WO2016086980A1 - Vaccine composition - Google Patents

Vaccine composition Download PDF

Info

Publication number
WO2016086980A1
WO2016086980A1 PCT/EP2014/076310 EP2014076310W WO2016086980A1 WO 2016086980 A1 WO2016086980 A1 WO 2016086980A1 EP 2014076310 W EP2014076310 W EP 2014076310W WO 2016086980 A1 WO2016086980 A1 WO 2016086980A1
Authority
WO
WIPO (PCT)
Prior art keywords
chikv
mva
vaccine
vector
cells
Prior art date
Application number
PCT/EP2014/076310
Other languages
French (fr)
Inventor
Peter LILJESTRÖM
Mariano Esteban RODRIGUEZ
Juan GARCÍA ARRIAZA
Original Assignee
Consejo Superior De Investigaciones Cientificas
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Consejo Superior De Investigaciones Cientificas filed Critical Consejo Superior De Investigaciones Cientificas
Priority to PCT/EP2014/076310 priority Critical patent/WO2016086980A1/en
Publication of WO2016086980A1 publication Critical patent/WO2016086980A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/36011Togaviridae
    • C12N2770/36111Alphavirus, e.g. Sindbis virus, VEE, EEE, WEE, Semliki
    • C12N2770/36134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a vaccine composition for protection against Chikungunya virus infection. Aspects of the invention relate to methods for protecting subjects against Chikungunya virus infection, and to compositions for use in the preparation of a vaccine composition.
  • Chikungunya is a disease caused by the Chikungunya virus (CHIKV) with a mortality rate of about 1 :1000.
  • the disease is characterized by skin rash, high fever, headache, vomiting, myalgia, and, mainly, polyarthralgia. Most of the symptoms resolve after 10 days, but the polyarthralgia can persist for months or years, and severe symptoms, such as encephalitis, hemorrhagic disease, and mortality, have also been described.
  • the disease is transmitted to humans by Aedes mosquito vectors.
  • CHIKV infection has been reported from Africa, islands in the Indian Ocean, India, Southeast Asia, and southern Europe, and recently in the Americas where over 700,000 individuals are estimated to have encountered CHIKV.
  • CHIKV contains a positive, single-stranded RNA genome of around 1 1 .8 kb which encodes four nonstructural and five structural proteins.
  • the nonstructural proteins (nsP1 , nsP2, nsP3, and nsP4) are required for virus replication.
  • the structural proteins are cleaved by capsid (C) autoproteinase and signalases from a polyprotein precursor to generate the C and envelope (pE2, 6K, and E1 ) proteins.
  • C capsid
  • pE2 precursor is cleaved to E3 and E2 by the furin protease in the trans-Golgi during transport of the envelope complex to the cell surface where virus assembly occurs.
  • pE2 is also known as p62.
  • Virions are 70-nm enveloped particles containing 240 heterodimers of E1/E2 glycoproteins on their surfaces.
  • Chikungunya nor are there currently any approved vaccines, although a number of candidate vaccines are in development. It would be desirable to develop a vaccine to protect against CHIKV.
  • RRV Ross River Virus
  • Described herein is a vaccine candidate against CHIKV, based on an attenuated poxvirus vector expressing the CHIKV structural genes.
  • the vaccine was described for the first time in J. Virol. 2014, 88(6):3527, Garcia-Arriaza et al, "A Novel Poxvirus Based Vaccine, MVA-CHIKV, Is Highly Immunogenic and Protects Mice against Chikungunya Infection", published ahead of print 8 January 2014. The contents of this publication are incorporated herein by reference.
  • compositions for use in the production of a vaccine comprising a nucleic acid comprising a nucleotide sequence encoding an attenuated poxvirus vector and one or more CHIKV structural proteins.
  • the nucleotide sequence encodes two, three, four, or five CHIKV structural proteins; most preferably five structural proteins.
  • the structural proteins may be selected from the group consisting of the C, E3, E2, 6K, and E1 proteins.
  • the nucleotide sequence encodes at least the E3 and E2 proteins; most preferably the nucleotide sequence encodes all of the C, E3, E2, 6K, and E1 proteins.
  • the CHIKV sequences can be derived from virus isolates which may differ in nucleotide sequence from one another and from neutralization resistant mutants.
  • the nucleotide sequence may further encode one or more replicase proteins (that is, one or more of nsP1 , nsP2, nsP3, and nsP4).
  • the sequence may encode two, three, or four of said replicase proteins. It is possible that host immune system responses against the replicase may further enhance protectivity of the vaccine, for example by reducing long term infection of macrophages by CHIKV.
  • the CHIKV nucleotide sequences can be derived from one or more virus isolates.
  • the isolates may be selected such that they differ from one another to the extent that a neutralisation response by sera generated from one isolate does not neutralise a second isolate. Including sequences from a number of isolates improves the chances of an immune response generated by the vaccine being effective against multiple strains of CHIKV. Sequences from different isolates may be combined in a single poxvirus vector, or multiple vectors representing different CHIKV isolates may be combined in a single vaccine composition.
  • the nucleic acid may be DNA, RNA, or a combination thereof, and can include any combination of naturally occurring, chemically modified or enzymatically modified nucleotides.
  • the nucleic acid of the invention may take the form of an expression vector that is capable of expression in an organism or in a cell of the organism, in culture or in vivo.
  • An organism or cell in which the coding sequence of the vector can be expressed can be eukaryotic or prokaryotic, and can be, without limitation, a bacterium, a yeast, an insect, a protozoan, or animals, such as a human, a bird, or a non-human mammal.
