Waiting
Login processing...

Trial ends in Request Full Access Tell Your Colleague About Jove

Immunology and Infection

Production of Human CRISPR-Engineered CAR-T Cells

Published: March 15, 2021 doi: 10.3791/62299
* These authors contributed equally

Summary

Here, we present a protocol for gene editing in primary human T cells using CRISPR Cas Technology to modify CAR-T cells.

Abstract

Adoptive cell therapies using chimeric antigen receptor T cells (CAR-T cells) have demonstrated remarkable clinical efficacy in patients with hematological malignancies and are currently being investigated for various solid tumors. CAR-T cells are generated by removing T cells from a patient's blood and engineering them to express a synthetic immune receptor that redirects the T-cells to recognize and eliminate target tumor cells. Gene editing of CAR-T cells has the potential to improve safety of current CAR-T cell therapies and further increase the efficacy of CAR-T cells. Here, we describe methods for the activation, expansion, and characterization of human CRISPR-engineered CD19 directed CAR-T cells. This comprises transduction of the CAR lentiviral vector and use of single guide RNA (sgRNA) and Cas9 endonuclease to target genes of interest in T cells. The methods described in this protocol can be universally applied to other CAR constructs and target genes beyond the ones used for this study. Furthermore, this protocol discusses strategies for gRNA design, lead gRNA selection and target gene knockout validation to reproducibly achieve high-efficiency, multiplex CRISPR-Cas9 engineering of clinical grade human T cells.

Introduction

Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the field of adoptive cell therapies and cancer immunotherapy. CAR-T-cells are engineered T-cells expressing a synthetic immune receptor that combines an antigen-specific single chain antibody fragment with signaling domains derived from the TCRzeta chain and costimulatory domains necessary and sufficient for T-cell activation and co-stimulation1,2,3,4. The manufacturing of CAR-T cells starts by extracting the patient's own T-cells, followed by ex vivo viral transduction of the CAR module and expansion of the CAR-T cell product with magnetic beads that function as artificial antigen presenting cells5. Expanded CAR-T cells are re-infused into the patient where they can engraft, eliminate target tumor cells and even persist for multiple years post infusion6,7,8. Although CAR-T cell therapy has resulted in remarkable response rates in B-cell malignancies, clinical success for solid tumors has been challenged by multiple factors including poor T-cell infiltration9, an immunosuppressive tumor microenvironment10, antigen coverage and specificity, and CAR-T cell dysfunction11,12. Another limitation of current CAR-T cell therapy includes the use of autologous T-cells. After multiple rounds of chemotherapy and high tumor burden, CAR-T cells can be of poor quality as compared to allogeneic CAR-T products from healthy donors in addition to the time and expense associated with manufacturing of autologous CAR-T cells. Gene-editing of the CAR-T cell product by CRISPR/Cas9 represents a new strategy to overcome current limitations of CAR-T cells13,14,15,16,17.

CRISPR/Cas9 is a two component system that can be used for targeted genome editing in mammalian cells18,19. The CRISPR-associated endonuclease Cas9 can induce site-specific double-strand breaks guided by small RNAs through base-pairing with the target DNA sequence20. In the absence of a repair template, double-strand breaks are repaired by the error prone nonhomologous end joining (NHEJ) pathway, resulting in frameshift mutations or premature stop codons through insertion and deletion mutations (INDELs)19,20,21. Efficiency, ease of use, cost-effectiveness and the ability for multiplex genome editing make CRISPR/Cas9 a powerful tool to enhance the efficacy and safety of autologous and allogeneic CAR-T cells. This approach can also be used to edit TCR directed T cells by replacing the CAR construct with a TCR. Additionally, allogeneic CAR-T cells that have limited potential to cause graft versus host disease can also be generated by gene editing the TCR, b2m, and HLA locus.

