TanCAR: A Novel Bispecific Chimeric Antigen Receptor for Cancer Immunotherapy

Zakaria Grada, Meenakshi Hegde, Tiara Byrd, Donald R Shaffer, Alexia Ghazi, Vita S Brawley, Amanda Corder, Kurt Schönfeld, Joachim Koch, Gianpietro Dotti, Helen E Heslop, Stephen Gottschalk, Winfried S Wels, Matthew L Baker, Nabil Ahmed, Zakaria Grada, Meenakshi Hegde, Tiara Byrd, Donald R Shaffer, Alexia Ghazi, Vita S Brawley, Amanda Corder, Kurt Schönfeld, Joachim Koch, Gianpietro Dotti, Helen E Heslop, Stephen Gottschalk, Winfried S Wels, Matthew L Baker, Nabil Ahmed

Abstract

Targeted T cells are emerging as effective non-toxic therapies for cancer. Multiple elements, however, contribute to the overall pathogenesis of cancer through both distinct and redundant mechanisms. Hence, targeting multiple cancer-specific markers simultaneously could result in better therapeutic efficacy. We created a functional chimeric antigen receptor-the TanCAR, a novel artificial molecule that mediates bispecific activation and targeting of T cells. We demonstrate the feasibility of cumulative integration of structure and docking simulation data using computational tools to interrogate the design and predict the functionality of such a complex bispecific molecule. Our prototype TanCAR induced distinct T cell reactivity against each of two tumor restricted antigens, and produced synergistic enhancement of effector functions when both antigens were simultaneously encountered. Furthermore, the TanCAR preserved the cytolytic ability of T cells upon loss of one of the target molecules and better controlled established experimental tumors by recognition of both targets in an animal disease model. This proof-of-concept approach can be used to increase the specificity of effector cells for malignant versus normal target cells, to offset antigen escape or to allow for targeting the tumor and its microenvironment.Molecular Therapy-Nucleic Acids (2013) 2, e105; doi:10.1038/mtna.2013.32; published online 9 July 2013.

