Chimeric antigen receptor- (CAR-) based immunotherapy has been under development for almost 25 years, over which period it has progressed from a new but cumbersome technology to an emerging therapeutic modality for malignant disease. The approach involves the genetic engineering of fusion receptors (CARs) that couple the HLA-independent binding of cell surface target molecules to the delivery of a tailored activating signal to host immune cells. Engineered CARs are delivered most commonly to peripheral blood T cells using a range of vector systems, most commonly integrating viral vectors. Preclinical refinement of this approach has proceeded over several years to the point that clinical testing is now being undertaken at several centres, using increasingly sophisticated and therapeutically successful genetic payloads. This paper considers several aspects of the pre-clinical and clinical development of CAR-based immunotherapy and how this technology is acquiring an increasing niche in the treatment of both solid and haematological malignancies. 1. Introduction to Chimeric Antigen Receptor Technology Tumour immunotherapy is one of the oldest branches of clinical immunology and has a long but checkered history. The overriding goal is to deploy the multiplicity of available immune effector mechanisms against tumour cells, but not against healthy counterparts. Unfortunately however, several obstacles render this a very difficult goal. Although hundreds of so-called tumour antigens have been identified, these are generally derived from self and thus are poorly immunogenic. Furthermore, tumours use several mechanisms to render themselves hostile to the initiation and propagation of immune attack. These immune subversive strategies include reduced expression of HLA molecules and target antigens coupled with the establishment of a microenvironment in which inhibitory cytokines and leukocytes abound (recently reviewed in [1]). Indeed, cancer cells can even dedifferentiate to evade detection in response to inflammatory cues provided by tumour-specific T cells [2]. Consequently, it is not surprising that attempts to harness tumour-specific T cells using a succession of vaccination-based approaches have not achieved striking success [3]. Recent developments using genetically enhanced T cells have led to renewed optimism in the quest to launch immune attack against malignant disease. One increasingly prominent technology in this arena involves the use of the so-called chimeric antigen receptors, or CARs. These bespoke fusion receptors are engineered as chimeric cDNAs
References
[1]
K. T?pfer, S. Kempe, N. Müller et al., “Tumor evasion from T cell surveillance,” Journal of Biomedicine and Biotechnology, vol. 2011, Article ID 918471, 2011.
[2]
J. Landsberg, J. Kohlmeyer, M. Renn et al., “Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation,” Nature, vol. 490, no. 7420, pp. 412–416, 2012.
[3]
S. A. Rosenberg, J. C. Yang, and N. P. Restifo, “Cancer immunotherapy: moving beyond current vaccines,” Nature Medicine, vol. 10, no. 9, pp. 909–915, 2004.
[4]
R. A. Willemsen, C. Ronteltap, P. Chames, R. Debets, and R. L. H. Bolhuis, “T cell retargeting with MHC class I-restricted antibodies: the CD28 costimulatory domain enhances antigen-specific cytotoxicity and cytokine production,” Journal of Immunology, vol. 174, no. 12, pp. 7853–7858, 2005.
[5]
G. Stewart-Jones, A. Wadle, A. Hombach et al., “Rational development of high-affinity T-cell receptor-like antibodies,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 14, pp. 5784–5788, 2009.
[6]
D. V. Tassev, M. Cheng, and N.-K. Cheung, “Retargeting NK92 cells using an HLA-A2-restricted, EBNA3C-specific chimeric antigen receptor,” Cancer Gene Therapy, vol. 19, no. 2, pp. 84–100, 2012.
[7]
Y. Kuwana, Y. Asakura, N. Utsunomiya et al., “Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions,” Biochemical and Biophysical Research Communications, vol. 149, no. 3, pp. 960–968, 1987.
[8]
G. Gross, G. Gorochov, T. Waks, and Z. Eshhar, “Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity,” Transplantation Proceedings, vol. 21, no. 1 I, pp. 127–130, 1989.
[9]
G. Gross, T. Waks, and Z. Eshhar, “Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity,” Proceedings of the National Academy of Sciences of the United States of America, vol. 86, no. 24, pp. 10024–10028, 1989.
[10]
Z. Eshhar, T. Waks, G. Gross, and D. G. Schindler, “Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the γ or ζ subunits of the immunoglobulin and T-cell receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 2, pp. 720–724, 1993.
[11]
U. Altenschmidt, R. Kahl, D. Moritz et al., “Cytolysis of tumor cells expressing the Neu/erbB-2, erbB-3, and erbB-4 receptors by genetically targeted naive T lymphocytes,” Clinical Cancer Research, vol. 2, no. 6, pp. 1001–1008, 1996.
[12]
A. Muniappan, B. Banapour, J. Lebkowski, and S. Talib, “Ligand-mediated cytolysis of tumor cells: use of heregulin-ζ chimeras to redirect cytotoxic T lymphocytes,” Cancer Gene Therapy, vol. 7, no. 1, pp. 128–134, 2000.
[13]
C. R. J. Pameijer, A. Navanjo, B. Meechoovet et al., “Conversion of a tumor-binding peptide identified by phage display to a functional chimeric T cell antigen receptor,” Cancer Gene Therapy, vol. 14, no. 1, pp. 91–97, 2007.
[14]
D. M. Davies, J. Foster, S. J. C. van der Stegen et al., “Flexible targeting of ErbB dimers that drive tumorigenesis by using genetically engineered T cells,” Molecular Medicine, vol. 18, no. 4, pp. 565–576, 2012.
[15]
J. Scholler, T. L. Brady, G. Binder-Scholl et al., “Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells,” Science Translational Medicine, vol. 4, no. 132, Article ID 132ra53, 2012.
[16]
T. Zhang, M.-R. Wu, and C. L. Sentman, “An NKp30-based chimeric antigen receptor promotes T cell effector functions and antitumor efficacy in vivo,” Journal of Immunology, vol. 189, no. 5, pp. 2290–2299, 2012.
[17]
Z. Sharifzadeh, F. Rahbarizadeh, M. A. Shokrgozar, et al., “Genetically engineered T cells bearing chimeric nanoconstructed receptors harboring TAG-72-specific camelid single domain antibodies as targeting agents,” Cancer Letters. In press.
[18]
K. Urbanska, E. Lanitis, M. Poussin et al., “A universal strategy for adoptive immunotherapy of cancer through use of a novel T-cell antigen receptor,” Cancer Research, vol. 72, no. 7, pp. 1844–1852, 2012.
[19]
S. O. Ang, C. Hartline, T. Mi, et al., “Generating a chimeric antigen receptor to redirect T-cell specificity after infusion,” Molecular Therapy, vol. 19, no. S137, abstract 353, 2011.
[20]
K. Tamada, D. Geng, Y. Sakoda, N. Bansal, R. Srivastava, and E. Davila, “Redirecting gene-modified T cells toward various cancer types using tagged antibodies,” Clinical Cancer Research. In press.
