A novel chimpanzee serotype-based adenoviral vector as delivery tool for cancer vaccines

Daniela Peruzzi, Sridhar Dharmapuri, Agostino Cirillo, Bruno Ercole Bruni, Alfredo Nicosia, Riccardo Cortese, Stefano Colloca, Gennaro Ciliberto, Nicola La Monica, Luigi Aurisicchio, Daniela Peruzzi, Sridhar Dharmapuri, Agostino Cirillo, Bruno Ercole Bruni, Alfredo Nicosia, Riccardo Cortese, Stefano Colloca, Gennaro Ciliberto, Nicola La Monica, Luigi Aurisicchio

Abstract

The use of adenovirus (Ad) as vaccine vectors is hindered by pre-existing immunity to human Ads in most of the human population. In order to overcome this limitation, uncommon alternative Ad serotypes need to be utilized. In this study, an E1-E3 deleted recombinant Ad based on the chimpanzee serotype 3 (ChAd3) was engineered to express human carcinoembryonic antigen (CEA) protein or rat neu extracellular/transmembrane domains (ECD.TM). ChAd3 vectors were tested in CEA transgenic (CEA.Tg) and BALB/NeuT mice, which show immunologic tolerance to these antigens. ChAd3 is capable of inducing an immune response comparable to that of hAd5 serotype-based vectors, thus breaking tolerance to tumor associated antigens (TAAs) and achieving anti-tumor effects. Of importance is that ChAd3 can overcome hAd5 pre-existing immunity and work in conjunction with DNA electroporation (DNA-EP) and other Ad vaccines based on common human serotypes.

Figures

Fig. 1
Fig. 1
Schematic representation of the vectors. CEA and rat neu ECD.TM expression cassettes were inserted by homologous recombination in E1–E3 deleted hAd5 and ChAd3 vectors as indicated.
Fig. 2
Fig. 2
ChAd3 and hAd5 show comparable expression of CEA in vitro and in vivo. (A) HeLa cells were infected at the indicated ratio vp/cell. 48 h later secreted CEA was measured in the supernatant by ELISA. Grey diamonds and white square symbols show average CEA expression mediated by hAd5-CEA and ChAd3-CEA, respectively. (B) CEA.Tg mice were injected i.m. with the indicated amount (vp) of either hAd5 or ChAd3. Three days later, mice were bled and circulating CEA levels were measured by ELISA. Grey dots indicate the geometric mean of the group.
Fig. 3
Fig. 3
Characterization of the immunogenic properties of ChAd3-CEA. (A) Kinetics of the immune response. Groups of 6–8 CEA.Tg mice were immunized with 1 × 1010 vp of either hAd5-CEA or ChAd3-CEA vector. At the indicated time point, mice were bled and PBMCs were stimulated with a pool of peptides covering the C-term of CEA (pool D) and analyzed by intracellular staining for IFNγ. Group average with standard deviations (error bar) is shown. (B) Vaccine potency of ChAd3-CEA. Groups of 6 CEA.Tg mice were vaccinated with the indicated vp of either hAd5-CEA or ChAd3-CEA vector. Thirty days after the injection, PBMCs were analyzed for CEA-specific response by intracellular staining for IFNγ. Histograms and error bars show group average and standard deviations, respectively. (C) hAd5 and ChAd3 vector mixed modality. Groups of 6 CEA.Tg mice were immunized either with 108 vp of hAd5-CEA or ChAd3-CEA vector or mock injected. Thirty days later, mice were boosted with the indicated vector and 2 weeks later, PBMCs were analyzed for CEA-specific response by intracellular staining for IFNγ. White circles indicate the immune response measured per single animal; gray dots represent the group geometric mean. Mice with responses above 0.1% CD8+/IFNγ+ (dashed line) are considered responders. Student's t-test, p = 0.05 for hAd5/hAd5 vs. hAd5/ChAd3 or ChAd3/ChAd3 groups.
Fig. 4
Fig. 4
ChAd3-CEA vaccine overcomes hAd5 immunity. (A) Top scheme. Groups of CEA.Tg mice were pre-injected with 1010 vp of wild type hAd5 and immunized either with hAd5-CEA or ChAd3-CEA (1010 vp). The average (±SD) anti-hAd5 titer of the group is shown. Two weeks later, the immune response against CEA was measured by intracellular staining for IFNγ⋅ (B) Pre-exposure to wt hAd5 does not impair ChAd3 vaccine potency. Groups of 10 CEA.Tg mice received 4 weekly DNA-EP with pV1J-CEA or left untreated (mock). Two weeks after, animals were injected with 109 vp of either of the two vectors and the immune response against CEA was measured 14 days later by intracellular staining for IFNγ. (C) Kinetics of immune response. Treated mice were bled at the indicated time points and PBMCs were analyzed by intracellular staining for IFNγ. Mice with a CEA-specific response >0.1% CD8+/IFNγ+ were considered responders.
Fig. 5
Fig. 5
ChAd3-_neu.ECD.TM immunogenic properties and anti-tumor effects. (A) T-cell responses. Groups of 10 BALB/NeuT mice were vaccinated at the 8th and 9th week of age. The immune response against rat neu 15.3 epitope was analyzed at week 11 and 14. Open circles indicate the response of individual mice, filled circle denotes the group mean. A mouse with T-cell reactivity of at least 0.1% was considered a responder. Student's t-test at week 11, p < 0.05 for hAd5wt+hAd5 vs. hAd5 alone or hAd5wt+ChAd3 or ChAd3 alone. (B) Antibody titer. Anti-rat HER-2/neu titers were determined as described in materials and methods. At least 10 mice were included in each group. Open circles indicate the response of individual mice, filled circle denotes the group mean. Student's t-test, p < 0.0003 for hAd5wt+hAd5 vs. hAd5 alone or hAd5wt+ChAd3 or ChAd3 alone. (C) Anti-tumor effect. Mice were immunized as described in the text and tumor development followed from week 15 onwards. At least 10 mice were included in each group. Percentage of tumor-free mice is shown. Group symbols are shown in graph legend. Log-Rank test, hAd5wt+ChAd3-ECD.TM vs. hAd5wt+hAd5-ECD.TM: p = 6.27 × 10−06.