  • the nucleotide sequence of the nucleic acid may be codon optimised for expression in a particular organism or cell; for example, humans.
  • nucleic acids of the invention can be single-stranded or double-stranded, and further that a single-stranded nucleic acid of the invention includes a polynucleotide fragment having a nucleotide sequence that is complementary to a nucleotide sequence that encodes said poxvirus vector and structural proteins according to the invention.
  • complementary refers to the ability of two single stranded polynucleotide fragments to base pair with each other.
  • a single-stranded nucleic acid of the invention also includes a polynucleotide fragment having a nucleotide sequence that is substantially complementary to a nucleotide sequence that encodes said poxvirus vector and structural proteins according to the invention, or to the complement of the nucleotide sequence that encodes said poxvirus vector and structural proteins.
  • Substantially complementary polynucleotide fragments can include at least one base pair mismatch, such that at least one nucleotide present on a first polynucleotide fragment will not base pair to at least one nucleotide present on a second polynucleotide fragment, however the two polynucleotide fragments will still have the capacity to hybridize.
  • the present invention therefore encompasses polynucleotide fragments that are substantially complementary.
  • Two polynucleotide fragments are substantially complementary if they hybridize under hybridization conditions exemplified by 2xSSC (SSC: 150 mM NaCI, 15 mM trisodium citrate, pH 7.6) at 55 e C.
  • Substantially complementary polynucleotide fragments for purposes of the present invention preferably share at least about 85% nucleotide identity, preferably at least about 90%, 95% or 99% nucleotide identity. Locations and levels of nucleotide sequence identity between two nucleotide sequences can be readily determined using, for example, CLUSTALW multiple sequence alignment software.
  • the poxvirus vector may be a vaccinia virus; preferably a modified vaccinia virus Ankara (MVA).
  • the MVA vector is further modified by deletion of one or more vaccinia immunomodulatory genes; preferably one, two, or three of the genes C6L, K7R, and A46R. Most preferably, all three genes are deleted. Deletion of these genes has been shown to enhance the innate immune response triggered by the viral vector, and potentially also specific immune response.
  • Alternative attenuated poxvirus vectors include NYVAC, an isolate of the Copenhagen vaccinia vaccine strain.
  • a further aspect of the invention provides an attenuated poxvirus vector expressing one or more CHIKV structural genes. Also provided is a vaccine composition comprising such an attenuated poxvirus vector.
  • the vaccine composition may also be used in combination with other vectors expressing CHIKV genes and protein components, and/or as a combination vaccine against other infections.
  • a combination vaccine may comprise one or more adjuvants. Preferred adjuvants include AS03, or Matrix - M.
  • a vaccine of the invention is conveniently administered to subjects using ingestion, topical administration, or direct injection, preferably subcutaneous or intramuscular injection.
  • the subject is preferably mammalian, more preferably human, although alternative subjects may be treated, for example, in the course of vaccine or disease research.
  • the dose of vaccine to be administered to a subject depends on the species and size of subject, the nature of the condition being treated, and can be readily determined by one of skill in the art.
  • the invention further provides an attenuated poxvirus vector expressing one or more CHIKV structural genes for use as a vaccine. Also provided is the use of an attenuated poxvirus vector expressing one or more CHIKV structural genes in the manufacture of a vaccine.
  • the vaccine is intended for use in protecting a subject against CHIKV infection.
  • the vaccine may also or instead be for use in protecting a subject against RRV infection.
  • a still further aspect of the invention provides a method of immunising a subject against CHIKV infection, the method comprising administering a vaccine comprising an attenuated poxvirus vector expressing one or more CHIKV structural genes to a subject.
  • a single administration may be sufficient to provide suitable immunization, but in preferred embodiments, more than one dose of vaccine is administered.
  • a first dose may be followed by a booster dose after one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months or longer intervals.
  • the first dose of the attenuated poxvirus vector vaccine may be followed by a second dose of an alternative vaccine; a so-called heterologous prime-boost protocol.
  • the attenuated poxvirus vector vaccine may be administered second, after an initial dose of an alternative vaccine.
  • the alternative vaccine may comprise a covalently linked p62-E1 heterodimer (E1 being the CHIKV E1 protein, and p62 being the CHIKV precursor protein which is cleaved during virus replication into the E2 and E3 proteins).
  • the alternative vaccine may comprise a DNA- replicon (DREP) based vaccine; this may comprise a nucleotide sequence comprising CHIKV genomic sequences operably linked to a promoter sequence.
  • DREP DNA- replicon
  • the genomic sequences may comprise genes coding for the envelope proteins (E3, E2, 6K, and E1 ). Preferably the genomic sequences do not include genes coding for the capsid (C) protein; in the absence of this protein, the DREP-CHIKV is unable to form viral particles, and is only able to express envelope proteins on the host cell surface.
  • the first dose is of DREP-CHIKV vaccine
  • the second is of the attenuated poxvirus vector.
  • the second dose may be combined with a dose of p62 - E1 vaccine.
  • the invention may further provide a kit comprising first and second doses of CHIKV vaccine.
  • the first dose is preferably a vaccine comprising an attenuated poxvirus vector expressing one or more CHIKV structural genes.
  • the second dose may be the same as the first dose, or may be a vaccine comprising a covalently linked p62-E1 heterodimer, or a DREP-CHIKV vaccine.
  • “First” and “second” here do not necessary refer to the order in which the vaccines are to be administered.
  • the doses may be provided in a suitable pharmaceutical container; for example, sealed vials, or alternatively as preloaded syringes.
  • FIG 1 Generation and in vitro characterization of recombinant MVA-CHIKV.