Figures

Figure 1
Figure 1
Designing a bispecific tandem chimeric antigen receptor (TanCAR) molecule. representation of the proposed chimeric antigen receptor molecule extracellular domains engaging the two targets; HER2 and CD19.
Figure 2
Figure 2
Docking platforms predict favorable binding potential of TanCAR to target molecules. Compilation of structure and docking data of (a) hypothetical structure of both FRP5-derived scFv and the CD19-specific scFv joined with a 20 amino acid Gly-Ser linker; (b) most favorable docking models of FRP5-derived scFv and the distal 200 amino acid residues of the extracellular domain of HER2; (c) most favorable docking models of CD19-scFv and the extracellular domain of CD19, and combined docking of (d) HER2 and (e) CD19 and the whole TanCAR exodomain, individually.
Figure 3
Figure 3
Construction and surface expression of the TanCAR molecule. (a) pSFG vector construct encoding the TanCAR; (b) detection of the surface expression of the TanCAR using a Fab-specific antibody and FRP5-specific HER2-Fc protein on 293T cells; and (c) on T cells. See Supplementary Figure S2 for description of the labeling strategy.
Figure 4
Figure 4
The TanCAR T cells distinctly recognize individual target molecules. (a) Flow cytometric analysis of the surface expression of the target antigens, HER2 and CD19, on a panel of human cancer cell lines used for functional testing; (b) cytotoxicity assay showing recognition and killing of HER2-positive Daoy cells and efficient blocking of this lysis using a soluble HER2 fragment; (c) similarly, TanCAR T cells recognized CD19-positive Raji cells and this lysis was blocked using the CD19 Ab 4G7; (d) in cocultures, TanCAR T cells secreted IFN-γ as well as IL-2 upon encounter of HER2- and CD19-positive target cell above the non-transduced T-cell control (NT). No cytokines were secreted in coculture with the HER2- and CD19-null target cell MDA-MB-468. (bd) Shown are representative plots of three or more experiments done in triplicates.
Figure 5
Figure 5
Enhanced cytolytic function upon simultaneous recognition of two antigens and preserved TanCAR T cell-induced cytolysis in a model of antigen loss. (a) Collective modeling of the simultaneous docking of the TanCAR to both HER2 and CD19. (b) In cytotoxicity assays, we saw consistently higher killing after the induction of CD19 at various tumor to T cell ratios. This was synergistic; with an exponential trend following a higher order equation; and was more prominent in higher tumor to T cell ratios (right panel). (c) Similarly, induction of CD19 (DOX+) in Daoy.TET.CD19 and T cell cocultures resulted in a more than fourfold increase in IFN-γ release as detected by ELISA (P < 0.01). (d) We modeled the scenario in which tumor cells downregulate the target antigen, by blocking HER2 in CD19-induced (DOX+) and CD19 null (DOX−) Daoy.TET.CD19 cells using a soluble HER2 fragment (s.HER2). Although soluble HER2 successfully induced substantial blocking of HER2-mediated killing in DOX− cells at various tumor to T cell ratios, it resulted only in a partial decrease in the cytolytic effect of TanCAR T cells in DOX+. P values were significant at T cell to tumor cell ratios of 1:20, 1:10, and 1:5. Shown are representative plots of three experiments done in triplicates.
Figure 6
Figure 6
Simultaneous targeting of two antigens enhances the in vivo antitumor activity of adoptively transferred TanCAR T cells. (a) Daoy.TET.CD19 xenografts were established for 3 weeks in the flanks of SCID mice, and then animals were randomized into four groups. Administration of PBS into the tumor and/or systemic doxycycline induced minimal or no alteration of the tumor growth pattern. By contrast, treatment with TanCAR T cells resulted in a significant delay in tumor progression that was further enhanced by induction of CD19 expression in the DOX+ group. (b) Kaplan–Meier survival curve: analysis performed 60 days after PBS or T-cells injection. Mice treated with TanCAR T cells had a significantly longer survival probability in comparison with control mice. Furthermore, induction of CD19 by the administration of doxycycline resulted in enhanced antitumor activity of adoptively transferred TanCAR T cells.