In this protocol, we show how CRISPR-engineering of T cells can be combined with viral vector mediated delivery of the CAR-Transgene to generate genome-edited CAR-T cell products with enhanced efficacy and safety. A schematic diagram of the entire process is shown in Figure 1. Using this approach, we have demonstrated high-efficiency gene knockout in primary human CAR-T cells. Figure 2A describes in detail the timeline of each step for editing and manufacturing T cells. Strategies for guide RNA design and knockout validation are also discussed to apply this approach to various target genes.

Subscription Required. Please recommend JoVE to your librarian.

Protocol

Human T cells were procured through the University of Pennsylvania Human Immunology Core, which operates under principles of Good Laboratory Practice with established standard operating procedures and/or protocols for sample receipt, processing, freezing, and analysis conform to MIATA and University of Pennsylvania ethics guidelines.

1. Lentiviral vector production

NOTE: The viral products have been made replication-defective by separation of packaging constructs (Rev, gag/pol/RRE, VSVg and transfer plasmid) into four separate plasmids, greatly reducing the likelihood of recombination events that may result in replication-competent virus.

  1. Prepare HEK293T cells one day prior to the transfection. Plate approximately 6 x 106 cells in T150 culture vessels in 30 mL of standard culture media (referred to as R10:RPMI 1640 supplemented with 10% fetal calf serum, 10 mM HEPES, 1% Pen/Strep, 1% L-glutamine) and incubate overnight at 37 °C. 18-24 h later, cells should be 60-70% confluent and look healthy (dendritic projections and uniform distribution across the flask). If cells look healthy, proceed to step 1.2.
  2. Prepare the transfection mix containing lipofection reagent (96 µL), pTRP gag/pol (Lot# RR13SEP19A) (18 µg), pTRP RSV-Rev (Lot# RR13SEP19B-3) (18 µg), pTRP VSVG (Lot# RR13SEP19C) (7 µg) packaging plasmids and 15 µg of expression plasmid (CD19bbz scFv cloned in pTRPE).
    1. For each transfection reaction, prepare one tube containing 1 mL of reduced-serum minimal essential media plus the four plasmids described in step 1.2 as well as one tube containing 2 mL of reduced-serum minimal essential media plus 90 µL of lipofection reagent. Combine these two solutions by dropwise addition of the 1 mL plasmid mix to the 2 mL of lipofection reagent mix. Be sure to vigorously mix the solution by pipetting up and down several times. Incubate the solution for 15 min at room temperature.
    2. In the meantime, aspirate the media from the HEK293T cell flask and gently wash cells with 10 mL of reduced-serum minimal essential media and aspirate again.
    3. Gently add the transfection mix (3 mL) from step 1.2.1 to the bottom corner of cell culture flask using a 5 mL serological pipette. Gently tilt the flask to evenly distribute the transfection mix across the cell culture flask and incubate for 10 min at room temperature. Make sure to not disturb the cell monolayer. After a 10 min incubation, add 35 mL of R10 media and return flask to the incubator for 24 h.
  3. After 24 h, collect the supernatant from the HEK293T cell culture flasks and transfer into 50 mL conical tubes. Add fresh R10 to the HEK29T cells and put the cells back into the incubator for another 24 h. Spin down the supernatant (300 x g) to remove cell debris and filter the supernatant through a 0.45 µm filter.
  4. Concentrate the filtrate from step 1.3 by high-speed ultracentrifugation (25,000 x g for 2.5 h or 8000 x g overnight (O/N)). Store the 24 h virus pellet at 4 °C while pooling the 48 h virus.
  5. Repeat steps 1.3-1.4 for the 48 h collection and combine 24 h and 48 h virus collections. Pellets from the 48 h collection will contain a combined harvest of the lentivirus.
  6. Resuspend viral pellet in approximately 1 mL of cold R10 and aliquot into 100 µL vials. Immediately snap freeze the aliquots using dry ice and store at -80 °C.
  7. Calculate functional viral titer in transducing units/mL by transducing a fixed amount of CD3/CD28 bead-activated primary human T-cells with serial dilutions of the lentiviral supernatant. Measure CAR-Transduction after 72 h by flow-cytometry.
    1. Stain for the CD19-directed CAR with an anti-FCM63 scFv antibody. For other chimeric antigen receptors, stain using fluorophore-labeled recombinant protein that is specific to the CAR scFv. Virus dilution that generates under 20% CAR positive cells by flow-cytometry is the most accurate dilution to calculate viral titer from. Above 20% CAR-positive cells, the chance for each positive cell to be transduced twice increases, resulting in an underestimation of the number of transducing particles. Calculate the transducing units per mL(TU/mL ) = (number of cells transduced x percent CAR positive cells x dilution factor)/(transduction volume in mL).