  • A Scheme of the MVA-CHIKV genome map. The different regions are indicated by capital letters. The right and left terminal regions are shown. Below the map, the deleted or fragmented genes are depicted as black boxes.
  • the CHIKV C, E3, E2, 6K, and E1 structural genes (from CHIKV isolate LR2006-OPY1 ), driven by the synthetic early/late (sE/L) virus promoter and inserted within the VACV TK viral locus (J2R), are indicated.
  • the deleted VACV C6L, K7R, and A46R genes are also indicated.
  • TK-L TK left; TK-R, TK right.
  • B PCR analysis of the VACV TK locus. Viral DNA was extracted from chick DF-1 cells mock infected or infected at 5 PFU/cell with MVA-CHIKV, MVA-GFP, or MVA-WT. Primers spanning the TK locus-flanking regions were used for PCR analysis of the CHIKV genes inserted within the TK locus. DNA products with their corresponding sizes (base pairs) are indicated by an arrow on the right. A molecular size marker (1 -kb ladder) with the corresponding sizes (base pairs) is indicated on the left.
  • C PCR analysis of the C6L, K7R, and A46R loci.
  • Viral DNA was extracted from DF-1 cells mock infected or infected at 5 PFU/cell with MVA-CHIKV, MVA-GFP, or MVA-WT. Primers spanning C6L-, K7R-, and A46R-flanking regions were used for PCR analysis of the C6L, K7R, and A46R loci, respectively. DNA products with the corresponding size (base pairs) of the parental virus (wt) or the virus with the C6L, K7R, and A46R deletions are indicated by an arrow on the right. A molecular size marker (1 -kb ladder) with the corresponding sizes (base pairs) is indicated on the left. (C) Expression of CHIKV E1 and E2 proteins.
  • DF-1 cells were mock infected or infected at 5 PFU/cell with MVA-CHIKV, MVA-GFP, or MVA-WT.
  • cells were lysed in Laemmli buffer, fractionated by 10% SDS-PAGE, and analyzed by Western blotting using rabbit polyclonal antibody against E1 or mouse polyclonal antibody against E2.
  • Arrows on the right indicate the positions of the CHIKV E1 and E2 proteins, with the corresponding sizes in kDa.
  • the sizes of standards (in kDa) Precision Plus protein standards; Bio-Rad Laboratories) are indicated on the left.
  • FIG 2 Viral growth kinetics and stability of MVA-CHIKV.
  • A Viral growth kinetics. Monolayers of DF-1 cells were infected at 0.01 PFU/cell with MVA-WT or MVA-CHIKV. At different times postinfection (0, 24, 48, and 72 h), cells were collected and virus titers in cell lysates were quantified by a plaque immunostaining assay with anti-WR antibodies. The means of results from two independent experiments are shown.
  • DF-1 cells were mock infected or infected with MVA-CHIKV from the different passages or with MVA-WT.
  • cells were lysed in Laemmli buffer, fractionated by 10% SDS-PAGE, and analyzed by Western blotting using rabbit polyclonal antibody against E1 or mouse polyclonal antibody against E2.
  • Rabbit anti-B-actin antibody was used as a protein loading control.
  • Rabbit anti-VACV early E3 protein antibody was used as a VACV loading control.
  • Arrows on the right indicate the positions of the CHIKV E1 and E2 proteins, B-actin, and the VACV E3 protein, with their corresponding sizes in kDa.
  • MVA-CHIKV Genetic stability of MVA-CHIKV, as determined by plaque immunostaining. Virus titers of MVA-CHIKV (P2 stock, P3 stock, and passage 9) were quantified by a plaque immunostaining assay with anti-VACV and anti-CHIKV antibodies. The means and standard deviations of results from two independent determinations are shown. It should be noted that the P3 stock represents virus purified with two sucrose cushions, while P2 and passage 9 stocks are virus titers from crude cell lysates.
  • FIG 3 Immunofluorescence analysis of CHIKV E2 protein produced in HeLa cells infected with MVA-CHIKV.
  • Subconfluent HeLa cells mock infected or infected with MVA-CHIKV or MVA-WT were fixed at 6 and 16 hpi (MVA-WT- and mock-infected cells are represented at 16 hpi), labeled with the corresponding primary antibodies or probes, monitored for appropriate fluorescence from protein-conjugated secondary antibodies, and visualized by confocal microscopy.
  • the antibodies or probes used were anti-E2 (D3-62; to detect CHIKV E2 protein, shown in green), phalloidin (to stain actin, shown in red), anti-calnexin (to detect the ER, shown in red), WGA(to detect Golgi apparatus membranes and the plasma membrane, shown in red), and DAPI (to mark DNA, shown in blue). Bar, 10 urn.
  • FIG 4 Architecture of HeLa cells following infection with MVA-CHIKV or MVA-WT.
  • HeLa cells infected with MVA-WT or MVA-CHIKV were chemically fixed at 6 hpi and then processed for conventional embedding in an epoxy resin and observed by electron microscopy.
  • A General overview of a cell infected with MVA-WT (left) and MVA-CHIKV (right);
  • B two high-magnification panels of a cell infected with MVA- CHIKV.
  • m mitochondria
  • IV immature virus
  • GM Golgi apparatus membranes. Bars:
  • FIG 5 MVA-CHIKV triggers an innate immune response in human macrophages and dendritic cells, upregulating type I IFN, proinflammatory cytokines, and chemokine expression.
  • Human THP-1 macrophages (A) and moDCs (B) were mock infected or infected with MVA-WT, MVA-GFP, or MVA-CHIKV at 5 PFU/cell (A) and 1 PFU/cell (B).
