Design of an Optimized Wilms' Tumor 1 (WT1) mRNA Construct for Enhanced WT1 Expression and Improved Immunogenicity In Vitro and In Vivo

Daphné Benteyn, Sébastien Anguille, Sandra Van Lint, Carlo Heirman, An Mt Van Nuffel, Jurgen Corthals, Sebastian Ochsenreither, Wim Waelput, Katrien Van Beneden, Karine Breckpot, Viggo Van Tendeloo, Kris Thielemans, Aude Bonehill, Daphné Benteyn, Sébastien Anguille, Sandra Van Lint, Carlo Heirman, An Mt Van Nuffel, Jurgen Corthals, Sebastian Ochsenreither, Wim Waelput, Katrien Van Beneden, Karine Breckpot, Viggo Van Tendeloo, Kris Thielemans, Aude Bonehill

Abstract

Tumor antigen-encoding mRNA for dendritic cell (DC)-based vaccination has gained increasing popularity in recent years. Within this context, two main strategies have entered the clinical trial stage: the use of mRNA for ex vivo antigen loading of DCs and the direct application of mRNA as a source of antigen for DCs in vivo. DCs transfected with mRNA-encoding Wilms' tumor 1 (WT1) protein have shown promising clinical results. Using a stepwise approach, we re-engineered a WT1 cDNA-carrying transcription vector to improve the translational characteristics and immunogenicity of the transcribed mRNA. Different modifications were performed: (i) the WT1 sequence was flanked by the lysosomal targeting sequence of dendritic cell lysosomal-associated membrane protein to enhance cytoplasmic expression; (ii) the nuclear localization sequence (NLS) of WT1 was deleted to promote shuttling from the nucleus to the cytoplasm; (iii) the WT1 DNA sequence was optimized in silico to improve translational efficiency; and (iv) this WT1 sequence was cloned into an optimized RNA transcription vector. DCs electroporated with this optimized mRNA showed an improved ability to stimulate WT1-specific T-cell immunity. Furthermore, in a murine model, we were able to show the safety, immunogenicity, and therapeutic activity of this optimized mRNA. This work is relevant for the future development of improved mRNA-based vaccine strategies K.Molecular Therapy-Nucleic Acids (2013) 2, e134; doi:10.1038/mtna.2013.54; published online 19 November 2013.

