Multiplex Genome Editing to Generate Universal CAR T Cells Resistant to PD1 Inhibition

Purpose: Using gene-disrupted allogeneic T cells as universal effector cells provides an alternative and potentially improves current chimeric antigen receptor (CAR) T-cell therapy against cancers and infectious diseases. Experimental Design: The CRISPR/Cas9 system has recently emerged as a simple and efficient way for multiplex genome engineering. By combining lentiviral delivery of CAR and electro-transfer of Cas9 mRNA and gRNAs targeting endogenous TCR, β-2 microglobulin (B2M) and PD1 simultaneously, to generate gene-disrupted allogeneic CAR T cells deficient of TCR, HLA class I molecule and PD1. Results: The CRISPR gene–edited CAR T cells showed potent antitumor activities, both in vitro and in animal models and were as potent as non-gene–edited CAR T cells. In addition, the TCR and HLA class I double deficient T cells had reduced alloreactivity and did not cause graft-versus-host disease. Finally, simultaneous triple genome editing by adding the disruption of PD1 led to enhanced in vivo antitumor activity of the gene-disrupted CAR T cells. Conclusions: Gene-disrupted allogeneic CAR and TCR T cells could provide an alternative as a universal donor to autologous T cells, which carry difficulties and high production costs. Gene-disrupted CAR and TCR T cells with disabled checkpoint molecules may be potent effector cells against cancers and infectious diseases. Clin Cancer Res; 23(9); 2255–66. ©2016 AACR.

[1]  David Bryder,et al.  Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. , 2014, Cell stem cell.

[2]  Alan Maréchal,et al.  A Multidrug-resistant Engineered CAR T Cell for Allogeneic Combination Immunotherapy , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[3]  Pamela A Shaw,et al.  Chimeric antigen receptor T cells for sustained remissions in leukemia. , 2014, The New England journal of medicine.

[4]  Aymeric Duclert,et al.  Multiplex Genome-Edited T-cell Manufacturing Platform for "Off-the-Shelf" Adoptive T-cell Immunotherapies. , 2015, Cancer research.

[5]  B. van Steensel,et al.  Easy quantitative assessment of genome editing by sequence trace decomposition , 2014, Nucleic acids research.

[6]  E. Leiter,et al.  Major Histocompatibility Complex Class I-Deficient NOD-B2mnull Mice are Diabetes and Insulitis Resistant , 1994, Diabetes.

[7]  Jin-Soo Kim,et al.  Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases , 2014, Genome research.

[8]  A. Wu,et al.  Engineered humanized diabodies for microPET imaging of prostate stem cell antigen-expressing tumors. , 2008, Protein engineering, design & selection : PEDS.

[9]  B. Levine,et al.  Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. , 2010, Cancer research.

[10]  Norman L. Letvin,et al.  In Vivo Natural Killer Cell Depletion during Primary Simian Immunodeficiency Virus Infection in Rhesus Monkeys , 2008, Journal of Virology.

[11]  D. Dolfi,et al.  Multifactorial T-cell Hypofunction That Is Reversible Can Limit the Efficacy of Chimeric Antigen Receptor–Transduced Human T cells in Solid Tumors , 2014, Clinical Cancer Research.

[12]  David A. Scott,et al.  Rationally engineered Cas9 nucleases with improved specificity , 2015, Science.

[13]  R. Shattock,et al.  Inhibition of HIV-1 infection of primary CD4+ T-cells by gene editing of CCR5 using adenovirus-delivered CRISPR/Cas9. , 2015, The Journal of general virology.

[14]  S. Rosenberg,et al.  Clinical Scale Zinc Finger Nuclease-mediated Gene Editing of PD-1 in Tumor Infiltrating Lymphocytes for the Treatment of Metastatic Melanoma. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[15]  S. Grupp,et al.  Treatment of advanced leukemia in mice with mRNA engineered T cells. , 2011, Human gene therapy.

[16]  Luigi Naldini,et al.  A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. , 2012, Blood.

[17]  Wei-Ting Hwang,et al.  Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. , 2014, The New England journal of medicine.

[18]  Randall J. Platt,et al.  Therapeutic genome editing: prospects and challenges , 2015, Nature Medicine.

[19]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[20]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[21]  I. Pastan,et al.  Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains , 2009, Proceedings of the National Academy of Sciences.

[22]  J. Orange,et al.  Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases , 2008, Nature Biotechnology.

[23]  W. Wilson,et al.  Construction and Preclinical Evaluation of an Anti-CD19 Chimeric Antigen Receptor , 2009, Journal of immunotherapy.

[24]  Jennifer A. Doudna,et al.  Sequence- and Structure-Specific RNA Processing by a CRISPR Endonuclease , 2010, Science.

[25]  Qing He,et al.  CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia , 2013, Science Translational Medicine.

[26]  D. Atanackovic,et al.  TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer , 2014, Gene Therapy.

[27]  Lei Zhang,et al.  Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer , 2012, Nature Medicine.

[28]  Yi Li,et al.  High-Affinity TCRs Generated by Phage Display Provide CD4+ T Cells with the Ability to Recognize and Kill Tumor Cell Lines1 , 2007, The Journal of Immunology.

[29]  Yu Chen,et al.  Genome editing of CXCR4 by CRISPR/cas9 confers cells resistant to HIV-1 infection , 2015, Scientific Reports.

[30]  D. Torigian,et al.  Mesothelin-Specific Chimeric Antigen Receptor mRNA-Engineered T Cells Induce Antitumor Activity in Solid Malignancies , 2013, Cancer Immunology Research.

[31]  M. Smyth,et al.  Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells , 2013, Clinical Cancer Research.

[32]  C. van Broeckhoven,et al.  Highly efficient gene delivery by mRNA electroporation in human hematopoietic cells: superiority to lipofection and passive pulsing of mRNA and to electroporation of plasmid cDNA for tumor antigen loading of dendritic cells. , 2001, Blood.

[33]  Bernd Hauck,et al.  Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. , 2013, The New England journal of medicine.

[34]  Di Zhang,et al.  CCR5 Gene Disruption via Lentiviral Vectors Expressing Cas9 and Single Guided RNA Renders Cells Resistant to HIV-1 Infection , 2014, PloS one.

[35]  N. Letvin,et al.  Use of an anti‐CD16 antibody for in vivo depletion of natural killer cells in rhesus macaques , 2008, Immunology.

[36]  J. Joung,et al.  High-fidelity CRISPR-Cas9 variants with undetectable genome-wide off-targets , 2015, Nature.

[37]  M. Raffeld,et al.  Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[38]  W. Burns,et al.  A novel chimeric antigen receptor against prostate stem cell antigen mediates tumor destruction in a humanized mouse model of pancreatic cancer. , 2014, Human gene therapy.

[39]  Andrew Sewell,et al.  Structural and kinetic basis for heightened immunogenicity of T cell vaccines , 2005, The Journal of experimental medicine.

[40]  J. Keith Joung,et al.  High frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells , 2013, Nature Biotechnology.

[41]  Rudolf Jaenisch,et al.  Toward eliminating HLA class I expression to generate universal cells from allogeneic donors. , 2013, Blood.

[42]  A. Bagg,et al.  Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. , 2011, The New England journal of medicine.