  • P values indicate significantly higher responses in comparisons of MVA-WT to MVA-GFP or MVA-CHIKV at the same hour ( * , P ⁇ 0.05; ** , P ⁇ 0.005; *** , P ⁇ 0.001 ).
  • Data are means +/- standard deviations of results with duplicate samples from one experiment and are representative of two independent experiments.
  • FIG 6 Immunization with MVA-CHIKV induces strong, broad, and polyfunctional adaptive CHIKV-specific T cell immune responses.
  • Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA-CHIKV 10 days after the last immunization.
  • adaptive, CHIKV-specific CD4 and CD8 T cell immune responses triggered by both immunization groups were measured by the ICS assay following stimulation of splenocytes with different CHIKV peptides (C, E1 , and E2). Values from unstimulated controls were subtracted in all cases.
  • Frequencies were calculated by reporting the number of CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 and are specific for each peptide to the total number of CD8 T cells in the MVA- CHIKV/MVA-CHIKV immunization group.
  • C Magnitudes of C, E1 , and E2 CHIKV- specific CD8 T cells. Frequencies represent the sums of the percentages of CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against the C, E1 , or E2 peptide.
  • Each slice corresponds to the proportion of CHIKV-specific CD8 T cells exhibiting one, two, three, or four functions within the total population of CHIKV- specific CD8 T cells.
  • FIG 7 Adaptive, VACV-specific T cell immune responses.
  • Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA- CHIKV 10 days after the last immunization.
  • adaptive, VACV-specific CD4 and CD8 T cell immune responses triggered by both immunization groups were measured by ICS assay following stimulation of splenocytes with MVA-infected EL4 cells. Values from unstimulated controls were subtracted in all cases. Data are from one experiment that is representative of two independent experiments.
  • A Overall magnitude of VACV- specific CD4 and CD8 T cells.
  • the values represent the sums of the percentages of T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells.
  • B Percentages of VACV-specific CD8 T cells producing CD107a, IFN- ⁇ , TNF-a, or IL-2. Frequencies represent percentages of CD8 T cells producing CD107a, IFN- ⁇ , TNF-a, or IL-2 against MVA-infected EL4 cells.
  • C Functional profiles of adaptive, VACV-specific CD8 T cell immune responses.
  • CHIKV-specific CD4 and CD8 memory T cell immune responses triggered by both immunization groups were measured by ICS assay as described in the legend to Fig. 6. Values from unstimulated controls were subtracted in all cases. P values indicate significantly higher responses in comparisons of MVA- WT/MVA-WT to MVA-CHIKV/MVA-CHIKV ( *** , P ⁇ 0.001 ). Data are from one experiment that is representative of two independent experiments. (A) Overall magnitudes of CHIKV-specific CD4 and CD8 T cells.
  • the values represent the sums of the percentages of T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against C plus E1 plus E2.
  • B Pattern of C, E1 , and E2 CHIKV-specific CD8 memory T cell immune responses. Frequencies were calculated by reporting the number of CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 and are specific for each peptide to the total number of CD8 T cells in the MVA-CHIKV/MVA-CHIKV immunization group.
  • C Magnitudes of C, E1 , and E2 CHIKV-specific CD8 T cells.
  • Frequencies represent the sums of the percentages of CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against the C, E1 , or E2 peptide.
  • D Magnitudes of CHIKV-specific CD8 T cells producing CD107a, IFN- ⁇ , TNF-a or IL-2. Frequencies represent percentages of CD8 T cells producing CD107a, IFN- ⁇ , TNF-a, or IL-2 against C plus E1 plus E2.
  • E Polyfunctional profile of CHIKV-specific CD8 memory T cell immune responses.
  • CD127 and CD62L expression was used to identify central memory (TCM, CD127 CD62), effector memory (TEM, CD127 CD62L), and effector (TE, CD127CD62L) subpopulations.
  • TCM, CD127 CD62 central memory
  • TEM, CD127 CD62L effector memory
  • TE, CD127CD62L effector subpopulations.
  • Magnitudes of TCM, TEM, and TE CHIKV-specific CD8 T cells are represented.
  • the values represent percentages of TCM, TEM, and TE populations producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against the C plus E1 plus E2 peptides.
  • the pie charts summarize the data.
  • Each slice corresponds to the proportion of the TCM, TEM, or TE population within the total CHIKV-specific CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2.
  • P values indicate significantly higher responses in comparisons of MVA-WT/MVA-WT to MVA-CHIKV/MVA-CHIKV ( *** , P ⁇ 0.001 ).
  • FIG 9 VACV-specific memory T cell immune responses. Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA- CHIKV 52 days after the last immunization.
  • VACV-specific CD4 and CD8 memory T cell immune responses triggered by both immunization groups were measured by ICS assay following stimulation of splenocytes with MVA-infected EL4 cells. Values from unstimulated controls were subtracted in all cases. P values indicate significantly higher responses in comparisons of MVA-WT/MVA-WT to MVA- CHIKV/MVA-CHIKV ( *** , P ⁇ 0.001 ). Data are from one experiment that is representative of two independent experiments. (A) Overall magnitude of VACV- specific CD4 and CD8 memory T cells.
  • the values represent the sums of the percentages of T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells.
  • B Percentage of VACV-specific CD8 T memory cells producing CD107a, IFN- ⁇ , TNF-a, or IL-2. Frequencies represent percentages of CD8 memory T cells producing CD107a, IFN- ⁇ , TNF-a, or IL-2 against MVA-infected EL4 cells.