References

    1. Kochenderfer JN, Rosenberg SA. Chimeric antigen receptor-modified T cells in CLL. N Engl J Med. 2011;365:1937–8; author reply 1938.
    1. 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 or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci USA. 1993;90:720–724.
    1. Sadelain M, Rivière I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer. 2003;3:35–45.
    1. Brentjens RJ, Latouche JB, Santos E, Marti F, Gong MC, Lyddane C, et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med. 2003;9:279–286.
    1. Pule M, Straathof K, Dotti G, Heslop HE, Rooney CM, Brenner MK. Three-module signaling endo-domain artificial T-cell receptor which transmits CD28, OX40 and CD3-zeta signals enhances IL-2 release and proliferative response in transduced primary T-cells. Blood. 2004;104:485a.
    1. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121:1822–1826.
    1. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, et al. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med. 2008;14:1264–1270.
    1. Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, 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. 2012;119:2709–2720.
    1. Jensen MC, Popplewell L, Cooper LJ, DiGiusto D, Kalos M, Ostberg JR, et al. Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant. 2010;16:1245–1256.
    1. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–733.
    1. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–360.
    1. Weijtens ME, Hart EH, Bolhuis RL. Functional balance between T cell chimeric receptor density and tumor associated antigen density: CTL mediated cytolysis and lymphokine production. Gene Ther. 2000;7:35–42.
    1. Buchner GS, Murphy RD, Buchete NV, Kubelka J. Dynamics of protein folding: probing the kinetic network of folding-unfolding transitions with experiment and theory. Biochim Biophys Acta. 2011;1814:1001–1020.
    1. Kuhlman B, Baker D. Exploring folding free energy landscapes using computational protein design. Curr Opin Struct Biol. 2004;14:89–95.
    1. Perez-Aguilar JM, Saven JG. Computational design of membrane proteins. Structure. 2012;20:5–14.
    1. Samish I, MacDermaid CM, Perez-Aguilar JM, Saven JG. Theoretical and computational protein design. Annu Rev Phys Chem. 2011;62:129–149.
    1. Kiel C, Beltrao P, Serrano L. Analyzing protein interaction networks using structural information. Annu Rev Biochem. 2008;77:415–441.
    1. Matsushima N, Yoshida H, Kumaki Y, Kamiya M, Tanaka T, Izumi Y, et al. Flexible structures and ligand interactions of tandem repeats consisting of proline, glycine, asparagine, serine, and/or threonine rich oligopeptides in proteins. Curr Protein Pept Sci. 2008;9:591–610.
    1. Ahmed N, Salsman VS, Kew Y, Shaffer D, Powell S, Zhang YJ, et al. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res. 2010;16:474–485.
    1. Pieper U, Webb BM, Barkan DT, Schneidman-Duhovny D, Schlessinger A, Braberg H, et al. ModBase, a database of annotated comparative protein structure models, and associated resources. Nucleic Acids Res. 2011;39 Database issue:D465–D474.
    1. Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney al. (2003Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature 421756–760.
    1. Shim AH, Liu H, Focia PJ, Chen X, Lin PC, He X. Structures of a platelet-derived growth factor/propeptide complex and a platelet-derived growth factor/receptor complex. Proc Natl Acad Sci USA. 2010;107:11307–11312.
    1. Schneidman-Duhovny D, Inbar Y, Nussinov R, Wolfson HJ. PatchDock and SymmDock: servers for rigid and symmetric docking. Nucleic Acids Res. 2005;33 Web Server issue:W363–W367.
    1. Andrusier N, Nussinov R, Wolfson HJ. FireDock: fast interaction refinement in molecular docking. Proteins. 2007;69:139–159.
    1. Gerstmayer B, Altenschmidt U, Hoffmann M, Wels W. Costimulation of T cell proliferation by a chimeric B7-2 antibody fusion protein specifically targeted to cells expressing the erbB2 proto-oncogene. J Immunol. 1997;158:4584–4590.
    1. Raab D, Graf M, Notka F, Schödl T, Wagner R. The GeneOptimizer Algorithm: using a sliding window approach to cope with the vast sequence space in multiparameter DNA sequence optimization. Syst Synth Biol. 2010;4:215–225.
    1. Rossig C, Bollard CM, Nuchtern JG, Merchant DA, Brenner MK. Targeting of G(D2)-positive tumor cells by human T lymphocytes engineered to express chimeric T-cell receptor genes. Int J Cancer. 2001;94:228–236.
    1. Ahmed N, Ratnayake M, Savoldo B, Perlaky L, Dotti G, Wels WS, et al. Regression of experimental medulloblastoma following transfer of HER2-specific T cells. Cancer Res. 2007;67:5957–5964.
