Abstract A37: Immuno-oncological efficacy of RXDX-106, a novel TAM (TYRO3, AXL, MER) family small-molecule kinase inhibitor

Y. Yokoyama, E. Lew, C. Walsh, Jack Y. Lee, Joanne Oh, E. Tindall, R. Nevarez, Amy Diliberto, Heather A. Ely, Ruth Seelige, A. Albert, J. Bui, Gary Li
{"title":"Abstract A37: Immuno-oncological efficacy of RXDX-106, a novel TAM (TYRO3, AXL, MER) family small-molecule kinase inhibitor","authors":"Y. Yokoyama, E. Lew, C. Walsh, Jack Y. Lee, Joanne Oh, E. Tindall, R. Nevarez, Amy Diliberto, Heather A. Ely, Ruth Seelige, A. Albert, J. Bui, Gary Li","doi":"10.1158/2326-6074.TUMIMM17-A37","DOIUrl":null,"url":null,"abstract":"Background: The TAM family of receptor tyrosine kinases (RTKs), TYRO3, AXL, and MER, have emerged as attractive targets for cancer therapy. In cancer cells, overexpression of TAM RTKs is associated with resistance and a mesenchymal phenotype. In immune cells, TAM RTKs play a key homeostatic role as negative regulators of immune responses, contributing to the evasion of cancer cells from immune surveillance. RXDX-106 is a potent and selective TAM family inhibitor in preclinical development. Previously, in several syngeneic tumor models, we have demonstrated that RXDX-106 alone inhibited tumor growth, whereas such benefit was reduced when the tumors grew in immunocompromised athymic nude mice. In addition, RXDX-106 induced favorable polarization of the tumor infiltrating immune cells towards an anti-cancer phenotype. Here we sought (1) to evaluate the efficacy of RXDX-106 as a single agent and in combination with immune checkpoint inhibitors; (2) to identify the immuno-modulatory mechanisms of action; (3) to explore the reciprocal regulation of TAM expression on cancer and immune cells in the tumor microenvironment; and (4) to decipher how pharmacological inhibition of TAM signaling pathways on both tumor and immune cells would be beneficial, given their complex regulation and intimate relationship in the tumor microenvironment. Methods and Results: In the present study, we demonstrated that tumor growth inhibition in an MC38 model was associated with a significant increase in tumor associated M1 macrophages, an increase in CD169hi antigen presenting macrophages, and upregulation of PD-L1. In addition, we observed a higher ratio of CD8+/CD4+ T cells and increased expression of CD69 and PD-1 on CD8+ T cells, all indicative of activation of cytotoxic T cells. Finally, we observed an increase in Granzyme B and IFNγ with a concomitant decrease in VEGF in tumor cell lysates, evidence supporting T cell activation and M1 polarization of macrophages. Furthermore, in a CT26 syngeneic model, we demonstrated that RXDX-106 inhibited tumor growth as a single agent, and the effect was further potentiated by combination therapy with immune checkpoint inhibitors, as evidenced by upregulation of anti-tumor gene expression patterns, upregulation of anti-tumor cytokines in the tumor cell lysates, and an increase in T cell function. Finally, in an AXL-driven tumor model, we demonstrated that AXL expressing tumors induced a pro-tumorigenic immune environment, and treatment with RXDX-106 resulted in complete tumor regression and re-polarization of macrophages towards an M1, anti-tumor phenotype. Conclusion: RXDX-106 has the potential to restore and enhance immune function in macrophages and T cells, resulting in repolarization of the immune response towards an anti-tumor microenvironment. The unique mechanism of activating both innate and adaptive immunity, plus regulating cross-talk between immune cells and tumor cells by RXDX-106 supports clinical development of RXDX-106 to potentially treat a wide variety of cancers. Citation Format: Yumi Yokoyama, Erin D. Lew, Colin Walsh, Jack Lee, Joanne Oh, Elizabeth A. Tindall, Robin Nevarez, Amy Diliberto, Heather Ely, Ruth Seelige, Amanda Albert, Jack Bui, Gary Li. Immuno-oncological efficacy of RXDX-106, a novel TAM (TYRO3, AXL, MER) family small-molecule kinase inhibitor [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 A37.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Checkpoints and Immunomodulation","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1158/2326-6074.TUMIMM17-A37","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background: The TAM family of receptor tyrosine kinases (RTKs), TYRO3, AXL, and MER, have emerged as attractive targets for cancer therapy. In cancer cells, overexpression of TAM RTKs is associated with resistance and a mesenchymal phenotype. In immune cells, TAM RTKs play a key homeostatic role as negative regulators of immune responses, contributing to the evasion of cancer cells from immune surveillance. RXDX-106 is a potent and selective TAM family inhibitor in preclinical development. Previously, in several syngeneic tumor