An Optimized GD2-Targeting Retroviral Cassette for More Potent and Safer Cellular Therapy of Neuroblastoma and Other Cancers

Simon Thomas, Karin Straathof, Nourredine Himoudi, John Anderson, Martin Pule, Simon Thomas, Karin Straathof, Nourredine Himoudi, John Anderson, Martin Pule

Abstract

Neuroblastoma is the commonest extra cranial solid cancer of childhood. Despite escalation of treatment regimens, a significant minority of patients die of their disease. Disialoganglioside (GD2) is consistently expressed at high-levels in neuroblastoma tumors, which have been targeted with some success using therapeutic monoclonal antibodies. GD2 is also expressed in a range of other cancer but with the exception of some peripheral nerves is largely absent from non-transformed tissues. Chimeric Antigen Receptors (CARs) are artificial type I proteins which graft the specificity of a monoclonal antibody onto a T-cell. Clinical data with early CAR designs directed against GD2 have shown some promise in Neuroblastoma. Here, we describe a GD2-targeting CAR retroviral cassette, which has been optimized for CAR T-cell persistence, efficacy and safety.

Conflict of interest statement

Competing Interests: MP has received contract research funding from Cellectis. He owns stock and receives salary contribution from Autolus Ltd. He is an inventor on patents filed by UCLB and has received and may receive royalties therefrom. He has received honoraria from Roche and Amgen for speaking. JA and ST own stock from Autolos Ltd. They are inventors on patents filed by UCLB and may receive royalties therefrom. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Comparison between huK666 and muK666…
Fig 1. Comparison between huK666 and muK666 based CARs.
(a) Comparison of amino acid sequences of huK666 and muK666 scFvs are compared. Complementarity determining regions are shown in red. The linker sequence is shown in green. On right, architectures of the CARs generated for initial comparison of huK666 with muK666. Both differ only in the scFvs used being either the original murine muK666 sequences, or the humanized (CDR grafted) huK666. Otherwise the CARs comprise of a human IgG1 hinge-CH2-CH3 spacer, a TM domain derived from CD28 and a compound endodomain comprising of fusions between endodomains from CD28, OX40 and CD3-Zeta. (b) Stability of muK666 vs huK666 CARs. T-cells from 5 donors were transduced with equal titers of retroviral supernatant coding for muK666 or huK666 CARs. CAR expression was detected by anti-human-Fc polyclonal antibodies. MFI was identical in both constructs. A representative example is shown with NT (green), muK666 (blue) and huK666 (red) histograms overlaid. These CAR T-cells were challenged with LAN-1 (a Neuroblastoma cell line expressing GD2) and A204 (a rhabdomyosarcoma cell line which is GD2 negative). Cytotoxicity is shown in (c), proliferation at day 7 is shown in (d), IFN-γ in (e) and IL-2 release in (f). Data shown as means +/-SEM from 6 independent experiments with different donors.
Fig 2. Effects of spacer on CAR…
Fig 2. Effects of spacer on CAR function.
Several huK666 CARs were cloned into an identical format whereby the scFv was tagged with an HA-tag, and the CAR was co-expressed with a truncated FMD-2A sequence with truncated CD34. The expression cassette is shown in (a) and cartoons of these formats are shown in (b). Co-expression of CAR (HA) and CD34 marker gene from the different cassettes. The four hinge regions were compared in terms of cytotoxicity (c) (Lan1 = GD2 positive target and A204 = GD2 negative target), IFN-γ secretion (d), IL-2 (e) and target-specific proliferation (f). Data shown as means +/-SEM from 4 independent experiments with different donors.
Fig 3. Removal of FcR binding motifs…
Fig 3. Removal of FcR binding motifs in IgG1 spacer.
(a) amino acid sequence of the wild type (top) and mutated (PVAA) CH2 regions responsible for FcγR binding. (b) flow staining with anti-Fc antibody to show comparable level of expression of receptor with or without the PVAA mutation. Identical CARs with and without PVAA were compared side by side in terms of cytotoxicity against GD2 engineered SupT1 and GD2 negative wild type SipT1 cells (c), cytotoxicity against GD2 positive Lan1 cells and FcRγ positive THP-1 cells (d), and IL-1β release on culture with THP-1 cells (e). Data shown as means +/-SEM from 4 independent experiments with different donors.
Fig 4. Optimization of expression cassette for…
Fig 4. Optimization of expression cassette for co-expression with iCasp9.
(a) Nine different expression cassettes were compared: Three different retroviral vectors were generated–wild-type SFG, SFG with scaffold-attachment region (SAR) inserted into the 3’UTR and SFG with CHS4 inserted into the 3’LTR U3 region. (retroviral vectors with both SAR and CHS4 were generated but produced very low vector titers and were not compared further); Into these retroviral vectors wild-type iCasp9-2A-huK666 CAR constructs were inserted and iCasp9 were inserted in 3 forms; codon-optimized, wild-type, and with the catalytic domain mutated. (b) Histogrammes at day 3 after transduction (blue lines). Overlaid T-cells cultured in 20nM CID are shown in red. Bargraphs of MFI of cells in the absence of CID at day 3 (c) and (d) day 7. Data shown as means +/-SEM from 5 independent experiments with different donors.
Fig 5. Consequence and function of iCasp9.
Fig 5. Consequence and function of iCasp9.
The SAR codon-optimized cassette was taken further and compared with the original cassette without iCasp9. (a) Expression of the CAR was unchanged. Depletion is shown by facs after addition of CID. The function of cassettes with and without iCasp9 were assessed by (b) Killing (c) IFN-γ release and (d) IL-2 release. Data shown as means +/-SEM from 4 independent experiments with different donors.
Fig 6. In vivo testing of GD2…
Fig 6. In vivo testing of GD2 CAR.
Balb-C mice were innocultaed with 1x106 CT26 or CT26-GD2 cell mixed in matrigel. 10 days post tumour inoculation, mice were sub-lethally irradiated (200 rads) and two weeks post tumour injection, mice were intravenously transplanted with a total of 1.5x106 transduced splenocytes or non transduced controls by tail vein injection. Wild type CT26 were used as antigen negative controls (a,c) whereas CT26 –GD2 tumors treated with untransduced T cells (b) did not regress whilst CT26-GD2 tumors challenged with CAR T cells regressed. Each line represents one mouse.