2. Designing of sgRNAs and gene disruption in primary human T cells

  1. Designing CRISPR sgRNA's
    NOTE: Several servers and programs facilitate the design of target-specific sgRNA's. In this protocol, CRISPR sgRNAs were designed using CHOPCHOP (https://chopchop.cbu.uib.no), and the gRNA design portal from the Broad institute (https://portals.broadinstitute.org/gpp/public/analysis-tools/sgrna-design).
    1. For each target gene, design six to ten sgRNA sequences to target early coding exon sequences.
      NOTE: sgRNA should have high on-target efficacy and low off target efficacy. Each machine learning algorithm for determining sgRNA efficacy works slightly differently. Therefore, comparing multiple sgRNA design tools and curating a list of six to ten sgRNA for screening is recommended.
  2. Gene disruption in primary human T-cells
    1. Obtain autologous peripheral blood mononuclear cells (PBMC's) from healthy volunteer donors. A schematic timeline of the protocol is described in Figure 2A and described below.
    2. Isolate CD4+ and CD8+ T-cells using commercially available CD4 and CD8 selection kits.
    3. Combine CD4+ and CD8+ T-cells at a 1:1 ratio and incubate overnight at 3x106 cells/mL in R10 supplemented with 5 ng/mL huIL-7 and huIL-15 each. Addition of IL-7 and IL-15 is recommended as they are known to promote a central memory phenotype and CAR-T cells expanded with IL-7 and IL-15 have superior anti-tumor efficacy compared to IL-2 expanded CAR-T cells22,23,24.
    4. The next day, count T-cells and centrifuge 5-10 x 106 cells at 300 x g for 5 min. Discard all the supernatant and wash the cell pellet in reduced-serum minimal essential media. Resuspend the pellet in 100 µL of nucleofection solution according to the manufacturer's instructions (Table of Materials).
    5. While washing the cells, prepare ribonucleoprotein (RNP) complex with Cas9 and gRNA by incubating 10 µg of Cas9 nuclease with 5 µg of sgRNA for 10 min at room temperature (RT). The molar ratio of Cas9 to sgRNA is 1:2.4. A mock control that contains Cas9 and electroporation enhancer, but no sgRNA, is recommended.
      NOTE: Multiplex gene editing can be performed at this step by making RNP complexes for each target separately and combining them with cells at the time nucleofection as described in the next step. Choosing the reagents to assemble the RNP complex are key to getting high KO efficiency. For example, Cas9 nucleases from different vendors have varying off-target effects and chemically modified gRNA decrease toxicity to T cells, thereby increasing KO efficiency.
    6. Combine the resuspended cells from step 2.2.4 with the RNP complex from step 2.2.5 and add 4.2 µL of 4 µM electroporation enhancer (ssDNA oligonucleotide that is non-homologous to human genome). Mix well and transfer into electroporation cuvettes. Avoid bubbles as they impair electroporation efficiency.
    7. Electroporate cells using pulse code EH111. After electroporation, incubate cells in R10 supplemented with 5 ng/mL huIL-7 and huIL-15 at 5x106 cells/mL at 30 °C for 48 h in 12-well plates. After 48 h, proceed with T-cell activation and expansion.

3. T cell activation, lentiviral transduction and expansion

NOTE: For screening sgRNA's, lentiviral transduction (Step 3.2) of the CAR construct is not necessary.