[21]
M. Chmielewski, A. Hombach, C. Heuser, G. P. Adams, and H. Abken, “T cell activation by antibody-like immunoreceptors: increase in affinity of the single-chain fragment domain above threshold does not increase T cell activation against antigen-positive target cells but decreases selectivity,” Journal of Immunology, vol. 173, no. 12, pp. 7647–7653, 2004.
[22]
S. Wilkie, G. Picco, J. Foster et al., “Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor,” Journal of Immunology, vol. 180, no. 7, pp. 4901–4909, 2008.
[23]
M. H. Kershaw, J. A. Westwood, L. L. Parker et al., “A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer,” Clinical Cancer Research, vol. 12, no. 20, pp. 6106–6115, 2006.
[24]
C. H. J. Lamers, R. Willemsen, P. Van Elzakker et al., “Immune responses to transgene and retroviral vector in patients treated with ex vivo-engineered T cells,” Blood, vol. 117, no. 1, pp. 72–82, 2011.
[25]
D. Moritz and B. Groner, “A spacer region between the single chain antibody-and the CDS ζ-chain domain of chimeric T cell receptor components is required for efficient ligand binding and signaling activity,” Gene Therapy, vol. 2, no. 8, pp. 539–546, 1995.
[26]
R. D. Guest, R. E. Hawkins, N. Kirillova et al., “The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens,” Journal of Immunotherapy, vol. 28, no. 3, pp. 203–211, 2005.
[27]
S. E. James, P. D. Greenberg, M. C. Jensen et al., “Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane,” Journal of Immunology, vol. 180, no. 10, pp. 7028–7038, 2008.
[28]
A. Hombach, A. A. Hombach, and H. Abken, “Adoptive immunotherapy with genetically engineered T cells: modification of the IgG1 Fc spacer domain in the extracellular moiety of chimeric antigen receptors avoids off-target activation and unintended initiation of an innate immune response,” Gene Therapy, vol. 17, no. 10, pp. 1206–1213, 2010.
[29]
J. S. Bridgeman, R. E. Hawkins, S. Bagley, M. Blaylock, M. Holland, and D. E. Gilham, “The optimal antigen response of chimeric antigen receptors harboring the CD3ζ transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex,” Journal of Immunology, vol. 184, no. 12, pp. 6938–6949, 2010.
[30]
B. A. Irving and A. Weiss, “The cytoplasmic domain of the T cell receptor ζ chain is sufficient to couple to receptor-associated signal transduction pathways,” Cell, vol. 64, no. 5, pp. 891–901, 1991.
[31]
N. M. Haynes, M. B. Snook, J. A. Trapani et al., “Redirecting mouse CTL against colon carcinoma: superior signaling efficacy of single-chain variable domain chimeras containing TCR-ζ vs FcεRI-γ,” Journal of Immunology, vol. 166, no. 1, pp. 182–187, 2001.
[32]
A. Krause, H. F. Guo, J. B. Latouche, C. Tan, N. K. V. Cheung, and M. Sadelain, “Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes,” Journal of Experimental Medicine, vol. 188, no. 4, pp. 619–626, 1998.
[33]
H. M. Finney, A. D. G. Lawson, C. R. Bebbington, and A. N. C. Weir, “Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product,” Journal of Immunology, vol. 161, no. 6, pp. 2791–2797, 1998.
[34]
J. Maher, R. J. Brentjens, G. Gunset, I. Rivière, and M. Sadelain, “Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ/CD28 receptor,” Nature Biotechnology, vol. 20, no. 1, pp. 70–75, 2002.
[35]
N. M. Haynes, J. A. Trapani, M. W. L. Teng et al., “Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors,” Blood, vol. 100, no. 9, pp. 3155–3163, 2002.
[36]
N. M. Haynes, J. A. Trapani, M. W. L. Teng et al., “Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation,” Journal of Immunology, vol. 169, no. 10, pp. 5780–5786, 2002.
[37]
C. Carpenito, M. C. Milone, R. Hassan et al., “Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 9, pp. 3360–3365, 2009.
[38]
H. M. Finney, A. N. Akbar, and A. D. G. Lawson, “Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCRζ chain,” Journal of Immunology, vol. 172, no. 1, pp. 104–113, 2004.
[39]
R. J. Brentjens, E. Santos, Y. Nikhamin et al., “Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts,” Clinical Cancer Research, vol. 13, no. 18, pp. 5426–5435, 2007.
[40]
C. Imai, K. Mihara, M. Andreansky et al., “Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia,” Leukemia, vol. 18, no. 4, pp. 676–684, 2004.
[41]
D.-G. Song, Q. Ye, M. Poussin, G. M. Harms, M. Figini, and D. J. Powell Jr., “CD27 costimulation augments the survival and antitumor activity of redirected human T cells in vivo,” Blood, vol. 119, no. 3, pp. 696–706, 2012.
[42]
B. Altvater, S. Landmeier, S. Pscherer et al., “2B4 (CD244) signaling via chimeric receptors costimulates tumor-antigen specific proliferation and in vitro expansion of human T cells,” Cancer Immunology, Immunotherapy, vol. 58, no. 12, pp. 1991–2001, 2009.
[43]
S. Guedan, C. Carpenito, and S. E. McGettigan, “Redirection of TH17 cells with a CAR containing the ICOS costimulatory domain enhances function, antitumor Activity and persistence of TH17 cells,” Molecular Therapy, vol. 20, no. S130, abstract 329, 2012.
[44]
A. Loskog, V. Giandomenico, C. Rossig, M. Pule, G. Dotti, and M. K. Brenner, “Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells,” Leukemia, vol. 20, no. 10, pp. 1819–1828, 2006.
[45]
H. Koehler, D. Kofler, A. Hombach, and H. Abken, “CD28 costimulation overcomes transforming growth factor-β-mediated repression of proliferation of redirected human CD4+ and CD8+ T cells in an antitumor cell attack,” Cancer Research, vol. 67, no. 5, pp. 2265–2273, 2007.
[46]
D. M. Kofler, M. Chmielewski, G. Rappl et al., “CD28 costimulation impairs the efficacy of a redirected T-cell antitumor attack in the presence of regulatory T cells which can be overcome by preventing lck activation,” Molecular Therapy, vol. 19, no. 4, pp. 760–767, 2011.
[47]
M. C. Milone, J. D. Fish, C. Carpenito et al., “Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo,” Molecular Therapy, vol. 17, no. 8, pp. 1453–1464, 2009.
[48]
C. M. Kowolik, M. S. Topp, S. Gonzalez 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, vol. 66, no. 22, pp. 10995–11004, 2006.
[49]
M. Kalos, B. L. Levine, D. L. Porter et al., “T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia,” Science Translational Medicine, vol. 3, no. 95, Article ID 95ra73, 2011.
[50]
D. L. Porter, B. L. Levine, M. Kalos, A. Bagg, and C. H. June, “Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia,” New England Journal of Medicine, vol. 365, no. 8, pp. 725–733, 2011.