References

    1. Maris JM. Recent advances in neuroblastoma. N Engl J Med. 2010;362: 2202–2211. 10.1056/NEJMra0804577
    1. London WB, Castel V, Monclair T, Ambros PF, Pearson ADJ, Cohn SL, et al. Clinical and biologic features predictive of survival after relapse of neuroblastoma: a report from the International Neuroblastoma Risk Group project. J Clin Oncol. 2011;29: 3286–3292. 10.1200/JCO.2010.34.3392
    1. Suzuki M, Cheung N-KV. Disialoganglioside GD2 as a therapeutic target for human diseases. Expert Opin Ther Targets. 2015;: 1–14. 10.1517/14728222.2014.986459
    1. Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15: 5323–5337. 10.1158/1078-0432.CCR-09-0737
    1. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med. 2010;363: 1324–1334. 10.1056/NEJMoa0911123
    1. Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol. American Society of Clinical Oncology; 2014;32: 798–808. 10.1200/JCO.2013.51.5304
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348: 62–68. 10.1126/science.aaa4967
    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. 10.1038/nm.1882
    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. 10.1172/JCI46110
    1. Pule MA, Straathof KC, Dotti G, Heslop HE, Rooney CM, Brenner MK. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther. 2005;12: 933–941. 10.1016/j.ymthe.2005.04.016
    1. Straathof KC, Pule MA, Yotnda P, Dotti G, Vanin EF, Brenner MK, et al. An inducible caspase 9 safety switch for T-cell therapy. Blood. 2005;105: 4247–4254. 10.1182/blood-2004-11-4564
    1. Nakamura K, Tanaka Y, Shitara K, Hanai N. Construction of humanized anti-ganglioside monoclonal antibodies with potent immune effector functions. Cancer Immunol Immunother. 2001;50: 275–284.
    1. Guest RD, Hawkins RE, Kirillova N, Cheadle EJ, Arnold J, O'Neill A, et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother. 2005;28: 203–211.
    1. Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, Rader C, et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res. 2015;3: 125–135. 10.1158/2326-6066.CIR-14-0127
    1. Almåsbak H, Walseng E, Kristian A, Myhre MR, Suso EM, Munthe LA, et al. Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model. Gene Ther. 2015;22: 391–403. 10.1038/gt.2015.4
    1. Hombach A, Hombach AA, Abken H. 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 Ther. Nature Publishing Group; 2010;17: 1206–1213. 10.1038/gt.2010.91
    1. Jonnalagadda M, Mardiros A, Urak R, Wang X, Hoffman LJ, Bernanke A, et al. Chimeric antigen receptors with mutated IgG4 Fc spacer avoid fc receptor binding and improve T cell persistence and antitumor efficacy. Mol Ther. 2015;23: 757–768. 10.1038/mt.2014.208
    1. Shields RL, Namenuk AK, Hong K, Meng YG, Rae J, Briggs J, et al. High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. J Biol Chem. American Society for Biochemistry and Molecular Biology; 2001;276: 6591–6604. 10.1074/jbc.M009483200
    1. Chung JH, Bell AC, Felsenfeld G. Characterization of the chicken beta-globin insulator. Proc Natl Acad Sci USA. 1997;94: 575–580.
    1. Kurre P, Morris J, Thomasson B, Kohn DB, Kiem H- P. Scaffold attachment region-containing retrovirus vectors improve long-term proviral expression after transplantation of GFP-modified CD34+ baboon repopulating cells. Blood. 2003;102: 3117–3119. 10.1182/blood-2003-03-0962