  1. Count electroporated T-cells and dilute to a concentration of 1 x 106 cells/mL using warm R10 supplemented with 5 ng/mL huIL-7 and huIL-15. Activate cells by adding anti-CD3/anti-CD28 monoclonal antibody coated magnetic beads at a ratio of 3 beads per live T-cell.
    NOTE: Electroporation causes significant cell death resulting in roughly 60% ± 15% viable cells compared to non-electroporated cells. Use a live/dead cell count to determine the appropriate amount of beads. Transfer the cells to 37 °C and incubate overnight.
  2. After overnight stimulation, transduce CRISPR-engineered T-cells with lentiviral supernatant from step 1. Add the appropriate volume based on the viral titer calculated in step 1.7 to achieve a multiplicity of infection (MOI) of 3 and incubate cells for 72 h at 37 °C.
    NOTE: The virus pellet resuspended in R10 is simply added on top of the cells according to the cell concentration, viral titer, and desired MOI.
  3. After 72 h, feed cells with 50% of the current culture volume R10 containing 10 ng/mL huIL-7 and huIL-15 and return them to the incubator for another 48 h. Do not disturb the clusters that have formed between the T-cells and the beads.
  4. After 48 h, remove the beads from the cells by gently resuspending the cell-bead mixture followed by magnetic separation. Count cells after bead removal and bring to a concentration of 0.8 x 106 cells/mL using R10 supplemented with 5 ng/mL huIL-7 and huIL-15. After de-beading, cell numbers should be similar to the cell count on Day 1 prior to electroporation (Figure 2B).
  5. After 24 h, count cells and feed to a concentration of 1 x 106 cells/mL with R10 + 5 ng/mL huIL-7 and huIL-15. Repeat this every 24 h until growth kinetics and cell size demonstrate cells have rested from stimulation.
    NOTE: From this point, T-cells double approximately every 24h. T-cell usually undergo 5-7 population doublings and have a cell volume of approximately 300 ± 50 fL when rested.
  6. Once rested, spin down CRISPR-engineered CAR-T cells and resuspend in freeze media (1:1 X-Vivo and FBS plus 10% DMSO) for cryopreservation. CAR-T cells used should be thawed and rested at 37 °C for 16 hours before an experiment.
    NOTE: Keep approximately 10x106 cells reserved for measuring knockout efficiency and CAR integration. Expected population doublings and volume changes during the expansion have been shown in Figure 2B,2C. Additionally, Figure 2D shows levels of CAR expression on both Mock and gene edited CAR-T cells.