  • C Functional profiles of VACV-specific CD8 memory T cell immune responses. Responses are grouped and color coded on the basis of the number of functions (4, 3, 2, or 1 ).
  • CD127 and CD62L expression was used to identify central memory (TCM, CD127 CD62), effector memory (TEM, CD127 CD62L), and effector (TE, CD127CD62L) subpopulations.
  • the memory T cell subpopulations are depicted as density plots. Blue dots represent T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2.
  • the graph represents the magnitudes of TCM, TEM, and TE VACV-specific CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2.
  • Frequencies represent percentages of TCM, TEM, and TE populations producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells.
  • the pie charts summarize the data. Each slice corresponds to the proportion of the TCM, TEM, or TE population within the total population of VACV-specific CD8 T cells producing CD107a and/or IFN- ⁇ and/or TNF-a and/or IL-2.
  • FIG 10 Immunization with MVA-CHIKV induces neutralizing antibodies against CHIKV and protects mice against CHIKV infection.
  • Five C57BL/6 mice per group were immunized as described in Materials and Methods. Data are from one experiment that is representative of two independent experiments.
  • CHIKV-neutralizing antibody titers detected at 6 weeks after the last immunization in sera of animals immunized with one dose of MVA- WT or one or two doses of MVA-CHIKV.
  • the detection limit is 100.
  • C Viremia detected in animals immunized with one dose of MVA-WT or one or two doses of MVA- CHIKV and challenged with a total of 10 6 PFU of CHIKV subcutaneously in both feet. Blood samples were collected at days 1 to 3 postchallenge from tail bleedings, and viremia was analyzed by plaque assay (PFU/ml). The detection limit is 250 PFU/ml.
  • FIG 1 CHIKV specific antibody responses induced by different CHIKV vaccine candidates using various prime-boost immunization strategies.
  • a Kruskal-Wallis test followed by Dunn's posttest was used to compare responses from CHIKV (WT). * , ** and *** indicate statistical differences of p ⁇ 0.05, p ⁇ 0.01 and p ⁇ 0.001 , respectively.
  • FIG 12 Viremia and foot swelling after CHIKV challenge. Mice were challenged seven weeks post the last immunization with 10 6 PFU of CHIKV in the feet.
  • A Viremia in serum collected two days after challenge.
  • a Kruskal-Wallis test followed by Dunn's post-test was used to compare responses from nal ' ve mice (PBS) and a Spearman rank test was used to examine correlation between the magnitude of antibody responses prior to challenge and the level of viremia and foot swelling post challenge.
  • FIG 13 Induction of CD8+ T cells with different CHIKV vaccine candidates.
  • Groups of 5 C57BL/6 mice were infected once with wild-type CHIKV, or primed with DREP-Env and boosted 3 weeks later with DREP-Env, p62-E1 protein antigen and/or MVA-CHIKV, as indicated. Mice were sacrificed, and spleens were collected 8 days after the last immunization.
  • CHIKV E1 peptide (HSMTNAVTI)-specific CD8+ T cells were identified by pentamer staining and characterized for the expression of CD62L/CD127 and CD27/CD43. DETAILED DESCRIPTION OF THE INVENTION
  • EXAMPLE 1 - MVA-CHIKV is highly immunogenic and protects mice against Chikungunya infection MATERIALS AND METHODS
  • the poxvirus strain used in this study as the parental virus for the generation of the recombinant MVA-CHIKV is a modified vaccinia virus Ankara (MVA) strain (obtained from the Ankara strain after 586 serial passages in CEFs; this strain is derived from clone F6 at passage 585, kindly provided by G. Sutter, Germany).
  • MVA modified vaccinia virus Ankara
  • GFP green fluorescent protein
  • TK thymidine kinase locus
  • VACV immunomodulatory vaccinia virus
  • the GFP cassette in the TK locus was replaced by the CHIKV cassette to generate MVA-CHIKV.
  • the parental attenuated MVA (referred to as wild-type MVA [MVA-WT]). All viruses were grown in primary CEFs, purified by centrifugation through two 36% (wt/vol) sucrose cushions in 10 mM Tris-HCI, pH 9, and titrated in DF-1 cells by a plaque immunostaining assay, as previously described. The titer determinations of the different viruses were performed at least three times. All viruses were free of contamination with mycoplasmas, fungi, or bacteria.
  • CHIKV infectious CHIKV clone LR2006-OPY1
  • a plasmid for production of CHIKV was generated by cloning the CHIKV cDNA under the control of the SP6 RNA polymerase promoter.
  • CHIKV was subsequently produced in BHK-21 cells as previously described.
  • the plasmid transfer vector pCyA20-ICRESl was generated and used for the construction of recombinant MVA-CHIKV, in which CHIKV structural genes (those for C, E3, E2, 6K, and E1 ) instead of the GFP gene were inserted into the TK locus of the parental MVA-GFP.
  • a 3.7-kbp DNA fragment encoding the CHIKV structural proteins (C, E3, E2, 6K, and E1 ) was amplified with oligonucleotides ICRES1 upper (5'-ATATA GTTTAAACATGGAGTTCATCCCAACCCAAAC-3': Pmel site underlined) and ICRES1 lower (5'-TATATAGATCTTTAGTGCCTGCTGAACGACACG-3'; Bglll site underlined) from plasmid pSP6-ICRES1 , which contains the structural genes from the CHIKV genome (CHIKV isolate LR2006-OPY1 ).