[51]
B. Savoldo, C. A. Ramos, E. Liu et al., “CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients,” Journal of Clinical Investigation, vol. 121, no. 5, pp. 1822–1826, 2011.
[52]
S. Wilkie, M. C. I. Van Schalkwyk, S. Hobbs et al., “Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling,” Journal of Clinical Immunology, vol. 32, no. 5, pp. 1059–1070, 2012.
[53]
J. Sun, G. Dotti, L. E. Huye et al., “T cells expressing constitutively active Akt resist multiple tumor-associated inhibitory mechanisms,” Molecular Therapy, vol. 18, no. 11, pp. 2006–2017, 2010.
[54]
N. Kunii, Y. Zhao, S. Jiang, et al., “Enhanced function of redirected human T cells expressing linker for activation of T-cells that is resistant to ubiquitylation,” Human Gene Therapy. In press.
[55]
T. L. Geiger, P. Nguyen, D. Leitenberg, and R. A. Flavell, “Integrated src kinase and costimulatory activity enhances signal transduction through single-chain chimeric receptors in T lymphocytes,” Blood, vol. 98, no. 8, pp. 2364–2371, 2001.
[56]
A. A. Hombach, J. Heiders, M. Foppe, M. Chmielewski, and H. Abken, “OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4+ T cells,” Oncoimmunology, vol. 1, pp. 458–466, 2012.
[57]
M. A. Pulè, K. C. Straathof, G. Dotti, H. E. Heslop, C. M. Rooney, and M. K. Brenner, “A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells,” Molecular Therapy, vol. 12, no. 5, pp. 933–941, 2005.
[58]
X. S. Zhong, M. Matsushita, J. Plotkin, I. Riviere, and M. Sadelain, “Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3 kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication,” Molecular Therapy, vol. 18, no. 2, pp. 413–420, 2010.
[59]
J. Wang, M. Jensen, Y. Lin et al., “Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains,” Human Gene Therapy, vol. 18, no. 8, pp. 712–725, 2007.
[60]
M. T. Stephan, V. Ponomarev, R. J. Brentjens et al., “T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection,” Nature Medicine, vol. 13, no. 12, pp. 1440–1449, 2007.
[61]
S. P. Zehntner, M. Brisebois, E. Tran, T. Owens, and S. Fournier, “Constitutive expression of a costimulatory ligand on antigen-presenting cells in the nervous system drives demyelinating disease,” FASEB Journal, vol. 17, pp. 1910–1912, 2003.
[62]
K. Murata, M. Nose, L. C. Ndhlovu, T. Sato, K. Sugamura, and N. Ishii, “Constitutive OX40/OX40 ligand interaction induces autoimmune-like diseases,” Journal of Immunology, vol. 169, no. 8, pp. 4628–4636, 2002.
[63]
G. F. Salinas, T. Cantaert, and M. Nolte, “Constitutive co-stimulation by B cells is sufficient to trigger T cell-driven autoimmune disease,” Annals of the Rheumatic Diseases, vol. 69, no. A71, 2010.
[64]
C. Imai, S. Iwamoto, and D. Campana, “Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells,” Blood, vol. 106, no. 1, pp. 376–383, 2005.
[65]
B. Altvater, S. Landmeier, S. Pscherer et al., “2B4 (CD244) signaling by recombinant antigen-specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells,” Clinical Cancer Research, vol. 15, no. 15, pp. 4857–4866, 2009.
[66]
C. J. Denman, V. V. Senyukov, S. S. Somanchi et al., “Membrane-bound IL-21 promotes sustained Ex Vivo proliferation of human natural killer cells,” PLoS ONE, vol. 7, no. 1, Article ID e30264, 2012.
[67]
N. Lapteva, A. G. Durett, J. Sun et al., “Large-scale ex vivo expansion and characterization of natural killer cells for clinical applications,” Cytotherapy, vol. 14, no. 9, pp. 1131–1143, 2012.
[68]
M. Rischer, S. Pscherer, S. Duwe, J. Vormoor, H. Jürgens, and C. Rossig, “Human γδ T cells as mediators of chimaeric-receptor redirected anti-tumour immunity,” British Journal of Haematology, vol. 126, no. 4, pp. 583–592, 2004.
[69]
H. Torikai, A. Reik, P.-Q. Liu 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,” Blood, vol. 119, no. 24, pp. 5697–5705, 2012.
[70]
M. E. Dudley, J. R. Wunderlich, P. F. Robbins et al., “Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes,” Science, vol. 298, no. 5594, pp. 850–854, 2002.
[71]
M. E. Dudley, J. C. Yang, R. Sherry et al., “Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens,” Journal of Clinical Oncology, vol. 26, no. 32, pp. 5233–5239, 2008.
[72]
A. Barber, A. Rynda, and C. L. Sentman, “Chimeric NKG2D expressing T cells eliminate immunosuppression and activate immunity within the ovarian tumor microenvironment,” Journal of Immunology, vol. 183, no. 11, pp. 6939–6947, 2009.
[73]
P. Spear, A. Barber, A. Rynda-Apple, and C. L. Sentman, “Chimeric antigen receptor T cells shape myeloid cell function within the tumor microenvironment through IFN-γ and GM-CSF,” Journal of Immunology, vol. 188, no. 12, pp. 6389–6398, 2012.
[74]
S. P. Kerkar, P. Muranski, A. Kaiser et al., “Tumor-specific CD8+ T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts,” Cancer Research, vol. 70, no. 17, pp. 6725–6734, 2010.
[75]
D. Chinnasamy, Z. Yu, S. P. Kerkar et al., “Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice,” Clinical Cancer Research, vol. 18, no. 6, pp. 1672–1683, 2012.
[76]
H. J. Pegram, J. C. Lee, E. G. Hayman et al., “Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning,” Blood, vol. 119, no. 18, pp. 4133–4141, 2012.
[77]
M. Chmielewski, C. Kopecky, A. A. Hombach, and H. Abken, “IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression,” Cancer Research, vol. 71, no. 17, pp. 5697–5706, 2011.
[78]
S. P. Kerkar, R. S. Goldszmid, P. Muranski et al., “IL-12 triggers a programmatic change in dysfunctional myeloid-derived cells within mouse tumors,” Journal of Clinical Investigation, vol. 121, no. 12, pp. 4746–4757, 2011.
[79]
A. C. Parente-Pereira, J. Burnet, D. Ellison et al., “Trafficking of CAR-engineered human T cells following regional or systemic adoptive transfer in SCID beige mice,” Journal of Clinical Immunology, vol. 31, no. 4, pp. 710–718, 2011.
[80]
R. J. Brentjens, I. Rivière, J. H. Park et al., “Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias,” Blood, vol. 118, no. 18, pp. 4817–4828, 2011.
