mechanisms of immune-mediated anti-tumor activity from combination blockade are ongoing. Analyses of tumor infiltrating immune cells suggest that the ratio of CD8 to G-MDSCs correlate with efficacy of combination blockade of PD-1 and CTLA-4. Depletion experiments are ongoing to address the role of CD8 and MDSCs in mediating anti-tumor immunity upon single and combination blockade of PD-1 and CTLA-4 in EGFR-driven GBM. Cita
背景与理由:多形性胶质母细胞瘤(GBM)是最常见的恶性胶质瘤类型,其预后较差,需要探索替代治疗方法。检查点阻断免疫疗法在一系列恶性肿瘤中产生了显著的临床反应。目前正在进行用检查点抑制剂治疗GBM的临床试验;但是,预计反应将是不同的。通过临床前GBM小鼠模型,我们系统地评估了靶向PD-1和CTLA-4的抗体在EGFR基因驱动的GBM免疫活性小鼠模型中作为单药单药或联合治疗的抗肿瘤效果。此外,我们对egfr驱动的GBM进行了全面的免疫分型。方法:将转基因EGFR、CDKN2A缺失、PTEN的荧光素酶报告基因固定的小鼠以立体定向方式注入TGFa- iCre慢病毒。对小鼠进行生物发光成像以检测生长的肿瘤,并在肿瘤检测后开始每3天使用小鼠抗PD-1、CTLA-4单克隆抗体或联合单克隆抗体治疗,共3次。对治疗小鼠和对照组进行总体存活和肿瘤浸润免疫细胞分析。另一组小鼠在治疗结束后3天处死,通过流式细胞术进行免疫表型分析。结果:我们观察到,与未治疗的对照组相比,只有联合阻断PD-1和CTLA-4的小鼠存活率有所提高。与对照组相比,单药单药治疗无效,没有改善生存。一小部分接受PD-1和CTLA-4联合阻断治疗的小鼠在肿瘤检测后显示出长达100天的长期生存。生物发光成像显示联合阻断PD-1和CTLA-4治疗的小鼠具有细胞抑制作用,并且在较小的亚群中持续消退。单药治疗显示持续的肿瘤生长与未治疗的对照组相似。肿瘤浸润免疫细胞的表征显示,PD-1和CTLA-4联合治疗与CD8 T细胞浸润数量增加和粒细胞MDSC (G-MDSC)浸润数量减少相关,而单药治疗不影响CD8与G-MDSC的比例。结论:PD-1和CTLA-4的免疫检查点阻断在我们的基因工程小鼠GBM模型中提供了生存益处,而单一治疗无效。旨在揭示免疫介导的联合阻断抗肿瘤活性机制的研究正在进行中。肿瘤浸润免疫细胞的分析表明,CD8与G-MDSCs的比值与联合阻断PD-1和CTLA-4的疗效相关。在egfr驱动的GBM中,CD8和MDSCs在单独或联合阻断PD-1和CTLA-4时介导抗肿瘤免疫的作用正在进行耗尽实验。引用格式:Alan T. Yeo, Alain Charest。了解egfr驱动的胶质母细胞瘤中检查点阻断机制[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫,2018;6(9增刊):摘要nr A34。
{"title":"Abstract A34: Understanding mechanisms of checkpoint blockade in EGFR-driven glioblastoma","authors":"Alan T Yeo, A. Charest","doi":"10.1158/2326-6074.TUMIMM17-A34","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A34","url":null,"abstract":"Background and Rationale: Glioblastoma multiforme (GBM) is the most common type of malignant glioma and has a poor prognosis, which calls for the exploration of alternative therapeutic approaches. Checkpoint blockade immunotherapies have produced significant clinical responses among a spectrum of malignancies. There are ongoing clinical trials for treatment of GBM with checkpoints inhibitors; however, it is anticipated that responses will be varied. With a preclinical GBM mouse model, we systematically evaluated the antitumor efficacy of antibodies targeting PD-1 and CTLA-4 administered as single agent monotherapy or combination in an EGFR genetically driven immunocompetent mouse model of GBM. In addition, we performed comprehensive immunophenotyping in EGFR-driven GBM. Methods: Transgenic EGFR, CDKN2A null, PTEN floxed mice with a floxed luciferase reporter were stereotactically injected intracranially with a TGFa- iCre lentivirus. Cohorts of mice were imaged with bioluminescence to detect growing tumors and were treated with murine monoclonal antibodies against PD-1, CTLA-4 and combination every 3 days for 3 doses beginning post tumor detection. Treated mice and controls were followed for overall survival and analysis of tumor infiltrating immune cells. Another cohort of mice was sacrificed 3 days post completion of treatment and immunophenotyping was performed by flow cytometry. Results: We observe that only mice treated with combination blockade of PD-1 and CTLA-4 had improved survival compared to untreated controls. Single agent monotherapies were ineffective as measured in no improvement in survival compared to controls. A small subset of mice treated with combination blockade of PD-1 