Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A076
Y. Gozlan, S. Hilgendorf, A. Aronin, Yehudith Sagiv, Liat Ben-gigi-Tamir, S. Amsili, A. Tamir, I. Pecker, Shirley Greenwald, A. Chajut, A. Foley-Comer, Y. Pereg, A. Peled, M. Dranitzki-Elhalel, E. Bremer
Background: The (re)activation of anticancer innate and adaptive immunity is at the forefront of developments in cancer therapy. Here, we report on a new immunotherapeutic fusion protein, termed Dual Signaling Protein 107 (DSP107). DSP107 was designed to combine activation of innate and adaptive immunity, by both blocking CD47/SIRPα interaction and activating 4-1BB. CD47 is overexpressed on cancer cells and upon binding to SIRPα on phagocytes transmits a “don’t eat me” signal, thereby suppressing innate immunity. 4-1BB is a costimulatory receptor that is transiently upregulated on tumor-infiltrating T-cells and is considered a surrogate marker for the tumor-reactive T-cell population. Activation of 4-1BB using its ligand or by agonistic antibodies reactivates anti-cancer T-cell immunity. In DSP107, the extracellular domains of SIRPα and 41BBL have been fused, yielding a dual function protein. DSP107 is produced as a homotrimer due to the trimerization property of 41BBL, an essential element for activating the 41BB receptor, a member of the TNF super-family of receptors. DSP107 was designed to bind to CD47 on tumor cells, thereby removing the inhibitory signal delivered to phagocytes. Simultaneously, CD47-mediated surface immobilization of DSP107 enables delivery of the 41BBL-4-1BB costimulatory signal to tumor localized T-cells. This dual immunomodulatory effect of DSP107 is designed to unleash both innate and adaptive immune responses targeted to the tumor site. Methods and Results: Trimeric DSP107 was successfully produced in a mammalian expression system. Both sides of DSP107 bound their cognate counterparts in kinetic Blitz binding assays and on human tumor and immune cell surfaces. The binding affinity of DSP107 was 1.6 nM for human CD47 and 0.69 nM for human 4-1BB as determined using BIAcore analysis. DSP107 blocked the interaction of SIRPα with CD47 in an ELISA-based competition assay (EC50 of 0.03 nM). DSP107 induced granulocyte- and macrophage-mediated phagocytosis of several lymphoma, leukemia and carcinoma cell lines in vitro. Further, DSP107 treatment triggered phagocytosis of primary AML cells by autologous macrophages. Co-treatment with DSP107 and therapeutic tumor-targeting antibodies, i.e., rituximab or cetuximab, resulted in enhanced phagocytosis of lymphoma or carcinoma cells, respectively. In a reporter assay measuring IL-8 secretion upon binding to/activation of 4-1BB, DSP107 activated 4-1BB signaling only in the presence of CD47-expressing cells. Further, DSP107 augmented the activation of purified T-cells activated by suboptimal concentrations of αCD3 + IL2 or αCD3/αCD28 Dynabeads in CD47 coated plates, as measured by percentage of CD25 expressing cells (up to 3-fold). When PBMCs were co-cultured with or without CD47-expressing cancer cells and stimulated with suboptimal concentrations of αCD3 + IL2, DSP107 treatment resulted in increased secretion of IFNg up to 2-fold), and increased T-cell proliferation (up to 2-fold). Con
背景:抗癌先天免疫和适应性免疫的(再)激活是癌症治疗发展的前沿。在这里,我们报道了一种新的免疫治疗融合蛋白,称为双信号蛋白107 (DSP107)。DSP107被设计为通过阻断CD47/SIRPα相互作用和激活4-1BB来联合激活先天免疫和适应性免疫。CD47在癌细胞上过表达,与吞噬细胞上的SIRPα结合后传递“不要吃我”的信号,从而抑制先天免疫。4-1BB是一种共刺激受体,在肿瘤浸润性t细胞上短暂上调,被认为是肿瘤反应性t细胞群的替代标记物。使用4-1BB配体或激动抗体激活4-1BB可重新激活抗癌t细胞免疫。在DSP107中,SIRPα和41BBL的细胞外结构域融合,产生双重功能蛋白。由于41BBL的三聚性,DSP107作为同源三聚体产生,41BBL是激活41BB受体的必要元素,41BB受体是TNF超家族受体的成员。DSP107被设计成与肿瘤细胞上的CD47结合,从而去除传递给吞噬细胞的抑制信号。同时,cd47介导的DSP107表面固定化能够将41BBL-4-1BB共刺激信号传递到肿瘤定位的t细胞。DSP107的这种双重免疫调节作用旨在释放针对肿瘤部位的先天和适应性免疫反应。方法与结果:三聚体DSP107在哺乳动物表达系统中获得成功。在动力学Blitz结合试验和人类肿瘤和免疫细胞表面上,DSP107的两边都与同源对应物结合。通过BIAcore分析,DSP107对人CD47的结合亲和力为1.6 nM,对人4-1BB的结合亲和力为0.69 nM。在elisa竞争实验中,DSP107阻断了SIRPα与CD47的相互作用(EC50为0.03 nM)。DSP107体外诱导粒细胞和巨噬细胞介导的多种淋巴瘤、白血病和癌细胞的吞噬作用。此外,DSP107治疗引发自体巨噬细胞吞噬原发性AML细胞。与DSP107和治疗性肿瘤靶向抗体(即利妥昔单抗或西妥昔单抗)共同治疗,分别导致淋巴瘤或癌细胞的吞噬能力增强。在一项测量结合/激活4-1BB时IL-8分泌的报告细胞实验中,DSP107仅在表达cd47的细胞存在时激活4-1BB信号。此外,DSP107增强了被αCD3 + IL2或αCD3/αCD28 Dynabeads在CD47包被板上的亚理想浓度激活的纯化t细胞的活化,通过CD25表达细胞的百分比来测量(高达3倍)。当PBMCs与表达cd47的癌细胞共培养或不与表达cd47的癌细胞共培养,并用次优浓度的αCD3 + IL2刺激时,DSP107治疗导致IFNg分泌增加高达2倍,t细胞增殖增加高达2倍。结论:我们证明了DSP107的可行性和功能活性,DSP107是一种结合先天和适应性免疫系统激活的新型治疗蛋白。DSP107的两个功能侧的双重靶向提供了同时起作用的多种功能,并可能产生协同效应。DSP107目前正处于ind研究和CMC开发阶段。DSP平台可设计为选择性肿瘤部位或微环境靶向,适用于大多数检查点靶点。引文格式:Yosi M. Gozlan, Susan Hilgendorf, Alexandra Aronin, Yehudith Sagiv, Liat Ben-gigi-Tamir, Shira Amsili, Ami Tamir, Iris Pecker, Shirley Greenwald, Ayelet Chajut, Adam Foley-Comer, Yaron Pereg, Amnon Peled, michael Dranitzki-Elhalel, Edwin Bremer。一种用于癌症免疫治疗的新型SIRPα-4-1BBL双信号蛋白(DSP)[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A076。
{"title":"Abstract A076: DSP107—a novel SIRPα-4-1BBL dual signaling protein (DSP) for cancer immunotherapy","authors":"Y. Gozlan, S. Hilgendorf, A. Aronin, Yehudith Sagiv, Liat Ben-gigi-Tamir, S. Amsili, A. Tamir, I. Pecker, Shirley Greenwald, A. Chajut, A. Foley-Comer, Y. Pereg, A. Peled, M. Dranitzki-Elhalel, E. Bremer","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A076","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A076","url":null,"abstract":"Background: The (re)activation of anticancer innate and adaptive immunity is at the forefront of developments in cancer therapy. Here, we report on a new immunotherapeutic fusion protein, termed Dual Signaling Protein 107 (DSP107). DSP107 was designed to combine activation of innate and adaptive immunity, by both blocking CD47/SIRPα interaction and activating 4-1BB. CD47 is overexpressed on cancer cells and upon binding to SIRPα on phagocytes transmits a “don’t eat me” signal, thereby suppressing innate immunity. 4-1BB is a costimulatory receptor that is transiently upregulated on tumor-infiltrating T-cells and is considered a surrogate marker for the tumor-reactive T-cell population. Activation of 4-1BB using its ligand or by agonistic antibodies reactivates anti-cancer T-cell immunity. In DSP107, the extracellular domains of SIRPα and 41BBL have been fused, yielding a dual function protein. DSP107 is produced as a homotrimer due to the trimerization property of 41BBL, an essential element for activating the 41BB receptor, a member of the TNF super-family of receptors. DSP107 was designed to bind to CD47 on tumor cells, thereby removing the inhibitory signal delivered to phagocytes. Simultaneously, CD47-mediated surface immobilization of DSP107 enables delivery of the 41BBL-4-1BB costimulatory signal to tumor localized T-cells. This dual immunomodulatory effect of DSP107 is designed to unleash both innate and adaptive immune responses targeted to the tumor site. Methods and Results: Trimeric DSP107 was successfully produced in a mammalian expression system. Both sides of DSP107 bound their cognate counterparts in kinetic Blitz binding assays and on human tumor and immune cell surfaces. The binding affinity of DSP107 was 1.6 nM for human CD47 and 0.69 nM for human 4-1BB as determined using BIAcore analysis. DSP107 blocked the interaction of SIRPα with CD47 in an ELISA-based competition assay (EC50 of 0.03 nM). DSP107 induced granulocyte- and macrophage-mediated phagocytosis of several lymphoma, leukemia and carcinoma cell lines in vitro. Further, DSP107 treatment triggered phagocytosis of primary AML cells by autologous macrophages. Co-treatment with DSP107 and therapeutic tumor-targeting antibodies, i.e., rituximab or cetuximab, resulted in enhanced phagocytosis of lymphoma or carcinoma cells, respectively. In a reporter assay measuring IL-8 secretion upon binding to/activation of 4-1BB, DSP107 activated 4-1BB signaling only in the presence of CD47-expressing cells. Further, DSP107 augmented the activation of purified T-cells activated by suboptimal concentrations of αCD3 + IL2 or αCD3/αCD28 Dynabeads in CD47 coated plates, as measured by percentage of CD25 expressing cells (up to 3-fold). When PBMCs were co-cultured with or without CD47-expressing cancer cells and stimulated with suboptimal concentrations of αCD3 + IL2, DSP107 treatment resulted in increased secretion of IFNg up to 2-fold), and increased T-cell proliferation (up to 2-fold). Con","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79798023","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.cricimteatiaacr18-a100
Andreia V. Pinho, M. V. Bulck, L. Chantrill, M. Arshi, D. Herrmann, C. Vennin, A. Gill, P. Timpson, A. Biankin, Jianmin Wu, I. Rooman