4. CRISPR Efficiency

  1. Genomic DNA extraction and target gene amplification
    1. From each screening culture, spin down 3-5 x 106 T-cells. Cells can be either frozen down as a dry pellet or one can proceed with genomic DNA extraction. At time of DNA extraction, resuspend pellets in 200 µL of Phosphate Buffer Saline (PBS) and extract genomic DNA from electroporated cells using a standard DNA extraction kit according to the manufacturer instruction.
      1. Briefly, lyse cells with proteinase K and load lysate onto a DNA binding column. During centrifugation, DNA is binding to the membrane while contaminants will pass through. After two washing steps to remove remaining contaminants and enzyme inhibitors, elute DNA in water.
    2. Amplify 200-300 ng of genomic DNA using standard PCR mix containing DNA polymerase, accessory proteins, salts, and dNTPs and 10 µM forward and reverse primers flanking the region of the intended double-strand break.
      NOTE: For PCR primer design, the reference genomic sequence (can be found using http://www.ensemble.org) flanking the gRNA cut site are entered into the NCBI primer blast tool (https://www.ncbi.nlm.nih.gov/tools/primer-blast). PCR primers should be designed such that the amplicon has a target size of 600-700bp. This length allows to design sequencing primers that bind within the amplicon with sufficient distance from the gRNA cut site (at least 150 bp) to ensure good sequencing quality around the Cas9 induced indels (see 4.2.1). Each gRNA requires a unique primer pair, unless the gRNA cut sites are in close proximity. 
    3. Run the entire PCR product on a 1% agarose gel and purify the amplicon using a standard agarose gel purification kit.
      NOTE: The process of determining the optimal Tm for PCR can take multiple rounds of troubleshooting. This is because the KO sequence has indels and the target gene should be primed for sequencing where there would be no indels usually 100 bp upstream or downstream of the cut site. This can vary widely due to indels resulting from non-homologous end joining. Designing nested sequencing primers to the PCR primers and increasing the concentration of product sequenced can eliminate problems with sequencing.
  2. Sequencing and indel detection
    1. Design sequencing primers that bind to the gel purified amplicon and send mock and knockout amplicons for Sanger sequencing. Once sequencing is complete, upload the trace files to the Desktop Genetics software (tide.deskgen.com, Desktop Genetics) for TIDE analysis.
      1. For sequencing primer design, enter the sequence of the PCR amplicon into a standard primer design software (NCBI, Eurofins or other publicly available tools). The design software will suggest multiple primer sequences that are suitable to sanger sequencing.
      2. Choose forward and reverse primers that bind within the amplicon at least 150 bp upstream or downstream of the gRNA cut site to ensure good sequencing quality around the indels. The sequencing chromatogram will be used in 4.2 for TIDE analysis, therefore it is important that sequencing quality is good for accurate indel decomposition (see Hultquist et al.)25.
    2. Use TIDE (Tracking of Indels by DEcomposition) analysis to detect knock out (KO) efficiency at the genomic level26. The algorithm accurately reconstructs the spectrum of indels from the sequence traces and calculates R2 values, reflecting goodness of fit after non-negative linear modeling by the TIDE software.
  3. Target protein detection
    1. For target genes whose gene product is expressed on the surface of T-cells, stain approximately 1 x 105 cells from each screening culture with a fluorochrome-tagged antibody specific for the target protein. Compare expression levels of the target protein between the mock control and knockout groups by flow cytometry as shown in Figure 3A,3B.
    2. For target genes whose gene product is expressed intracellularly, lyse approximately 3 x 106 million cells per screening group in lysis buffer and follow standard protocols for SDS-PAGE and western blotting. Alternatively, surface or intracellular flow-cytometry can be used to probe for the target protein.

5. Monitoring Off-target effects using iGUIDE - Library preparation, DNA sequencing, and analysis

NOTE: iGUIDE technique allows for detection of locations of Cas9 guided cleavage and quantify the distributions of those DNA double-stranded breaks.

  1. Perform iGUIDE as described by Nobles et al.21. The off-target effects of sgRNA targeting TRAC locus using the iGUIDE technique have been shown in Stadtmauer et al, 202013.

Subscription Required. Please recommend JoVE to your librarian.

Representative Results

We describe here a protocol to genetically engineer T cells, that can be used to generate both autologous and allogeneic CAR-T cells, as well as TCR redirected T cells.

Figure 1 provides a detailed description of the stages involved in the process of manufacturing CRISPR edited T cells. The process begins by designing sgRNA to the gene of interest. Once the sgRNA are designed and synthesized they are then used to make RNP complexes with the appropriate Cas protein. T cells are isolated from either a healthy donor or a patient apheresis and RNP complexes are delivered either by electroporation or nucleofection. Post editing, the T cells are activated and transduced with the lentiviral vector coding for the CAR or the TCR construct. After activation, T cells are expanded in culture and cryopreserved for future studies. The detailed protocol followed in the laboratory is described in Figure 2. During the expansion, the population doubling and volume changes are tracked throughout the protocol and an example is shown in Figure 2B and C for both mock and edited CAR-T cells. Figure 2B,C show that the KO of the gene of interest did not cause any significant changes in the proliferation and activation during the expansion. These results, however, depend on the target gene being edited and hence may or may not lead to changes in proliferation and expansion.