and CTLA-4 displayed long term survival up to 100 days post tumor detection. Bioluminescence imaging revealed cytostatic effects in mice treated with combination blockade of PD-1 and CTLA-4 and in a smaller subset, sustained regression. Monotherapy treatment displayed continuing tumor growth similar to untreated controls. Characterization of tumor infiltrating immune cells displayed evidence that only combination treatment of PD-1 and CTLA-4 correlates with an increase in the number of CD8 T cell infiltrate and a decrease in the number of granulocytic MDSC (G-MDSC) infiltrate while monotherapy does not affect the ratio of CD8 to G-MDSC. Conclusions: Immune checkpoint blockade of both PD-1 and CTLA-4 provides survival benefit whereas monotherapies are ineffective in our genetically engineered mouse model of GBM. Studies aimed at revealing the mechanisms of immune-mediated anti-tumor activity from combination blockade are ongoing. Analyses of tumor infiltrating immune cells suggest that the ratio of CD8 to G-MDSCs correlate with efficacy of combination blockade of PD-1 and CTLA-4. Depletion experiments are ongoing to address the role of CD8 and MDSCs in mediating anti-tumor immunity upon single and combination blockade of PD-1 and CTLA-4 in EGFR-driven GBM. Cita","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79833902","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A13: Optimization of a dendritic cell-targeting MIP3α-antigen fusion vaccine in the B16F10 mouse melanoma model A13:树突状细胞靶向mip3 α-抗原融合疫苗在B16F10小鼠黑色素瘤模型中的优化
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A13
James T. Gordy, K. Luo, R. Markham
Background: The chemokine MIP-3α (CCL20) binds to CCR6 found on immature dendritic cells. Vaccines fusing MIP-3α to gp100 have been shown to be effective in therapeutically alleviating melanoma in mouse models. However, that protection was not complete. To optimize the therapy, our laboratory is exploring two avenues. First, we added agents designed to modulate the tolerogenic tumor microenvironment. Here, we report that neutralizing IL-10 at the tumor site enhances the therapeutic anti-melanoma efficacy of a MIP-3α-gp100 DNA vaccine. Secondly, we are optimizing the antigenic profile of the vaccine to help reduce the probability of immunoediting processes leading to tumor therapy escape. Methods: The current studies utilize the B16F10 syngeneic, transplantable, mouse melanoma model system. The MIP-3α-antigen DNA vaccine is administered intramuscularly (i.m.) into the tibialis muscle, followed immediately by i.m. electroporation. Constructs utilized include MIP-3α fused to gp100 alone or in addition to TRP-2. Vaccinations are given therapeutically, beginning at day 3 or 5 post challenge. Tumor sizes, growth, and survival were all assessed. Treatment responses were characterized by flow cytometric analysis of tumor infiltrate. Vaccine-specific T-cells were delineated by gp10025-33 stimulation followed by intracellular cytokine staining for IFN-γ and assessment by flow cytometry. The mechanism of αIL-10 efficacy was explored by RT-PCR and confirmed with a knockout mouse model. Results: With this therapeutic protocol, we demonstrate for the first time that by either neutralizing IL-10 or adding a second antigen to our construct, we are able to enhance the antitumor efficacy of a MIP-3α-gp100 vaccine, leading to significantly smaller tumors, slower