We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Lentiviral vectors in cancer immunotherapy

    Robyn AA Oldham

    Department of Medical Biophysics, University of Toronto, 27 King's College Circle, Toronto, ON M5S, Canada

    Authors contributed equally

    Search for more papers by this author

    ,
    Elliot M Berinstein

    Department of Medical Biophysics, University of Toronto, 27 King's College Circle, Toronto, ON M5S, Canada

    Authors contributed equally

    Search for more papers by this author

    &
    Jeffrey A Medin

    *Author for correspondence:

    E-mail Address: jmedin@uhnres.utoronto.ca

    Department of Medical Biophysics, University of Toronto, 27 King's College Circle, Toronto, ON M5S, Canada

    Institute of Medical Sciences, University of Toronto, 27 King's College Circle, Toronto, ON M5S, Canada

    University Health Network, Max Bell Research Centre, 101 College Street, Room 5R406, Toronto, ON, M5G 1L7, Canada

    Published Online:https://doi.org/10.2217/imt.14.108

    Basic science advances in cancer immunotherapy have resulted in various treatments that have recently shown success in the clinic. Many of these therapies require the insertion of genes into cells to directly kill them or to redirect the host's cells to induce potent immune responses. Other analogous therapies work by modifying effector cells for improved targeting and enhanced killing of tumor cells. Initial studies done using γ-retroviruses were promising, but safety concerns centered on the potential for insertional mutagenesis have highlighted the desire to develop other options for gene delivery. Lentiviral vectors (LVs) have been identified as potentially more effective and safer alternative delivery vehicles. LVs are now in use in clinical trials for many different types of inherited and acquired disorders, including cancer. This review will discuss current knowledge of LVs and the applications of this viral vector-based delivery vehicle to cancer immunotherapy.