models, we have demonstrated that RXDX-106 alone inhibited tumor growth, whereas such benefit was reduced when the tumors grew in immunocompromised athymic nude mice. In addition, RXDX-106 induced favorable polarization of the tumor infiltrating immune cells towards an anti-cancer phenotype. Here we sought (1) to evaluate the efficacy of RXDX-106 as a single agent and in combination with immune checkpoint inhibitors; (2) to identify the immuno-modulatory mechanisms of action; (3) to explore the reciprocal regulation of TAM expression on cancer and immune cells in the tumor microenvironment; and (4) to decipher how pharmacological inhibition of TAM signaling pathways on both tumor and immune cells would be beneficial, given their complex regulation and intimate relationship in the tumor microenvironment. Methods and Results: In the present study, we demonstrated that tumor growth inhibition in an MC38 model was associated with a significant increase in tumor associated M1 macrophages, an increase in CD169hi antigen presenting macrophages, and upregulation of PD-L1. In addition, we observed a higher ratio of CD8+/CD4+ T cells and increased expression of CD69 and PD-1 on CD8+ T cells, all indicative of activation of cytotoxic T cells. Finally, we observed an increase in Granzyme B and IFNγ with a concomitant decrease in VEGF in tumor cell lysates, evidence supporting T cell activation and M1 polarization of macrophages. Furthermore, in a CT26 syngeneic model, we demonstrated that RXDX-106 inhibited tumor growth as a single agent, and the effect was further potentiated by combination therapy with immune checkpoint inhibitors, as evidenced by upregulation of anti-tumor gene expression patterns, upregulation of anti-tumor cytokines in the tumor cell lysates, and an increase in T cell function. Finally, in an AXL-driven tumor model, we demonstrated that AXL expressing tumors induced a pro-tumorigenic immune environment, and treatment with RXDX-106 resulted in complete tumor regression and re-polarization of macrophages towards an M1, anti-tumor phenotype. Conclusion: RXDX-106 has the potential to restore and enhance immune function in macrophages and T cells, resulting in repolarization of the immune response towards an anti-tumor microenvironment. The unique mechanism of activating both innate and adaptive immunity, plus regulating cross-talk between immune cells and tumor cells by RXDX-106 supports clinical development of RXDX-106 to potentially treat a wide variety of cancers. Citation Format: Yumi Yokoyama, Erin D. Lew, Colin Walsh, Jack Lee, Joanne Oh, Elizabeth A. Tindall, Robin Nevarez, Amy Diliberto, Heather Ely, Ruth Seelige, Amanda Albert, Jack Bui, Gary Li. Immuno-oncological efficacy of RXDX-106, a novel TAM (TYRO3, AXL, MER) family small-molecule kinase inhibitor [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 A37.
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
摘要:新型TAM (TYRO3, AXL, MER)家族小分子激酶抑制剂RXDX-106的免疫肿瘤学疗效
背景:TAM家族的受体酪氨酸激酶(RTKs), TYRO3, AXL和MER,已经成为癌症治疗的有吸引力的靶点。在癌细胞中,TAM rtk的过表达与耐药性和间充质表型相关。在免疫细胞中,TAM rtk作为免疫反应的负调节因子起着关键的稳态作用,有助于癌细胞逃避免疫监视。RXDX-106是一种有效的、选择性的TAM家族抑制剂,处于临床前开发阶段。先前,在几个同基因肿瘤模型中,我们已经证明RXDX-106单独抑制肿瘤生长,而当免疫功能低下的胸腺裸鼠肿瘤生长时,这种益处降低。此外,RXDX-106诱导肿瘤浸润免疫细胞向抗癌表型的有利极化。在这里,我们寻求(1)评估RXDX-106作为单一药物和与免疫检查点抑制剂联合使用的疗效;(2)确定免疫调节作用机制;(3)探讨肿瘤微环境中TAM表达对肿瘤和免疫细胞的相互调节作用;(4)考虑到TAM信号通路在肿瘤微环境中的复杂调控和密切关系,解释TAM信号通路对肿瘤和免疫细胞的药理抑制如何有益。方法和结果:在本研究中,我们证明了MC38模型中肿瘤生长抑制与肿瘤相关M1巨噬细胞的显著增加、CD169hi抗原呈递巨噬细胞的增加以及PD-L1的上调有关。此外,我们观察到CD8+/CD4+ T细胞比例增加,CD69和PD-1在CD8+ T细胞上的表达增加,这些都表明细胞毒性T细胞的活化。最后,我们观察到颗粒酶B和IFNγ的增加,同时肿瘤细胞裂解物中VEGF的减少,证据支持T细胞活化和巨噬细胞的M1极化。此外,在CT26同基因模型中,我们证明了RXDX-106作为单一药物抑制肿瘤生长,并且通过与免疫检查点抑制剂联合治疗进一步增强了这种作用,证明了抗肿瘤基因表达模式上调,肿瘤细胞裂解物中抗肿瘤细胞因子上调,T细胞功能增加。最后,在AXL驱动的肿瘤模型中,我们证明了表达AXL的肿瘤诱导了促瘤性免疫环境,RXDX-106治疗导致肿瘤完全消退,巨噬细胞向M1抗肿瘤表型重新极化。结论:RXDX-106具有恢复和增强巨噬细胞和T细胞免疫功能的潜力,导致免疫应答对抗肿瘤微环境的复极化。RXDX-106激活先天免疫和适应性免疫的独特机制,以及调节免疫细胞和肿瘤细胞之间的串导,支持RXDX-106的临床开发,可能治疗多种癌症。引文格式:横山由美、艾琳·d·卢、科林·沃尔什、杰克·李、乔安妮·欧、伊丽莎白·a·廷德尔、罗宾·内瓦雷斯、艾米·迪利贝托、希瑟·伊莉、露丝·西利格、阿曼达·阿尔伯特、杰克·布伊、加里·李。新型TAM (TYRO3, AXL, MER)家族小分子激酶抑制剂RXDX-106的免疫肿瘤疗效研究[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 A37。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Abstract A21: Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27 dependent pathway Abstract A11: Ipilimumab protects T cells from the antiproliferative effects of dexamethasone Abstract A29: Multimeric anti-human OX40 induces robust immune responses Abstract A33: Tumor derived T cell clones for evaluation of check point inhibitor therapeutics Abstract A40: Prevalence of TIGIT expression in normal tissues, inflammation, and cancer
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
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
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
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