[81]
M. H. Kershaw, G. Wang, J. A. Westwood et al., “Redirecting migration of T cells to chemokine secreted from tumors by genetic modification with CXCR2,” Human Gene Therapy, vol. 13, no. 16, pp. 1971–1980, 2002.
[82]
A. Di Stasi, B. De Angelis, C. M. Rooney et al., “T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting CD30 have improved homing and antitumor activity in a Hodgkin tumor model,” Blood, vol. 113, no. 25, pp. 6392–6402, 2009.
[83]
E. K. Moon, C. Carpenito, J. Sun et al., “Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor,” Clinical Cancer Research, vol. 17, no. 14, pp. 4719–4730, 2011.
[84]
A. S. Y. Lo, J. R. Taylor, F. Farzaneh, D. M. Kemeny, N. J. Dibb, and J. Maher, “Harnessing the tumour-derived cytokine, CSF-1, to co-stimulate T-cell growth and activation,” Molecular Immunology, vol. 45, no. 5, pp. 1276–1287, 2008.
[85]
J. H. Pinthus, T. Waks, V. Malina et al., “Adoptive immunotherapy of prostate cancer bone lesions using redirected effector lymphocytes,” Journal of Clinical Investigation, vol. 114, no. 12, pp. 1774–1781, 2004.
[86]
R. Sackstein, J. S. Merzaban, D. W. Cain et al., “Ex vivo glycan engineering of CD44 programs human multipotent mesenchymal stromal cell trafficking to bone,” Nature Medicine, vol. 14, no. 2, pp. 181–187, 2008.
[87]
S. N. Robinson, P. J. Simmons, M. W. Thomas et al., “Ex vivo fucosylation improves human cord blood engraftment in NOD-SCID IL-2Rγnull mice,” Experimental Hematology, vol. 40, no. 6, pp. 445–456, 2012.
[88]
R. Brentjens, R. Yeh, Y. Bernal, I. Riviere, and M. Sadelain, “Treatment of chronic lymphocytic leukemia with genetically targeted autologous t cells: case report of an unforeseen adverse event in a phase i clinical trial,” Molecular Therapy, vol. 18, no. 4, pp. 666–668, 2010.
[89]
R. A. Morgan, J. C. Yang, M. Kitano, M. E. Dudley, C. M. Laurencot, and S. A. Rosenberg, “Case report of a serious adverse event following the administration of t cells transduced with a chimeric antigen receptor recognizing ERBB2,” Molecular Therapy, vol. 18, no. 4, pp. 843–851, 2010.
[90]
F. Thistlethwaite, D. Rothwell, A. Byatte, et al., “A CRUK phase I trial of adoptive transfer of autologous tumour antigen-specific T cells with pre-conditioning chemotherapy and intravenous IL2 in patients with advanced CEA positive tumours,” UK NCRI Conference, Abstract LB14, 2010, http://www.ncri.org.uk/ncriconference/2010abstracts/abstracts/LB14.htm.
[91]
A. Murphy, J. A. Westwood, L. E. Brown et al., “Antitumor activity of dual-specific T cells and influenza virus,” Cancer Gene Therapy, vol. 14, no. 5, pp. 499–508, 2007.
[92]
M. A. Pule, B. Savoldo, G. D. Myers et al., “Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma,” Nature Medicine, vol. 14, no. 11, pp. 1264–1270, 2008.
[93]
S. Terakura, T. N. Yamamoto, R. A. Gardner, C. J. Turtle, M. C. Jensen, and S. R. Riddell, “Generation of CD19-chimeric antigen receptor modified CD8+ T cells derived from virus-specific central memory T cells,” Blood, vol. 119, no. 1, pp. 72–82, 2012.
[94]
A. Dutour, V. Marin, I. Pizzitola et al., “In vitro and in vivo antitumor effect of anti-CD33 chimeric receptor-expressing EBV-CTL against CD 33+ acute myeloid leukemia,” Advances in Hematology, vol. 2012, Article ID 683065, 2012.
[95]
C. Rossig, A. B?r, S. Pscherer et al., “Target antigen expression on a professional antigen-presenting cell induces superior proliferative antitumor T-cell responses via chimeric T-cell receptors,” Journal of Immunotherapy, vol. 29, no. 1, pp. 21–31, 2006.
[96]
K. P. Micklethwaite, B. Savoldo, P. J. Hanley et al., “Derivation of human T lymphocytes from cord blood and peripheral blood with antiviral and antileukemic specificity from a single culture as protection against infection and relapse after stem cell transplantation,” Blood, vol. 115, no. 13, pp. 2695–2703, 2010.
[97]
B. G. Till, M. C. Jensen, J. Wang et al., “Adoptive immunotherapy for indolent non-hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells,” Blood, vol. 112, no. 6, pp. 2261–2271, 2008.
[98]
M. T. Lotze, W. J. Buchser, and X. Liang, “Blocking the interleukin 2 (IL2)-induced systemic autophagic syndrome promotes profound antitumor effects and limits toxicity,” Autophagy, vol. 8, no. 8, pp. 1264–1266, 2012.
[99]
C. Quintarelli, J. F. Vera, B. Savoldo et al., “Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes,” Blood, vol. 110, no. 8, pp. 2793–2802, 2007.
[100]
B. Heemskerk, K. Liu, M. E. Dudley et al., “Adoptive cell therapy for patients with melanoma, using tumor-infiltrating lymphocytes genetically engineered to secrete interleukin-2,” Human Gene Therapy, vol. 19, no. 5, pp. 496–510, 2008.
[101]
L. S. Evans, P. R. Witte, A. L. Feldbaus et al., “Expression of chimeric granulocyte-macrophage colony-stimulating factor/interleukin 2 receptors in human cytotoxic T lymphocyte clones results in granulocyte-macrophage colony-stimulating factor-dependent growth,” Human Gene Therapy, vol. 10, no. 12, pp. 1941–1951, 1999.
[102]
J. F. Vera, V. Hoyos, B. Savoldo et al., “Genetic manipulation of tumor-specific cytotoxic T lymphocytes to restore responsiveness to IL-7,” Molecular Therapy, vol. 17, no. 5, pp. 880–888, 2009.
[103]
S. Wilkie, S. E. Burbridge, L. Chiapero-Stanke et al., “Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4,” Journal of Biological Chemistry, vol. 285, no. 33, pp. 25538–25544, 2010.
[104]
R. Goldstein, C. Hanley, J. Morris et al., “Clinical investigation of the role of interleukin-4 and interleukin-13 in the evolution of prostate cancer,” Cancers, vol. 3, no. 4, pp. 4281–4293, 2011.
[105]
H. Singh, L. M. Serrano, T. Pfeiffer et al., “Combining adoptive cellular and immunocytokine therapies to improve treatment of B-lineage malignancy,” Cancer Research, vol. 67, no. 6, pp. 2872–2880, 2007.
[106]
L. Gattinoni, C. A. Klebanoff, and N. P. Restifo, “Paths to stemness: building the ultimate antitumour T cell,” Nature Reviews Cancer, vol. 12, no. 10, pp. 671–684, 2012.