References

    1. Wold W.S.M., Tollefson A.E.E. 2nd ed. Humana Press; 2007. Adenovirus methods and protocols adenoviruses, Ad vectors, quantitation, and animal models.
    1. Hierholzer J.C., Wigand R., Anderson L.J., Adrian T., Gold J.W. Adenoviruses from patients with AIDS: a plethora of serotypes and a description of five new serotypes of subgenus D (types 43–47) J Infect Dis. 1988;158(4):804–813.
    1. Schnurr D., Dondero M.E. Two new candidate adenovirus serotypes. Intervirology. 1993;36(2):79–83.
    1. De Jong J.C., Wermenbol A.G., Verweij-Uijterwaal M.W., Slaterus K.W., Wertheim-Van Dillen P., Van Doornum G.J. Adenoviruses from human immunodeficiency virus-infected individuals, including two strains that represent new candidate serotypes Ad50 and Ad51 of species B1 and D, respectively. J Clin Microbiol. 1999;37(12):3940–3945.
    1. Jager L., Ehrhardt A. Emerging adenoviral vectors for stable correction of genetic disorders. Curr Gene Ther. 2007;7(4):272–283.
    1. Hartman Z.C., Appledorn D.M., Amalfitano A. Adenovirus vector induced innate immune responses: impact upon efficacy and toxicity in gene therapy and vaccine applications. Virus Res. 2008;132(1–2):1–14.
    1. He Z., Wlazlo A.P., Kowalczyk D.W., Cheng J., Xiang Z.Q., Giles-Davis W. Viral recombinant vaccines to the E6 and E7 antigens of HPV-16. Virology. 2000;270(1):146–161.
    1. Xiang Z.Q., Yang Y., Wilson J.M., Ertl H.C. A replication-defective human adenovirus recombinant serves as a highly efficacious vaccine carrier. Virology. 1996;219(1):220–227.
    1. Tims T., Briggs D.J., Davis R.D., Moore S.M., Xiang Z., Ertl H.C. Adult dogs receiving a rabies booster dose with a recombinant adenovirus expressing rabies virus glycoprotein develop high titers of neutralizing antibodies. Vaccine. 2000;18(25):2804–2807.
    1. Letvin N.L., Mascola J.R., Sun Y., Gorgone D.A., Buzby A.P., Xu L. Preserved CD4+ central memory T cells and survival in vaccinated SIV-challenged monkeys. Science. 2006;312(5779):1530–1533.
    1. Shiver J.W., Fu T.M., Chen L., Casimiro D.R., Davies M.E., Evans R.K. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature. 2002;415(6869):331–335.
    1. Sullivan N.J., Geisbert T.W., Geisbert J.B., Xu L., Yang Z.Y., Roederer M. Accelerated vaccination for Ebola virus haemorrhagic fever in non-human primates. Nature. 2003;424(6949):681–684.
    1. Priddy F.H., Brown D., Kublin J., Monahan K., Wright D.P., Lalezari J. Safety and immunogenicity of a replication-incompetent adenovirus type 5 HIV-1 clade B gag/pol/nef vaccine in healthy adults. Clin Infect Dis. 2008;46(11):1769–1781.
    1. Cox K.S., Clair J.H., Prokop M.T., Sykes K.J., Dubey S.A., Shiver J.W. DNA gag/adenovirus type 5 (Ad5) gag and Ad5 gag/Ad5 gag vaccines induce distinct T-cell response profiles. J Virol. 2008;82(16):8161–8171.
    1. Casimiro D.R., Chen L., Fu T.M., Evans R.K., Caulfield M.J., Davies M.E. Comparative immunogenicity in rhesus monkeys of DNA plasmid, recombinant vaccinia virus, and replication-defective adenovirus vectors expressing a human immunodeficiency virus type 1 gag gene. J Virol. 2003;77(11):6305–6313.
    1. Fitzgerald J.C., Gao G.P., Reyes-Sandoval A., Pavlakis G.N., Xiang Z.Q., Wlazlo A.P. A simian replication-defective adenoviral recombinant vaccine to HIV-1 gag. J Immunol. 2003;170(3):1416–1422.