    Papers of special note have been highlighted as: • of interest

    References

    • 1 Morgan RA, Dudley ME, Wunderlich JR et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314(5796), 126–129 (2006).
    • 2 Gene therapy clinical trials worldwide. www.wiley.co.uk/genmed/clinical
    • 3 Wilson JM. Adenoviruses as gene-delivery vehicles. N. Engl. J. Med. 334, 1185–1187 (1996)
    • 4 Janovitz T, Klein IA, Oliveira T et al. High-throughput sequencing reveals principles of adeno-associated virus serotype 2 integration. J. Virol. 87(15), 8559–8568 (2013).
    • 5 Dong JY, Fan PD, Frizzell RA. Quantitative analysis of the packaging capacity of recombinant adeno-associated virus. Hum. Gene Ther. 7(17), 2101–2112 (1996).
    • 6 Malim MH, Emerman M. HIV-1 accessory proteins – ensuring viral survival in a hostile environment. Cell Host Microbe 3(6), 388–398 (2008).
    • 7 Jones IM, Morikawa Y. The molecular basis of HIV capsid assembly. Rev. Med. Virol. 8(2), 87–95 (1998).
    • 8 Finkelshtein D, Werman A, Novick D, Barak S, Rubinstein M. LDL receptor and its family members serve as the cellular receptors for vesicular stomatitis virus. Proc. Natl Acad. Sci. USA 110(18), 7306–7311 (2013).
    • 9 Zennou V, Petit C, Guetard D, Nerhbass U, Montagnier L, Charneau P. HIV-1 genome nuclear import is mediated by a central DNA flap. Cell 101(2), 173–185 (2000).
    • 10 Palù G, Parolin C, Takeuchi Y, Pizzato M. Progress with retroviral gene vectors. Rev. Med. Virol. 10(3), 185–202 (2000).
    • 11 Watanabe S, Temin HM. Encapsidation sequences for spleen necrosis virus, an avian retrovirus, are between the 5’ long terminal repeat and the start of the gag gene. Proc. Natl Acad. Sci. USA 79(19), 5986–5990 (1982).
    • 12 Dull T, Zufferey R, Kelly M et al. A third-generation lentivirus vector with a conditional packaging system. J. Virol. 72(11), 8463–8471 (1998).
    • 13 Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat. Biotechnol. 15(9), 871–875 (1997).
    • 14 Zufferey R, Dull T, Mandel RJ et al. Self-inactivating lentivirus vector for safe and efficient in vivo gene delivery. J. Virol. 72(12), 9873–9880 (1998).
    • 15 Miyoshi H, Blömer U, Takahashi M, Gage FH, Verma IM. Development of a self-inactivating lentivirus vector. J. Virol. 72(10), 8150–8157 (1998).
    • 16 Sirven A, Pflumio F, Zennou V et al. The human immunodeficiency virus type-1 central DNA flap is a crucial determinant for lentiviral vector nuclear import and gene transduction of human hematopoietic stem cells. Blood 96(13), 4103–4110 (2000).
    • 17 Zufferey R, Donello JE, Trono D, Hope TJ. Woodchuck hepatitis virus posttranscriptional regulatory element enhances expression of transgenes delivered by retroviral vectors. J. Virol. 73(4), 2886–2892 (1999).
    • 18 Barry SC, Harder B, Brzezinski M, Flint LY, Seppen J, Osborne WR. Lentivirus vectors encoding both central polypurine tract and posttranscriptional regulatory element provide enhanced transduction and transgene expression. Hum. Gene Ther. 12(9), 1103–1108 (2001).
    • 19 Gossen M, Freundlieb S, Bender G, Müller G, Hillen W, Bujard H. Transcriptional activation by tetracyclines in mammalian cells. Science 268(5218), 1766–1769 (1995).
    • 20 Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 288(5466), 669–672 (2000).
    • 21 Cattoglio C, Facchini G, Sartori D et al. Hot spots of retroviral integration in human CD34+ hematopoietic cells. Blood 110(6), 1770–1778 (2007).
    • 22 Ronen K, Negre O, Roth S et al. Distribution of lentiviral vector integration sites in mice following therapeutic gene transfer to treat β-thalassemia. Mol. Ther. 19(7), 1273–1286 (2011).
    • 23 Rittelmeyer I, Rothe M, Brugman MH et al. Hepatic lentiviral gene transfer is associated with clonal selection, but not with tumor formation in serially transplanted rodents. Hepatology 58(1), 397–408 (2013).
    • 24 Philippe S, Sarkis C, Barkats M et al. Lentiviral vectors with a defective integrase allow efficient and sustained transgene expression in vitro and in vivo. Proc. Natl Acad. Sci. USA 103(47), 17684–17689 (2006).