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, being predicted to become the second leading cause of cancer-related death by 2030. Chronic pancreatitis is a risk factor for PDAC and both diseases are characterized by a strong desmoplastic response, comprised of activated myofibroblasts and immune cell infiltrates. Genomic aberrations in the SLIT-ROBO pathway are frequent in PDAC. Nevertheless, their role in the pancreas is unclear. We have used an integrative approach combining the study of murine models and PDAC patients with the objective of unraveling the function of the SLIT-ROBO signaling pathway in pancreatic disease. RNA expression of SLIT-ROBO genes was analyzed in murine normal pancreas, pancreatitis and PDAC. Primary cell cultures and experimental pancreatitis were studied using pancreas-specific Robo2 (Pdx1-Cre;Robo2F/F) and whole-body Slit1 (Slit1-/-) knockout mice. Gene and protein expression were assessed in a cohort of PDAC patients (n=109). In mouse pancreatitis and PDAC, epithelial Robo2 expression is lost while Robo1 expression becomes most prominent in the stroma. Pdx1Cre;Robo2F/F pancreatic cell cultures showed increased activation of Robo1-positive myofibroblasts and induction of TGF-β and Wnt pathways. Likewise, induction of pancreatitis in Pdx1Cre;Robo2F/F mice enhanced myofibroblast activation, collagen crosslinking, T-cell infiltration and tumorigenic immune markers. Similar results were obtained using Slit1-/- animals. Moreover, TGF-β inhibition using galunisertib treatment suppressed Robo2-mediated effects in the microenvironment. In patients, ROBO2 expression is overall low in PDAC, while ROBO1 is variably expressed in epithelium and high in the stroma. ROBO1 expression is correlated with markers of activated stroma, Wnt and TGF-β pathways. ROBO2low;ROBO1high subpopulation of patients present the poorest survival rates. In conclusion, Robo2 acts nonautonomously as a stroma suppressor gene by restraining myofibroblast activation and inflammation in the pancreatic microenvironment. ROBO1/2 expression is prognostic in PDAC patients and may guide therapy with TGF-β inhibitors or immunotherapies, currently being tested in clinical trials for advanced pancreatic cancer. Citation Format: Andreia V. Pinho, Mathias Van Bulck, Lorraine Chantrill, Mehreen Arshi, David Herrmann, Claire Vennin, APGI - Australian Pancreatic Cancer Genome Initiative, Anthony Gill, Paul Timpson, Andrew Biankin, Jianmin Wu, Ilse Rooman. ROBO2 is a stroma suppressor gene in the pancreas through regulation of TGF-β [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A100.
胰腺导管腺癌(PDAC)预后不佳,预计到2030年将成为癌症相关死亡的第二大原因。慢性胰腺炎是PDAC的危险因素,这两种疾病的特征都是强烈的纤维组织增生反应,包括活化的肌成纤维细胞和免疫细胞浸润。在PDAC中,SLIT-ROBO通路的基因组畸变是常见的。然而,它们在胰腺中的作用尚不清楚。我们采用了一种综合方法,结合小鼠模型和PDAC患者的研究,目的是揭示SLIT-ROBO信号通路在胰腺疾病中的功能。分析小鼠正常胰腺、胰腺炎和PDAC中SLIT-ROBO基因的RNA表达。使用胰腺特异性Robo2 (Pdx1-Cre;Robo2F/F)和全身Slit1 (Slit1-/-)敲除小鼠,研究原代细胞培养和实验性胰腺炎。在一组PDAC患者(n=109)中评估基因和蛋白表达。在小鼠胰腺炎和PDAC中,上皮细胞中Robo2表达缺失,而间质中Robo1表达最为突出。胰腺细胞培养显示robo1阳性肌成纤维细胞活化增加,TGF-β和Wnt通路诱导增加。同样,诱导Pdx1Cre;Robo2F/F小鼠胰腺炎可增强肌成纤维细胞活化、胶原交联、t细胞浸润和致瘤性免疫标志物。用Slit1-/-动物也得到了类似的结果。此外,使用galunisertib治疗抑制TGF-β可抑制微环境中robo2介导的作用。在患者中,ROBO2在PDAC中的表达总体较低,而ROBO1在上皮中表达不同,在间质中表达较高。ROBO1的表达与活化基质、Wnt和TGF-β通路的标志物相关。ROBO2low;ROBO1high亚群患者生存率最低。总之,在胰腺微环境中,Robo2非自主地作为基质抑制基因抑制肌成纤维细胞的激活和炎症。ROBO1/2的表达是PDAC患者的预后,可能指导TGF-β抑制剂或免疫疗法的治疗,目前正在晚期胰腺癌的临床试验中进行测试。引文格式:Andreia V. Pinho, Mathias Van Bulck, Lorraine Chantrill, Mehreen Arshi, David Herrmann, Claire Vennin, APGI - Australian Pancreatic Cancer Genome Initiative, Anthony Gill, Paul Timpson, Andrew Biankin, Jianmin Wu, Ilse Rooman。ROBO2是通过调控TGF-β在胰腺中发挥基质抑制作用的基因[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志,2019;7(2增刊):摘要nr A100。
{"title":"Abstract A100: ROBO2 is a stroma suppressor gene in the pancreas through regulation of TGF-β","authors":"Andreia V. Pinho, M. V. Bulck, L. Chantrill, M. Arshi, D. Herrmann, C. Vennin, A. Gill, P. Timpson, A. Biankin, Jianmin Wu, I. Rooman","doi":"10.1158/2326-6074.cricimteatiaacr18-a100","DOIUrl":"https://doi.org/10.1158/2326-6074.cricimteatiaacr18-a100","url":null,"abstract":"Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, being predicted to become the second leading cause of cancer-related death by 2030. Chronic pancreatitis is a risk factor for PDAC and both diseases are characterized by a strong desmoplastic response, comprised of activated myofibroblasts and immune cell infiltrates. Genomic aberrations in the SLIT-ROBO pathway are frequent in PDAC. Nevertheless, their role in the pancreas is unclear. We have used an integrative approach combining the study of murine models and PDAC patients with the objective of unraveling the function of the SLIT-ROBO signaling pathway in pancreatic disease. RNA expression of SLIT-ROBO genes was analyzed in murine normal pancreas, pancreatitis and PDAC. Primary cell cultures and experimental pancreatitis were studied using pancreas-specific Robo2 (Pdx1-Cre;Robo2F/F) and whole-body Slit1 (Slit1-/-) knockout mice. Gene and protein expression were assessed in a cohort of PDAC patients (n=109). In mouse pancreatitis and PDAC, epithelial Robo2 expression is lost while Robo1 expression becomes most prominent in the stroma. Pdx1Cre;Robo2F/F pancreatic cell cultures showed increased activation of Robo1-positive myofibroblasts and induction of TGF-β and Wnt pathways. Likewise, induction of pancreatitis in Pdx1Cre;Robo2F/F mice enhanced myofibroblast activation, collagen crosslinking, T-cell infiltration and tumorigenic immune markers. Similar results were obtained using Slit1-/- animals. Moreover, TGF-β inhibition using galunisertib treatment suppressed Robo2-mediated effects in the microenvironment. In patients, ROBO2 expression is overall low in PDAC, while ROBO1 is variably expressed in epithelium and high in the stroma. ROBO1 expression is correlated with markers of activated stroma, Wnt and TGF-β pathways. ROBO2low;ROBO1high subpopulation of patients present the poorest survival rates. In conclusion, Robo2 acts nonautonomously as a stroma suppressor gene by restraining myofibroblast activation and inflammation in the pancreatic microenvironment. ROBO1/2 expression is prognostic in PDAC patients and may guide therapy with TGF-β inhibitors or immunotherapies, currently being tested in clinical trials for advanced pancreatic cancer. Citation Format: Andreia V. Pinho, Mathias Van Bulck, Lorraine Chantrill, Mehreen Arshi, David Herrmann, Claire Vennin, APGI - Australian Pancreatic Cancer Genome Initiative, Anthony Gill, Paul Timpson, Andrew Biankin, Jianmin Wu, Ilse Rooman. ROBO2 is a stroma suppressor gene in the pancreas through regulation of TGF-β [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A100.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85074994","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A065
O. Dufva, J. Klievink, K. Saeed, M. Kankainen, Mette Ilander, Tiiina Hannunen, S. Lagström, P. Ellonen, Dean Anthony Lee, S. Mustjoki
Harnessing natural killer (NK) cells to attack tumors could improve immune-based cancer treatment strategies. However, mechanisms regulating sensitivity or resistance of cancer cells to the effector function of NK cells are incompletely understood. Here, we performed genome-scale CRISPR-Cas9 loss-of-function screens in human cancer cells to discover genes that influence susceptibility to primary human NK cells. To screen for genes essential for the interaction between NK cells and cancer cells, we infected human cancer cells expressing Cas9 with a genome-scale lentiviral guide RNA library. The resulting pool of knockout cells was exposed to NK cells expanded from peripheral blood of healthy donors. Enriched and depleted knockout clones were detected by next-generation sequencing of the integrated sgRNA cassettes, enabling identification of genes conferring resistance or susceptibility to NK cell-mediated lysis. The screens were performed in cell lines from diverse cancer types, including chronic myeloid leukemia (CML), B cell acute lymphoblastic leukemia, diffuse large B-cell lymphoma (DLBCL), and multiple myeloma. We recovered several known mechanisms of NK cell/cancer cell interactions, demonstrating feasibility of the screening approach. Loss of genes encoding components of the MHC class I complex (B2M, HLA-A, HLA-C, HLA-E) sensitized multiple cancer cell lines to NK cell-mediated lysis. This is consistent with missing-self recognition as a fundamental mechanism of NK cell activation. Furthermore, knockout