[107]
Z. Li, J. Düllmann, B. Schiedlmeier et al., “Murine leukemia induced by retroviral gene marking,” Science, vol. 296, no. 5567, p. 497, 2002.
[108]
M. G. Ott, M. Schmidt, K. Schwarzwaelder et al., “Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1,” Nature Medicine, vol. 12, no. 4, pp. 401–409, 2006.
[109]
S. Stein, M. G. Ott, S. Schultze-Strasser et al., “Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease,” Nature Medicine, vol. 16, no. 2, pp. 198–204, 2010.
[110]
S. Newrzela, K. Cornils, T. Heinrich et al., “Retroviral insertional mutagenesis can contribute to immortalization of mature T lymphocytes,” Molecular Medicine, vol. 17, no. 11, pp. 1223–1232, 2011.
[111]
C. Traversari, S. Marktel, Z. Magnani et al., “The potential immunogenicity of the TK suicide gene does not prevent full clinical benefit associated with the use of TK-transduced donor lymphocytes in HSCT for hematologic malignancies,” Blood, vol. 109, no. 11, pp. 4708–4715, 2007.
[112]
A. Recchia, C. Bonini, Z. Magnani et al., “Retroviral vector integration deregulates gene expression but has no consequence on the biology and function of transplanted T cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 5, pp. 1457–1462, 2006.
[113]
C. Bonini, A. Bondanza, S. K. Perna et al., “The suicide gene therapy challenge: how to improve a successful gene therapy approach,” Molecular Therapy, vol. 15, no. 7, pp. 1248–1252, 2007.
[114]
M. K. Brenner, “Gene transfer and the treatment of haematological malignancy,” Journal of Internal Medicine, vol. 249, no. 4, pp. 345–358, 2001.
[115]
P. Tiberghien, C. Ferrand, B. Lioure et al., “Administration of herpes simplex-thymidine kinase-expressing donor T cells with a T-cell-depleted allogeneic marrow graft,” Blood, vol. 97, no. 1, pp. 63–72, 2001.
[116]
L. M. Muul, L. M. Tuschong, S. L. Soenen et al., “Persistence and expression of the adenosine deaminase gene for 12 years and immune reaction to gene transfer components: long-term results of the first clinical gene therapy trial,” Blood, vol. 101, no. 7, pp. 2563–2569, 2003.
[117]
S. H. Yoon, J. M. Lee, H. I. Cho et al., “Adoptive immunotherapy using human peripheral blood lymphocytes transferred with RNA encoding Her-2neu-specific chimeric immune receptor in ovarian cancer xenograft model,” Cancer Gene Therapy, vol. 16, no. 6, pp. 489–497, 2009.
[118]
Y. Zhao, Z. Zheng, C. J. Cohen et al., “High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation,” Molecular Therapy, vol. 13, no. 1, pp. 151–159, 2006.
[119]
D. M. Barrett, Y. Zhao, X. Liu et al., “Treatment of advanced leukemia in mice with mRNA engineered T cells,” Human Gene Therapy, vol. 22, no. 12, pp. 1575–1586, 2011.
[120]
Y. Zhao, E. Moon, C. Carpenito et al., “Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor,” Cancer Research, vol. 70, no. 22, pp. 9053–9061, 2010.
[121]
Y. Nakazawa, L. E. Huye, V. S. Salsman et al., “PiggyBac-mediated cancer immunotherapy using EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor,” Molecular Therapy, vol. 19, pp. 2133–2143, 2011.
[122]
H. Singh, P. R. Manuri, S. Olivares et al., “Redirecting specificity of T-cell populations for CD19 using the sleeping beauty system,” Cancer Research, vol. 68, no. 8, pp. 2961–2971, 2008.
[123]
D. Hollyman, J. Stefanski, M. Przybylowski et al., “Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy,” Journal of Immunotherapy, vol. 32, no. 2, pp. 169–180, 2009.
[124]
M. M. Suhoski, T. N. Golovina, N. A. Aqui et al., “Engineering artificial antigen-presenting cells to express a diverse array of co-stimulatory molecules,” Molecular Therapy, vol. 15, no. 5, pp. 981–988, 2007.
[125]
L. X. J. Wang, J. A. Westwood, M. Moeller et al., “Tumor ablation by gene-modified T cells in the absence of autoimmunity,” Cancer Research, vol. 70, no. 23, pp. 9591–9598, 2010.
[126]
R. Bos, S. Van Duikeren, H. Morreau et al., “Balancing between antitumor efficacy and autoimmune pathology in T-cell-mediated targeting of carcinoembryonic antigen,” Cancer Research, vol. 68, no. 20, pp. 8446–8455, 2008.
[127]
M. R. Parkhurst, J. C. Yang, R. C. Langan et al., “T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis,” Molecular Therapy, vol. 19, no. 3, pp. 620–626, 2011.
[128]
C. Berger, M. E. Flowers, E. H. Warren, and S. R. Riddell, “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, vol. 107, no. 6, pp. 2294–2302, 2006.
[129]
K. C. Straathof, M. A. Pulè, P. Yotnda et al., “An inducible caspase 9 safety switch for T-cell therapy,” Blood, vol. 105, no. 11, pp. 4247–4254, 2005.
[130]
A. Di Stasi, S.-K. Tey, G. Dotti et al., “Inducible apoptosis as a safety switch for adoptive cell therapy,” New England Journal of Medicine, vol. 365, no. 18, pp. 1673–1683, 2011.
[131]
Y. Chu, N. Senghaas, R. W. K?ster, W. Wurst, and R. Kühn, “Novel Caspase-suicide proteins for tamoxifen-inducible apoptosis,” Genesis, vol. 46, no. 10, pp. 530–536, 2008.
[132]
B. Philip, S. Thomas, and V. Marin, “A highly compact epitope-based marker-suicide gene for more convenient and safer T-cell adoptive immunotherapy,” Blood, vol. 116, abstract 1473, pp. 629–630, 2010.
[133]
F. Perosa, E. Favoino, C. Vicenti, F. Merchionne, and F. Dammacco, “Identification of an antigenic and immunogenic motif expressed by two 7-Mer rituximab-specific cyclic peptide mimotopes: implication for peptide-based active immunotherapy,” Journal of Immunology, vol. 179, no. 11, pp. 7967–7974, 2007.
[134]
G. Suntharalingam, M. R. Perry, S. Ward et al., “Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412,” New England Journal of Medicine, vol. 355, no. 10, pp. 1018–1028, 2006.
[135]
M. Chmielewski, O. Hahn, G. Rappl et al., “T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice,” Gastroenterology, vol. 143, no. 4, pp. 1095–1107, 2012.
[136]
S. Kong, S. Sengupta, B. Tyler et al., “Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells,” Clinical Cancer Research, vol. 18, no. 21, pp. 5949–5960, 2012.