    1. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, et al. UCSF Chimera–a visualization system for exploratory research and analysis. J Comput Chem. 2004;25:1605–1612.
    1. Zhou X, Vink M, Klaver B, Berkhout B, Das AT. Optimization of the Tet-On system for regulated gene expression through viral evolution. Gene Ther. 2006;13:1382–1390.
    1. Ahmed N, Salsman VS, Yvon E, Louis CU, Perlaky L, Wels WS, et al. Immunotherapy for osteosarcoma: genetic modification of T cells overcomes low levels of tumor antigen expression. Mol Ther. 2009;17:1779–1787.
    1. Bargou R, Leo E, Zugmaier G, Klinger M, Goebeler M, Knop S, et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science. 2008;321:974–977.
    1. Hagemeyer CE, von Zur Muhlen C, von Elverfeldt D, Peter K. Single-chain antibodies as diagnostic tools and therapeutic agents. Thromb Haemost. 2009;101:1012–1019.
    1. Wu C, Ying H, Grinnell C, Bryant S, Miller R, Clabbers A, et al. Simultaneous targeting of multiple disease mediators by a dual-variable-domain immunoglobulin. Nat Biotechnol. 2007;25:1290–1297.
    1. Gu J, Ghayur T. Generation of dual-variable-domain immunoglobulin molecules for dual-specific targeting. Meth Enzymol. 2012;502:25–41.
    1. Doppalapudi VR, Huang J, Liu D, Jin P, Liu B, Li L, et al. Chemical generation of bispecific antibodies. Proc Natl Acad Sci USA. 2010;107:22611–22616.
    1. Duchnowska R, Szczylik C. Central nervous system metastases in breast cancer patients administered trastuzumab. Cancer Treat Rev. 2005;31:312–318.
    1. Shmueli E, Wigler N, Inbar M. Central nervous system progression among patients with metastatic breast cancer responding to trastuzumab treatment. Eur J Cancer. 2004;40:379–382.
    1. Verneris MR, Arshi A, Edinger M, Kornacker M, Natkunam Y, Karami M, et al. Low levels of Her2/neu expressed by Ewing's family tumor cell lines can redirect cytokine-induced killer cells. Clin Cancer Res. 2005;11:4561–4570.
    1. Hong JJ, Rosenberg SA, Dudley ME, Yang JC, White DE, Butman JA, et al. Successful treatment of melanoma brain metastases with adoptive cell therapy. Clin Cancer Res. 2010;16:4892–4898.
    1. Study of administration of CMV-specific cytotoxic T lymphocytes expressing CAR targeting HER2 in patients with GBM (HERT-GBM). 2011 ) < >.
    1. Patel SD, Moskalenko M, Tian T, Smith D, McGuinness R, Chen L, et al. T-cell killing of heterogenous tumor or viral targets with bispecific chimeric immune receptors. Cancer Gene Ther. 2000;7:1127–1134.
    1. Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365:1673–1683.
    1. Wilkie S, van Schalkwyk MC, Hobbs S, Davies DM, van der Stegen SJ, Pereira AC, et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J Clin Immunol. 2012;32:1059–1070.
    1. Calarese DA, Scanlan CN, Zwick MB, Deechongkit S, Mimura Y, Kunert R, et al. Antibody domain exchange is an immunological solution to carbohydrate cluster recognition. Science. 2003;300:2065–2071.
    1. Kleinjung J, Petit MC, Orlewski P, Mamalaki A, Tzartos SJ, Tsikaris V, et al. The third-dimensional structure of the complex between an Fv antibody fragment and an analogue of the main immunogenic region of the acetylcholine receptor: a combined two-dimensional NMR, homology, and molecular modeling approach. Biopolymers. 2000;53:113–128.
    1. Leysath CE, Monzingo AF, Maynard JA, Barnett J, Georgiou G, Iverson BL, et al. Crystal structure of the engineered neutralizing antibody M18 complexed to domain 4 of the anthrax protective antigen. J Mol Biol. 2009;387:680–693.
    1. Wilkinson IC, Hall CJ, Veverka V, Shi JY, Muskett FW, Stephens PE, et al. High resolution NMR-based model for the structure of a scFv-IL-1beta complex: potential for NMR as a key tool in therapeutic antibody design and development. J Biol Chem. 2009;284:31928–31935.
    1. Ginalski K, Elofsson A, Fischer D, Rychlewski L. 3D-Jury: a simple approach to improve protein structure predictions. Bioinformatics. 2003;19:1015–1018.
    1. Zola H, MacArdle PJ, Bradford T, Weedon H, Yasui H, Kurosawa Y. Preparation and characterization of a chimeric CD19 monoclonal antibody. Immunol Cell Biol. 1991;69 (Pt 6):411–422.
    1. Wels W, Harwerth IM, Zwickl M, Hardman N, Groner B, Hynes NE. Construction, bacterial expression and characterization of a bifunctional single-chain antibody-phosphatase fusion protein targeted to the human erbB-2 receptor. Biotechnology (NY) 1992;10:1128–1132.

Source: PubMed

3
Tilaa