  • the amplification reaction was performed with Platinum Pfx DNA polymerase (Invitrogen) according to the manufacturer's recommendations. Then, this fragment was digested with Pmel and Bglll and cloned into the VACV insertion vector pCyA-20 (7,582 bp), previously digested with the same restriction enzymes and dephosphorylated by incubation with shrimp alkaline phosphatase, to generate the plasmid transfer vector pCyA-ICRES1 (1 1 ,324 bp).
  • the plasmid pCyA-20 used for the generation of plasmid pCyA-ICRES1 , was previously described and contains a synthetic band containing the viral early/late promoter and a multiple-cloning site in the plasmid pLZAWI (provided by Sanofi-Pasteur). The correct generation of pCyA-ICRES1 was further confirmed by DNA sequence analysis.
  • This plasmid was used in transient infection and transfection assays for the insertion of CHIKV structural genes (those for C, E3, E2, 6K, and E1 ) into the TK locus of the MVA genome under the transcriptional control of the viral synthetic early/late (sE/L) promoter.
  • B-Gal B-galactosidase
  • DF-1 cells (3 x 10 6 cells) were infected with parental MVA-GFP at a multiplicity of infection (MOI) of 0.05 PFU/cell and transfected 1 h later with 10 ug of DNA of plasmid pCyA-ICRES1 , using Lipofectamine reagent according to the manufacturer's recommendations (Invitrogen).
  • MOI multiplicity of infection
  • hpi the cells were harvested, lysed by freeze-thaw cycling, sonicated, and used for recombinant-virus screening.
  • Recombinant MVAs containing the 3.7-kb CHIKV DNA fragment (encoding the structural proteins of CHIKV, C, E3, E2, 6K, and E1 ), with the GFP gene deleted, and transiently coexpressing the B-Gal marker gene (MVA-CHIKV, X-Gal+) were selected by consecutive rounds of plaque purification in DF-1 cells stained with X-Gal (5-bromo-4-chloro-3-indolyl-B-D-galactopyranoside, 400 ug/ml, for three passages in total).
  • recombinant MVAs containing the 3.7-kb CHIKV DNA fragment and with the B-Gal gene deleted by homologous recombination from between the left TK arm and the short left TK arm repeat flanking the marker (MVA-CHIKV, X-Gal-) were isolated by three additional consecutive rounds of plaque purification screening for nonstaining viral foci in DF-1 cells in the presence of X-Gal (400 ug/ml).
  • the isolated plaques were expanded in DF-1 cells for 3 days, and the crude viruses obtained were used for the next plaque purification round.
  • the resulting recombinant virus, MVA-CHIKV (passage 2 [P2] stock) was grown in CEFs, purified through two 36% (wt/vol) sucrose cushions, and titrated by plaque immunostaining assay.
  • CHIKV proteins from recombinant MVA-CHIKV by Western blotting.
  • monolayers of DF-1 cells were mock infected or infected at 5 PFU/cell with MVA-WT, MVA-GFP, or MVACHIKV.
  • mice were lysed in Laemmli buffer, and cells extracts were fractionated in 10% SDS-PAGE and analyzed by Western blotting with mouse polyclonal antibody against E2 (antibody 3E4, kindly provided by Philippe Despres from the Pasteur Institute, Paris, France; diluted 1 :500), mouse monoclonal antibody against E1 -E2 (antibody D3-62; kindly provided by Marc Lecuit and Therese Couderc from the Pasteur Institute, Paris, France; diluted 1 :500), or rabbit polyclonal antibody against E1 (kindly provided by Lisa F. P.
  • CHIKV peptides representative of the capsid (C), E1 , and E2 CHIKV proteins were used in the immunological analysis. Peptides sequences are as follows: ACLVGDJVM (capsid), HSMTNAAVTI (E1 ), and IILYYYELY (E2).
  • mice received 2 x 10 7 PFU of MVA-WT or MVA-CHIKV by the i.p. route in 200ul of PBS.
  • C02 carbon dioxide
  • ICS intracellular cytokine staining
  • mice Groups of 5- to 6-week-old female C57BL/6 mice (n 5) were immunized with 1 x10 7 PFU of MVA-WT or MVA-CHIKV by the i.p. route in 200 ul PBS. MVA-WT was administered once, whereas MVA-CHIKV was administered once or twice with a 3-week immunization interval. Serum was collected by tail bleedings 3 weeks after the first immunization and 6 weeks after the second immunization, and antibody responses were analyzed by enzyme-linked immunosorbent assay (ELISA) and a neutralization assay.
  • ELISA enzyme-linked immunosorbent assay
  • CHIKV challenge study A challenge study to evaluate the efficacy of the recombinant MVA-CHIKV was performed in a biosafety level 3 laboratory at the Astrid Fagraeus Laboratory at Karolinska Institutet, Sweden.
  • Female C57BL/6 mice previously immunized with MVA-WT or MVA-CHIKV to study antibody responses were used (see "C57BL/6 mouse immunogenicity study" above [5 mice/group]).
  • mice were challenged with a total of 10 6 PFU of CHIKV (CHIKV isolate LR2006-OPY1 ) via the subcutaneous route in the dorsal side of each hind foot.
  • ICS assay The magnitudes, polyfunctionality, and phenotypes of the VACV- and CHIKV-specific adaptive and memory T cell responses were analyzed by ICS, as previously described, with some modifications.