[137]
E. B. Santos, R. Yeh, J. Lee et al., “Sensitive in vivo imaging of T cells using a membrane-bound Gaussia princeps luciferase,” Nature Medicine, vol. 15, no. 3, pp. 338–344, 2009.
[138]
G. Koehne, M. Doubrovin, E. Doubrovina et al., “Serial in vivo imaging of the targeted migration of human HSV-TK-transduced antigen-specific lymphocytes,” Nature Biotechnology, vol. 21, no. 4, pp. 405–413, 2003.
[139]
S. S. Yaghoubi, M. C. Jensen, N. Satyamurthy et al., “Noninvasive detection of therapeutic cytolytic T cells with 18 F-FHBG PET in a patient with glioma,” Nature Reviews Clinical Oncology, vol. 6, no. 1, pp. 53–58, 2009.
[140]
M. M. Doubrovin, E. S. Doubrovina, P. Zanzonico, M. Sadelain, S. M. Larson, and R. J. O'Reilly, “In vivo imaging and quantitation of adoptively transferred human antigen-specific T cells transduced to express a human norepinephrine transporter gene,” Cancer Research, vol. 67, no. 24, pp. 11959–11969, 2007.
[141]
E. Sharif-Paghaleh, K. Sunassee, R. Tavaré et al., “In vivo SPECT reporter gene imaging of regulatory T cells,” PLoS ONE, vol. 6, no. 10, Article ID e25857, 2011.
[142]
R. S. Warren, E. K. Bergsland, R. Pennathur-Das, J. Nemunaitis, A. P. Venook, and K. M. Hege, “Clinical studies of regional and systemic gene therapy with autologous CC49-z modified T cells in colorectal cancer metastatic to the liver,” Cancer Gene Therapy, vol. 5, pp. S1–S2, 1998.
[143]
Q. Ma, R. M. Gonzalo-Daganzo, and R. P. Junghans, “Genetically engineered T cells as adoptive immunotherapy of cancer,” Cancer Chemotherapy and Biological Response Modifiers, vol. 20, pp. 315–341, 2002.
[144]
C. H. Lamers, S. Sleijfer, A. G. Vulto et al., “Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience,” Journal of Clinical Oncology, vol. 24, no. 13, pp. e20–22, 2006.
[145]
J. R. Park, D. L. DiGiusto, M. Slovak et al., “Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma,” Molecular Therapy, vol. 15, no. 4, pp. 825–833, 2007.
[146]
C. U. Louis, B. Savoldo, G. Dotti et al., “Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma,” Blood, vol. 118, no. 23, pp. 6050–6056, 2011.
[147]
M. C. Jensen, L. Popplewell, L. J. Cooper et al., “Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans,” Biology of Blood and Marrow Transplantation, vol. 16, no. 9, pp. 1245–1256, 2010.
[148]
J. N. Kochenderfer, M. E. Dudley, S. A. Feldman 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, vol. 119, no. 12, pp. 2709–2720, 2012.
[149]
B. G. Till, M. C. Jensen, J. Wang et al., “CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results,” Blood, vol. 119, no. 17, pp. 3940–3950, 2012.
[150]
H. E. Heslop, “Safer cars,” Molecular Therapy, vol. 18, no. 4, pp. 661–662, 2010.
[151]
R. J. Brentjens, J. B. Latouche, E. Santos et al., “Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15,” Nature Medicine, vol. 9, no. 3, pp. 279–286, 2003.
[152]
J. N. Kochenderfer, S. A. Feldman, Y. Zhao et al., “Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor,” Journal of Immunotherapy, vol. 32, no. 7, pp. 689–702, 2009.
[153]
L. J. N. Cooper, M. S. Topp, L. M. Serrano et al., “T-cell clones can be rendered specific for CD19: toward the selective augmentation of the graft-versus-B-lineage leukemia effect,” Blood, vol. 101, no. 4, pp. 1637–1644, 2003.
[154]
J. Wang, O. W. Press, C. G. Lindgren et al., “Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes,” Molecular Therapy, vol. 9, no. 4, pp. 577–586, 2004.
[155]
J. Vera, B. Savoldo, S. Vigouroux et al., “T lymphocytes redirected against the κ light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells,” Blood, vol. 108, no. 12, pp. 3890–3897, 2006.
[156]
B. Savoldo, C. M. Rooney, A. Di Stasi et al., “Epstein Barr virus-specific cytotoxic T lymphocytes expressing the anti-CD30ζ artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease,” Blood, vol. 110, no. 7, pp. 2620–2630, 2007.
[157]
V. Marin, I. Pizzitola, V. Agostoni et al., “Cytokine-induced killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors,” Haematologica, vol. 95, no. 12, pp. 2144–2152, 2010.
[158]
R. Thokala, H. Singh, S. Olivares, R. Champlin, and L. J. N. Cooper, “Targeting leukemias by CD123 specific chimeric antigen receptor,” Blood, vol. 118, abstract 1908, 2011.
[159]
K. Mihara, K. Yanagihara, M. Takigahira et al., “Synergistic and persistent effect of T-cell immunotherapy with anti-CD19 or anti-CD38 chimeric receptor in conjunction with rituximab on B-cell non-Hodgkin lymphoma,” British Journal of Haematology, vol. 151, no. 1, pp. 37–46, 2010.
[160]
K. Mihara, K. Yanagihara, M. Takigahira et al., “Activated T-cell-mediated immunotherapy with a chimeric receptor against CD38 in b-cell non-hodgkin lymphoma,” Journal of Immunotherapy, vol. 32, no. 7, pp. 737–743, 2009.
[161]
M. Hudecek, T. M. Schmitt, S. Baskar et al., “The B-cell tumor-associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor,” Blood, vol. 116, no. 22, pp. 4532–4541, 2010.
[162]
D. Moritz, W. Wels, J. Mattern, and B. Groner, “Cytotoxic T lymphocytes with a grafted recognition specificity for ERBB2- expressing tumor cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 91, no. 10, pp. 4318–4322, 1994.
[163]
U. Altenschmidt, E. Klundt, and B. Groner, “Adoptive transfer of in vitro-targeted, activated T lymphocytes results in total tumor regression,” Journal of Immunology, vol. 159, no. 11, pp. 5509–5515, 1997.
[164]
N. M. Haynes, M. J. Smyth, M. H. Kershaw, J. A. Trapani, and P. K. Darcy, “Fas-ligand-mediated lysis of erbB-2-expressing tumour cells by redirected cytotoxic T lymphocytes,” Cancer Immunology Immunotherapy, vol. 47, no. 5, pp. 278–286, 1999.
[165]
J. H. Pinthus, T. Waks, K. Kaufman-Francis et al., “Immuno-gene therapy of established prostate tumors using chimeric receptor-redirected human lymphocytes,” Cancer Research, vol. 63, no. 10, pp. 2470–2476, 2003.