growing tumors, and overall increases in mouse survival. Surprisingly, the additive effects of αIL-10 were not shown to be directly mediated by any T-cell parameter tested, including vaccine-specific tumor infiltrating lymphocytes (TILs), total TILs of either CD4+ or CD8+ subset, regulatory T-cells, granzyme positive T-cells, and others. We discovered, however, that IFNα-4 transcripts in the tumor were significantly upregulated in mice given vaccine and αIL-10 compared to vaccine alone. A mouse model with IFNαR1 knocked out eliminated the protection provided by αIL-10, demonstrating that the additional therapeutic value of αIL-10 is primarily mediated by type-I interferons. The immunologic details of the added TRP2 antigen are currently being explored. Conclusions: Efficient targeting of antigen to immature dendritic cells with a chemokine fusion vaccine offers a potential alternative approach to the ex vivo dendritic cell antigen loading protocols currently undergoing clinical investigation. Adding multiple antigens or combining this approach with an IL-10 neutralizing antibody therapy that modulates the tolerogenic tumor microenvironment both enhance vaccine efficacy. Further potential therapy optimizatio
背景:趋化因子MIP-3α (CCL20)与未成熟树突状细胞上发现的CCR6结合。在小鼠模型中,融合MIP-3α与gp100的疫苗已被证明在治疗性缓解黑色素瘤方面是有效的。然而,这种保护并不完全。为了优化治疗,我们的实验室正在探索两种途径。首先,我们添加了旨在调节耐受性肿瘤微环境的药物。在这里,我们报道在肿瘤部位中和IL-10可以增强MIP-3α-gp100 DNA疫苗的抗黑色素瘤疗效。其次,我们正在优化疫苗的抗原谱,以帮助降低免疫编辑过程导致肿瘤治疗逃逸的可能性。方法:本研究采用B16F10同基因、可移植的小鼠黑色素瘤模型系统。mip -3α-抗原DNA疫苗肌内注射(i.m)到胫骨肌,随后立即i.m电穿孔。所使用的构建体包括MIP-3α单独与gp100或与TRP-2融合。疫苗接种是治疗性的,从感染后第3或5天开始。评估肿瘤大小、生长和生存。通过肿瘤浸润的流式细胞术分析来表征治疗效果。通过gp10025-33刺激、细胞内细胞因子IFN-γ染色和流式细胞术评估来描绘疫苗特异性t细胞。采用RT-PCR方法探讨αIL-10的作用机制,并建立敲除小鼠模型进行验证。结果:通过这种治疗方案,我们首次证明,通过中和IL-10或在我们的构建中添加第二抗原,我们能够增强MIP-3α-gp100疫苗的抗肿瘤功效,导致肿瘤显着变小,肿瘤生长缓慢,并且总体上提高了小鼠存活率。令人惊讶的是,αIL-10的累加效应并未被任何测试的t细胞参数直接介导,包括疫苗特异性肿瘤浸润淋巴细胞(til)、CD4+或CD8+亚群的总til、调节性t细胞、颗粒酶阳性t细胞等。然而,我们发现,与单独接种疫苗相比,接种疫苗和αIL-10的小鼠肿瘤中IFNα-4转录物显著上调。敲除IFNαR1的小鼠模型消除了αIL-10提供的保护作用,表明αIL-10的附加治疗价值主要是由i型干扰素介导的。目前正在探索添加的TRP2抗原的免疫学细节。结论:利用趋化因子融合疫苗将抗原有效靶向未成熟树突状细胞,为目前正在进行临床研究的体外树突状细胞抗原加载方案提供了一种潜在的替代方法。添加多种抗原或将这种方法与调节耐受性肿瘤微环境的IL-10中和抗体治疗相结合,都能提高疫苗的效力。目前正在研究的进一步潜在的治疗优化为这条研究路线提供了成为一种新的黑色素瘤治疗方法的希望。引文格式:James T. Gordy, Kun Luo, Richard B. Markham。树突状细胞靶向mip3 α-抗原融合疫苗在B16F10小鼠黑色素瘤模型中的优化[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A13。
{"title":"Abstract A13: Optimization of a dendritic cell-targeting MIP3α-antigen fusion vaccine in the B16F10 mouse melanoma model","authors":"James T. Gordy, K. Luo, R. Markham","doi":"10.1158/2326-6074.TUMIMM17-A13","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A13","url":null,"abstract":"Background: The chemokine MIP-3α (CCL20) binds to CCR6 found on immature dendritic cells. Vaccines fusing MIP-3α to gp100 have been shown to be effective in therapeutically alleviating melanoma in mouse models. However, that protection was not complete. To optimize the therapy, our laboratory is exploring two avenues. First, we added agents designed to modulate the tolerogenic tumor microenvironment. Here, we report that neutralizing IL-10 at the tumor site enhances the therapeutic anti-melanoma efficacy of a MIP-3α-gp100 DNA vaccine. Secondly, we are optimizing the antigenic profile of the vaccine to help reduce the probability of immunoediting processes leading to tumor therapy escape. Methods: The current studies utilize the B16F10 syngeneic, transplantable, mouse melanoma