of IFN-JAK-STAT signaling mediators led to increased tumor cell lysis, suggesting that MHC class I induction in response to NK cell-derived IFN gamma enables NK cell evasion by tumor cells. We also identified genes essential for effective NK cell-mediated lysis. NCR3LG1, encoding the B7-H6 ligand for the NKp30 activating NK cell receptor, was essential for NK cell lysis of CML cells. In contrast, knockout of apoptotic mediators and TRAIL pathway components conferred resistance to NK cell cytotoxicity in DLBCL cells, indicating heterogeneity in NK cell/cancer cell interactions between cancer types. Our data support a view that distinct mechanisms regulate sensitivity to NK cell cytotoxicity in different cancers. Importantly, our results indicate that loss-of-function mutations in the antigen-presenting machinery and the IFN-JAK-STAT pathway sensitize tumors to NK cell effector function. As alterations in these genes are associated with resistance to T-cell immunotherapies such as PD-1 blockade, NK cell-based therapies could be employed to overcome resistance in these patients. In summary, we suggest that systematic identification of mechanisms governing tumor immune susceptibility has the potential to uncover novel immunotherapy targets. Citation Format: Olli Dufva, Jay Klievink, Khalid Saeed, Matti Kankainen, Mette Ilander, Tiiina Hannunen, Sonja Lagstrom, Pekka Ellonen, Dean A Lee, Satu Mustjoki. Genome-scale CRISPR screens identify essenti
利用自然杀伤(NK)细胞攻击肿瘤可以改善基于免疫的癌症治疗策略。然而,调节癌细胞对NK细胞效应功能的敏感性或抗性的机制尚不完全清楚。在这里,我们在人类癌细胞中进行了基因组规模的CRISPR-Cas9功能丧失筛选,以发现影响对原代人类NK细胞易感性的基因。为了筛选NK细胞与癌细胞相互作用所必需的基因,我们用基因组级慢病毒引导RNA文库感染表达Cas9的人类癌细胞。由此产生的敲除细胞池暴露于从健康供者外周血扩增的NK细胞。通过对整合的sgRNA磁带进行下一代测序,可以检测到富集和缺失的敲除克隆,从而鉴定出对NK细胞介导的裂解具有抗性或易感性的基因。筛选来自不同癌症类型的细胞系,包括慢性髓性白血病(CML)、B细胞急性淋巴细胞白血病、弥漫性大B细胞淋巴瘤(DLBCL)和多发性骨髓瘤。我们恢复了NK细胞/癌细胞相互作用的几个已知机制,证明了筛选方法的可行性。编码MHC I类复合体(B2M, HLA-A, HLA-C, HLA-E)成分的基因缺失使多种癌细胞对NK细胞介导的裂解敏感。这与缺失自我识别作为NK细胞激活的基本机制是一致的。此外,IFN- jak - stat信号介质的敲除导致肿瘤细胞裂解增加,这表明MHC I类诱导对NK细胞来源的IFN γ的反应使NK细胞逃避肿瘤细胞。我们还确定了有效NK细胞介导的裂解所必需的基因。NCR3LG1编码NKp30激活NK细胞受体的B7-H6配体,是CML细胞NK细胞裂解所必需的。相比之下,敲除凋亡介质和TRAIL通路成分使DLBCL细胞对NK细胞的细胞毒性产生抗性,这表明NK细胞/癌细胞在不同癌症类型之间的相互作用存在异质性。我们的数据支持一种观点,即在不同的癌症中,不同的机制调节对NK细胞毒性的敏感性。重要的是,我们的研究结果表明,抗原呈递机制和IFN-JAK-STAT通路的功能缺失突变使肿瘤对NK细胞效应物功能敏感。由于这些基因的改变与对t细胞免疫疗法(如PD-1阻断)的耐药性有关,因此可以采用基于NK细胞的疗法来克服这些患者的耐药性。总之,我们认为系统地识别肿瘤免疫易感性的机制有可能发现新的免疫治疗靶点。引文格式:Olli Dufva, Jay Klievink, Khalid Saeed, Matti Kankainen, Mette Ilander, Tiiina Hannunen, Sonja Lagstrom, Pekka Ellonen, Dean A Lee, Satu Mustjoki。基因组级CRISPR筛选鉴定肿瘤对NK细胞敏感性的必要基因[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A065。
{"title":"Abstract A065: Genome-scale CRISPR screens identify essential genes for tumor sensitivity to NK cells","authors":"O. Dufva, J. Klievink, K. Saeed, M. Kankainen, Mette Ilander, Tiiina Hannunen, S. Lagström, P. Ellonen, Dean Anthony Lee, S. Mustjoki","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A065","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A065","url":null,"abstract":"Harnessing natural killer (NK) cells to attack tumors could improve immune-based cancer treatment strategies. However, mechanisms regulating sensitivity or resistance of cancer cells to the effector function of NK cells are incompletely understood. Here, we performed genome-scale CRISPR-Cas9 loss-of-function screens in human cancer cells to discover genes that influence susceptibility to primary human NK cells. To screen for genes essential for the interaction between NK cells and cancer cells, we infected human cancer cells expressing Cas9 with a genome-scale lentiviral guide RNA library. The resulting pool of knockout cells was exposed to NK cells expanded from peripheral blood of healthy donors. Enriched and depleted knockout clones were detected by next-generation sequencing of the integrated sgRNA cassettes, enabling identification of genes conferring resistance or susceptibility to NK cell-mediated lysis. The screens were performed in cell lines from diverse cancer types, including chronic myeloid leukemia (CML), B cell acute lymphoblastic leukemia, diffuse large B-cell lymphoma (DLBCL), and multiple myeloma. We recovered several known mechanisms of NK cell/cancer cell interactions, demonstrating feasibility of the screening approach. Loss of genes encoding components of the MHC class I complex (B2M, HLA-A, HLA-C, HLA-E) sensitized multiple cancer cell lines to NK cell-mediated lysis. This is consistent with missing-self recognition as a fundamental mechanism of NK cell activation. Furthermore, knockout of IFN-JAK-STAT signaling mediators led to increased tumor cell lysis, suggesting that MHC class I induction in response to NK cell-derived IFN gamma enables NK cell evasion by tumor cells. We also identified genes essential for effective NK cell-mediated lysis. NCR3LG1, encoding the B7-H6 ligand for the NKp30 activating NK cell receptor, was essential for NK cell lysis of CML cells. In contrast, knockout of apoptotic mediators and TRAIL pathway components conferred resistance to NK cell cytotoxicity in DLBCL cells, indicating heterogeneity in NK cell/cancer cell interactions between cancer types. Our data support a view that distinct mechanisms regulate sensitivity to NK cell cytotoxicity in different cancers. Importantly, our results indicate that loss-of-function mutations in the antigen-presenting machinery and the IFN-JAK-STAT pathway sensitize tumors to NK cell effector function. As alterations in these genes are associated with resistance to T-cell immunotherapies such as PD-1 blockade, NK cell-based therapies could be employed to overcome resistance in these patients. In summary, we suggest that systematic identification of mechanisms governing tumor immune susceptibility has the potential to uncover novel immunotherapy targets. Citation Format: Olli Dufva, Jay Klievink, Khalid Saeed, Matti Kankainen, Mette Ilander, Tiiina Hannunen, Sonja Lagstrom, Pekka Ellonen, Dean A Lee, Satu Mustjoki. Genome-scale CRISPR screens identify essenti","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86597924","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A053
T. Bruno, A. Ruffin, A. Cillo, R. Ferris, D. Vignali
Immunotherapy, specifically anti-PD1, has improved patient survival in a range of tumor types including head and neck squamous cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC). Despite the success of anti-PD1 therapy, only 20% of patients produce a durable response to this treatment. Thus, a need exists to develop additional therapeutic strategies to treat these patients, which includes evaluation of other tumor-infiltrating immune cells that could further augment the CD8+ and CD4+ T-cell response. Tumor-infiltrating B cells (TIL-B) represent a possible target for immunotherapy due to their predominance in the tumor microenvironment (TME) and crucial role in the immune response. However, TIL-B function in cancer and in the context of immunotherapy has been understudied. In fact, conclusions on an anti- or protumor role for TIL-Bs in the TME is dependent on the study. However, in HNSCC and NSCLC patients, current evidence suggests an antitumor role for TIL-Bs. Specifically, detection of TIL-Bs within tertiary lymphoid structures (TLS) correlates with better prognosis. While TIL-Bs have been identified in HNSCC and NSCLC patients, their complete phenotypic signature and function in the TME has been understudied with no focus on their role as antigen presenting cells (APCs) and their influence on CD8+ and CD4+ tumor infiltrating lymphocytes (TILs). We hypothesize that TIL-Bs help generate potent, long-term immune responses against cancer by presenting tumor antigens to CD4 TILs within TLS.Using unmanipulated, primary human B cells from fresh tumor, we quantified and further characterized TIL-Bs in HNSCC and NSCLC utilizing single-cell RNAseq and multiparameter flow cytometry. We observed increased numbers of activated TIL-Bs in these primary tumors compared to other immune subsets, specifically CD27+ TIL-Bs. We further assessed the TIL-Bs by correlating phenotype of the TIL-B with its location in the TME, predominantly separating out differences between TIL-Bs within TLS and outside TLS. In addition, we generated a specific antigen presentation assay in vitro, and we observed three types of CD4+ TIL responses when TIL-Bs presented autologous tumor antigens. There were activated responder CD4+ TILs that proliferated when combined with TIL-Bs alone, which indicates stimulation with endogenous tumor antigens. There were antigen-associated responders that required exogenous autologous tumor lysate to elicit a CD4+ TIL response, and there were patient CD4 TILs that did not respond to antigen presentation by TIL-Bs. Within the activated and antigen-associated responders, the TIL-B phenotype influenced the CD4+ TIL phenotype; if the TIL-Bs were activated (CD27+), the CD4+ TILs were T helper (antitumor) CD4+ T-cells and if the TIL-Bs were non-activated (CD27-), the CD4+ TILs were T regulatory cells (protumor). These data suggest that TIL-Bs influence the phenotype and function of CD4+ TILs in patient tumors. In conclusion, activated TIL-Bs are