    • 25 Yáñez-Muñoz RJ, Balaggan KS, MacNeil A et al. Effective gene therapy with nonintegrating lentiviral vectors. Nat. Med. 12(3), 348–353 (2006).
    • 26 Bayer M, Kantor B, Cockrell A et al. A large U3 deletion causes increased in vivo expression from a nonintegrating lentiviral vector. Mol. Ther. 16(12), 1968–1976 (2008).
    • 27 Cornu TI, Cathomen T. Targeted genome modifications using integrase-deficient lentiviral vectors. Mol. Ther. 15(12), 2107–2113 (2007).
    • 28 Kim Y, Kweon J, Kim A et al. A library of TAL effector nucleases spanning the human genome. Nat. Biotechnol. 31(3), 251–258 (2013).
    • 29 Karwacz K, Mukherjee S, Apolonia L et al. Nonintegrating lentivector vaccines stimulate prolonged T-cell and antibody responses and are effective in tumor therapy. J. Virol. 83(7), 3094–3103 (2009).
    • 30 Provasi E, Genovese P, Lombardo A et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat. Med. 18(5), 807–815 (2012).
    • 31 Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337(6096), 816–821 (2012).
    • 32 Koike-Yusa H, Li Y, Tan EP, Velasco-Herrera Mdel C, Yusa K. Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nat. Biotechnol. 32(3), 267–273 (2014).
    • 33 Gilbert LA, Larson MH, Morsut L et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154(2), 442–451 (2013).
    • 34 Fu Y, Foden JA, Khayter C et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 9, 822–826 (2013).
    • 35 Hsu PD, Scott DA, Weinstein JA et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).
    • 36 Pattanayak V, Lin S, Guilinger JP, Ma E, Doudna JA, Liu DR. High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nat. Biotechnol. 31, 839–843 (2013).
    • 37 Cong L, Ran FA, Cox D et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).
    • 38 Kim E, Kim S, Kim DH, Choi BS, Choi IY, Kim JS. Precision genome engineering with programmable DNA-nicking enzymes. Genome Res. 22(7), 1327–1333 (2012).
    • 39 Wang J, Friedman G, Doyon Y et al. Targeted gene addition to a predetermined site in the human genome using a ZFN-based nicking enzyme. Genome Res. 22(7), 1316–1326 (2012).
    • 40 Ramirez CL, Certo MT, Mussolino C et al. Engineered zinc finger nickases induce homology-directed repair with reduced mutagenic effects. Nucleic Acids Res. 40(12), 5560–5568 (2012).
    • 41 Holkers M, Maggio I, Liu J et al. Differential integrity of TALE nuclease genes following adenoviral and lentiviral vector gene transfer into human cells. Nucleic Acids Res. 41(5), e63 (2013).
    • 42 Black ME, Newcomb TG, Wilson HM, Loeb LA. Creation of drug-specific herpes simplex virus type 1 thymidine kinase mutants for gene therapy. Proc. Natl Acad. Sci. USA 93(8), 3525–3529 (1996).
    • 43 Casucci M, Bondanza A. Suicide gene therapy to increase the safety of chimeric antigen receptor-redirected T lymphocytes. J. Cancer 2, 378–382 (2001).
    • 44 Berger C, Flowers ME, Warren EH, Riddell SR. Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation. Blood 107(6), 2294–2302 (2006).
    • 45 Sato T, Neschadim A, Konrad AM, Fowler DH, Lavie A, Medin JA. Engineered human tmpk/AZT as a novel enzyme/prodrug axis for suicide gene therapy. Mol. Ther. 15(5), 962–970 (2007).• This paper presents the development of a novel cell-fate control (aka ‘suicide’) system based on a minimally modified human kinase that converts the prodrug azidothymidine to a cytotoxic version. Prodrug conversion is efficient and cell killing is effected through DNA chain termination and mitochondria disruption effects.
    • 46 Neschadim A, Wang JC, Sato T, Fowler DH, Lavie A, Medin JA. Cell fate control gene therapy based on engineered variants of human deoxycytidine kinase. Mol. Ther. 20(5), 1002–1013 (2012).• A human dCK based cell-fate control (aka ‘suicide’) system is described here, which is capable of inducing apoptosis in transduced cells exposed to a variety of normally nontoxic prodrugs such as bromovinyl-deoxyuridine or l-deoxythymidine. As this modified safety enzyme adds the first phosphate to the prodrug, it is also effective in imaging studies.
