A subscription to JoVE is required to view this content. Sign in or start your free trial.
Method Article
This protocol outlines a detailed procedure for generating transient chimeric antigen receptor (CAR) T cells using non-integrating mRNA for cancer immunotherapy and provides reliable methods for evaluating CAR-T cells and their cytotoxic function.
Chimeric antigen receptor (CAR) T cell therapy has emerged as a pioneering cancer treatment, achieving unprecedented success in treating certain hematological malignancies such as lymphomas and leukemias. However, as more cancer patients receive CAR-T cell therapies, treatment-associated secondary primary malignancies are increasingly being reported partly due to unexpected CAR transgene insertion, raising serious safety concerns. To address this issue, we describe here a nonviral, non-integrating approach to generate transient CAR-T cells using mRNA. We electroporated T cells with modified mRNA encoding a human epidermal growth factor receptor 2 (HER2)-specific CAR and generated transient HER2-targeted CAR-T cells. The CAR was efficiently expressed on the T cell surface 1 day after electroporation, increased by day 2, and dramatically declined by day 5. The transient CAR-T cells exhibited potent cytotoxicity against HER2-positive SKOV-3 ovarian cancer cells and secreted high levels of IFN-ϒ. This protocol provides a step-by-step guide for developing small-scale transient CAR-T cells without permanent CAR transgene integration, describing detailed procedures for preparation of CAR mRNA, activation and transfection of T cells, assessment of CAR expression, and in vitro analysis of CAR-T cell function. This method is suitable for transient CAR-T cell generation in both preclinical and clinical studies.
Cancer is an increasingly important threat to human health, with an annually estimated 23.6 million newly diagnosed cases and 10 million deaths globally1. Surgical treatment combined with radiation and chemotherapy remains the gold standard of care for various types of localized noninvasive and invasive cancers2,3,4. Although the traditional systematic treatment has achieved tremendous success in managing early-stage cancers, it is very toxic and has limited effect on metastatic and hematological cancers5. Patients with these cancers undergo frequent treatments and endure significant toxicity in the hope of stabilizing and delaying disease progression. Chimeric antigen receptor (CAR) T cell therapy has recently emerged as a revolutionary targeted therapy that demonstrates potent efficacy for B cell hematological cancers and offers hope to these desperate patients6.
CAR-T cells are engineered T lymphocytes expressing a CAR on the cell surface that specifically recognizes tumor-associated antigens (TAAs) and activates the cells without requiring antigenic presentation from major histocompatibility complex proteins (MHCs)7. Upon activation, the CAR-T cells secrete a panel of cytokines and lyse tumor cells independently of MHCs, thus enhancing the destruction of tumors even when MHCs are downregulated or lost due to the immunosuppressive tumor microenvironment8,9. CARs consist of at least three distinct domains—an extracellular single-chain fragment variant (scFv) containing the antigen-recognizing variable regions of a TAA-specific antibody, a transmembrane domain that anchors the CAR to the cell surface, and an intracellular domain that mediates adaptor protein recruitment and downstream signaling10. Based on variations in the intracellular domains, the CAR structures have evolved through five generations so far, with the first generation containing only the CD3ζ activation domain and subsequent generations possessing one or more extra costimulatory domains from molecules such as 4-1BB and CD2811. These intracellular domains affect the CAR-T cell signaling mechanisms, proliferation, survival, and toxicity, which together determine the clinical antitumor efficacy. The clinical success of CAR-T cell therapy comes from second-generation CAR-T cells that specifically target CD19, a biomarker highly expressed on B-lineage cells, for treating B-cell leukemias and lymphomas12,13. To date, the Food and Drug Administration (FDA) has approved six second-generation CAR-T cell therapies targeting either CD-19 or the B-cell maturation antigen (BCMA) for relapsed or refractory hematological malignancies, including large B-cell lymphoma, mantle cell lymphoma, and multiple myeloma 6. The newer generations of CAR-T cells are currently undergoing intensive preclinical and clinical studies14,15.
Clinical production of CAR-T cells is a complex, expensive, and time-consuming process. Currently, the leukocytes used for CAR-T cell preparation come from the patients themselves to avoid allogeneic reactions, although donor-derived universal (or off-the-shelf) CAR-T cells are under active development and clinical evaluation16. Following isolation of the leukocytes from the patient's peripheral blood by leukapheresis, a foreign gene fragment encoding the CAR is introduced into the activated T cells using either viral or nonviral approaches17,18,19. Retroviruses and lentiviruses are the most common viral vectors for CAR gene delivery, which results in random and permanent integration of the CAR-encoding DNA into the T cell genome20. Nonviral approaches, including mRNA-lipid nanoparticles, transposon systems, and CRISPR/Cas9 genome editing, are less common21. Subsequently, the CAR-expressing T cells are expanded ex vivo on a large scale and then collected and reinfused back into the patients22,23. Manufacturing viral vectors that meet stringent Good Manufacturing Practice (GMP) standards are very difficult, complex, and costly, placing a significant burden on manufacturers, regulatory agencies, and patients, thereby limiting its widespread clinical application.