  • 4 x 10 6 splenocytes (depleted of red blood cells) were seeded on M96 plates and stimulated for 6 h incomplete RPMI 1640 medium supplemented with 10% FCS containing 1 ul/ml GolgiPlug (BD Biosciences) (to inhibit cytokine secretion), anti-CD107a-Alexa Fluor 488 (BD Biosciences), Monensin (1 x; eBioscience), and 1 ug/ml of the different CHIKV peptides (C, E1 , or E2) or MVA-infected EL4 cells (4 x 10 5 EL4 cells in 1 x 10 6 splenocytes; the ratio of MVA infected EL4 cells to splenocytes was equal to 1 :
  • fluorochrome-conjugated antibodies were used: CD3-phycoerythrin (PE)-CF594, CD4-allophycocyanin (APC)-Cy7, CD8- V500, IFN-Y-PE-Cy7, tumor necrosis factor alpha (TNF-a)-PE, and interleukin 2 (IL- 2)-APC.
  • CD62L-Alexa 700 and CD127-PerCP-Cy5.5 were used: CD62L-Alexa 700 and CD127-PerCP-Cy5.5. All antibodies were from BD Biosciences. Cells were acquired using a Gallios flow cytometer (Beckman Coulter). Analyses of the data were performed using FlowJo software version 8.5.3 (Tree Star, Ashland, OR). The number of lymphocyte- gated events was 10 6 . After gating, Boolean combinations of single functional gates were then created using FlowJo software to determine the frequency of each response based on all possible combinations of cytokine.
  • FIG. 1 A shows a diagram of the MVA-CHIKV genome.
  • the correct insertion and purity of recombinant MVA-CHIKV was confirmed by PCR and DNA sequence analysis.
  • PCR using primers annealing in the VACV TK gene-flanking regions confirmed the presence of the CHIKV structural genes in MVA- CHIKV, with no wild-type contamination in the preparation (Fig. 1 B); in the parental virus MVA-GFP, the GFP was present, and in the MVA-WT, the VACV TK locus was amplified (Fig. 1 B).
  • PCRs using primers annealing in the C6L-, K7R-, and A46R-flanking regions confirmed the deletion of the VACV genes C6L, K7R, and A46R, respectively, from MVA-CHIKV (Fig. 1 C), also with no wild-type contamination in the preparation.
  • the proper insertion and correct sequence of the CHIKV structural genes in the TK locus and the correct deletions of the VACV genes C6L, K7R, and A46R were also confirmed by DNA sequencing (data not shown).
  • MVA-CHIKV replicates in cell culture.
  • CHIKV structural proteins affects MVA replication in cell culture.
  • the results showed that the kinetics of viral growth were similar between the two viruses (Fig. 2A), indicating that the constitutive expression of CHIKV structural proteins does not impair vector replication under permissive conditions.
  • MVA-CHIKV is stable in cell culture.
  • a stability test was performed.
  • the recombinant MVA-CHIKV was further grown in DF-1 cells infected at a low multiplicity during 9 consecutive passages, and expression of the CHIKV E1 and E2 proteins during the different passages was determined by Western blotting (Fig. 2B). The results showed that MVA-CHIKV efficiently expresses the CHIKV E1 and E2 proteins after successive passages, indicating that the recombinant MVACHIKV is genetically stable.
  • MVA-CHIKV titers in the P2 stock, in the P3 stock, and at passage 9 were similar when plaques expressing VACV antigens or plaques expressing CHIKV antigens were analyzed, confirming the high genetic stability of MVA-CHIKV.
  • MVA-CHIKV expresses the CHIKV E2 protein in the cytoplasm and in the cell membrane.
  • the expression and intracellular localization of the structural CHIKV E2 protein was also analyzed by immunofluorescence in human HeLa cells infected with MVA-WT or MVA-CHIKV using antibodies specific to E2 and specific probes or antibodies for different proteins and compartments, such as actin (phalloidin), ER (anticalnexin), and the Golgi apparatus or Golgi apparatus-derived membranes, as well as for the plasma membrane (WGA).
  • actin phalloidin
  • ER anti-exin
  • WGA plasma membrane
  • the CHIKV E2 protein is expressed mainly at the plasma membrane and throughout the cytoplasm, where it is localized in discrete accumulations at the viral factories.
  • MVA-CHIKV induces specific morphological cell alterations, with the formation and accumulation of Golgi apparatus derived membranes.
  • ultrathin sections of HeLa cells infected with MVA-WT or MVA-CHIKV were visualized by electron microscopy at low and high magnifications (Fig. 4).
  • Fig. 4 Both in MVA-WT and in MVA-CHIKV-infected cells, the assembly of immature virus (IV) forms of MVA was detected, and remarkably, in MVA-CHIKV- infected cells, an atypical growth and accumulation of Golgi apparatus-like membranes was observed.
  • MVA-CHIKV triggers an innate immune response in human macrophages and dendritic cells, inducing type I IFN, proinflammatory cytokines, and chemokine expression.
  • Type I IFN innate immune responses play a critical role in controlling CHIKV viral replication.
  • CHIKV structural genes in the MVA genome is able to impair the response of innate immune cells to MVA infection
  • IFN- ⁇ type I IFN
  • TNF-a proinflammatory cytokines
  • MIP-1 a chemokines
  • IFIT1 and IFIT2 IFN-inducible genes
  • Fig. 5A some key cytosolic sensors that lead to antiviral IFN production
  • MVA-CHIKV promotes a robust innate immune response in human macrophages and moDCs by inducing the expression of type I IFN (IFN- ⁇ ), proinflammatory cytokines (TNF-a), chemokines (MIP-1 a, IP-10, and RANTES), IFN- in-ducible genes (IFIT1 and IFIT2), and some key cytosolic sensors that lead to antiviral IFN production (RIG-I and MDA-5).