    1. Roberts D.M., Nanda A., Havenga M.J., Abbink P., Lynch D.M., Ewald B.A. Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature. 2006;441(7090):239–243.
    1. Kresge K.J. Renewed promise. Annual AIDS vaccine meeting highlights recent data from clinical trials and lessons on recruitment and retention of volunteers. IAVI Rep. 2005;9(4):18–20.
    1. Guibinga G.H., Lochmuller H., Massie B., Nalbantoglu J., Karpati G., Petrof B.J. Combinatorial blockade of calcineurin and CD28 signaling facilitates primary and secondary therapeutic gene transfer by adenovirus vectors in dystrophic (mdx) mouse muscles. J Virol. 1998;72(6):4601–4609.
    1. Ye X., Robinson M.B., Pabin C., Batshaw M.L., Wilson J.M. Transient depletion of CD4 lymphocyte improves efficacy of repeated administration of recombinant adenovirus in the ornithine transcarbamylase deficient sparse fur mouse. Gene Ther. 2000;7(20):1761–1767.
    1. Singh R., Tian B., Kostarelos K. Artificial envelopment of nonenveloped viruses: enhancing adenovirus tumor targeting in vivo. FASEB J. 2008;22(9):3389–3402.
    1. Sakurai F., Kawabata K., Mizuguchi H. Adenovirus vectors composed of subgroup B adenoviruses. Curr Gene Ther. 2007;7(4):229–238.
    1. Fattori E., Zampaglione I., Arcuri M., Meola A., Ercole B.B., Cirillo A. Efficient immunization of rhesus macaques with an HCV candidate vaccine by heterologous priming-boosting with novel adenoviral vectors based on different serotypes. Gene Ther. 2006;13(14):1088–1096.
    1. Douglas J.T., Miller C.R., Kim M., Dmitriev I., Mikheeva G., Krasnykh V. A system for the propagation of adenoviral vectors with genetically modified receptor specificities. Nat Biotechnol. 1999;17(5):470–475.
    1. Belousova N., Krendelchtchikova V., Curiel D.T., Krasnykh V. Modulation of adenovirus vector tropism via incorporation of polypeptide ligands into the fiber protein. J Virol. 2002;76(17):8621–8631.
    1. Sarkioja M., Pesonen S., Raki M., Hakkarainen T., Salo J., Ahonen M.T. Changing the adenovirus fiber for retaining gene delivery efficacy in the presence of neutralizing antibodies. Gene Ther. 2008;15(12):921–929.
    1. Sailaja G., HogenEsch H., North A., Hays J., Mittal S.K. Encapsulation of recombinant adenovirus into alginate microspheres circumvents vector-specific immune response. Gene Ther. 2002;9(24):1722–1729.
    1. Barouch D.H., Pau M.G., Custers J.H., Koudstaal W., Kostense S., Havenga M.J. Immunogenicity of recombinant adenovirus serotype 35 vaccine in the presence of pre-existing anti-Ad5 immunity. J Immunol. 2004;172(10):6290–6297.
    1. Lemckert A.A., Sumida S.M., Holterman L., Vogels R., Truitt D.M., Lynch D.M. Immunogenicity of heterologous prime-boost regimens involving recombinant adenovirus serotype 11 (Ad11) and Ad35 vaccine vectors in the presence of anti-ad5 immunity. J Virol. 2005;79(15):9694–9701.
    1. Shiver J.W., Emini E.A. Recent advances in the development of HIV-1 vaccines using replication-incompetent adenovirus vectors. Annu Rev Med. 2004;55:355–372.
    1. Bangari D.S., Mittal S.K. Development of nonhuman adenoviruses as vaccine vectors. Vaccine. 2006;24(7):849–862.
    1. Wuest T., Both G.W., Prince A.M., Hofmann C., Loser P. Recombinant ovine atadenovirus induces a strong and sustained T cell response against the hepatitis C virus NS3 antigen in mice. Vaccine. 2004;22(21–22):2717–2721.