免疫疗法,特别是抗pd1,已经改善了包括头颈部鳞状细胞癌(HNSCC)和非小细胞肺癌(NSCLC)在内的一系列肿瘤类型的患者生存率。尽管抗pd1治疗取得了成功,但只有20%的患者对这种治疗产生了持久的反应。因此,需要开发额外的治疗策略来治疗这些患者,其中包括评估其他肿瘤浸润性免疫细胞,这些免疫细胞可以进一步增强CD8+和CD4+ t细胞反应。肿瘤浸润性B细胞(TIL-B)由于其在肿瘤微环境(TME)中的优势和在免疫应答中的关键作用而成为免疫治疗的可能靶点。然而,TIL-B在癌症和免疫治疗中的功能尚未得到充分研究。事实上,关于til - b在TME中的抗或抗肿瘤作用的结论取决于研究结果。然而,在HNSCC和NSCLC患者中,目前的证据表明til - b具有抗肿瘤作用。具体来说,三级淋巴结构(TLS)中til - b的检测与较好的预后相关。虽然已经在HNSCC和NSCLC患者中发现了til - b,但它们在TME中的完整表型特征和功能尚未得到充分研究,没有重点研究它们作为抗原提呈细胞(APCs)的作用及其对CD8+和CD4+肿瘤浸润淋巴细胞(TILs)的影响。我们假设til - b通过将肿瘤抗原呈递给TLS内的CD4 til,帮助产生有效的、长期的抗癌免疫反应。利用新鲜肿瘤中未经处理的人原代B细胞,我们利用单细胞RNAseq和多参数流式细胞术定量并进一步表征了HNSCC和NSCLC中的til -B。我们观察到,与其他免疫亚群相比,这些原发性肿瘤中激活的til - b数量增加,特别是CD27+ til - b。我们通过将TIL-B的表型与其在TME中的位置相关联来进一步评估TIL-B,主要分离出TLS内和TLS外TIL-B之间的差异。此外,我们在体外建立了特异性抗原呈递实验,当TIL- b呈递自体肿瘤抗原时,我们观察到三种类型的CD4+ TIL反应。当单独与til - b联合时,应答性CD4+ til被激活增殖,这表明受到内源性肿瘤抗原的刺激。有抗原相关应答者需要外源性自体肿瘤裂解液来引发CD4+ TIL应答,并且有患者CD4 TIL对TIL- b的抗原呈递没有应答。在活化和抗原相关应答者中,TIL- b表型影响CD4+ TIL表型;如果til - b被激活(CD27+), CD4+ til是T辅助(抗肿瘤)CD4+ T细胞,如果til - b未被激活(CD27-), CD4+ til是T调节细胞(肿瘤)。这些数据表明,til - b影响患者肿瘤中CD4+ til的表型和功能。综上所述,活化的til - b在人类原发肿瘤中增加,它们可以向CD4+ til呈递抗原并影响其整体表型。确定til - b在HNSCC和NSCLC患者中的完全激活特征将决定其在这些癌症中的抗肿瘤功能程度。由于目前对不同肿瘤类型间TIL-B功能的统一研究并不多,因此比较HNSCC和NSCLC中TIL-B的差异具有重要意义。此外,由于HNSCC有两种病因(病毒或致癌物诱导),我们能够更好地研究激活和非激活的til - b在实体肿瘤中的差异功能。最终,本研究的结果将有助于预测如何在未来的TIL-B特异性免疫疗法中靶向TIL-B功能,或与目前针对HNSCC和NSCLC患者的免疫疗法(如阻断抑制受体PD-1)联合使用。引文格式:Tullia C. Bruno, Ayana T. Ruffin, Anthony R. Cillo, Robert L. Ferris, Dario A.A. Vignali。人原发性肿瘤中活化的B细胞呈递抗原,增强三级淋巴结构中CD4 t细胞的抗肿瘤功能[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A053。
{"title":"Abstract A053: Activated B cells in human primary tumors present antigen and increase antitumor function of CD4 T-cells in tertiary lymphoid structures","authors":"T. Bruno, A. Ruffin, A. Cillo, R. Ferris, D. Vignali","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A053","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A053","url":null,"abstract":"Immunotherapy, specifically anti-PD1, has improved patient survival in a range of tumor types including head and neck squamous cell carcinoma (HNSCC) and non-small cell lung cancer (NSCLC). Despite the success of anti-PD1 therapy, only 20% of patients produce a durable response to this treatment. Thus, a need exists to develop additional therapeutic strategies to treat these patients, which includes evaluation of other tumor-infiltrating immune cells that could further augment the CD8+ and CD4+ T-cell response. Tumor-infiltrating B cells (TIL-B) represent a possible target for immunotherapy due to their predominance in the tumor microenvironment (TME) and crucial role in the immune response. However, TIL-B function in cancer and in the context of immunotherapy has been understudied. In fact, conclusions on an anti- or protumor role for TIL-Bs in the TME is dependent on the study. However, in HNSCC and NSCLC patients, current evidence suggests an antitumor role for TIL-Bs. Specifically, detection of TIL-Bs within tertiary lymphoid structures (TLS) correlates with better prognosis. While TIL-Bs have been identified in HNSCC and NSCLC patients, their complete phenotypic signature and function in the TME has been understudied with no focus on their role as antigen presenting cells (APCs) and their influence on CD8+ and CD4+ tumor infiltrating lymphocytes (TILs). We hypothesize that TIL-Bs help generate potent, long-term immune responses against cancer by presenting tumor antigens to CD4 TILs within TLS.Using unmanipulated, primary human B cells from fresh tumor, we quantified and further characterized TIL-Bs in HNSCC and NSCLC utilizing single-cell RNAseq and multiparameter flow cytometry. We observed increased numbers of activated TIL-Bs in these primary tumors compared to other immune subsets, specifically CD27+ TIL-Bs. We further assessed the TIL-Bs by correlating phenotype of the TIL-B with its location in the TME, predominantly separating out differences between TIL-Bs within TLS and outside TLS. In addition, we generated a specific antigen presentation assay in vitro, and we observed three types of CD4+ TIL responses when TIL-Bs presented autologous tumor antigens. There were activated responder CD4+ TILs that proliferated when combined with TIL-Bs alone, which indicates stimulation with endogenous tumor antigens. There were antigen-associated responders that required exogenous autologous tumor lysate to elicit a CD4+ TIL response, and there were patient CD4 TILs that did not respond to antigen presentation by TIL-Bs. Within the activated and antigen-associated responders, the TIL-B phenotype influenced the CD4+ TIL phenotype; if the TIL-Bs were activated (CD27+), the CD4+ TILs were T helper (antitumor) CD4+ T-cells and if the TIL-Bs were non-activated (CD27-), the CD4+ TILs were T regulatory cells (protumor). These data suggest that TIL-Bs influence the phenotype and function of CD4+ TILs in patient tumors. In conclusion, activated TIL-Bs are ","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"86631772","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A089
Mayuresh Mane, Khalid Shalaby, Ivan J. Cohen, Avi S. Albeg, Jenny N. Ijoma, M. Ko, Masatomo Maeda, K. Vemuri, J. Satagopan, A. Moroz, J. Zurita, L. Shenker, E. Ackerstaff, M. Shindo, Ekaterina Moroz, Maxim A. Moroz, I. Serganova, J. Koutcher, V. Ponomarev, R. Blasberg
Objective: Determine whether LDH-A knockdown enhances CAR T-cell tumor-targeting and treatment response.Background: It has been shown that T-cells are restricted from entering many human and murine solid tumors, including both murine and human prostate tumors. CD3(+) T-cells are restricted from Myc-CaP murine prostate tumors and localize around the periphery of the tumor nodules (1). Human prostate specific membrane antigen (hPSMA) targeted CAR T-cells are also restricted from hPSMA(+) Myc-CaP tumors and this is only partially reversed by anti-PD1 treatment (1). This report describes the benefit of LDH-A depletion and a greater response of CAR T-cells targeting Myc-CaP tumors with low expression of LDH-A. Methods: LDH-A depletion in hPSMA(+) Myc-CaP cells bearing a bioluminescence reporter (Renilla Luciferase), was achieved by shRNA knockdown (KD) (1). A scrambled IgG shRNA was used as a control (NC). LDH-A expression was quantified by digital droplet PCR (ddPCR), Western blotting and LDH enzyme activity. The glycolytic activity of KD and NC cells was measured using Seahorse XF96 and XFp analyzers. Intratumoral lactate levels were monitored by magnetic resonance spectroscopy (MRS). The preparation and characterization of “second generation” hPSMA-directed CAR T-cells and the hPSMA(+) Myc-CaP tumor models in SCID mice have been described (1). To monitor CAR T-cell trafficking, T-cells were transduced with a Click Beetle Red luciferase reporter to enable efficient visualization by bioluminescence imaging (BLI) of CAR T-cell trafficking, persistence and viability within the hPSMA(+)Myc-CaP tumor mass. Tumor volume was calculated from caliper measurements. CD31 and PD-L1 expression was quantified with immunofluorescent staining and Metamorph Offline image analysis. Results and Discussion: These studies demonstrated that LDH-A KD was the dominant factor in reducing tumor growth. The difference in tumor doubling time (DT) between KD and NC tumors in the presence of CAR T-cell therapy was significant (p Citation Format: Mayuresh M. Mane, Khalid Shalaby, Ivan Cohen, Avi Albeg, Jenny Ijoma, Myat Ko, Masatomo Maeda, Kiranmayi Vemuri, Jaya Satagopan, Anna Moroz, Juan Zurita, Larissa Shenker, Ellen Ackerstaff, Masahiro Shindo, Ekaterina Moroz, Maxim A. Moroz, Inna Serganova, Jason Koutcher, Vladimir Ponomarev, Ronald G. Blasberg. The effect of lactate dehydrogenase-A (LDH-A) knockdown and human prostate-specific membrane antigen (hPSMA) directed CAR T-cell treatment on hPSMA(+) Myc-CaP tumors [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A089.