Once the cells are cryopreserved, levels of CAR expression are also determined for further functional studies. In this case, as shown in Figure 2D we checked CD19 CAR expression on both the Mock edited and KO CAR-T cells and did not see any significant changes. This will again depend on the gene of interest being edited. Finally, the KO efficiency can be determined using multiple techniques such as flow cytometry and western blot for protein level detection and also Sanger sequencing for gene level detection of the KO. Figure 3A and 3B show representative flow cytometry plots where PDCD1 and TRAC locus is targeted using sgRNA, showing an efficiency of 90% for the PDCD1 sgRNA and 98% for the TRAC sgRNA across multiple healthy donors. Thus, this protocol can achieve high efficiency knockout with minimal loss in viability.

Figure 1
Figure 1. Schematic diagram showing T cell editing using CRISPR Cas9 Technology and manufacturing of primary human CAR-T cells. Please click here to view a larger version of this figure.

Figure 2
Figure 2. Expansion of edited CAR-T cells and their population doublings. (A) Timeline of CRISPR editing and manufacturing in primary human CART cells. (B) Population Doublings in Mock and CRISPR edited CD19 CAR-T cells measured using a Coulter Counter during the expansion of the CAR-T cells (n=3 healthy donors; KO=knockout) (C) Cell size (µm3) measured using a Coulter Counter during the expansion of the CAR-T cells (n=3 healthy donors). (D) Representative flow cytometry plots showing CAR staining and average in multiple donors showing CAR expression in both mock and edited CAR-T cells. CAR expression was detected using an anti-idiotype antibody conjugated to a fluorophore and gated on Lymphocytes/Singlets/Live Cells (n=3 healthy donors, UTD=untransduced,). Please click here to view a larger version of this figure.

Figure 3
Figure 3. Characterizing KO efficiency in edited CAR-T Cells using flow cytometry. (A) Representative flow cytometry plots showing PD-1 staining and average in multiple donors showing PD-1 KO efficiency using a gRNA targeting the TRAC locus in mock and edited CAR-T cells. PD-1 expression was detected using PD-1 antibody (Clone EH12.2H7) conjugated to fluorophore and gated on Lymphocytes/Singlets/Live Cells (n=3 healthy donors). Error bars indicate mean±standard error of the mean (SEM). **** p<0.0001, *** p=0.0005, ** p=0.001 by Welch's t test. (B) Representative flow cytometry plots showing CD3 staining and average in multiple donors showing CD3 KO efficiency using a gRNA targeting the TRAC locus in mock and edited CAR-T cells. CD3 expression was detected using CD3 antibody (Clone OKT3) conjugated to fluorophore and gated on Lymphocytes/Singlets/Live Cells (n=3 healthy donors). Error bars indicate mean±standard error of the mean (SEM). **** p<0.0001, *** p=0.0005, ** p=0.001 by Welch's t test. Please click here to view a larger version of this figure.

Subscription Required. Please recommend JoVE to your librarian.

Discussion

Here we describe approaches to gene edit CAR-T cells using CRISPR Cas9 technology and manufacture products to further test for function and efficacy. The above protocol has been optimized for performing CRIPSR gene editing in primary human T cells combined with engineering T cells with chimeric antigen receptors. This protocol allows high knockout efficiency with minimal donor-to-donor variability. Modification using CRISPR can improve both the efficacy and safety of CAR-T cells by eliminating receptors that inhibit T cells function and manufacturing allogeneic CAR-T cells.

For small scale expansion protocols, starting with 106 cells per group leads to around 5006 CAR-T cells with an average of 6 population doublings in a healthy normal donor. However, this can vary depending upon if the gene of interest affects T cell activation, transduction and proliferation. Five hundred million cells are sufficient for confirming KO efficiency, in vitro assays and in vivo assays. There are different variations to the protocol wherein CRISPR editing could be performed before or after activation and CAR-Transduction. Ren et al described one such variation where cells are edited after bead stimulation and CAR-Transduction15. The advantage of CRISPR editing before bead stimulation is lower cell quantities need to be edited since the T cells have not proliferated yet, making the procedure less time consuming and more cost-efficient. Additionally, performing editing upfront is directly translatable to the clinic. In fact, many steps in this protocol have been informed by what can be adopted in the clinic which increases the consistency of the KO efficiency when moving from bench to bedside.