model system. The MIP-3α-antigen DNA vaccine is administered intramuscularly (i.m.) into the tibialis muscle, followed immediately by i.m. electroporation. Constructs utilized include MIP-3α fused to gp100 alone or in addition to TRP-2. Vaccinations are given therapeutically, beginning at day 3 or 5 post challenge. Tumor sizes, growth, and survival were all assessed. Treatment responses were characterized by flow cytometric analysis of tumor infiltrate. Vaccine-specific T-cells were delineated by gp10025-33 stimulation followed by intracellular cytokine staining for IFN-γ and assessment by flow cytometry. The mechanism of αIL-10 efficacy was explored by RT-PCR and confirmed with a knockout mouse model. Results: With this therapeutic protocol, we demonstrate for the first time that by either neutralizing IL-10 or adding a second antigen to our construct, we are able to enhance the antitumor efficacy of a MIP-3α-gp100 vaccine, leading to significantly smaller tumors, slower growing tumors, and overall increases in mouse survival. Surprisingly, the additive effects of αIL-10 were not shown to be directly mediated by any T-cell parameter tested, including vaccine-specific tumor infiltrating lymphocytes (TILs), total TILs of either CD4+ or CD8+ subset, regulatory T-cells, granzyme positive T-cells, and others. We discovered, however, that IFNα-4 transcripts in the tumor were significantly upregulated in mice given vaccine and αIL-10 compared to vaccine alone. A mouse model with IFNαR1 knocked out eliminated the protection provided by αIL-10, demonstrating that the additional therapeutic value of αIL-10 is primarily mediated by type-I interferons. The immunologic details of the added TRP2 antigen are currently being explored. Conclusions: Efficient targeting of antigen to immature dendritic cells with a chemokine fusion vaccine offers a potential alternative approach to the ex vivo dendritic cell antigen loading protocols currently undergoing clinical investigation. Adding multiple antigens or combining this approach with an IL-10 neutralizing antibody therapy that modulates the tolerogenic tumor microenvironment both enhance vaccine efficacy. Further potential therapy optimizatio","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85202643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A22: Augmentation of a novel adenoviral vaccine strategy by checkpoint inhibitors A22:通过检查点抑制剂增强一种新的腺病毒疫苗策略
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A22
Erika J. Crosby, Gang-jun Lei, Junping Wei, X. Yang, Tao Wang, Congxiao Liu, H. Lyerly, Z. Hartman
The immunologic hurdles for a vaccine targeting cancer are much higher than for those targeting an infectious disease. The profoundly immunosuppressive tumor microenvironment, the lack of microbial danger signals, and the need to break tolerance without causing catastrophic autoimmunity are all considerations that must be made when designing an effective anti-cancer vaccine. Immune checkpoint blockade (ICB) including programmed death 1 (PD1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) monoclonal antibodies have revolutionized cancer treatment as a whole, including the potential for a successful cancer vaccine. Human epidermal growth factor receptor 2 (HER2) is an oncogene that is overexpressed in 20-25% of breast cancers and has been successfully targeted with therapeutic anti-HER2 therapies, particularly