355 Directing a high avidity KRAS G12D-specific TCR engineered with a CD8αβ co-receptor and chimeric cytokine receptor using non-viral knock-in enhances anti-tumor responses

Allison P Drain, Nicholas Rouillard, Nathaniel Swanson, Santosh Narayan, Tyler Warner, Nicole Danek, Ken Gareau, Cheryl Black, James Parsons, Anthony Thomas, Jinsheng Liang, Luhua Shen, Tanya Tetrault, Vince Nguyen, Iqraa Priyata, Sarah Vidyasagar, Joshua Francis, Xingyue He, Patrick J Browne, Rebecca C Lamothe, Meghan D Storlie, Gregory J Cost, Thomas M Schmitt, Philip D Greenberg, Smita S Chandran, Christopher A Klebanoff, Ankit Gupta, Damien Hallet, Gary Shapiro, Kim Nguyen, Loic Vincent
{"title":"355 Directing a high avidity KRAS G12D-specific TCR engineered with a CD8αβ co-receptor and chimeric cytokine receptor using non-viral knock-in enhances anti-tumor responses","authors":"Allison P Drain, Nicholas Rouillard, Nathaniel Swanson, Santosh Narayan, Tyler Warner, Nicole Danek, Ken Gareau, Cheryl Black, James Parsons, Anthony Thomas, Jinsheng Liang, Luhua Shen, Tanya Tetrault, Vince Nguyen, Iqraa Priyata, Sarah Vidyasagar, Joshua Francis, Xingyue He, Patrick J Browne, Rebecca C Lamothe, Meghan D Storlie, Gregory J Cost, Thomas M Schmitt, Philip D Greenberg, Smita S Chandran, Christopher A Klebanoff, Ankit Gupta, Damien Hallet, Gary Shapiro, Kim Nguyen, Loic Vincent","doi":"10.1136/jitc-2023-sitc2023.0355","DOIUrl":null,"url":null,"abstract":"<h3>Background</h3> Adoptive T cell therapy has demonstrated clinical activity in a subset of patients with solid tumors; however, consistent responses will require further optimization. T cell receptor (TCR)-engineered T cells recognize peptides derived from intracellular and surface proteins presented in the context of MHC class I. Immunologic targeting of recurrently mutated oncogenic drivers, such as KRAS, overcomes many of the major obstacles of this modality because the resulting epitope is: 1) tumor-specific, 2) essential for cancer cell fitness, and 3) derived from a stably expressed non-self protein. AFNT-212 is a next-generation engineered T cell therapy that uses non-viral targeted knock-in (KI) at the TCRα constant (<i>TRAC</i>) locus to express a multi-cistronic cassette that includes 1) a high-affinity TCR specific for KRAS<sub>G12D</sub> mutation, 2) a CD8αβ coreceptor, and 3) a chimeric cytokine receptor. <h3>Methods</h3> Human CD4<sup>+</sup> and CD8<sup>+</sup> T cells were genetically engineered by a novel CRISPR-Cas nuclease and gRNAs targeting <i>TRAC</i> and the TCRβ constant (<i>TRBC</i>) genes allowing for knock-out of the endogenous TCR loci and simultaneous integration of the non-viral plasmid-based transgene cassette. Engineered T cells were assessed for specificity and potency, including activation, proliferation, and cytotoxicity, against KRAS G12D peptide presented by HLA-A*11:01 and a panel of KRAS G12D-expressing tumor cell lines. <i>In vitro</i> safety studies were performed along with <i>in vivo</i> efficacy studies in multiple human xenograft models. <h3>Results</h3> Engineered primary T cells showed specific recognition of KRAS G12D peptide, demonstrated cytotoxicity against endogenously expressing HLA-A*11:01<sup>+</sup>/KRAS G12D<sup>+</sup> cell lines in tumor cell re-challenge assays <i>in vitro</i>, and mediated robust anti-tumor activity <i>in vivo</i>. Inclusion of the chimeric cytokine receptor allowed for a more potent anti-tumor response stemming from improved T cell expansion and resistance to exhaustion. No off-target liabilities were identified upon co-incubation of AFNT-212 with all possible peptides in the human proteome matching the xScan-defined epitope recognition motif for the TCR, demonstrating specificity. Gene editing safety evaluation did not reveal any off-target activity for the CRISPR-Cas nucleases and engineered T cells did not show cytokine-independent proliferation, collectively supporting a favorable pre-clinical safety profile for AFNT-212. <h3>Conclusions</h3> We report a novel TCR gene therapy approach targeting mutant KRAS G12D-expressing tumors with a coordinated CD4/CD8 T cell response that has a promising efficacy and safety profile. Our work supports the planned clinical development of AFNT-212 as a novel non-viral KI TCR-engineered T cell therapy for KRAS-mutant solid tumors.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Regular and Young Investigator Award Abstracts","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1136/jitc-2023-sitc2023.0355","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

Adoptive T cell therapy has demonstrated clinical activity in a subset of patients with solid tumors; however, consistent responses will require further optimization. T cell receptor (TCR)-engineered T cells recognize peptides derived from intracellular and surface proteins presented in the context of MHC class I. Immunologic targeting of recurrently mutated oncogenic drivers, such as KRAS, overcomes many of the major obstacles of this modality because the resulting epitope is: 1) tumor-specific, 2) essential for cancer cell fitness, and 3) derived from a stably expressed non-self protein. AFNT-212 is a next-generation engineered T cell therapy that uses non-viral targeted knock-in (KI) at the TCRα constant (TRAC) locus to express a multi-cistronic cassette that includes 1) a high-affinity TCR specific for KRASG12D mutation, 2) a CD8αβ coreceptor, and 3) a chimeric cytokine receptor.