Figures

Figure 1
Figure 1
Schematic of the different WT1-encoding vectors and quality control of the in vitro transcribed mRNA. (a) The T7 promoter, 5′-UTR, 3′-UTR, human β-globin UTR, poly(A)-tail (A64 or A120), the NLS, sig, and the lysosomal targeting sequence (DC-LAMP) are shown. All pGEM constructs have an overhang of nucleotides derived from the vector backbone, which remains as 3′ attachment to the poly(A)-tail after linearization with SpeI. The pST1 vector has a free-ending poly(A)-tail after linearization with SapI. The original WT1 sequence is depicted in white and the optimized WT1 sequence in gray. (b) Electropherogram of the wild-type WT1 mRNA, representative of all mRNA samples, and (c) gel-like image of mRNA derived from the different WT1 constructs. NLS, nuclear localization sequence; sig, signal peptide; UTR, untranslated regions; WT1, Wilms' tumor 1.
Figure 2
Figure 2
Expression and presentation of WT1 protein after electroporation with mRNA derived from the different WT1-encoding vectors. (a) Intracellular staining of WT1 in K562 cells. K562 cells were electroporated with the indicated WT1 mRNA constructs and analyzed for WT1 expression by intracellular staining 4-, 24-, and 48-hour postelectroporation (open histograms). As a control for nonspecific WT1 immunoreactivity, parallel electroporations were performed with a control mRNA (pGEM/NEF-DC-LAMP), which yielded similar expression levels as the isotype controls (gray-filled histograms). This figure is representative of three independent experiments. (b) Comparison of WT1 ΔMFI (MFI of positive population substracted by the basal WT1 epxression) values of the total K562 population and the total immature DC population after electroporation with WT1-sh-DC-L-OPT mRNA encoded by the pGEM vector. Data are presented as mean ± SEM of three independent experiments (**P = 0.0081). (c) Immunocytochemical detection of WT1 in TriMix-matured, WT1 mRNA-electroporated DCs. Immunocytochemistry for WT1 was performed on DCs fixated 24 hours after coelectroporation with WT1-encoding mRNA or control mRNA. Immunocytochemical staining patterns are shown from one representative experiment out of five (upper panel). Expression data were scored according to the staining intensity (middle panel) and according to the percentage of WT1-positive cells (lower panel; data are expressed as mean ± SEM of five independent experiments). (d) TriMix-DCs were coelectroporated with the indicated WT1 mRNA constructs or with the control mRNA. Twenty-four hours later, electroporated DCs were cocultured with the WT1-specific T-cell clone for 20 hours. IFN-γ release during this coculture was measured. Results are shown as mean ± SEM of three independent experiments (*P < 0.05). (e) Human HLA-A2+ TriMix-DCs were loaded with the WT1 antigen either by electroporation with the pST1-derived WT1-sh-DC-L-OPT mRNA or by pulsing with the HLA-A2-restricted WT1126–134 peptide. Negative controls included TriMix-DCs electroporated with the control mRNA specified above or pulsed with the HLA-A2-restricted Melan-A26-35 peptide. Four, 24, or 48 hours after antigen loading, cells were used to stimulate a human HLA-A2-restricted WT1126-134-specific CD8+ T-cell clone. IFN-γ release was measured by ELISA after a 20-hour coculture. DC, dendritic cell; ELISA, enzyme-linked immunosorbent assay; IFN-γ, interferon-γ MFI, mean fluorescence intensity; SEM, standard error of the mean; WT1, Wilms' tumor 1.
Figure 3
Figure 3
Functionality of the WT1 protein encoded by the optimized vector in an in vivo mouse model. (a) In vivo CTL assay after immunization with wild-type WT1 mRNA or mRNA encoded by the optimized vector. In vivo cytolysis of the target cells was assessed ex vivo by flow cytometry analysis of CFSElo or CFSEhi cells in the spleen, injected and noninjected lymph nodes, and the peripheral blood. Data shown are from the blood and are representative for the other compartments. This figure is representative of three independent experiments. Data are presented as the percentage of specific lysis. (b) C57BL/6 mice (n = 7) were inoculated with 5 × 105 C1498-WT1 cells s.c. 7 days later, and mice were immunized intranodally with control, wild-type WT1+TriMix, or optimized WT1+TriMix mRNA. Tumor volume was assessed (**P < 0.01; ***P < 0.001). WT1, Wilms' tumor 1.
Figure 4
Figure 4
Autotoxicity-related side effects after WT1 immunization. (a) WT1 expression in the kidney under physiological conditions. (b) No proteinuria was observed in mice immunized with the optimized WT1 mRNA, indicating no damage to the kidney (n = 8 mice/group). (c) No pathological changes, such as lymphocyte infiltration or tissue destruction and repair, were observed in the kidney, lung, or liver (negative control) of WT1 mRNA-immunized mice. WT1, Wilms' tumor 1.

