Synthetic cytokine circuits that drive T cells into immune-excluded tumors

Author:

Allen Greg M.12ORCID,Frankel Nicholas W.13ORCID,Reddy Nishith R.13,Bhargava Hersh K.14ORCID,Yoshida Maia A.13,Stark Sierra R.13,Purl Megan13,Lee Jungmin13ORCID,Yee Jacqueline L.5ORCID,Yu Wei13ORCID,Li Aileen W.13ORCID,Garcia K. Christopher6ORCID,El-Samad Hana17ORCID,Roybal Kole T.158ORCID,Spitzer Matthew H.589ORCID,Lim Wendell A.1378ORCID

Affiliation:

1. Cell Design Institute; University of California San Francisco, San Francisco, CA 94158, USA.

2. Department of Medicine, University of California San Francisco; San Francisco, CA 94158, USA.

3. Department of Cellular and Molecular Pharmacology, University of California San Francisco; San Francisco, CA 94158, USA.

4. Biophysics Graduate Program, University of California San Francisco; San Francisco, CA 94158, USA.

5. Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA 94158, USA.

6. Department of Molecular and Cellular Physiology and Structural Biology, Howard Hughes Medical Institute, Stanford University; Stanford, USA.

7. Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA 94158, USA.

8. Parker Institute for Cancer Immunotherapy, University of California San Francisco; San Francisco, CA 94158, USA.

9. Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco; San Francisco, CA 94158, USA.

Abstract

Chimeric antigen receptor (CAR) T cells are ineffective against solid tumors with immunosuppressive microenvironments. To overcome suppression, we engineered circuits in which tumor-specific synNotch receptors locally induce production of the cytokine IL-2. These circuits potently enhance CAR T cell infiltration and clearance of immune-excluded tumors, without systemic toxicity. The most effective IL-2 induction circuit acts in an autocrine and T cell receptor (TCR)- or CAR-independent manner, bypassing suppression mechanisms including consumption of IL-2 or inhibition of TCR signaling. These engineered cells establish a foothold in the target tumors, with synthetic Notch–induced IL-2 production enabling initiation of CAR-mediated T cell expansion and cell killing. Thus, it is possible to reconstitute synthetic T cell circuits that activate the outputs ultimately required for an antitumor response, but in a manner that evades key points of tumor suppression.

Publisher

American Association for the Advancement of Science (AAAS)

Subject

Multidisciplinary

Cited by 47 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3