    • 47 Scaife M, Pacienza N, Au BC et al. Engineered human Tmpk fused with truncated cell-surface markers: versatile cell-fate control safety cassettes. Gene Ther. 20(1), 24–34 (2013).
    • 48 Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 392(6673), 245–252 (1998).
    • 49 Nestle FO, Alijagic S, Gilliet M et al. Vaccination of melanoma patients with peptide or tumor lysate-pulsed dendritic cells. Nat. Med. 4(3), 328–332 (1998).
    • 50 Dawson NA, Roesch EE. Sipuleucel-T and immunotherapy in the treatment of prostate cancer. Expert Opin. Biol. Ther. 14(5), 709–719 (2014).
    • 51 Breckpot K, Dullaers M, Bonehill A et al. Lentivirally transduced dendritic cells as a tool for cancer immunotherapy. J. Gene Med. 5(8), 654–667 (2003).
    • 52 Jeon, MS, Atfield, A., Venuprasad, K et al. Essential role of the E3 ubiquitin ligase Cbl-b in T cell anergy induction. Immunity 21(2), 167–177 (2004).
    • 53 Nurieva, R., Thomas, S., Nguyen, T et al. T-cell tolerance or function is determined by combinatorial costimulatory signals. EMBO J. 25(11), 2623–2633 (2006).
    • 54 Curtsinger, JM, Johnson, CM, Mescher, MF. CD8 T cell clonal expansion and development of effector function require prolonged exposure to antigen, costimulation, and signal 3 cytokine. J. Immunol. 171(10), 5165–5171 (2003).
    • 55 Karwacz, K, Bricogne, C, MacDonald, D et al. PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8(+) T cells. EMBO Mol. Med. 3(10), 581–592 (2011).
    • 56 Pen, JJ, Keersmaecker, BD, Heirman, C et al. (2014). Interference with PD-L1/PD-1 co-stimulation during antigen presentation enhances the multifunctionality of antigen-specific T cells. Gene Ther. 21(3), 262–271 (2014).
    • 57 Fife, BT, Pauken, KE, Eagar, TN et al. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat. Immunol. 10(11), U1185–U1170 (2009).
    • 58 Escors, D, Lopes, L, Lin, R et al. Targeting dendritic cell signaling to regulate the response to immunization. Blood 111(6), 3050–3061 (2008).
    • 59 Felizardo TC, Wang JCM, Mcgray RA et al. Differential immune responses mediated by adenovirus- and lentivirus-transduced DCs in a HER-2/neu overexpressing tumor model. Gene Ther. 18(10), 986–995 (2011).
    • 60 Brown BD, Sitia G, Annoni A et al. In vivo administration of lentiviral vectors triggers a type I interferon response that restricts hepatocyte gene transfer and promotes vector clearance. Blood 109(7), 2797–2805 (2007).
    • 61 Breckpot K, Escors D, Arce F et al. HIV-1 lentiviral vector immunogenicity is mediated by Toll-Like Receptor 3 (TLR3) and TLR7. J. Virol. 84(11), 5627–5636 (2010).
    • 62 Metharom P, Ellem KA, Schmidt C, Wei MQ. Lentiviral vector-mediated tyrosinase-related protein 2 gene transfer to dendritic cells for the therapy of melanoma. Hum. Gene Ther. 12(18), 2203–2213 (2001).
    • 63 Metharom P, Ellem KA, Wei MQ. Gene transfer to dendritic cells induced a protective immunity against melanoma. Cell Mol. Immunol. 2(4), 281–288 (2005).
    • 64 Koya RC, Kimura T, Ribas A et al. Lentiviral vector-mediated autonomous differentiation of mouse bone marrow cells into immunologically potent dendritic cell vaccines. Mol. Ther. 15(5), 971–980 (2007).
    • 65 Wang B, He J, Liu C, Chang LJ. An effective cancer vaccine modality: Lentiviral modification of dendritic cells expressing multiple cancer-specific antigens. Vaccine 24(17), 3477–3489 (2006).
    • 66 Mossoba ME, Walia JS, Rasaiah VI et al. Tumor protection following vaccination with low doses of lentivirally transduced DCs expressing the self-antigen erbB2. Mol. Ther. 16(3), 607–617 (2008).
    • 67 Xiao L, Joo K-I, Lim M, Wang P. Dendritic cell directed vaccination with a lentivector encoding PSCA for prostate cancer in mice. PLoS ONE 7(11) e48866, (2012).• Describes lentiviral vector (LV)-transduced dendritic cells (DCs) engineered to express the prostate cancer tumor-associated antigen PSCA. Such LV transduced DCs were able to induce a cancer-specific immune response and provide significant tumor suppression.