References

    1. Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med. 1996;184:465–472.
    1. Nair SK, Boczkowski D, Morse M, Cumming RI, Lyerly HK, Gilboa E. Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nat Biotechnol. 1998;16:364–369.
    1. Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2012;1:1111–1134.
    1. Van Nuffel AM, Benteyn D, Wilgenhof S, Corthals J, Heirman C, Neyns B, et al. Intravenous and intradermal TriMix-dendritic cell therapy results in a broad T-cell response and durable tumor response in a chemorefractory stage IV-M1c melanoma patient. Cancer Immunol Immunother. 2012;61:1033–1043.
    1. Wilgenhof S, Van Nuffel AM, Corthals J, Heirman C, Tuyaerts S, Benteyn D, et al. Therapeutic vaccination with an autologous mRNA electroporated dendritic cell vaccine in patients with advanced melanoma. J Immunother. 2011;34:448–456.
    1. Benteyn D, Van Nuffel AM, Wilgenhof S, Corthals J, Heirman C, Neyns B, et al. Characterization of CD8+ T-cell responses in the peripheral blood and skin injection sites of melanoma patients treated with mRNA electroporated autologous dendritic cells (TriMixDC-MEL). Biomed Res Int. 2013;2013:976383.
    1. Van Lint S, Goyvaerts C, Maenhout S, Goethals L, Disy A, Benteyn D, et al. Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy. Cancer Res. 2012;72:1661–1671.
    1. Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, et al. Direct injection of protamine-protected mRNA: results of a phase ½ vaccination trial in metastatic melanoma patients. J Immunother. 2009;32:498–507.
    1. Weide B, Carralot JP, Reese A, Scheel B, Eigentler TK, Hoerr I, et al. Results of the first phase I/II clinical vaccination trial with direct injection of mRNA. J Immunother. 2008;31:180–188.
    1. Bonehill A, Tuyaerts S, Van Nuffel AM, Heirman C, Bos TJ, Fostier K, et al. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Mol Ther. 2008;16:1170–1180.
    1. Bonehill A, Van Nuffel AM, Corthals J, Tuyaerts S, Heirman C, François V, et al. Single-step antigen loading and activation of dendritic cells by mRNA electroporation for the purpose of therapeutic vaccination in melanoma patients. Clin Cancer Res. 2009;15:3366–3375.
    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.
    1. Hohenstein P, Hastie ND. The many facets of the Wilms' tumour gene, WT1. Hum Mol Genet. 2006;15 Spec No 2:R196–R201.
    1. Wilsher M, Cheerala B. WT1 as a complementary marker of malignant melanoma: an immunohistochemical study of whole sections. Histopathology. 2007;51:605–610.
    1. Wagner N, Panelos J, Massi D, Wagner KD. The Wilms' tumor suppressor WT1 is associated with melanoma proliferation. Pflugers Arch. 2008;455:839–847.
    1. Yang L, Han Y, Suarez Saiz F, Saurez Saiz F, Minden MD. A tumor suppressor and oncogene: the WT1 story. Leukemia. 2007;21:868–876.
    1. Mundlos S, Pelletier J, Darveau A, Bachmann M, Winterpacht A, Zabel B. Nuclear localization of the protein encoded by the Wilms' tumor gene WT1 in embryonic and adult tissues. Development. 1993;119:1329–1341.
    1. Sugiyama H. WT1 (Wilms' tumor gene 1): biology and cancer immunotherapy. Jpn J Clin Oncol. 2010;40:377–387.
    1. Oka Y, Tsuboi A, Taguchi T, Osaki T, Kyo T, Nakajima H, et al. Induction of WT1 (Wilms' tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc Natl Acad Sci USA. 2004;101:13885–13890.
    1. Anguille S, Van Tendeloo VF, Berneman ZN. Leukemia-associated antigens and their relevance to the immunotherapy of acute myeloid leukemia. Leukemia. 2012;26:2186–2196.
    1. Van Driessche A, Berneman ZN, Van Tendeloo VF. Active specific immunotherapy targeting the Wilms' tumor protein 1 (WT1) for patients with hematological malignancies and solid tumors: lessons from early clinical trials. Oncologist. 2012;17:250–259.
    1. Oka Y, Elisseeva OA, Tsuboi A, Ogawa H, Tamaki H, Li H, et al. Human cytotoxic T-lymphocyte responses specific for peptides of the wild-type Wilms' tumor gene (WT1) product. Immunogenetics. 2000;51:99–107.
    1. Gao L, Bellantuono I, Elsässer A, Marley SB, Gordon MY, Goldman JM, et al. Selective elimination of leukemic CD34(+) progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood. 2000;95:2198–2203.
    1. Van Tendeloo VF, Van de Velde A, Van Driessche A, Cools N, Anguille S, Ladell K, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci USA. 2010;107:13824–13829.
    1. Smits EL, Anguille S, Cools N, Berneman ZN, Van Tendeloo VF. Dendritic cell-based cancer gene therapy. Hum Gene Ther. 2009;20:1106–1118.
    1. Anguille S, Lion E, Smits E, Berneman ZN, van Tendeloo VF. Dendritic cell vaccine therapy for acute myeloid leukemia: questions and answers. Hum Vaccin. 2011;7:579–584.
    1. Van Nuffel AM, Benteyn D, Wilgenhof S, Corthals J, Heirman C, Neyns B, et al. Intravenous and intradermal TriMix-dendritic cell therapy results in a broad T-cell response and durable tumor response in a chemorefractory stage IV-M1c melanoma patient. Cancer Immunol Immunother. 2012;61:1033–1043.