Despite the exciting success of CAR-T therapy, there is growing concern about its safety. Given the random nature of CAR transgene integration into the T cell genome during viral or transposon transduction, disruption of tumor suppressor genes or activation of oncogenes may occur, leading to malignant transformation of the T cells24,25. Since the FDA's approval of the first CAR-T cell therapy product in 2017, follow-up studies show that as many as 15% of patients who receive the therapy develop secondary primary malignancies (SPMs) such as T cell lymphoma (TCL), non-small cell lung cancer (NSCLC) and skin cancer26,27,28,29. Strikingly, CAR transgenes were highly detected in some SPMs30,31, and some cases of TCL were directly caused by abnormal expansion of the CAR-T cells24,32,33, indicating a direct role of the CAR-T products in inducing some SPMs. Other studies further identified CAR transgene insertions in critical genes such as TET234, JAK135, and PBX236 in the SPM tumor cells. In light of the accumulating evidence of T-cell malignancies following CAR-T cell therapy, the FDA recently concluded that a boxed warning highlighting a serious risk of T-cell malignancy is required for all currently approved CD19-directed and BCMA-targeted CAR-T cell therapies37. However, large-cohort studies following the long-term effects of CAR-T treatment report an incidence of SPMs ranging from 3.8% to 15% among treated patients26,27,28,38,39. The incidence is not significantly higher than that observed in blood cancer patients receiving traditional systematic treatment40,41, suggesting a relatively safe profile for CAR-T cell therapy. Nonetheless, there is an urgent need to develop safe and efficacious CAR-T cells that minimize the risk of SPMs through more cost-effective approaches.
Here, we describe a detailed protocol for a nonviral, non-integrating approach to generate CAR-T cells. The goal is to provide a straightforward, step-by-step guide for generating small-scale, effective CAR-T cells with transient CAR expression, thereby avoiding insertional mutagenesis and minimizing the risk of SPMs. We show that the electroporation of T cells with modified mRNA encoding a CAR specific to the TAA HER2 resulted in robust and transient expression of the anti-HER2 CAR on the T cell surface. While expressing the CAR, the T cells potently killed HER2-positive tumor cells and secreted a high level of IFN-ϒ. The protocol is described in four separate but continuous sections that include preparation of CAR mRNA, activation and electroporation of T cell, assessment of CAR expression, and analysis of CAR-T cell function. This approach is suitable for developing and producing transient CAR-T cells for both academic research and clinical cell therapies.
The PBMCs used here were previously isolated from whole blood from the Stanford Hospital Blood Center according to the Institutional Review Board (IRB)-approved protocol (13942) using the Ficoll-Paque density gradient described before42. The participants' informed consent was not applicable as the blood we used to collect the PBMCs was obtained commercially from the Stanford Hospital Blood Center.
1. Preparation of CAR mRNA
2. Activation and electroporation of T Cell
3. Assessment of CAR expression
4. Analysis of CAR-T cell function
We constructed a second-generation HER2-targeted CAR containing a scFv derived from the humanized anti-HER2 mouse monoclonal antibody (mAb) 4D547, a transmembrane region, and an intracellular 4-1BB costimulatory domain followed by the CD3ζ activation domain (Figure 1A). The DNA sequence encoding the CAR contained a 5' SP6 promoter to drive transcription of the CAR mRNA in vitro (Figure 1B). The CAR mRNA was approximately...
In this study, we describe a detailed nonviral, non-integrating approach for generating transient CAR-T cells and provide technical procedures for assessing CAR-T cell function. This approach avoids the use of conventional retroviral and lentiviral vectors for permanent and random CAR transgene delivery, instead leveraging electroporation to efficiently introduce in vitro modified CAR mRNA into T cells. This method enables high-level, transient CAR expression on the T cell surface, significantly reducing the ris...
The authors Liang Hu, Robert Berahovich, Yanwei Huang, Shiming Zhang, Jinying Sun, Xianghong Liu, Hua Zhou, Shirley, Xu, Haoqi Li, and Vita Golubovskaya are employees of ProMab Biotechnologies. Lijun Wu is an employee and shareholder of ProMab Biotechnologies.
This work was supported by ProMab Biotechnologies.
Name | Company | Catalog Number | Comments |
7-AAD viability dye | Biolegend | 420404 | |
ACEA Novocyte flow cytometer | Agilent | NovoCyte 3000 | |
AIM-V medium | Gibco | 12055083 | |
APC goat anti-human IgG F(ab')2 antibody | Jackson ImmunoResearch Laboratories | 109-136-097 | |
BglII Restriction Enzyme | New England BioLabs (NEB) | R0144L | |
3´-O-Me-m7G(5')ppp(5')G RNA Cap Structure Analog | NEB | S1411S | |
DMEM, high glucose | Gibco | 11965092 | |
Dynabeads Human T-Activator CD3/CD28 beads | Gibco | 11131D | |
E-Plate 96 | Agilent | 5232376001 | |
Ethanol | Sigma | 459844-4L | |
FBS | Lonza.com | 14-503F | |
HiScribe SP6 RNA Synthesis Kit | New England BioLabs (NEB) | E2070S | |
Human IL-2 Recombinant Protein | Gibco | 15140122 | |
Millennium RNA Markers | Invitrogen | AM7150 | |
Monarch RNA Cleanup Kit (500 μg) | NEB | T2050L | |
N1-Methylpseudo-UTP | Trilink | N-1081-10 | |
Neon Transfection Instrument | Invitrogen | MPK5000 | |
Neon Transfection System 100-μL Kit | Invitrogen | MPK10096 | |
Penicillin-Streptomycin (10000 U/mL) | Gibco | 14-503F | |
RPMI 1640 Medium | Gibco | 11875135 | |
Trypsin-EDTA (0.05%), phenol red | Gibco | 25300120 | |
UltraPure Phenol:Chloroform:Isoamyl Alcohol (25:24:1, v/v) | Invitrogen | 15593049 | |
xCELLigence Real-Time Cell Analysis (RTCA) instrument | Agilent | RTCA MP | |
ZymoPURE II Plasmid Midiprep Kit | Zymo Research | D4201 |
Request permission to reuse the text or figures of this JoVE article
Request PermissionThis article has been published
Video Coming Soon
Copyright © 2025 MyJoVE Corporation. All rights reserved