    • 68 Romani N, Gruner S, Brang D et al. Proliferating dendritic cell progenitors in human blood. J. Exp. Med. 180(1), 83–93 (1994).
    • 69 Sallusto F, Lanzavecchia A. Efficient presentation of soluble-antigen by cultured human dendritic cells is maintained by granulocyte-macrophage colony-stimulating factor plus interleukin-4 and down-regulated by Tumor-Necrosis-Factor-alpha. J. Exp. Med. 179(4), 1109–1118 (1994).
    • 70 Reid CDL, Stackpoole A, Meager A, Tikerpae J. Interactions of tumor-necrosis-factor with granulocyte-macrophage colony-stimulating factor and other cytokines in the regulation of dendritic cell-growth in vitro from early bipotent CD34+ progenitors in human bone-marrow. J. Immunol. 149(8), 2681–2688 (1992).
    • 71 Salmon P, Arrighi JF, Piguet V et al. Transduction of CD34+ cells with lentiviral vectors enables the production of large quantities of transgene-expressing immature and mature dendritic cells. J. Gene Med. 3(4), 311–320 (2001).
    • 72 Chinnasamy N, Chinnasamy D, Toso JF et al. Efficient gene transfer to human peripheral blood monocyte-derived dendritic cells using human immunodeficiency virus type 1-based lentiviral vectors. Hum. Gene Ther. 11(13), 1901–1909 (2000).
    • 73 Senju S, Haruta M, Matsumura K et al. Generation of dendritic cells and macrophages from human induced pluripotent stem cells aiming at cell therapy. Gene Ther. 18(9), 874–883 (2011).• Describes the LV-mediated transformation of donor dermal fibroblasts by transduction of OCT3/4, SOX2, c-MYC and Klf4 transgenes and their subsequent differentiation into functional DCs.
    • 74 Zhan XC, Dravid G, Ye ZH et al. Functional antigen-presenting leucocytes derived from human embryonic stem cells in vitro. Lancet 364(9429), 163–171 (2004).
    • 75 Kamath AT, Henri S, Battye F, Tough DF, Shortman K. Developmental kinetics and lifespan of dendritic cells in mouse lymphoid organs. Blood 100(5), 1734–1741 (2002).
    • 76 Wang JCM, Felizardo TC, Au BCY, Fowler DH, Dekaban GA, Medin JA. Engineering lentiviral vectors for modulation of dendritic cell apoptotic pathways. Virol. J. 10, 240 (2013).
    • 77 He YK, Zhang JY, Donahue C, Falo LD. Skin-derived dendritic cells induce potent CD8(+) T cell immunity in recombinant lentivector-mediated genetic immunization. Immunity 24(5), 643–656 (2006).
    • 78 Negri DRM, Michelini Z, Baroncelli S et al. Successful immunization with a single injection of non-integrating lentiviral vector. Mol. Ther. 15(9), 1716–1723 (2007).
    • 79 Dai B, Yang L, Yang H, Hu B, Baltimore D, Wang P. HIV-1 Gag-specific immunity induced by a lentivector-based vaccine directed to dendritic cells. Proc. Natl Acad. Sci. USA 106(48), 20382–20387 (2009).
    • 80 Hirano N, Takahashi T, Ohtake S et al. Expression of costimulatory molecules in human leukemias. Leukemia 10(7), 1168–1176 (1996).
    • 81 Brunda MJ, Luistro L, Warrier RR et al. Anti-tumor and anti-metastatic activity of interleukin-12 against murine tumors. J. Exp. Med. 178(4), 1223–1230 (1993).
    • 82 Cohen J. IL-12 deaths: explanation and a puzzle. Science 270(5238), 908–908 (1995).
    • 83 Gollob JA, Mier JW, Veenstra K et al. Phase I trial of twice-weekly intravenous interleukin 12 in patients with metastatic renal cell cancer or malignant melanoma: Ability to maintain IFN-gamma induction is associated with clinical response. Clin. Cancer Res. 6(5), 1678–1692 (2000).
    • 84 Haicheur N, Escudier B, Dorval T et al. Cytokines and soluble cytokine receptor induction after IL-12 administration in cancer patients. Clin. Exp. Immunol. 119(1), 28–37 (2000).
    • 85 Portielje JEA, Lamers CHJ, Kruit WHJ et al. Repeated administrations of interleukin (IL)-12 are associated with persistently elevated plasma levels of IL-10 and declining IFN-gamma, tumor necrosis factor-alpha, IL-6, and IL-8 responses. Clin. Cancer Res. 9(1), 76–83 (2003).
    • 86 Pizzoferrato E, Chu NR, Hawley TS et al. Enhanced immunogenicity of B cell lymphoma genetically engineered to express both B7–1 and interleukin-12. Hum. Gene Ther. 8(18), 2217–2228 (1997).