目的:探讨ldl - a敲低是否能增强CAR -t细胞的肿瘤靶向性和治疗反应。背景:研究表明,t细胞在许多人类和小鼠实体肿瘤(包括小鼠和人类前列腺肿瘤)中是受限制的。CD3(+) t细胞在Myc-CaP小鼠前列腺肿瘤中受到限制,并局限于肿瘤结节周围(1)。人前列腺特异性膜抗原(hPSMA)靶向CAR - t细胞也在hPSMA(+) Myc-CaP肿瘤中受到限制,这只能通过抗pd1治疗部分逆转(1)。该报告描述了LDH-A消耗的益处以及靶向低表达LDH-A的CAR - t细胞对Myc-CaP肿瘤的更大反应。方法:通过shRNA敲低(KD),在携带生物发光报告基因(Renilla Luciferase)的hPSMA(+) Myc-CaP细胞中实现LDH-A的缺失(1)。用一个重组的IgG shRNA作为对照(NC)。采用数字液滴PCR (ddPCR)、Western blotting和LDH酶活性检测LDH- a表达。采用Seahorse XF96和XFp分析仪测定KD和NC细胞的糖酵解活性。用磁共振波谱(MRS)监测瘤内乳酸水平。已经描述了“第二代”hsma定向CAR - t细胞和hPSMA(+)Myc-CaP肿瘤模型在SCID小鼠中的制备和表征(1)。为了监测CAR - t细胞的运输,t细胞用Click Beetle Red荧光素酶报告基因转导,以便通过生物发光成像(BLI)有效地可视化hPSMA(+)Myc-CaP肿瘤块内CAR - t细胞的运输、持久性和活力。通过卡尺测量计算肿瘤体积。采用免疫荧光染色和Metamorph Offline图像分析定量检测CD31和PD-L1的表达。结果与讨论:这些研究表明ldl - a KD是抑制肿瘤生长的主要因素。在CAR - t细胞治疗下,KD和NC肿瘤的肿瘤翻倍时间(DT)有显著差异(p引文格式:Mayuresh M. Mane, Khalid Shalaby, Ivan Cohen, Avi Albeg, Jenny Ijoma, Myat Ko, Masatomo Maeda, Kiranmayi Vemuri, Jaya Satagopan, Anna Moroz, Juan Zurita, Larissa Shenker, Ellen Ackerstaff, Masahiro Shindo, Ekaterina Moroz, Maxim A. Moroz, Inna Serganova, Jason Koutcher, Vladimir Ponomarev, Ronald G. Blasberg)。乳酸脱氢酶a (LDH-A)敲低和人前列腺特异性膜抗原(hPSMA)靶向CAR - t细胞治疗hPSMA(+) Myc-CaP肿瘤的作用[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A089。
{"title":"Abstract A089: The effect of lactate dehydrogenase-A (LDH-A) knockdown and human prostate-specific membrane antigen (hPSMA) directed CAR T-cell treatment on hPSMA(+) Myc-CaP tumors","authors":"Mayuresh Mane, Khalid Shalaby, Ivan J. Cohen, Avi S. Albeg, Jenny N. Ijoma, M. Ko, Masatomo Maeda, K. Vemuri, J. Satagopan, A. Moroz, J. Zurita, L. Shenker, E. Ackerstaff, M. Shindo, Ekaterina Moroz, Maxim A. Moroz, I. Serganova, J. Koutcher, V. Ponomarev, R. Blasberg","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A089","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A089","url":null,"abstract":"Objective: Determine whether LDH-A knockdown enhances CAR T-cell tumor-targeting and treatment response.Background: It has been shown that T-cells are restricted from entering many human and murine solid tumors, including both murine and human prostate tumors. CD3(+) T-cells are restricted from Myc-CaP murine prostate tumors and localize around the periphery of the tumor nodules (1). Human prostate specific membrane antigen (hPSMA) targeted CAR T-cells are also restricted from hPSMA(+) Myc-CaP tumors and this is only partially reversed by anti-PD1 treatment (1). This report describes the benefit of LDH-A depletion and a greater response of CAR T-cells targeting Myc-CaP tumors with low expression of LDH-A. Methods: LDH-A depletion in hPSMA(+) Myc-CaP cells bearing a bioluminescence reporter (Renilla Luciferase), was achieved by shRNA knockdown (KD) (1). A scrambled IgG shRNA was used as a control (NC). LDH-A expression was quantified by digital droplet PCR (ddPCR), Western blotting and LDH enzyme activity. The glycolytic activity of KD and NC cells was measured using Seahorse XF96 and XFp analyzers. Intratumoral lactate levels were monitored by magnetic resonance spectroscopy (MRS). The preparation and characterization of “second generation” hPSMA-directed CAR T-cells and the hPSMA(+) Myc-CaP tumor models in SCID mice have been described (1). To monitor CAR T-cell trafficking, T-cells were transduced with a Click Beetle Red luciferase reporter to enable efficient visualization by bioluminescence imaging (BLI) of CAR T-cell trafficking, persistence and viability within the hPSMA(+)Myc-CaP tumor mass. Tumor volume was calculated from caliper measurements. CD31 and PD-L1 expression was quantified with immunofluorescent staining and Metamorph Offline image analysis. Results and Discussion: These studies demonstrated that LDH-A KD was the dominant factor in reducing tumor growth. The difference in tumor doubling time (DT) between KD and NC tumors in the presence of CAR T-cell therapy was significant (p Citation Format: Mayuresh M. Mane, Khalid Shalaby, Ivan Cohen, Avi Albeg, Jenny Ijoma, Myat Ko, Masatomo Maeda, Kiranmayi Vemuri, Jaya Satagopan, Anna Moroz, Juan Zurita, Larissa Shenker, Ellen Ackerstaff, Masahiro Shindo, Ekaterina Moroz, Maxim A. Moroz, Inna Serganova, Jason Koutcher, Vladimir Ponomarev, Ronald G. Blasberg. The effect of lactate dehydrogenase-A (LDH-A) knockdown and human prostate-specific membrane antigen (hPSMA) directed CAR T-cell treatment on hPSMA(+) Myc-CaP tumors [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A089.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91130071","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A056
Chang-Suk Chae
Harnessing the intrinsic ability of our immune system to recognize and eliminate malignanT-cells represents the most promising anticancer strategy since the development of chemotherapy. However, hostile microenvironmental conditions within aggressive solid tumors inhibit the optimal activity of protective immune cells. Targeting immunosuppression and re-programming immune cell function in the tumor microenvironment are thus fundamental requirements for developing successful cancer immunotherapies. Our CRI postdoctoral project aims at identifying, understanding and disabling the molecular mechanisms by which endoplasmic reticulum (ER) stress responses inhibit the natural function of dendritic cells (DCs) in the ovarian cancer microenvironment. The central hypothesis of this study is that ovarian tumors trigger ER stress and aberrant activation of the IRE1-XBP1 pathway in infiltrating DCs to cripple key immuno-metabolic processes and impede the development of protective T-cell responses. To determine how IRE1α-XBP1 overactivation defines regulatory DC phenotypes in the ovarian cancer microenvironment, metastatic ovarian tumors were developed in ER stress-Activated Indicator (ERAI) reporter mice. We found that DCs demonstrating IRE1 activation in the tumor microenvironment overexpress tolerogenic and immunosuppressive molecules. Next, to determine whether tumor-derived factors may affect DC metabolism, we optimized an ex vivo culture system that recreates the tumor microenvironment using malignant ascites samples from ovarian cancer patients. We treated human monocyte-derived DCs from healthy donors with patient-derived ovarian cancer malignant ascites supernatants and assessed the bioenergetic profile of DCs. Oxygen consumption rate (OCR), which measures mitochondrial respiration, ATP production and spare respiratory capacity, was increased in DCs exposed to ascites supernatants. We found that these metabolic changes upon ascites treatment relied on IRE1a-XBP1 pathway. Further, since therapeutic DC-based vaccines have shown limited effects in ovarian cancer patients, we tested the novel translational hypothesis that XBP1-deficient bone marrow-derived DCs (BMDCs) would be better equipped to endure and function in the tumor microenvironment when used as therapeutic vaccines. While adoptive transfer of WT BMDCs did not induce any therapeutic effect, treatment with XBP1-deficient BMDCs elicited a marked increase in overall host survival. This result suggest that compared with WT BMDCs, their XBP1-deficient counterparts were resistant to the immunosuppressive effects of the tumor microenvironment. Our results provide a unique mechanistic rationale for targeting the IRE1-XBP1 arm of the ER stress response as a potent approach to reprogram and enhance antitumor immune cell function in cancer. These findings should also pave the way for devising a new generation of cancer immunotherapies that may improve the dismal survival of >21,000 American women affect
{"title":"Abstract A056: Incessant ER stress responses promote dendritic cell dysfunction in ovarian cancer","authors":"Chang-Suk Chae","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A056","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A056","url":null,"abstract":"Harnessing the intrinsic ability of our immune system to recognize and eliminate malignanT-cells represents the most promising anticancer strategy since the development of chemotherapy. However, hostile microenvironmental conditions within aggressive solid tumors inhibit the optimal activity of protective immune cells. Targeting immunosuppression and re-programming immune cell function in the tumor microenvironment are thus fundamental requirements for developing successful cancer immunotherapies. Our CRI postdoctoral project aims at identifying, understanding and disabling the molecular mechanisms by which endoplasmic reticulum (ER) stress responses inhibit the natural function of dendritic cells (DCs) in the ovarian cancer microenvironment. The central hypothesis of this study is that ovarian tumors trigger ER stress and aberrant activation of the IRE1-XBP1 pathway in infiltrating DCs to cripple key immuno-metabolic processes and impede the development of protective T-cell responses. To determine how IRE1α-XBP1 overactivation defines regulatory DC phenotypes in the ovarian cancer microenvironment, metastatic ovarian tumors were developed in ER stress-Activated Indicator (ERAI) reporter mice. We found that DCs demonstrating IRE1 activation in the tumor microenvironment overexpress tolerogenic and immunosuppressive molecules. Next, to determine whether tumor-derived factors may affect DC metabolism, we optimized an ex vivo culture system that recreates the tumor microenvironment using malignant ascites samples from ovarian cancer patients. We treated human monocyte-derived DCs from healthy donors with patient-derived ovarian cancer malignant ascites supernatants and assessed the bioenergetic profile of DCs. Oxygen consumption rate (OCR), which measures mitochondrial respiration, ATP production and spare respiratory capacity, was increased in DCs exposed to ascites supernatants. We found that these metabolic changes upon ascites treatment relied on IRE1a-XBP1 pathway. Further, since therapeutic DC-based vaccines have shown limited effects in ovarian cancer patients, we tested the novel translational hypothesis that XBP1-deficient bone marrow-derived DCs (BMDCs) would be better equipped to endure and function in the tumor microenvironment when used as therapeutic vaccines. While adoptive transfer of WT BMDCs did not induce any therapeutic effect, treatment with XBP1-deficient BMDCs elicited a marked increase in overall host survival. This result suggest that compared with WT BMDCs, their XBP1-deficient counterparts were resistant to the immunosuppressive effects of the tumor microenvironment. Our results provide a unique mechanistic rationale for targeting the IRE1-XBP1 arm of the ER stress response as a potent approach to reprogram and enhance antitumor immune cell function in cancer. These findings should also pave the way for devising a new generation of cancer immunotherapies that may improve the dismal survival of >21,000 American women affect","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88130813","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A112