    1. Jensen MC, Riddell SR. Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev. 2014;257: 127–144. 10.1111/imr.12139
    1. Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol. 1998;161: 2791–2797.
    1. Davila ML, Rivière I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6: 224ra25–224ra25. 10.1126/scitranslmed.3008226
    1. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368: 1509–1518. 10.1056/NEJMoa1215134
    1. Turatti F, Figini M, Alberti P, Willemsen RA, Canevari S, Mezzanzanica D. Highly efficient redirected anti-tumor activity of human lymphocytes transduced with a completely human chimeric immune receptor. J Gene Med. John Wiley & Sons, Ltd; 2005;7: 158–170. 10.1002/jgm.647
    1. Westwood JA, Smyth MJ, Teng MWL, Moeller M, Trapani JA, Scott AM, et al. Adoptive transfer of T cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice. Proc Natl Acad Sci USA. National Acad Sciences; 2005;102: 19051–19056. 10.1073/pnas.0504312102
    1. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015. 10.1038/nm.3838
    1. Hudecek M, Lupo-Stanghellini MT, Kosasih PL, Sommermeyer D, Jensen MC, Rader C, et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin Cancer Res. American Association for Cancer Research; 2013;19: 3153–3164. 10.1158/1078-0432.CCR-13-0330
    1. Zhao Z, Condomines M, van der Stegen SJC, Perna F, Kloss CC, Gunset G, et al. Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T Cells. Cancer Cell. 2015;28: 415–428. 10.1016/j.ccell.2015.09.004
    1. Zhong X-S, Matsushita M, Plotkin J, Rivière I, Sadelain M. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther. 2010;18: 413–420. 10.1038/mt.2009.210
    1. Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther. 2013;21: 904–912. 10.1038/mt.2013.17
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18: 843–851. 10.1038/mt.2010.24
    1. Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36: 133–151. 10.1097/CJI.0b013e3182829903
    1. Macarthur CC, Xue H, Van Hoof D, Lieu PT, Dudas M, Fontes A, et al. Chromatin insulator elements block transgene silencing in engineered human embryonic stem cell lines at a defined chromosome 13 locus. Stem Cells Dev. 2012;21: 191–205. 10.1089/scd.2011.0163
    1. Moreno R, Martínez I, Petriz J, Nadal M, Tintoré X, Gonzalez JR, et al. The β-interferon scaffold attachment region confers high-level transgene expression and avoids extinction by epigenetic modifications of integrated provirus in adipose tissue-derived human mesenchymal stem cells. Tissue Eng Part C Methods. 2011;17: 275–287. 10.1089/ten.TEC.2010.0383
    1. Rivière I, Brose K, Mulligan RC. Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells. Proc Natl Acad Sci USA. 1995;92: 6733–6737.
    1. Donnelly ML, Hughes LE, Luke G, Mendoza H, Dam ten E, Gani D, et al. The 'cleavage' activities of foot-and-mouth disease virus 2A site-directed mutants and naturally occurring “2A-like” sequences. J Gen Virol. 2001;82: 1027–1041. 10.1099/0022-1317-82-5-1027
    1. Cosset FL, Takeuchi Y, Battini JL, Weiss RA, Collins MK. High-titer packaging cells producing recombinant retroviruses resistant to human serum. J Virol. 1995;69: 7430–7436.

Source: PubMed

3
订阅