antibody combinations like trastuzumab and pertuzumab. However, even the most potent anti-HER2 therapy available is often accompanied by a high rate of recurrence, with the many responders eventually becoming resistant. Given the relative success of combination therapy using antibodies targeting different epitopes of HER2, we hypothesized that a HER2 targeting vaccine approach could further broaden the immune repertoire and reduce rates of resistance and recurrence. We developed both an implantable and a mammary specific spontaneous tumor model driven by an oncogenic isoform of HER2 (HER2Δ16). Using these models we tested a novel adenoviral vaccine platform encoding an inactive HER2Δ16 variant. We have shown that this isoform is significantly more oncogenic than full length HER2 and plays a role in anti-HER2 therapeutic resistance. Using the implantable tumor model, we found that therapeutic vaccination elicits a robust anti-HER2 specific cellular and humoral response, as well as significantly inhibits tumor growth of HER2Δ16-positive tumors. While effective at reducing tumor growth, we observed that our vaccine was typically not capable of eliciting tumor regression in mice, due to the immunosuppressive tumor microenvironment of established tumors. As such, we tested our vaccine platform in combination with two recently approved checkpoint inhibitors anti-CTLA-4 and anti-PD-1. This combination greatly enhanced the HER2-specific immune response as well as the antitumor effect seen post vaccination, with many tumors exhibiting complete regression. Our spontaneous model provides the ideal setting to test our vaccine platform as it is tolerant to human HER2, driven by HER2 expression, and grows at a rate that provides sufficient time to intervene with an immune targeting therapy. Using this model we have further shown that vaccination against HER2Δ16 can prevent spontaneous tumor formation and work is ongoing to test therapeutic vaccine strategies in combination with ICB. Future studies will be focused on determining the exact mechanism of regression and evaluating the impact on de novo and acquired resistance by combining this novel therapeutic
针对癌症的疫苗的免疫障碍要比针对传染病的疫苗高得多。肿瘤微环境的深度免疫抑制,微生物危险信号的缺乏,以及在不引起灾难性自身免疫的情况下打破耐受性的需要,都是设计有效抗癌疫苗时必须考虑的因素。免疫检查点阻断(ICB),包括程序性死亡1 (PD1)和细胞毒性t淋巴细胞抗原4 (CTLA-4)单克隆抗体,从整体上彻底改变了癌症治疗,包括成功开发癌症疫苗的潜力。人表皮生长因子受体2 (HER2)是一种致癌基因,在20-25%的乳腺癌中过度表达,并且已经成功地靶向治疗性抗HER2疗法,特别是曲妥珠单抗和帕妥珠单抗等抗体组合。然而,即使是最有效的抗her2治疗也常常伴随着高复发率,许多应答者最终会变得耐药。鉴于使用针对不同HER2表位的抗体进行联合治疗的相对成功,我们假设HER2靶向疫苗方法可以进一步扩大免疫库,降低耐药和复发率。我们开发了一种可植入的和乳腺特异性的自发肿瘤模型,该模型由HER2的致癌异构体驱动(HER2Δ16)。利用这些模型,我们测试了一种新的腺病毒疫苗平台,该平台编码一种失活的HER2Δ16变体。我们已经证明,这种亚型明显比全长HER2更具致癌性,并在抗HER2治疗耐药性中发挥作用。通过植入式肿瘤模型,我们发现治疗性疫苗可引起强大的抗her2特异性细胞和体液反应,并显著抑制HER2Δ16-positive肿瘤的肿瘤生长。虽然能有效抑制肿瘤生长,但我们观察到,由于已建立肿瘤的免疫抑制肿瘤微环境,我们的疫苗通常不能引起小鼠肿瘤消退。因此,我们测试了我们的疫苗平台与两种最近批准的抗ctla -4和抗pd -1检查点抑制剂的组合。这种组合极大地增强了her2特异性免疫反应以及接种后的抗肿瘤效果,许多肿瘤表现出完全消退。我们的自发模型为测试我们的疫苗平台提供了理想的环境,因为它对人类HER2具有耐受性,由HER2表达驱动,并且以足够的速度生长,为免疫靶向治疗提供了足够的时间进行干预。使用该模型,我们进一步表明,针对HER2Δ16的疫苗接种可以预防自发肿瘤的形成,并且正在进行与ICB结合的治疗性疫苗策略的测试。未来的研究将集中于确定回归的确切机制,并通过将这种新的治疗平台与当前标准的HER2靶向治疗相结合,评估对新生和获得性耐药的影响。我们的结论是,ICB的结合可以帮助克服免疫障碍,并增加治疗性癌症疫苗的效用。引用格式:Erika J. Crosby,雷刚军,魏俊平,杨晓毅,王涛,刘从晓,H Kim Lyerly, Zachary C. Hartman。检查点抑制剂增强一种新型腺病毒疫苗策略[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A22。