Xiaoshan Shi, M. Manoharan, Nitin Mandloi, S. Priyadharshini, L. Iyer, R. Gupta, Papia Chakraborty, Amitabha Chaudhuri, Ravi Gupta
Cancer immunotherapy is now an established treatment option for many cancers. Cancer immunotherapy boosts host antitumor immunity to provide long-term benefit; however, only a small fraction of the treated patients show a durable clinical response. The tumor microenvironment ecosystem, with its complex mixture of non-malignant and malignant cells, is a major contributor in regulating the response to checkpoint blockade and development of resistance. Ongoing efforts to characterize the tumor microenvironment to stratify patients for immunotherapy and find biomarkers of response often use methods that are limited by the availability of adequate tumor tissue from needle biopsy material and loss of tissue viability during sample processing that precludes the use of single-cell sequencing platforms. Therefore, genomic methods that use deconvolution to assess the relative proportion of different cell types and their phenotypes in the tumor microenvironment are desirable for clinical use. To this end, we have developed OncoPeptTUME, a novel computational approach that utilizes a proprietary minimal gene expression signature to assign immune scores for eight broad categories of immune cells present in the tumor microenvironment. To validate the approach, we used 9,640 TCGA gene expression datasets from 33 different tumors, defined their immune cell content, organized samples into clusters based on their immune cell content, and identified the molecular differences that predict survival in samples belonging to different clusters. We further performed a deeper analysis of samples enriched in infiltrating CD8+ T-cells to identify T-cell phenotype that correlated with a long-term survival benefit. A small set of genes associated with functional T-cell phenotype was used on a dataset of melanoma samples to show that higher expression of the genes discriminated responders from the nonresponders to nivolumab treatment. In conclusion, our analysis demonstrates that OncoPeptTUME is a powerful immunogenomic tool to predict patient prognosis, stratify patients who will benefit from cancer immunotherapy and identify pathways and novel biomarkers of long-term benefit from the use of cancer immunotherapy drugs. Citation Format: Xiaoshan Shi, Malini Manoharan, Nitin Mandloi, Sushri Priyadharshini, Laxman Iyer, Rohit Gupta, Papia Chakraborty, Amitabha Chaudhuri, Ravi Gupta. OncoPeptTUME: A novel computational approach analyzes the tumor microenvironment to predict response to checkpoint inhibitors [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A112.
{"title":"Abstract A112: OncoPeptTUME: A novel computational approach analyzes the tumor microenvironment to predict response to checkpoint inhibitors","authors":"Xiaoshan Shi, M. Manoharan, Nitin Mandloi, S. Priyadharshini, L. Iyer, R. Gupta, Papia Chakraborty, Amitabha Chaudhuri, Ravi Gupta","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A112","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A112","url":null,"abstract":"Cancer immunotherapy is now an established treatment option for many cancers. Cancer immunotherapy boosts host antitumor immunity to provide long-term benefit; however, only a small fraction of the treated patients show a durable clinical response. The tumor microenvironment ecosystem, with its complex mixture of non-malignant and malignant cells, is a major contributor in regulating the response to checkpoint blockade and development of resistance. Ongoing efforts to characterize the tumor microenvironment to stratify patients for immunotherapy and find biomarkers of response often use methods that are limited by the availability of adequate tumor tissue from needle biopsy material and loss of tissue viability during sample processing that precludes the use of single-cell sequencing platforms. Therefore, genomic methods that use deconvolution to assess the relative proportion of different cell types and their phenotypes in the tumor microenvironment are desirable for clinical use. To this end, we have developed OncoPeptTUME, a novel computational approach that utilizes a proprietary minimal gene expression signature to assign immune scores for eight broad categories of immune cells present in the tumor microenvironment. To validate the approach, we used 9,640 TCGA gene expression datasets from 33 different tumors, defined their immune cell content, organized samples into clusters based on their immune cell content, and identified the molecular differences that predict survival in samples belonging to different clusters. We further performed a deeper analysis of samples enriched in infiltrating CD8+ T-cells to identify T-cell phenotype that correlated with a long-term survival benefit. A small set of genes associated with functional T-cell phenotype was used on a dataset of melanoma samples to show that higher expression of the genes discriminated responders from the nonresponders to nivolumab treatment. In conclusion, our analysis demonstrates that OncoPeptTUME is a powerful immunogenomic tool to predict patient prognosis, stratify patients who will benefit from cancer immunotherapy and identify pathways and novel biomarkers of long-term benefit from the use of cancer immunotherapy drugs. Citation Format: Xiaoshan Shi, Malini Manoharan, Nitin Mandloi, Sushri Priyadharshini, Laxman Iyer, Rohit Gupta, Papia Chakraborty, Amitabha Chaudhuri, Ravi Gupta. OncoPeptTUME: A novel computational approach analyzes the tumor microenvironment to predict response to checkpoint inhibitors [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A112.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87399570","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A114
M. Suarez-Carmona, N. Valous, P. Charoentong, Jakob Nikolas Kather, M. Hampel, B. Lenoir, D. Ferber, S. Schott, S. Kess, I. Zoernig, D. Jaeger, N. Halama
Epithelial ovarian cancer (EOC) is an immunogenic tumor entity, as evidenced by multiple correlative studies indicating the impact of intraepithelial tumor-infiltrating T lymphocyte (TIL) density on patient outcome. Immunotherapy trials have generated modest results so far and chemotherapy remains standard of care. In this study, we focus on EOC metastases invading the omentum. Our aim is to characterize their immune landscape with the goal of identifying specific targetable characteristics of these metastases, which are the most prevalent and relate to high morbidity.We used a cohort of 120 ovarian cancer specimens for histologic analysis, cytokine, or metabolic profiling. Furthermore, we developed an EOC tissue explant culture model and treated whole-tissue explants with drugs before assessing immune cell density, distribution, and activation status. Finally, we cultured macrophages isolated from patient-derived ascites to study their response to various treatments.Samples were classified into omental metastases and primary tumors (based on the presence of fat on histologic sections). We observed that omental metastases are characterized by an inflamed microenvironment orchestrated by macrophages and are infiltrated by high amounts of TILs with low expression of activation markers, and a skewed localization around fat patches. These TILs express high amounts of the tumor-supporting CCL5 chemokine. Macrophages storing fatty acids in the form of big vacuoles were found in these fatty tumors as well. Targeting macrophages using the CCR5 inhibitor maraviroc in whole tissue explants effectively restored T-cell distribution across the tissue and slightly affected macrophage polarization specifically in fat-containing tumors. Inhibiting fatty acid import in macrophages more dramatically affected the cytokine landscape, also specifically in fat-containing tumors, and established a Th1-supporting environment permeable to T-cell expansion and activation. In brief, omental metastases are characterized by: (a) a smoldering inflammatory reaction and high macrophage density, (b) increased T-cell accumulation around fatty areas away from cancer cells, and (c) T-cell exhaustion accompanied by CCL5 expression. Treatment of EOC explants revealed that macrophages infiltrating omental metastases can be repolarized in situ, leading to TIL expansion and activation. Citation Format: Meggy Suarez-Carmona, Nektarios A. Valous, Pornpimol Charoentong, Jakob N. Kather, Mareike Hampel, Benedicte M.A. Lenoir, Dyke Ferber, Sarah Schott, Sabine Kess, Inka Zoernig, Dirk Jaeger, Niels Halama. Omental fat in ovarian cancer induces metabolic and immune alterations [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A114.