[166]
F. Turatti, M. Figini, P. Alberti, R. A. Willemsen, S. Canevari, and D. Mezzanzanica, “Highly efficient redirected anti-tumor activity of human lymphocytes transduced with a completely human chimeric immune receptor,” Journal of Gene Medicine, vol. 7, no. 2, pp. 158–170, 2005.
[167]
N. Ahmed, M. Ratnayake, B. Savoldo et al., “Regression of experimental medulloblastoma following transfer of HER2-specific T cells,” Cancer Research, vol. 67, no. 12, pp. 5957–5964, 2007.
[168]
S. Li, J. Yang, F. A. Urban et al., “Genetically engineered T cells expressing a HER2-specific chimeric receptor mediate antigen-specific tumor regression,” Cancer Gene Therapy, vol. 15, no. 6, pp. 382–392, 2008.
[169]
H. Wang, H. Wei, R. Zhang et al., “Genetically targeted T cells eradicate established breast cancer in syngeneic mice,” Clinical Cancer Research, vol. 15, no. 3, pp. 943–950, 2009.
[170]
N. Ahmed, V. S. Salsman, E. Yvon et al., “Immunotherapy for osteosarcoma: genetic modification of T cells overcomes low levels of tumor antigen expression,” Molecular Therapy, vol. 17, no. 10, pp. 1779–1787, 2009.
[171]
Y. Zhao, Q. J. Wang, S. Yang et al., “A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity,” Journal of Immunology, vol. 183, no. 9, pp. 5563–5574, 2009.
[172]
N. Ahmed, V. S. Salsman, Y. Kew et al., “HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors,” Clinical Cancer Research, vol. 16, no. 2, pp. 474–485, 2010.
[173]
N. Rainusso, V. S. Brawley, A. Ghazi et al., “Immunotherapy targeting HER2 with genetically modified T cells eliminates tumor-initiating cells in osteosarcoma,” Cancer Gene Therapy, vol. 19, no. 3, pp. 212–217, 2012.
[174]
R. A. Morgan, L. A. Johnson, and J. Davis, “Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma,” Human Gene Therapy, vol. 23, no. 10, pp. 1043–1053, 2012.
[175]
P. K. Darcy, N. M. Haynes, M. B. Snook et al., “Redirected perforin-dependent lysis of colon carcinoma by ex vivo genetically engineered CTL,” Journal of Immunology, vol. 164, no. 7, pp. 3705–3712, 2000.
[176]
L. Ren-Heidenreich, G. T. Hayman, and K. T. Trevor, “Specific targeting of EGP-2+ tumor cells by primary lymphocytes modified with chimeric T cell receptors,” Human Gene Therapy, vol. 11, no. 1, pp. 9–19, 2000.
[177]
T. Daly, R. E. Royal, M. H. Kershaw et al., “Recognition of human colon cancer by T cells transduced with a chimeric receptor gene,” Cancer Gene Therapy, vol. 7, no. 2, pp. 284–291, 2000.
[178]
E. Lanitis, M. Poussin, I. S. Hagemann et al., “Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor,” Molecular Therapy, vol. 20, no. 3, pp. 633–643, 2012.
[179]
A. Hombach, C. Heuser, R. Sircar et al., “T cell targeting of TAG72+ tumor cells by a chimeric receptor with antibody-like specificity for a carbohydrate epitope,” Gastroenterology, vol. 113, no. 4, pp. 1163–1170, 1997.
[180]
M. C. Gong, J. B. Latouche, A. Krause, W. D. W. Heston, N. H. Bander, and M. Sadelain, “Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen,” Neoplasia, vol. 1, no. 2, pp. 123–127, 1999.
[181]
T. Zhang, B. A. Lemoi, and C. L. Sentman, “Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy,” Blood, vol. 106, no. 5, pp. 1544–1551, 2005.
[182]
K. S. Kahlon, C. Brown, L. J. N. Cooper, A. Raubitschek, S. J. Forman, and M. C. Jensen, “Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells,” Cancer Research, vol. 64, no. 24, pp. 9160–9166, 2004.
[183]
C. E. Brown, R. Starr, B. Aguilar et al., “Stem-like tumor-initiating cells isolated from IL13Rα2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T cells,” Clinical Cancer Research, vol. 18, no. 8, pp. 2199–2209, 2012.
[184]
A. A. Chekmasova, T. D. Rao, Y. Nikhamin et al., “Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen,” Clinical Cancer Research, vol. 16, no. 14, pp. 3594–3606, 2010.
[185]
M. E. M. Weijtens, R. A. Willemsen, D. Valerio, K. Stam, and R. L. H. Bolhuis, “Single chain Ig/γ gene-redirected human T lymphocytes produce cytokines, specifically lyse tumor cells, and recycle lytic capacity,” Journal of Immunology, vol. 157, no. 2, pp. 836–843, 1996.
[186]
C. Rossig, C. M. Bollard, J. G. Nuchtern, D. A. Merchant, and M. K. Brenner, “Targeting of GD2-positive tumor cells by human T lymphocytes engineered to express chimeric T-cell receptor genes,” International Journal of Cancer, vol. 94, no. 2, pp. 228–236, 2001.
[187]
S. Kailayangiri, B. Altvater, J. Meltzer et al., “The ganglioside antigen G D2 is surface-expressed in Ewing sarcoma and allows for MHC-independent immune targeting,” British Journal of Cancer, vol. 106, no. 6, pp. 1123–1133, 2012.
[188]
C. O. Yun, K. F. Nolan, E. J. Beecham, R. A. Reisfeld, and R. P. Junghans, “Targeting of T lymphocytes to melanoma cells through chimeric anti-GD3 immunoglobulin t-cell receptors,” Neoplasia, vol. 2, no. 5, pp. 449–459, 2000.
[189]
H. Abken, A. Hombach, C. Heuser, and U. Reinhold, “A novel strategy in the elimination of disseminated melanoma cells: chimeric receptors endow T cells with tumor specificity,” Recent Results in Cancer Research, vol. 158, pp. 249–264, 2001.
[190]
J. A. Westwood, M. J. Smyth, M. W. L. Teng et al., “Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 52, pp. 19051–19056, 2005.
[191]
P. C. Schuberth, G. Jakka, S. M. Jensen, et al., “Effector memory and central memory NY-ESO-1-specific re-directed T cells for treatment of multiple myeloma,” Gene Therapy. In press.
[192]
A. Morgenroth, M. Cartellieri, M. Schmitz et al., “Targeting of tumor cells expressing the prostate stem cell antigen (PSCA) using genetically engineered T-cells,” Prostate, vol. 67, no. 10, pp. 1121–1131, 2007.
[193]
P. Hwu, J. C. Yang, R. Cowherd et al., “In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes,” Cancer Research, vol. 55, no. 15, pp. 3369–3373, 1995.
[194]
M. H. Kershaw, J. A. Westwood, and P. Hwu, “Dual-specific T cells combine proliferation and antitumor activity,” Nature Biotechnology, vol. 20, no. 12, pp. 1221–1227, 2002.
