Loading...

Preprints

Manufacturing CD20/CD19-targeted iCasp9 regulatable CAR-TSCMcells usingqCART, theQuantum pBac-based CAR-T system

Chang PS, Chen Y, Hua W, Hsu JC, Tsai J, Huang Y, Kao Y, Wu P, Chang Y, Chang M, Chang Y, Jian S, Lai J, Lai M, Yang W, Shen C, Wen KK, Wu SC.
Preprint from
bioRxiv
4 May 2022
PPR
PPR488835
Abstract

ABSTRACT

Background

CD19-targeted chimeric antigen receptor therapies (CAR19) have driven a paradigm shift in the treatment of relapsed/refractory B-cell malignancies. However, >50% of CAR19-treated patients experienced progressive disease mainly due to antigen escape and low persistence. Clinical prognosis is heavily influenced by CAR-T cell function and systemic cytokine toxicities. Furthermore, it remains a challenge to efficiently, cost-effectively, and consistently manufacture clinically relevant number of virally engineered CAR-T cells.

Methods

Using a highly efficient piggyBac transposon-based vector, Quantum pBac , we developed a virus-free cell engineering system, Quantum CART (qCART ™ ) , for development and production of multiplex CAR-T therapies.

Results

Here, we demonstrated in vitro and in vivo that consistent, robust, and functional CD20/CD19 dual-targeted CAR-T stem cell memory (T SCM ) cells can be efficiently manufactured using the qCART ™ system for clinical application. qCART ™-manufactured CAR-T cells from cancer patients expanded efficiently, rapidly eradicated tumors, and can be safely controlled via an iCasp9 suicide gene-inducing drug.

Conclusions

The qCART ™ system is an elegant system for the manufacturing of CAR-T products having all the desired CAR-T therapy attributes. We believe that the simplicity of manufacturing multiplex CAR-T cells using the qCART ™ system will not only significantly enhance the accessibility of CAR-T therapy but also unlock the full potential of armored CAR-T therapy for the treatment of solid tumors in the future.

What is already known on this topic

Despite the considerable success which has been achieved with CD19-targeted chimeric antigen receptor therapies (CAR19), >50% of CAR19-treated patients still experienced progressive disease. Therefore, there is a need to further improve CAR19 therapies. Current CAR19 therapies commonly utilize virus-based cell engineering methods. CAR-T production using these methods face multiple hurdles, including difficulties to efficiently, cost-effectively, and consistently manufacture clinically relevant number of CAR-T cells. We have previously used a highly efficient piggyBac transposon-based vector, Quantum pBac , to establish Quantum CART ( qCART ™) which is a virus-free cell engineering system for development and production of multiplex CAR-T therapies.

What this study adds

In this report, we further demonstrate in vitro and in vivo that consistent, robust, and functional iCasp9-regulatable, CD20/CD19 dual-targeted CAR-T stem cell memory (T SCM ) cells can be efficiently manufactured using the qCART ™ system for clinical application. These cells possess all the desired attributes for ensuring therapeutic efficacy in CAR-T therapy, including high CAR-T SCM , balanced CD8/CD4 ratio, low exhaustion and senescence marker expressions, and high ex vivo and in vivo expansion capacity. Importantly, we show that qCART ™-manufactured CAR-T cells from hematological cancer patients expanded efficiently, effectively eradicated tumors, and can be safely controlled via an iCasp9 suicide gene-inducing drug. We believe that the simplicity of manufacturing multiplex CAR-T cells using the qCART ™ system will not only significantly enhance the accessibility of CAR-T therapy but also unlock the full potential of armored CAR-T therapy for the treatment of solid tumors in the future.

How this study might affect research, practice or policy

Our findings demonstrate that qCART ™ is a virus-free CAR-T engineering system for manufacturing CAR-T SCM cells from either healthy donors or hematological cancer patients, that possess all the desired attributes for a successful CAR-T therapy. These cells expanded efficiently, rapidly eradicated tumors, and can be safely controlled via activation of iCasp9. We expect that this simple yet robust system for manufacturing multiplex CAR-T cells will advance the CAR-T field.