上皮性卵巢癌(EOC)是一种免疫原性肿瘤实体,多项相关研究表明上皮内肿瘤浸润T淋巴细胞(TIL)密度对患者预后的影响。到目前为止,免疫治疗试验取得了适度的结果,化疗仍然是标准的治疗方法。在这项研究中,我们主要关注侵袭网膜的EOC转移。我们的目的是表征它们的免疫景观,目的是确定这些转移瘤的特定靶向特征,这些转移瘤是最普遍的,与高发病率有关。我们对120例卵巢癌标本进行组织学分析、细胞因子分析或代谢分析。此外,我们建立了EOC组织外植体培养模型,并在评估免疫细胞密度、分布和激活状态之前对整个组织外植体进行药物处理。最后,我们培养了从患者来源的腹水中分离的巨噬细胞,以研究它们对各种治疗的反应。样本被分为网膜转移瘤和原发性肿瘤(基于组织学切片上脂肪的存在)。我们观察到,大网膜转移的特征是巨噬细胞介导的炎症微环境,并被大量低表达激活标志物的til浸润,以及脂肪斑块周围的倾斜定位。这些til表达大量的肿瘤支持CCL5趋化因子。在这些脂肪性肿瘤中也发现巨噬细胞以大液泡的形式储存脂肪酸。在整个组织外植体中使用CCR5抑制剂maraviroc靶向巨噬细胞,有效地恢复了t细胞在组织中的分布,并轻微影响巨噬细胞极化,特别是在含脂肪的肿瘤中。抑制巨噬细胞中脂肪酸的输入更显著地影响了细胞因子景观,特别是在含脂肪的肿瘤中,并建立了一个支持th1的环境,可渗透到t细胞的扩增和活化中。简而言之,大网膜转移的特点是:(a)阴燃炎症反应和高巨噬细胞密度,(b)远离癌细胞的脂肪区周围t细胞积聚增加,(c) t细胞衰竭伴CCL5表达。EOC外植体的治疗表明,浸润大网膜转移瘤的巨噬细胞可以原位再极化,导致TIL扩张和激活。引文格式:Meggy Suarez-Carmona, Nektarios A. Valous, Pornpimol Charoentong, Jakob N. Kather, Mareike Hampel, Benedicte ma. Lenoir, Dyke Ferber, Sarah Schott, Sabine Kess, Inka zoerning, Dirk Jaeger, Niels Halama。卵巢癌大网膜脂肪诱导代谢和免疫改变[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫,2019;7(2增刊):摘要nr A114。
{"title":"Abstract A114: Omental fat in ovarian cancer induces metabolic and immune alterations","authors":"M. Suarez-Carmona, N. Valous, P. Charoentong, Jakob Nikolas Kather, M. Hampel, B. Lenoir, D. Ferber, S. Schott, S. Kess, I. Zoernig, D. Jaeger, N. Halama","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A114","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A114","url":null,"abstract":"Epithelial ovarian cancer (EOC) is an immunogenic tumor entity, as evidenced by multiple correlative studies indicating the impact of intraepithelial tumor-infiltrating T lymphocyte (TIL) density on patient outcome. Immunotherapy trials have generated modest results so far and chemotherapy remains standard of care. In this study, we focus on EOC metastases invading the omentum. Our aim is to characterize their immune landscape with the goal of identifying specific targetable characteristics of these metastases, which are the most prevalent and relate to high morbidity.We used a cohort of 120 ovarian cancer specimens for histologic analysis, cytokine, or metabolic profiling. Furthermore, we developed an EOC tissue explant culture model and treated whole-tissue explants with drugs before assessing immune cell density, distribution, and activation status. Finally, we cultured macrophages isolated from patient-derived ascites to study their response to various treatments.Samples were classified into omental metastases and primary tumors (based on the presence of fat on histologic sections). We observed that omental metastases are characterized by an inflamed microenvironment orchestrated by macrophages and are infiltrated by high amounts of TILs with low expression of activation markers, and a skewed localization around fat patches. These TILs express high amounts of the tumor-supporting CCL5 chemokine. Macrophages storing fatty acids in the form of big vacuoles were found in these fatty tumors as well. Targeting macrophages using the CCR5 inhibitor maraviroc in whole tissue explants effectively restored T-cell distribution across the tissue and slightly affected macrophage polarization specifically in fat-containing tumors. Inhibiting fatty acid import in macrophages more dramatically affected the cytokine landscape, also specifically in fat-containing tumors, and established a Th1-supporting environment permeable to T-cell expansion and activation. In brief, omental metastases are characterized by: (a) a smoldering inflammatory reaction and high macrophage density, (b) increased T-cell accumulation around fatty areas away from cancer cells, and (c) T-cell exhaustion accompanied by CCL5 expression. Treatment of EOC explants revealed that macrophages infiltrating omental metastases can be repolarized in situ, leading to TIL expansion and activation. Citation Format: Meggy Suarez-Carmona, Nektarios A. Valous, Pornpimol Charoentong, Jakob N. Kather, Mareike Hampel, Benedicte M.A. Lenoir, Dyke Ferber, Sarah Schott, Sabine Kess, Inka Zoernig, Dirk Jaeger, Niels Halama. Omental fat in ovarian cancer induces metabolic and immune alterations [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A114.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83003261","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA07
J. Ishihara, Ako Ishihara, Koichi Saskai, S. S. Lee, Mariko Yasui, Hiroyuki Abe, L. Potin, Peyman Hosseinchi, Kazuto Fukunaga, Michal M. Raczy, L. Gray, John-Michael Williford, M. Fukayama, Tiffany M. Marchell, A. Mansurov, A. T. Alpar, S. Kron, M. Swartz, J. Hubbell
Cancer immunotherapy with immune checkpoint inhibitors (CPI) and interleukin (IL)-2 has demonstrated clinical efficacy but is frequently accompanied with severe adverse events caused by excessive and systemic immune system activation. Here, we addressed this need by targeting both the CPI antibodies anti-cytotoxic T-lymphocyte antigen 4 antibody (CTLA4) + anti-programmed death-ligand 1 antibody (PD-L1) and the cytokine IL-2 to tumors via conjugation (for the antibodies) or recombinant fusion (for the cytokine) to a collagen-binding domain (CBD) derived from the blood protein von Willebrand factor (VWF) A3 domain, harnessing the exposure of tumor stroma collagen to blood components due to the leakiness of the tumor vasculature. We show that intravenously (i.v.) administered CBD protein accumulated mainly in tumors, for example 57% of total injected dose depositing in an orthotopic breast tumor model. The CBD was observed to localize throughout the tumor stroma, not merely in the subendothelial space. CBD conjugation or fusion decreased the systemic toxicity of both CTLA4+PD-L1 combination therapy and IL-2, for example eliminating hepatotoxicity with the CPI molecules and ameliorating capillary leak syndrome and pulmonary edema with IL-2. Both CBD-CPI and CBD-IL-2 significantly suppressed tumor growth compared to their unmodified forms in multiple murine cancer models, and both CBD-CPI and CBD-IL-2 increased tumor-infiltrating CD8+ T cells; increases in the ratio of effector CD8+ T cells to T regulatory cells were observed. In an orthotopic breast tumor model, combination treatment with CPI and IL-2 eradicated tumors in 9/13 animals with the CBD-modified drugs, whereas it did so in only 1/13 animals with the unmodified drugs. Thus, the A3 domain of von Willebrand factor can be used to engineer immunotherapies with high translational promise as systemically-administered tumor targeting drugs with improved safety and efficacy compared to their native forms. The targeting approach exploits vascular permeability in the tumor to render the ubiquitous extracellular matrix protein accessible in the tumor, while sparing most other tissues. Citation Format: Jun Ishihara, Ako Ishihara, Koichi Saskai, Steve Seung-Young Lee, Mariko Yasui, Hiroyuki Abe, Lambert Potin, Peyman Hosseinchi, Kazuto Fukunaga, Michal M. Raczy, Laura T. Gray, John-Michael Williford, Masashi Fukayama, Tiffany M. Marchell, Aslan Mansurov, Aaron T. Alpar, Stephen J. Kron, Melody Swartz, Jeffrey A. Hubbell. Using matrix protein affinity to modulate the tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr IA07.