    1. Bonehill A, Heirman C, Tuyaerts S, Michiels A, Breckpot K, Brasseur F, et al. Messenger RNA-electroporated dendritic cells presenting MAGE-A3 simultaneously in HLA class I and class II molecules. J Immunol. 2004;172:6649–6657.
    1. De Keersmaecker B, Heirman C, Allard S, Bonehill A, Corthals J, Thielemans K, et al. Lumenal part of the DC-LAMP protein is not required for induction of antigen-specific T cell responses by means of antigen-DC-LAMP messenger RNA-electroporated dendritic cells. Hum Gene Ther. 2010;21:479–485.
    1. Van Nuffel AM, Benteyn D, Wilgenhof S, Pierret L, Corthals J, Heirman C, et al. Dendritic cells loaded with mRNA encoding full-length tumor antigens prime CD4+ and CD8+ T cells in melanoma patients. Mol Ther. 2012;20:1063–1074.
    1. Fath S, Bauer AP, Liss M, Spriestersbach A, Maertens B, Hahn P, et al. Multiparameter RNA and codon optimization: a standardized tool to assess and enhance autologous mammalian gene expression. PLoS ONE. 2011;6:e17596.
    1. Turksma AW, Bontkes HJ, Ruizendaal JJ, van den Heuvel H, Scholten KB, Santegoets SJ, et al. Increased cytotoxic capacity of tumor antigen specific human T cells after in vitro stimulation with IL21 producing dendritic cells. Hum Immunol. 2013;74:506–513.
    1. Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, et al. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood. 2006;108:4009–4017.
    1. Van Driessche A, Van de Velde AL, Nijs G, Braeckman T, Stein B, De Vries JM, et al. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy. 2009;11:653–668.
    1. Bruening W, Moffett P, Chia S, Heinrich G, Pelletier J. Identification of nuclear localization signals within the zinc fingers of the WT1 tumor suppressor gene product. FEBS Lett. 1996;393:41–47.
    1. Coosemans A, Wölfl M, Berneman ZN, Van Tendeloo V, Vergote I, Amant F, et al. Immunological response after therapeutic vaccination with WT1 mRNA-loaded dendritic cells in end-stage endometrial carcinoma. Anticancer Res. 2010;30:3709–3714.
    1. Niksic M, Slight J, Sanford JR, Caceres JF, Hastie ND. The Wilms' tumour protein (WT1) shuttles between nucleus and cytoplasm and is present in functional polysomes. Hum Mol Genet. 2004;13:463–471.
    1. Van Driessche A, Van de Velde AL, Nijs G, Braeckman T, Stein B, De Vries JM, et al. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy. 2009;11:653–668.
    1. Kreiter S, Selmi A, Diken M, Sebastian M, Osterloh P, Schild H, et al. Increased antigen presentation efficiency by coupling antigens to MHC class I trafficking signals. J Immunol. 2008;180:309–318.
    1. Rock KL, Farfán-Arribas DJ, Shen L. Proteases in MHC class I presentation and cross-presentation. J Immunol. 2010;184:9–15.
    1. Apcher S, Manoury B, Fåhraeus R. The role of mRNA translation in direct MHC class I antigen presentation. Curr Opin Immunol. 2012;24:71–76.
    1. Apcher S, Daskalogianni C, Lejeune F, Manoury B, Imhoos G, Heslop L, et al. Major source of antigenic peptides for the MHC class I pathway is produced during the pioneer round of mRNA translation. Proc Natl Acad Sci USA. 2011;108:11572–11577.
    1. Kuhn AN, Diken M, Kreiter S, Vallazza B, Türeci Ö, Sahin U. Determinants of intracellular RNA pharmacokinetics: Implications for RNA-based immunotherapeutics. RNA Biol. 2011;8:35–43.
    1. Depping R, Schindler SG, Jacobi C, Kirschner KM, Scholz H. Nuclear transport of Wilms' tumour protein Wt1 involves importins a and ß. Cell Physiol Biochem. 2012;29:223–232.
    1. Van der Bruggen PS, Vigneron N, Van den Eynde B.Peptide database at < >
    1. Reits E, Griekspoor A, Neijssen J, Groothuis T, Jalink K, van Veelen P, et al. Peptide diffusion, protection, and degradation in nuclear and cytoplasmic compartments before antigen presentation by MHC class I. Immunity. 2003;18:97–108.
    1. Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, et al. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res. 2010;70:9031–9040.
    1. Tuyaerts S, Noppe SM, Corthals J, Breckpot K, Heirman C, De Greef C, et al. Generation of large numbers of dendritic cells in a closed system using Cell Factories. J Immunol Methods. 2002;264:135–151.
    1. Holtkamp S, Kreiter S, Selmi A, Simon P, Koslowski M, Huber C, et al. Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells. Blood. 2006;108:4009–4017.
    1. Michiels A, Tuyaerts S, Bonehill A, Corthals J, Breckpot K, Heirman C, et al. Electroporation of immature and mature dendritic cells: implications for dendritic cell-based vaccines. Gene Ther. 2005;12:772–782.
    1. Allard SD, Pletinckx K, Breckpot K, Heirman C, Bonehill A, Michiels A, et al. Functional T-cell responses generated by dendritic cells expressing the early HIV-1 proteins Tat, Rev and Nef. Vaccine. 2008;26:3735–3741.
    1. Ho WY, Nguyen HN, Wolfl M, Kuball J, Greenberg PD. In vitro methods for generating CD8+ T-cell clones for immunotherapy from the naïve repertoire. J Immunol Methods. 2006;310:40–52.
    1. Van Beneden K, Geers C, Pauwels M, Mannaerts I, Wissing KM, Van den Branden C, et al. Comparison of trichostatin A and valproic acid treatment regimens in a mouse model of kidney fibrosis. Toxicol Appl Pharmacol. 2013;271:276–284.

Source: PubMed

3
Suscribir