Skip to main content
eScholarship
Open Access Publications from the University of California

UC San Diego

UC San Diego Electronic Theses and Dissertations bannerUC San Diego

Using Light to Improve CAR T Cell Immunotherapy Development and Applications

Abstract

Cancer is the second-leading cause of death worldwide. Over the past two decades, chimeric antigen receptor (CAR) T cell therapy has emerged as a promising alternative to traditional surgical, radiation and chemotherapy cancer treatments. Genetically engineered CAR T cells are designed to target and eradicate cancer cells in vivo. However, it remains difficult to identify a set of truly cancer-specific surface antigens to target—a critical requirement to prevent potentially fatal CAR T cell on-target off-tumor toxicity against other healthy tissues elsewhere in the body. I develop a variety of CARs and Receptors and assess their function using genetically encoded fluorescent protein-based biosensors to rapidly detect the pre-transcriptional molecular events leading to CAR-mediated T cell activation. I next propose the novel concept of using light to spatially and temporally limit CAR expression in T cells localized to the tumor site in order to limit on-target off-tumor toxicity in distant healthy tissues. After creating and evaluating a variety of light-sensitive protein-based optogenetic systems to control CAR expression, I uncover three limitations. First, even when kept in the dark, some light-sensitive engineered T cells prematurely express CAR. Second, engineered T cells stimulated with light only weakly upregulate CAR expression. Third, the amount of blue light exposure necessary to induce CAR expression is phototoxic to the T cells. To overcome these limitations, I create the first light-inducible optogenetic system capable of driving robust CAR expression in T cells only following stimulation with minimal, non-toxic amounts of blue light. To do so, I create and optimize a novel genetic AND-gate by integrating components of tamoxifen-inducible Cre recombinase systems with a blue light-inducible split Cre system driven by heterodimerization between the highly sensitive Magnet system protein domains, nMag and pMag. To prevent premature CAR expression, the cytosol-localizing mutant T2 estrogen receptor ligand binding domain (ERT2) is fused to the N-terminal half of the CreN-nMag fusion protein, thus physically separating it from its nuclear-localized binding partner NLS-pMag-CreC. Without tamoxifen to drive ERT2-CreN-nMag protein translocation into the nucleus, the high levels of spontaneous, premature Cre-loxP recombination native to the original photoactivatable split Cre system is significantly suppressed. Upon stimulation with both tamoxifen and blue light, T cells engineered with this novel optogenetic system undergo efficient Cre-loxP recombination to express CAR, with high sensitivity to low-intensity, short-duration blue light exposure. I demonstrate that the new tamoxifen- and photo-activatable split-Cre recombinase system, called TamPA-Cre, can be applied to strictly control localized CAR expression and subsequent T cell activation. The TamPA-Cre system has the potential to limit on-target off-tumor toxicity against distant healthy tissues in a way that was not previously possible.

Main Content
For improved accessibility of PDF content, download the file to your device.
Current View