{"title":"Abstract A22: Augmentation of a novel adenoviral vaccine strategy by checkpoint inhibitors","authors":"Erika J. Crosby, Gang-jun Lei, Junping Wei, X. Yang, Tao Wang, Congxiao Liu, H. Lyerly, Z. Hartman","doi":"10.1158/2326-6074.TUMIMM17-A22","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A22","url":null,"abstract":"The immunologic hurdles for a vaccine targeting cancer are much higher than for those targeting an infectious disease. The profoundly immunosuppressive tumor microenvironment, the lack of microbial danger signals, and the need to break tolerance without causing catastrophic autoimmunity are all considerations that must be made when designing an effective anti-cancer vaccine. Immune checkpoint blockade (ICB) including programmed death 1 (PD1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) monoclonal antibodies have revolutionized cancer treatment as a whole, including the potential for a successful cancer vaccine. Human epidermal growth factor receptor 2 (HER2) is an oncogene that is overexpressed in 20-25% of breast cancers and has been successfully targeted with therapeutic anti-HER2 therapies, particularly antibody combinations like trastuzumab and pertuzumab. However, even the most potent anti-HER2 therapy available is often accompanied by a high rate of recurrence, with the many responders eventually becoming resistant. Given the relative success of combination therapy using antibodies targeting different epitopes of HER2, we hypothesized that a HER2 targeting vaccine approach could further broaden the immune repertoire and reduce rates of resistance and recurrence. We developed both an implantable and a mammary specific spontaneous tumor model driven by an oncogenic isoform of HER2 (HER2Δ16). Using these models we tested a novel adenoviral vaccine platform encoding an inactive HER2Δ16 variant. We have shown that this isoform is significantly more oncogenic than full length HER2 and plays a role in anti-HER2 therapeutic resistance. Using the implantable tumor model, we found that therapeutic vaccination elicits a robust anti-HER2 specific cellular and humoral response, as well as significantly inhibits tumor growth of HER2Δ16-positive tumors. While effective at reducing tumor growth, we observed that our vaccine was typically not capable of eliciting tumor regression in mice, due to the immunosuppressive tumor microenvironment of established tumors. As such, we tested our vaccine platform in combination with two recently approved checkpoint inhibitors anti-CTLA-4 and anti-PD-1. This combination greatly enhanced the HER2-specific immune response as well as the antitumor effect seen post vaccination, with many tumors exhibiting complete regression. Our spontaneous model provides the ideal setting to test our vaccine platform as it is tolerant to human HER2, driven by HER2 expression, and grows at a rate that provides sufficient time to intervene with an immune targeting therapy. Using this model we have further shown that vaccination against HER2Δ16 can prevent spontaneous tumor formation and work is ongoing to test therapeutic vaccine strategies in combination with ICB. Future studies will be focused on determining the exact mechanism of regression and evaluating the impact on de novo and acquired resistance by combining this novel therapeutic ","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79618782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
期刊
Checkpoints and Immunomodulation
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