    • 87 Labbe A, Nelles M, Walia J et al. IL-12 immunotherapy of murine leukaemia: comparison of systemic versus gene modified cell therapy. J. Cell Mol. Med. 13(8B), 1962–1976 (2009).• Describes the how leukemia cells themselves can be turned into anticancer vaccines by LV-mediated transduction of the cDNA encoding a fusion form of IL-12 subunits. Importantly, here it was shown that what is vital to the success of this approach is the threshold of fused IL-12 produced by the transduced tumor cells; above a certain level all animals were protected from tumor challenge, below that threshold no animals were protected.
    • 88 Wei LZ, Xu Y, Nelles ME et al. Localized interleukin-12 delivery for immunotherapy of solid tumours. J. Cell Mol. Med. 17(11), 1465–1474 (2013).
    • 89 Stripecke R, Cardoso AA, Pepper KA et al. Lentiviral vectors for efficient delivery of CD80 and granulocyte-macrophage-colony-stimulating factor in human acute lymphoblastic leukemia and acute myeloid leukemia cells to induce anti-leukemic immune responses. Blood 96(4), 1317–1326 (2000).
    • 90 Chan L, Hardwick N, Darling D et al. IL-2/B7.1 (CD80) fusagene transduction of AML blasts by a self-inactivating lentiviral vector stimulates T cell responses in vitro: a strategy to generate whole cell vaccines for AML. Mol. Ther. 11(1), 120–131 (2005).
    • 91 Cortes JE, Kantarjian HM, O'Brien S et al. A pilot study of interleukin-2 for adult patients with acute myelogenous leukemia in first complete remission. Cancer 85(7), 1506–1513 (1999).
    • 92 Suhoski MM, Golovina TN, Aqui NA et al. Engineering artificial antigen-presenting cells to express a diverse array of co-stimulatory molecules. Mol. Ther. 15(5), 981–988 (2007).
    • 93 Dembic Z, Haas W, Weiss S et al. Transfer of specificity by murine alpha-T-cell and beta-T-cell receptor genes. Nature 320(6059), 232–238 (1986).
    • 94 Yang S, Cohen CJ, Peng PD et al. Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition. Gene Ther. 15(21), 1411–1423 (2008).
    • 95 Clinicaltrials.gov NCT01795976. https://clinicaltrials.gov/ct2/show/NCT01795976
    • 96 Clinicaltrials.gov NCT00910650. https://clinicaltrials.gov/ct2/show/NCT00910650
    • 97 Bendle GM, Linnemann C, Hooijkaas AI et al. Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat. Med. 16(5), 565–570 (2010).
    • 98 Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365(8), 725–733 (2011).• Describes the ability of a CD19-directed chimeric antigen receptor in autologous T cells to treat patients with chronic lymphocytic leukemia. Chimeric antigen receptor T cells were able to provide a robust and specific immune response against CD19+ B-cell malignancies, clearing them from patients.
    • 99 Kochenderfer JN, Dudley ME, Feldman SA et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119(12), 2709–2720 (2012).
    • 100 Finney HM, Akbar AN, Lawson ADG. Activation of resting human primary T cells with chimeric receptors: Costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J. Immunol. 172(1), 104–113 (2004).
    • 101 Imai C, Mihara K, Andreansky M et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18(4), 676–684 (2004).
    • 102 Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma-subunit or zeta-subunit of the immunoglobulin and T-cell receptors. Proc. Natl Acad. Sci. USA 90(2), 720–724 (1993).
    • 103 Boissel L, Betancur-Boissel M, Lu W et al. Retargeting NK-92 cells by means of CD19-and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology 2(10), e26527 (2013).
    • 104 Frigault MJ, June CH. Predicting cytokine storms: it's about density. Blood 118(26), 6724–6726 (2011).
    • 105 Di Stasi A, Tey S-K, Dotti G et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365(18), 1673–1683 (2011).
    • 106 Barrett DM, Zhao Y, Liu X et al. Treatment of advanced leukemia in mice with mRNA engineered T cells. Hum. Gene Ther. 22(12), 1575–1586 (2011).
    • 107 Maus MV, Haas AR, Beatty GL et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 1(1), 26–31 (2013).