    1. McCoy K., Tatsis N., Korioth-Schmitz B., Lasaro M.O., Hensley S.E., Lin S.W. Effect of preexisting immunity to adenovirus human serotype 5 antigens on the immune responses of nonhuman primates to vaccine regimens based on human- or chimpanzee-derived adenovirus vectors. J Virol. 2007;81(12):6594–6604.
    1. Tatsis N., Tesema L., Robinson E.R., Giles-Davis W., McCoy K., Gao G.P. Chimpanzee-origin adenovirus vectors as vaccine carriers. Gene Ther. 2006;13(5):421–429.
    1. Zhi Y., Figueredo J., Kobinger G.P., Hagan H., Calcedo R., Miller J.R. Efficacy of severe acute respiratory syndrome vaccine based on a nonhuman primate adenovirus in the presence of immunity against human adenovirus. Hum Gene Ther. 2006;17(5):500–506.
    1. Zhou D., Cun A., Li Y., Xiang Z., Ertl H.C. A chimpanzee-origin adenovirus vector expressing the rabies virus glycoprotein as an oral vaccine against inhalation infection with rabies virus. Mol Ther. 2006;14(5):662–672.
    1. Perreau M., Mennechet F., Serratrice N., Glasgow J.N., Curiel D.T., Wodrich H. Contrasting effects of human, canine, and hybrid adenovirus vectors on the phenotypical and functional maturation of human dendritic cells: implications for clinical efficacy. J Virol. 2007;81(7):3272–3284.
    1. Perreau M., Kremer E.J. Frequency, proliferation, and activation of human memory T cells induced by a nonhuman adenovirus. J Virol. 2005;79(23):14595–14605.
    1. Bangari D.S., Mittal S.K. Porcine adenoviral vectors evade preexisting humoral immunity to adenoviruses and efficiently infect both human and murine cells in culture. Virus Res. 2004;105(2):127–136.
    1. Ryan S.O., Gantt K.R., Finn O.J. Tumor antigen-based immunotherapy and immunoprevention of cancer. Int Arch Allergy Immunol. 2007;142(3):179–189.
    1. Facciabene A., Aurisicchio L., Elia L., Palombo F., Mennuni C., Ciliberto G. DNA and adenoviral vectors encoding carcinoembryonic antigen fused to immunoenhancing sequences augment antigen-specific immune response and confer tumor protection. Hum Gene Ther. 2006;17(1):81–92.
    1. Facciabene A., Aurisicchio L., Elia L., Palombo F., Mennuni C., Ciliberto G. Vectors encoding carcinoembryonic antigen fused to the B subunit of heat-labile enterotoxin elicit antigen-specific immune responses and antitumor effects. Vaccine. 2007;26(1):47–58.
    1. Gallo P., Dharmapuri S., Nuzzo M., Maldini D., Cipriani B., Forni G. Adenovirus vaccination against neu oncogene exerts long-term protection from tumorigenesis in BALB/neuT transgenic mice. Int J Cancer. 2007;120(3):574–584.
    1. Mennuni C., Calvaruso F., Facciabene A., Aurisicchio L., Storto M., Scarselli E. Efficient induction of T-cell responses to carcinoembryonic antigen by a heterologous prime-boost regimen using DNA and adenovirus vectors carrying a codon usage optimized cDNA. Int J Cancer. 2005;117(3):444–455.
    1. Aurisicchio L., Mennuni C., Giannetti P., Calvaruso F., Nuzzo M., Cipriani B. Immunogenicity and safety of a DNA prime/adenovirus boost vaccine against rhesus CEA in nonhuman primates. Int J Cancer. 2007;120(11):2290–2300.
    1. Yang Z.-Y., Wyatt L.S., Kong W.-P., Moodie Z., Moss B., Nabel G.J. Overcoming immunity to a viral vaccine by DNA priming before vector boosting. J Virol. 2003;77:799–803.
