301 Preclinical development of AB-2100, a PSMA neovasculature-inducible CA9 CAR resistant to FASL and TGFb mediated suppression for the treatment of ccRCC

Ivan Chan,Laura Lim,Irene Scarfò, Darrian Moskowitz,Beatriz Millare,Kevin Dang,Jeremy Chen,Nickolas Attanasio,Angela Boroughs,Michelle Nguyen,Suchismita Mohanty, Jenessa Smith,James Zhang,Jennifer McDevitt,Thomas J Gardner, Alma Gomez, Vince Thomas, Marvin Chew, Rakesh Sudhakah, Amanda Fearon, Vibhavari Sail,Stanley Zhou

Journal for ImmunoTherapy of Cancer(2023)

引用 0|浏览3
暂无评分
摘要

Background

Clinically effective CAR T cell therapy for solid tumors may require substantial T cell engineering to increase specificity and potency. We have developed AB-2100, an autologous, integrated circuit T (ICT) cell that encodes multiple synthetic ‘modules’ to overcome challenges in the treatment of clear cell renal cell carcinoma (ccRCC). AB-2100 includes a sequential ‘AND’ logic gate designed to limit off-tumor toxicity through dual tumor antigen recognition, a short hairpin ribonucleic acid-microRNA (shRNA-miR) module for the constitutive expression of shRNA-miRs for knockdown of FAS and TGFBR2, and a constitutive synthetic pathway activator (SPA) that drives constitutive STAT3 signaling for enhanced T cell cytotoxicity and expansion.

Methods

On-target, off-tumor toxicity was previously observed with constitutive CA9 CAR T cell therapy. To overcome this, the AB-2100 sequential ‘AND’ logic gate consists of a priming receptor (PrimeR) specific for PSMA and an inducible CA9-targeted CAR that is expressed upon PrimeR engagement with PSMA on the tumor neovasculature of ccRCC. This unique feature of the logic gate increases the safety profile of AB-2100 given that PSMA and CA9 are not expressed in the same normal tissues. Dual-antigen specificity of the logic gate was assessed in vitro and in vivo via CA9+ and PSMA+CA9+-786-O tumors established on contralateral flanks. To model vascular priming, AB-2100 cells were co-cultured with PSMA-expressing endothelial HUVEC cells and K562-CA9 cells. An in vitro FAS cross-linking assay was conducted to assess the impact of FAS knockdown on FAS-mediated apoptosis. The enhanced anti-tumor activity conferred by TGFBR2 shRNA and SPA modules were assessed in a subcutaneous 786-O xenograft model. Lastly, AB-2100 potency was measured in a subcutaneous renal A498 xenograft tumor model.

Results

In vitro cytotoxicity against single or dual antigen expressing tumor cell lines, as well as a dual flank xenograft model demonstrate that AB-2100 selectively kills tumors that express both CA9 and PSMA, and not tumors that express CA9 alone. Furthermore, we confirmed that AB-2100 was able to prime off of PSMA-expressing endothelial HUVEC cells and kill K562-CA9 tumor cells. Finally, AB-2100 containing shRNA-miR and SPA modules demonstrated enhanced anti-tumor activity in xenograft RCC models (786-O and A498).

Conclusions

Preclinical data demonstrate that AB-2100 can selectively target antigens that cannot be safely targeted by conventional CARs, and overcome multiple suppressive mechanisms in the tumor microenvironment. These results support the evaluation of AB-2100 in the clinic for the treatment of advanced or metastatic ccRCC.
更多
查看译文
关键词
ccrcc,neovasculature-inducible
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要