There are multiple variables that can be chosen at each step of the protocol. For example, T cells can be electroporated or nucleofected. While both achieve comparable KO efficiencies, in our experience using the nucleofector has higher viability post editing and hence is preferred. However, using the electroporator may prove to be more cost-effective in the long run. Both of these equipments are used for small scale T cell expansions. For large scale and clinical scale expansions, protocols must be re-optimized to perform editing and requires different equipment for nucleofection. There are multiple technological platforms that can be used for both small scale and large-scale editing and expansion depending on the needs of the user.

Subscription Required. Please recommend JoVE to your librarian.

Disclosures

The authors have no disclosures.

Acknowledgments

We acknowledge the Human Immunology Core for providing normal donor T cells and the Flow Cytometry Core at University of Pennsylvania.

Materials

Name Company Catalog Number Comments
4D-Nucleofactor Core Unit Lonza AAF-1002B
4D-Nucleofactor X-Unit Lonza AAF-1002X
Accuprime Pfx Supermix ThermoFisher 12344040
Beckman Optima XPN ultracentrifuge Beckman Coulter
Brilliant Violet 605 anti-human CD3 Antibody Biolegend 317322 Clone OKT3
BV711 Anti-human PD1 Biolegend Clone EH12.2H7
Cas9-Electroporation enhancers IDT 1075915
CD3/CD28 Dynabeads ThermoFisher 40203D
CD4+ T cell isolation Kit StemCell technologies 15062
CD8+ T cell isolation Kit StemCell technologies 15063
Corning 0.45 micron vacuum filter/bottle Corning 430768
Corning T150 cell culture flask Millipore Sigma CLS430825
DMSO Millipore Sigma D2650
DNAeasy Blood and Tissue Kit Qiagen 69504
DynaMag Magnet ThermoFisher 12321D
Glutamax supplement ThermoFisher 35050061
HEK293T cells ATCC CRL-3216
HEPES (1 M) ThermoFisher 15630080
huIL-15 PeproTech 200-15
huIL-7 PeproTech 200-07
Lipofectamine 2000 ThermoFisher 11668019
Nucleospin Gel and PCR cleanup Takara 740609.25
Opti-MEM ThermoFisher 31985062
P3 Primary cell 4D-nucleofactor X Kit L Lonza V4XP-3024
Penicilin-Streptomycin-Glutamine ThermoFisher 10378016
pTRPE expression Plasmid in house
Rabbit Anti-Mouse FMC63 scFv Monoclonal Antibody, (R19M), PE CytoArt 200105
RPMI1640 ThermoFisher 12633012
sgRNA IDT
Spy Fi Cas9 Aldevron 9214
Ultracentrifuge tubes Beckman Coulter 326823
Viral packaging mix in house
X-Vivo-15 Media Lonza BE02-060F