    1. Thompson J.A. Carcinoembryonic antigen gene family: molecular biology and clinical perspective. J Clin Lab Anal. 1991;5:344.
    1. Slamon D.J., Clark G.M., Wong S.G., Levin W.J., Ullrich A., McGuire W.L. Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 1987;235(4785):177–182.
    1. Slamon D.J., Godolphin W., Jones L.A., Holt J.A., Wong S.G., Keith D.E. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244(4905):707–712.
    1. Cipriani B., Fridman A., Bendtsen C., Dharmapuri S., Mennuni C., Pak I. Therapeutic vaccination halts disease progression in BALB-neuT mice: the amplitude of elicited immune response is predictive of vaccine efficacy. Hum Gene Ther. 2008;19(7):670–680.
    1. Lucchini F., Sacco M.G., Hu N., Villa A., Brown J., Cesano L. Early and multifocal tumors in breast, salivary, harderian and epididymal tissues developed in MMTY-Neu transgenic mice. Cancer Lett. 1992;64(3):203–209.
    1. UKCCR Co-ordinating Committee on Cancer Research (UKCCCR) guidelines for the welfare of animals in experimental neoplasia second edition, London, WC2A 3PX. Br J Cancer. 1998
    1. Giannetti P., Facciabene A., La Monica N., Aurisicchio L. Individual mouse analysis of the cellular immune response to tumor antigens in peripheral blood by intracellular staining for cytokines. J Immunol Methods. 2006;316(1–2):84–96.
    1. Aste-Amezaga M., Bett A.J., Wang F., Casimiro D.R., Antonello J.M., Patel D.K. Quantitative adenovirus neutralization assays based on the secreted alkaline phosphatase reporter gene: application in epidemiologic studies and in the design of adenovector vaccines. Hum Gene Ther. 2004;15(3):293–304.
    1. Maxwell P. Carcinoembryonic antigen: cell adhesion molecule and useful diagnostic marker. Br J Biomed Sci. 1999;56(3):209–214.
    1. Eades-Perner A.M., van der Putten H., Hirth A., Thompson J., Neumaier M., von Kleist S. Mice transgenic for the human carcinoembryonic antigen gene maintain its spatiotemporal expression pattern. Cancer Res. 1994;54(15):4169–4176.
    1. Hance K.W., Zeytin H.E., Greiner J.W. Mouse models expressing human carcinoembryonic antigen (CEA) as a transgene: evaluation of CEA-based cancer vaccines. Mutat Res. 2005;576(1–2):132–154.
    1. Coyne C.B., Bergelson J.M. CAR: a virus receptor within the tight junction. Adv Drug Deliv Rev. 2005;57(6):869–882.
    1. Dechecchi M.C., Melotti P., Bonizzato A., Santacatterina M., Chilosi M., Cabrini G. Heparan sulfate glycosaminoglycans are receptors sufficient to mediate the initial binding of adenovirus types 2 and 5. J Virol. 2001;75(18):8772–8780.
    1. Baker A.H., McVey J.H., Waddington S.N., Di Paolo N.C., Shayakhmetov D.M. The influence of blood on in vivo adenovirus bio-distribution and transduction. Mol Ther. 2007;15(8):1410–1416.
    1. Waddington S.N., McVey J.H., Bhella D., Parker A.L., Barker K., Atoda H. Adenovirus serotype 5 hexon mediates liver gene transfer. Cell. 2008;132(3):397–409.
    1. Liu J., O’Brien K.L., Lynch D.M., Simmons N.L., La Porte A., Riggs A.M. Immune control of an SIV challenge by a T-cell-based vaccine in rhesus monkeys. Nature. 2009;457(7225):87–91.
    1. Roy S., Kobinger G.P., Lin J., Figueredo J., Calcedo R., Kobasa D. Partial protection against H5N1 influenza in mice with a single dose of a chimpanzee adenovirus vector expressing nucleoprotein. Vaccine. 2007;25(39–40):6845–6851.

Source: PubMed

3
購読する