Cao Zhiyuan, Pu Chengfei, Jiang Xianyang, Han Guiting, Shen Xiaogang, Wang Wensheng, Ding Wei, Huang Zhipeng, Huang Xi, Jia Beibei, Lu Victor X, Tian Le, Wu Zhao, Xiao Lei
Innovative Cellular Therapeutics, Shanghai, China.
Innovative Cellular Therapeutics Holdings Limited, Rockville, Maryland, USA.
J Immunother Cancer. 2025 May 31;13(5):e011238. doi: 10.1136/jitc-2024-011238.
BACKGROUND: The challenges that remain in the treatment of solid tumors with chimeric antigen receptor (CAR)-T cells include limited solid tumor-specific targets and poor CAR-T cell expansion and function due to limited availability of solid tumor antigens outside the tumor microenvironment. Prostate cancer is the second most common cancer among men worldwide. Current CAR-T therapies for prostate cancer lack specific targets, posing safety risks. To overcome these problems, we identified prostatic acid phosphatase (PAP, also known as ACPP or ACP3) as a feasible CAR-T target for prostate cancer and developed CoupledCAR, a novel approach for expanding tumor-targeting CAR-T cells without tumor antigens. METHODS: We analyzed the expression of PAP from The Cancer Genome Atlas database and validated its expression in normal and cancer tissues through immunohistochemistry staining. To generate anti-PAP specific antibodies, we screened the human single-chain antibody library using transmembrane PAP-His antigen and selected antibodies based on their binding ability and specificity. We constructed PAP-targeted CAR and evaluated their antitumor efficacy both in vitro and in vivo. We validated the function of PAP CoupledCAR in both in vitro and in vivo experiments, and further analyzed its mechanism using single-cell RNA sequencing (scRNA-Seq). RESULTS: PAP was specifically expressed in prostate epithelial and prostate cancer cells, with no expression in other tissues. Seven single-chain variable fragments were screened from the human single-chain antibody library, with S5D1 showing the highest binding ability for the PAP. PAP CAR-T cells demonstrated strong antitumor efficacy both in vitro and in vivo. Furthermore, the CoupledCAR system significantly expanded PAP CAR-T cells, promoting memory-like status, reducing exhaustion, and enhancing their antitumor efficacy. The scRNA-Seq demonstrated that the expansion of PAP CAR-T cells in the CoupledCAR system is mediated by costimulatory signals and cytokine signals, rather than T-cell receptor signals. CONCLUSIONS: Our study is the first to demonstrate that PAP is a specific target for CAR-T therapy in prostate cancer, both in vitro and in vivo. We developed the CoupledCAR platform technology for solid tumor CAR-T cell therapy, enabling the expansion of tumor-targeting CAR-T cells without requiring tumor antigens and thereby enhancing their functionality against solid tumors.
背景:嵌合抗原受体(CAR)-T细胞治疗实体瘤仍面临诸多挑战,包括实体瘤特异性靶点有限,以及由于肿瘤微环境外实体瘤抗原可用性有限导致CAR-T细胞扩增和功能不佳。前列腺癌是全球男性中第二常见的癌症。目前用于前列腺癌的CAR-T疗法缺乏特异性靶点,存在安全风险。为克服这些问题,我们确定前列腺酸性磷酸酶(PAP,也称为ACPP或ACP3)作为前列腺癌可行的CAR-T靶点,并开发了CoupledCAR,这是一种无需肿瘤抗原即可扩增肿瘤靶向CAR-T细胞的新方法。 方法:我们分析了癌症基因组图谱数据库中PAP的表达,并通过免疫组织化学染色验证其在正常组织和癌组织中的表达。为产生抗PAP特异性抗体,我们使用跨膜PAP-His抗原筛选人单链抗体文库,并根据其结合能力和特异性选择抗体。我们构建了靶向PAP的CAR,并在体外和体内评估其抗肿瘤疗效。我们在体外和体内实验中验证了PAP CoupledCAR的功能,并使用单细胞RNA测序(scRNA-Seq)进一步分析其机制。 结果:PAP在前列腺上皮细胞和前列腺癌细胞中特异性表达,在其他组织中无表达。从人单链抗体文库中筛选出7个单链可变片段,其中S5D1对PAP的结合能力最强。PAP CAR-T细胞在体外和体内均表现出强大的抗肿瘤疗效。此外,CoupledCAR系统显著扩增了PAP CAR-T细胞,促进了记忆样状态,减少了耗竭,并增强了其抗肿瘤疗效。scRNA-Seq表明,CoupledCAR系统中PAP CAR-T细胞扩增是由共刺激信号和细胞因子信号介导的,而非T细胞受体信号。 结论:我们的研究首次证明,PAP在体外和体内均是前列腺癌CAR-T治疗的特异性靶点。我们开发了用于实体瘤CAR-T细胞治疗的CoupledCAR平台技术,无需肿瘤抗原即可扩增肿瘤靶向CAR-T细胞,从而增强其对实体瘤的功能。
J Immunother Cancer. 2025-5-31
Cancer Biol Ther. 2020-6-2
J Immunother Cancer. 2021-4
J Immunother Cancer. 2024-10-4
J Exp Clin Cancer Res. 2025-6-5
J Hematol Oncol. 2023-10-25
Front Immunol. 2022
Sci Transl Med. 2022-6-22
Int J Mol Sci. 2021-10-7
Curr Oncol Rep. 2021-6-14
J Immunother Cancer. 2021-5