使用免疫检查点抑制剂(CPI)和白细胞介素(IL)-2进行癌症免疫治疗已经证明了临床疗效,但经常伴随着过度和全身免疫系统激活引起的严重不良事件。在这里,我们通过将CPI抗体(抗细胞毒性t淋巴细胞抗原4抗体(CTLA4) +抗程序性死亡配体1抗体(PD-L1)和细胞因子IL-2结合(针对抗体)或重组融合(针对细胞因子)到来自血液蛋白血管性血液病因子(VWF) A3结构域的胶原结合结构域(CBD)靶向肿瘤来解决这一需求。利用肿瘤间质胶原暴露于由于肿瘤血管渗漏的血液成分。我们发现静脉(i.v.)给药的CBD蛋白主要在肿瘤中积累,例如,57%的总注射剂量沉积在原位乳腺肿瘤模型中。观察到CBD在整个肿瘤基质中都有分布,而不仅仅是在内皮下空间。CBD结合或融合降低了CTLA4+PD-L1联合治疗和IL-2的全身毒性,例如CPI分子消除肝毒性,IL-2改善毛细血管渗漏综合征和肺水肿。在多种小鼠癌症模型中,与未修饰的形式相比,CBD-CPI和CBD-IL-2均显著抑制肿瘤生长,并且CBD-CPI和CBD-IL-2均增加肿瘤浸润性CD8+ T细胞;观察到效应CD8+ T细胞与T调节性细胞的比例增加。在原位乳腺肿瘤模型中,CPI和IL-2联合治疗在9/13只使用cbd修饰药物的动物中根除了肿瘤,而使用未修饰药物的动物中只有1/13的动物根除了肿瘤。因此,血管性血液病因子的A3结构域可用于设计具有高翻译前景的免疫疗法,作为系统给药的肿瘤靶向药物,与天然形式相比,具有更高的安全性和有效性。靶向方法利用肿瘤中的血管通透性,使无处不在的细胞外基质蛋白在肿瘤中可接近,同时保留大多数其他组织。引文形式:石原慎子、石原慎子、笹井浩一、Steve seseyoung Lee、Mariko Yasui、Hiroyuki Abe、Lambert Potin、Peyman Hosseinchi、Kazuto Fukunaga、michael M. Raczy、Laura T. Gray、John-Michael Williford、Masashi fuyamama、Tiffany M. Marchell、Aslan Mansurov、Aaron T. Alpar、Stephen J. Kron、Melody Swartz、Jeffrey A. Hubbell。利用基质蛋白亲和力调节肿瘤微环境[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志,2019;7(2增刊):摘要1 - 7。
{"title":"Abstract IA07: Using matrix protein affinity to modulate the tumor microenvironment","authors":"J. Ishihara, Ako Ishihara, Koichi Saskai, S. S. Lee, Mariko Yasui, Hiroyuki Abe, L. Potin, Peyman Hosseinchi, Kazuto Fukunaga, Michal M. Raczy, L. Gray, John-Michael Williford, M. Fukayama, Tiffany M. Marchell, A. Mansurov, A. T. Alpar, S. Kron, M. Swartz, J. Hubbell","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA07","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA07","url":null,"abstract":"Cancer immunotherapy with immune checkpoint inhibitors (CPI) and interleukin (IL)-2 has demonstrated clinical efficacy but is frequently accompanied with severe adverse events caused by excessive and systemic immune system activation. Here, we addressed this need by targeting both the CPI antibodies anti-cytotoxic T-lymphocyte antigen 4 antibody (CTLA4) + anti-programmed death-ligand 1 antibody (PD-L1) and the cytokine IL-2 to tumors via conjugation (for the antibodies) or recombinant fusion (for the cytokine) to a collagen-binding domain (CBD) derived from the blood protein von Willebrand factor (VWF) A3 domain, harnessing the exposure of tumor stroma collagen to blood components due to the leakiness of the tumor vasculature. We show that intravenously (i.v.) administered CBD protein accumulated mainly in tumors, for example 57% of total injected dose depositing in an orthotopic breast tumor model. The CBD was observed to localize throughout the tumor stroma, not merely in the subendothelial space. CBD conjugation or fusion decreased the systemic toxicity of both CTLA4+PD-L1 combination therapy and IL-2, for example eliminating hepatotoxicity with the CPI molecules and ameliorating capillary leak syndrome and pulmonary edema with IL-2. Both CBD-CPI and CBD-IL-2 significantly suppressed tumor growth compared to their unmodified forms in multiple murine cancer models, and both CBD-CPI and CBD-IL-2 increased tumor-infiltrating CD8+ T cells; increases in the ratio of effector CD8+ T cells to T regulatory cells were observed. In an orthotopic breast tumor model, combination treatment with CPI and IL-2 eradicated tumors in 9/13 animals with the CBD-modified drugs, whereas it did so in only 1/13 animals with the unmodified drugs. Thus, the A3 domain of von Willebrand factor can be used to engineer immunotherapies with high translational promise as systemically-administered tumor targeting drugs with improved safety and efficacy compared to their native forms. The targeting approach exploits vascular permeability in the tumor to render the ubiquitous extracellular matrix protein accessible in the tumor, while sparing most other tissues. Citation Format: Jun Ishihara, Ako Ishihara, Koichi Saskai, Steve Seung-Young Lee, Mariko Yasui, Hiroyuki Abe, Lambert Potin, Peyman Hosseinchi, Kazuto Fukunaga, Michal M. Raczy, Laura T. Gray, John-Michael Williford, Masashi Fukayama, Tiffany M. Marchell, Aslan Mansurov, Aaron T. Alpar, Stephen J. Kron, Melody Swartz, Jeffrey A. Hubbell. Using matrix protein affinity to modulate the tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr IA07.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79547561","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A092
I. Tirado-González, A. Nevmerzhitskaya, Arnaud Descot, D. Soetopo, E. Członka, Maresa Weitmann, C. Wachtel, J. Slotta-Huspenina, Christine Tran-Quang, K. Götze, E. Alberto, C. Rothlin, J. Ghysdael, H. Medyouf
Background: TAM receptor tyrosine kinases—Tyro3, Axl and Mertk—and their ligands, Gas6 and Pros1, have been extensively studied for their cell-intrinsic pro-oncogenic function in cancer cells, including leukemia. However, much less is known about their indirect impact on tumor growth through their function as modulators of the immune system. In particular, no study has yet explored this aspect in the context of hematological malignancies. Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) is the most aggressive human ALL subtype, in particular in adults, where it represents 30% of all ALL cases. The disease responds poorly to standard chemotherapy and has a very high risk of recurrence. Treatment relies on the use of a BCR-ABL1 tyrosine kinase inhibitor (TKI) with or without chemotherapy followed by an allogenic-SCT. Despite this intensive regimen a significant fraction (40%) of adult patients fails to achieve long-term disease-free survival, clearly pointing to an unmet clinical need. Methods: To explore the importance of TAM receptors and their ligands in the immune response against leukemia, we developed a very aggressive syngeneic model of Ph+ B-ALL. In this model, leukemia is driven by the expression of the fusion oncoprotein BCR-ABL1 and the loss of the Arf gene, often altered in human Ph+ ALL. Functional studies were carried out using small-molecule inhibitors and genetic mouse models. The latter allowed us to interrogate the importance of specific TAM receptors in defined immune cellular subsets. Lastly, we used pharmacologic inhibitors to explore the potential therapeutic benefit of targeting TAMR signaling alone or in combination with standard of care treatment (TKI) and checkpoint inhibitors. Results: Our study for the first time shows that Gas6, a high-affinity Axl ligand, promotes the establishment of an immune-suppressive milieu that contributes to the aggressive phenotype associated with Ph+ B-ALL. Importantly, Gas6 is not expressed by leukemic cells but rather produced by bone marrow associated stromal cells. Using genetic approaches, we demonstrate that Gas6 primarily acts through its high-affinity receptor, Axl, specifically on myeloid cells, to inhibit the anti-leukemic immune response. This immune-suppressive effect can be effectively blocked using an orally available selective Axl inhibitor or the genetic deletion of Axl in Csf1r expressing myeloid cells, leading to reduced leukemic burden and significantly prolonged survival of leukemia-challenged mice. In a subset of long-term survivors, progressive increase in PD1 expression limited the antileukemic effects associated with Axl deficiency, an effect that can be efficiently reverted by combination treatment with anti-PD1 checkpoint inhibitor.Mechanistically, the antileukemic effects promoted by Gas6/Axl blockade are mediated by an enhanced inflammatory response, followed by a potent adaptive immune response. This can be further potentiated by combinati
{"title":"Abstract A092: TAM receptors targeting unleashes antileukemic immunity and enables checkpoint blockade leading to eradication of leukemic cells","authors":"I. Tirado-González, A. Nevmerzhitskaya, Arnaud Descot, D. Soetopo, E. Członka, Maresa Weitmann, C. Wachtel, J. Slotta-Huspenina, Christine Tran-Quang, K. Götze, E. Alberto, C. Rothlin, J. Ghysdael, H. Medyouf","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A092","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A092","url":null,"abstract":"Background: TAM receptor tyrosine kinases—Tyro3, Axl and Mertk—and their ligands, Gas6 and Pros1, have been extensively studied for their cell-intrinsic pro-oncogenic function in cancer cells, including leukemia. However, much less is known about their indirect impact on tumor growth through their function as modulators of the immune system. In particular, no study has yet explored this aspect in the context of hematological malignancies. Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL) is the most aggressive human ALL subtype, in particular in adults, where it represents 30% of all ALL cases. The disease responds poorly to standard chemotherapy and has a very high risk of recurrence. Treatment relies on the use of a BCR-ABL1 tyrosine kinase inhibitor (TKI) with or without chemotherapy followed by an allogenic-SCT. Despite this intensive regimen a significant fraction (40%) of adult patients fails to achieve long-term disease-free survival, clearly pointing to an unmet clinical need. Methods: To explore the importance of TAM receptors and their ligands in the immune response against leukemia, we developed a very aggressive syngeneic model of Ph+ B-ALL. In this model, leukemia is driven by the expression of the fusion oncoprotein BCR-ABL1 and the loss of the Arf gene, often altered in human Ph+ ALL. Functional studies were carried out using small-molecule inhibitors and genetic mouse models. The latter allowed us to interrogate the importance of specific TAM receptors in defined immune cellular subsets. Lastly, we used pharmacologic inhibitors to explore the potential therapeutic benefit of targeting TAMR signaling alone or in combination with standard of care treatment (TKI) and checkpoint inhibitors. Results: Our study for the first time shows that Gas6, a high-affinity Axl ligand, promotes the establishment of an immune-suppressive milieu that contributes to the aggressive phenotype associated with Ph+ B-ALL. Importantly, Gas6 is not expressed by leukemic cells but rather produced by bone marrow associated stromal cells. Using genetic approaches, we demonstrate that Gas6 primarily acts through its high-affinity receptor, Axl, specifically on myeloid cells, to inhibit the anti-leukemic immune response. This immune-suppressive effect can be effectively blocked using an orally available selective Axl inhibitor or the genetic deletion of Axl in Csf1r expressing myeloid cells, leading to reduced leukemic burden and significantly prolonged survival of leukemia-challenged mice. In a subset of long-term survivors, progressive increase in PD1 expression limited the antileukemic effects associated with Axl deficiency, an effect that can be efficiently reverted by combination treatment with anti-PD1 checkpoint inhibitor.Mechanistically, the antileukemic effects promoted by Gas6/Axl blockade are mediated by an enhanced inflammatory response, followed by a potent adaptive immune response. This can be further potentiated by combinati","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73622617","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}