Methods

Human CD4+ and CD8+ T cells were genetically engineered by a novel CRISPR-Cas nuclease and gRNAs targeting TRAC and the TCRβ constant (TRBC) genes allowing for knock-out of the endogenous TCR loci and simultaneous integration of the non-viral plasmid-based transgene cassette. Engineered T cells were assessed for specificity and potency, including activation, proliferation, and cytotoxicity, against KRAS G12D peptide presented by HLA-A*11:01 and a panel of KRAS G12D-expressing tumor cell lines. In vitro safety studies were performed along with in vivo efficacy studies in multiple human xenograft models.

Results

Engineered primary T cells showed specific recognition of KRAS G12D peptide, demonstrated cytotoxicity against endogenously expressing HLA-A*11:01+/KRAS G12D+ cell lines in tumor cell re-challenge assays in vitro, and mediated robust anti-tumor activity in vivo. Inclusion of the chimeric cytokine receptor allowed for a more potent anti-tumor response stemming from improved T cell expansion and resistance to exhaustion. No off-target liabilities were identified upon co-incubation of AFNT-212 with all possible peptides in the human proteome matching the xScan-defined epitope recognition motif for the TCR, demonstrating specificity. Gene editing safety evaluation did not reveal any off-target activity for the CRISPR-Cas nucleases and engineered T cells did not show cytokine-independent proliferation, collectively supporting a favorable pre-clinical safety profile for AFNT-212.

Conclusions

We report a novel TCR gene therapy approach targeting mutant KRAS G12D-expressing tumors with a coordinated CD4/CD8 T cell response that has a promising efficacy and safety profile. Our work supports the planned clinical development of AFNT-212 as a novel non-viral KI TCR-engineered T cell therapy for KRAS-mutant solid tumors.
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
355利用非病毒敲入介导CD8αβ共受体和嵌合细胞因子受体工程的高亲和度KRAS g12d特异性TCR增强抗肿瘤反应
过继性T细胞疗法已经证明对一部分实体瘤患者具有临床活性;然而,一致的响应需要进一步优化。T细胞受体(TCR)工程T细胞识别来自MHC i类背景下的细胞内和表面蛋白的肽。针对复发突变的致癌驱动因子(如KRAS)的免疫靶向克服了这种模式的许多主要障碍,因为所产生的表位是:1)肿瘤特异性,2)癌细胞适应性必需的,3)来源于稳定表达的非自身蛋白。AFNT-212是一种新一代工程化T细胞疗法,它使用TCRα常数(TRAC)位点的非病毒靶向敲入(KI)来表达一个多顺反子盒,其中包括1)KRASG12D突变的高亲和力TCR, 2) CD8αβ辅助受体,以及3)嵌合细胞因子受体。方法利用一种新的CRISPR-Cas核酸酶和靶向TRAC和TCRβ常数(TRBC)基因的gRNAs对人CD4+和CD8+ T细胞进行基因工程改造,从而敲除内源性TCR位点并同时整合基于非病毒质粒的转基因盒。通过HLA-A*11:01和一组表达KRAS G12D的肿瘤细胞系,评估工程T细胞对KRAS G12D肽的特异性和效力,包括活化、增殖和细胞毒性。在多种人类异种移植模型中进行了体外安全性研究和体内有效性研究。结果经工程改造的原代T细胞对KRAS G12D肽具有特异性识别能力,在体外肿瘤细胞再攻毒实验中对内源性表达HLA-A*11:01+/KRAS G12D+的细胞系显示出细胞毒性,并介导了较强的体内抗肿瘤活性。嵌合细胞因子受体的包含允许更有效的抗肿瘤反应,源于改善T细胞扩增和抵抗衰竭。在将AFNT-212与与xscan定义的TCR表位识别基序匹配的所有可能的人类蛋白质组肽共孵育时,没有发现脱靶性,证明了特异性。基因编辑安全性评估没有显示CRISPR-Cas核酸酶的任何脱靶活性,工程T细胞没有显示细胞因子不依赖性增殖,共同支持AFNT-212良好的临床前安全性。我们报道了一种新的TCR基因治疗方法,靶向表达KRAS g12d的突变肿瘤,具有协调的CD4/CD8 T细胞反应,具有良好的疗效和安全性。我们的工作支持AFNT-212作为一种新的非病毒性KI tcr工程T细胞治疗kras突变实体瘤的计划临床开发。
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
1455 Characterization of gene expression signatures of tumor immunogenicity and cellular proliferation from murine cancer models grownin vitroandin vivo 1193 Synergistic targeting of multiple activating pathways with natural killer cell engagers 1413 Immunogenicity of SARS-CoV-2 mRNA vaccines in individuals with thymic epithelial tumors 1150 DM005, an EGFR × MET bispecific antibody-drug conjugate, showed robust anti-tumor activity in PDX models 1274 Infliximab for decompensated diabetes following immune checkpoint inhibitor therapy
×
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