[195]
A. Hekele, P. Dall, D. Moritz et al., “Growth retardation of tumors by adoptive transfer of cytotoxic T lymphocytes reprogrammed by CD44V6-specific SCFV:ζ-chimera,” International Journal of Cancer, vol. 68, no. 2, pp. 232–238, 1996.
[196]
P. Dall, I. Herrmann, B. Durst et al., “In vivo cervical cancer growth inhibition by genetically engineered cytotoxic T cells,” Cancer Immunology, Immunotherapy, vol. 54, no. 1, pp. 51–60, 2005.
[197]
N. K. V. Cheung, H. F. Guo, S. Modak, and I. Y. Cheung, “Anti-idiotypic antibody facilitates scFv chimeric immune receptor gene transduction and clonal expansion of human lymphocytes for tumor therapy,” Hybridoma and Hybridomics, vol. 22, no. 4, pp. 209–218, 2003.
[198]
D. E. Gilham, A. O'Neil, C. Hughes et al., “Primary polyclonal human T lymphocytes targeted to carcino-embryonic antigens and neural cell adhesion molecule tumor antigens by CD3ζ-based chimeric immune receptors,” Journal of Immunotherapy, vol. 25, no. 2, pp. 139–151, 2002.
[199]
T. M. J. Niederman, Z. Ghogawala, B. S. Carter, H. S. Tompkins, M. M. Russell, and R. C. Mulligan, “Antitumor activity of cytotoxic T lymphocytes engineered to target vascular endothelial growth factor receptors,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 10, pp. 7009–7014, 2002.
[200]
M. H. Kershaw, J. A. Westwood, Z. Zhu, L. Witte, S. K. Libutti, and P. Hwu, “Generation of gene-modified T cells reactive against the angiogenic kinase insert domain-containing receptor (KDR) found on tumor vasculature,” Human Gene Therapy, vol. 11, no. 18, pp. 2445–2452, 2000.
[201]
H. R. Jiang, D. E. Gilham, K. Mulryan, N. Kirillova, R. E. Hawidns, and P. L. Stern, “Combination of vaccination and chimeric receptor expressing T cells provides improved active therapy of tumors,” Journal of Immunology, vol. 177, no. 7, pp. 4288–4298, 2006.
[202]
S. Gattenl?hner, A. Marx, B. Markfort et al., “Rhabdomyosarcoma lysis by T cells expressing a human autoantibody-based chimeric receptor targeting the fetal acetylcholine receptor,” Cancer Research, vol. 66, no. 1, pp. 24–28, 2006.
[203]
M. Casucci, L. Falcone, and B. Nicolis di Robilant, “Dual transgenesis of T cells with a CD44v6 CAR and a suicide gene for the safe eradication of myeloid leukemia and myeloma,” Molecular Therapy, vol. 20, no. S1, abstract 465, p. S180, 2012.
[204]
S. D. Patel, M. Moskalenko, T. Tian et al., “T-cell killing of heterogenous tumor or viral targets with bispecific chimeric immune receptors,” Cancer Gene Therapy, vol. 7, no. 8, pp. 1127–1134, 2000.
[205]
C. Uherek, T. Tonn, B. Uherek et al., “Retargeting of natural killer-cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction,” Blood, vol. 100, no. 4, pp. 1265–1273, 2002.
[206]
H. E. Daldrup-Link, R. Meier, M. Rudelius et al., “In vivo tracking of genetically engineered, anti-HER2/neu directed natural killer cells to HER2/neu positive mammary tumors with magnetic resonance imaging,” European Radiology, vol. 15, no. 1, pp. 4–13, 2005.
[207]
R. Meier, M. Piert, G. Piontek et al., “Tracking of [18F]FDG-labeled natural killer cells to HER2/neu-positive tumors,” Nuclear Medicine and Biology, vol. 35, no. 5, pp. 579–588, 2008.
[208]
H. J. Pegram, J. T. Jackson, M. J. Smyth, M. H. Kershaw, and P. K. Darcy, “Adoptive transfer of gene-modified primary NK cells can specifically inhibit tumor progression in vivo,” Journal of Immunology, vol. 181, no. 5, pp. 3449–3455, 2008.
[209]
A. Kruschinski, A. Moosmann, I. Poschke et al., “Engineering antigen-specific primary human NK cells against HER-2 positive carcinomas,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 45, pp. 17481–17486, 2008.
[210]
A. Biglari, T. D. Southgate, L. J. Fairbairn, and D. E. Gilham, “Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo,” Gene Therapy, vol. 13, no. 7, pp. 602–610, 2006.
[211]
S. Tammana, X. Huang, M. Wong et al., “4-1BB and CD28 signaling plays a synergistic role in redirecting umbilical cord blood t cells against b-cell malignancies,” Human Gene Therapy, vol. 21, no. 1, pp. 75–86, 2010.
[212]
M. R. Roberts, K. S. Cooke, A. C. Tran et al., “Antigen-specific cytolysis by neutrophils and NK cells expressing chimeric immune receptors bearing or signaling domains,” Journal of Immunology, vol. 161, no. 1, pp. 375–384, 1998.
[213]
G. Wang, R. K. Chopra, R. E. Royal, J. C. Yang, S. A. Rosenberg, and P. Hwu, “A T cell-independent antitumor response in mice with bone marrow cells retrovirally transduced with an antibody/Fc-γ chain chimetic receptor gene recognizing a human ovarian cancer antigen,” Nature Medicine, vol. 4, no. 2, pp. 168–172, 1998.
[214]
Y. Xu, P. K. Darcy, and M. H. Kershaw, “Tumor-specific dendritic cells generated by genetic redirection of Toll-like receptor signaling against the tumor-associated antigen, erbB2,” Cancer Gene Therapy, vol. 14, no. 9, pp. 773–780, 2007.
[215]
C. Cruz, K. Micklethwaite, B. Savoldo, et al., “Infusion of CD19-directed/multivirus-specific CTLs post HSCT for B cell malignancies,” Molecular Therapy, vol. 20, no. S1, abstract 536, p. S207, 2012.
[216]
S. F. Slovin, X. Wang, and O. Borquez-Ojeda, “Targeting Castration Resistant Prostate Cancer (CRPC) with autologous PSMA-directed chimeric antigen receptor T cells,” Molecular Therapy, vol. 20, no. S1, abstract 81, p. S33, 2012.
[217]
R. P. Junghans, R. Rathore, and Q. Ma, “Phase 1 trial of anti-PSMA designed T cells in advanced prostate cancer,” Journal of Clinical Oncology, vol. 29, no. S7, abstract 130, 2011.
[218]
L. E. Kandalaft, D. J. Powell, and G. Coukos, “A phase I clinical trial of adoptive transfer of folate receptor-alpha redirected autologous T cells for recurrent ovarian cancer,” Journal of Translational Medicine, vol. 10, no. 1, article 157, 2012.