DOWNLOAD MATERIALS LIST

References

  1. Kowolik, C. M., et al. CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Research. 66 (22), 10995-11004 (2006).
  2. Krause, A., et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. The Journal of Experimental Medicine. 188 (4), 619-626 (1998).
  3. Kuwana, Y., et al. Expression of chimeric receptor composed of immunoglobulin-derived V resions and T-cell receptor-derived C regions. Biochemical and biophysical research Communications. 149 (3), 960-968 (1987).
  4. Maher, J., Brentjens, R. J., Gunset, G., Rivière, I., Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor. Nature Biotechnology. 20 (1), 70-75 (2002).
  5. Levine, B. L., Miskin, J., Wonnacott, K., Keir, C. Global manufacturing of CAR-T cell therapy. Molecular Therapy-Methods & Clinical Development. 4, 92-101 (2017).
  6. Neelapu, S. S., et al. Axicabtagene ciloleucel CAR-T-cell therapy in refractory large B-cell lymphoma. New England Journal of Medicine. 377 (26), 2531-2544 (2017).
  7. Porter, D. L., et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Science Translational Medicine. 7 (303), 139 (2015).
  8. Maude, S. L., et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. New England Journal of Medicine. 371 (16), 1507-1517 (2014).
  9. Melero, I., Rouzaut, A., Motz, G. T., Coukos, G. T-cell and NK-cell infiltration into solid tumors: a key limiting factor for efficacious cancer immunotherapy. Cancer Discovery. 4 (5), 522-526 (2014).
  10. Mohammed, S., et al. Improving chimeric antigen receptor-modified T cell function by reversing the immunosuppressive tumor microenvironment of pancreatic cancer. Molecular Therapy. 25 (1), 249-258 (2017).
  11. Moon, E. K., et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clinical Cancer Research. 20 (16), 4262-4273 (2014).
  12. Beatty, G. L., O'Hara, M. Chimeric antigen receptor-modified T cells for the treatment of solid tumors: defining the challenges and next steps. Pharmacology & Therapeutics. 166, 30-39 (2016).
  13. Stadtmauer, E. A., et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 367 (6481), (2020).
  14. MacLeod, D. T., et al. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR-T cells. Molecular Therapy. 25 (4), 949-961 (2017).
  15. Ren, J., et al. Multiplex genome editing to generate universal CAR-T cells resistant to PD1 inhibition. Clinical Cancer Research. 23 (9), 2255-2266 (2017).
  16. Ureña-Bailén, G., et al. CRISPR/Cas9 technology: towards a new generation of improved CAR-T cells for anticancer therapies. Briefings in Functional Genomics. 19 (3), 191-200 (2020).
  17. Li, C., Mei, H., Hu, Y. Applications and explorations of CRISPR/Cas9 in CAR-T-cell therapy. Briefings in Functional Genomics. 19 (3), 175-182 (2020).
  18. Ran, F. A., et al. Genome engineering using the CRISPR-Cas9 system. Nature Protocols. 8 (11), 2281-2308 (2013).
  19. Cong, L., et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 339 (6121), 819-823 (2013).
  20. Jinek, M., et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 337 (6096), 816-821 (2012).
  21. Jinek, M., et al. RNA-programmed genome editing in human cells. Elife. 2, 00471 (2013).
  22. Schluns, K. S., Kieper, W. C., Jameson, S. C., Lefrançois, L. Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nature Immunology. 1 (5), 426-432 (2000).
  23. Prlic, M., Lefrancois, L., Jameson, S. C. Multiple choices: regulation of memory CD8 T cell generation and homeostasis by interleukin (IL)-7 and IL-15. The Journal of Experimental Medicine. 195 (12), 49-52 (2002).
  24. Zhou, J., et al. Chimeric antigen receptor T (CAR-T) cells expanded with IL-7/IL-15 mediate superior antitumor effects. Protein & Cell. 10 (10), 764-769 (2019).
  25. Hultquist, J. F., et al. CRISPR-Cas9 genome engineering of primary CD4+ T cells for the interrogation of HIV-host factor interactions. Nature Protocols. 14 (1), 1-27 (2019).
  26. Brinkman, E. K., Chen, T., Amendola, M., van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Research. 42 (22), 168 (2014).

Tags

Human CRISPR-engineered CAR-T Cells Laboratory Protocol Genome Editing CRISPR-Cas9 Clinical Trials CAR Constructs Target Genes Guide RNA Screen Culture Conditions Seeding Density Peripheral Blood Mononuclear Cells (PBMCs) CD4 Positive T Cells CD8 Positive T Cells Autologous T Cells IL7 And IL15 Supplementation Cell Counting Centrifugation Nuclear Affectio
Production of Human CRISPR-Engineered CAR-T Cells
Play Video
PDF DOI DOWNLOAD MATERIALS LIST

Cite this Article

Agarwal, S., Wellhausen, N., Levine, More

Agarwal, S., Wellhausen, N., Levine, B. L., June, C. H. Production of Human CRISPR-Engineered CAR-T Cells. J. Vis. Exp. (169), e62299, doi:10.3791/62299 (2021).

Less
Copy Citation Download Citation Reprints and Permissions
View Video

Get cutting-edge science videos from JoVE sent straight to your inbox every month.

Waiting X
Simple Hit Counter