  • IFN- ⁇ type I IFN
  • TNF-a proinflammatory cytokines
  • MIP-1 a chemokines
  • IFIT1 and IFIT2 IFN- in-ducible genes
  • RAG-I and MDA-5 some key cytosolic sensors that lead to antiviral IFN production
  • MVA-CHIKV induces strong, broad, and polyfunctional adaptive CHIKV-specific T cell immune responses.
  • the role of T cell responses in controlling CHIKV infection is not well known.
  • Adaptive, CHIKV-specific T cell immune responses elicited by both immunization groups were measured 10 days after the last immunization by a polychromatic intracellular cytokine staining (ICS) assay, after the stimulation of splenocytes with specific peptides representative of the C, E1 , and E2 CHIKV proteins.
  • ICS polychromatic intracellular cytokine staining
  • the MVA-CHIKV/MVA-CHIKV immunization group triggered an overall CHIKV-specific immune response mediated only by CD8 + T cells, with no CHIKV-specific CD4 + T cells detected, indicating the selectivity for CD8 + T cells of the peptides used in the ICS assay (Fig. 6A).
  • the pattern of adaptive, CHIKV-specific T cell immune responses showed that CD8+ T cell responses induced by MVACHIKV/ MVA-CHIKV were broad, with most of the responses directed mainly against the E1 and E2 peptides (80%) and to a lesser extent against C (Fig. 6B).
  • MVA-CHIKV/MVA-CHIKV significantly enhanced the magnitude of C-, E1 - and E2-specific CD8+ T cell responses (P ⁇ 0.001 ) (Fig. 6C).
  • CHIKV-specific CD8+ T cells producing CD107a, IFN- ⁇ , or TNF-a are the populations most induced by the MVACHIKV/MVA-CHIKV immunization group (Fig. 6D); levels were also of a significantly higher magnitude than those induced by MVAWT/MVA-WT (P ⁇ 0.001 ) (Fig. 6D).
  • the quality of the adaptive, CHIKV-specific T cell immune response was characterized in part by the pattern of cytokine production and its cytotoxic potential.
  • 15 different CHIKV-specific CD8 + T cell populations could be identified (Fig. 6E).
  • MVA-CHIKV/MVACHIKV induced a high polyfunctional profile, with 97% of CD8 + T cells exhibiting two, three, or four functions (Fig.6E).
  • CD8 + T cells producing CD107a plus IFN-y plus TNF-a plus IL- 2, CD107a plus IFN-y plus TNF-a, or IFN-y plus TNF-a were the most abundant populations elicited by MVA-CHIKV/MVA-CHIKV, which, compared to MVA-WT/MVA- WT, also induced significantly higher increases in the percentages of most of the populations (P ⁇ 0.001 ) (Fig. 6E).
  • MVA-WT and MVA-CHIKV induce similar magnitudes and polyfunctionalities of adaptive, VACV-specific T cell immune responses.
  • MVA-WT/MVA-WT and MVA-CHIV/MVA-CHIKV it was of interest to analyze the responses to the MVA vector.
  • ICS assay similar to the protocol followed in the analysis of adaptive CHIKV-specific T cell immune responses after the stimulation of splenocytes with MVA-infected EL4 cells.
  • Both immunization groups triggered an overall adaptive, VACV-specific immune response mediated only by CD8 + T cells (determined as the sum of the individual responses producing IFN- ⁇ , TNF-a, and/or IL-2 cytokines, as well as the expression of CD107a on the surfaces of activated T cells as an indirect marker of cytotoxicity); this experiment was carried out with MVA-infected EL4 cells, and the magnitudes of adaptive, VACV-specific CD8 + T cell immune responses were similar in the two immunization groups (Fig. 7A). Moreover, VACV-specific CD8 + T cells producing CD107a, IFN- ⁇ , or TNF a-were the most induced populations in both immunization groups, and they were induced at similar magnitudes (Fig.
  • VACV-specific CD8 + T cell immune responses were characterized by the production of CD107a, IFN- ⁇ , TNF-a, and/or IL-2, and 15 distinct VACV-specific CD8 + T cell populations could be identified (Fig. 7C).
  • VACV-specific CD8 + T cell responses were similarly polyfunctional in the two immunization groups, with 88 to 91 % of the CD8 + T cells exhibiting two, three, or four functions.
  • CD8+ T cells producing CD107a plus IFN-Y plus TNF-a plus IL-2, CD107a plus IFN- ⁇ plus TNF-a, IFN- ⁇ plus TNF-a, CD107a plus IFN- ⁇ , or only CD107a were the most induced populations elicited by both immunization groups (at similar percentages) (Fig. 7C).
  • these results showed that MVA-WT and MVACHIKV elicited similar magnitudes and levels of quality of adaptive, VACV-specific CD8 + T cell immune responses. Similar findings were observed in two independent experiments.
  • MVA-CHIKV induces strong, broad, polyfunctional, and durable CHIKV-specific memory T cell immune responses.
  • the durability of a vaccine-induced T cell response is an important feature, since long-term protection is a requirement for prophylactic vaccination.
  • CHIKV-specific memory T cell immune responses elicited by both immunization groups 52 days after the last immunization by the ICS assay in a manner similar to that of the protocol followed in the adaptive phase.
  • the MVA-CHIKV/MVA-CHIKV immunization group triggered an overall CHIKV-specific memory immune response mediated only by CD8 + T cells, with no CHIKV-specific CD4 + T cells detected, indicating the selectivity for CD8 + T cells of the peptides used in the ICS assay (Fig. 8A).
  • the pattern of CHIKV-specific memory T cell immune responses showed that CD8 T cell responses induced by MVACHIKV/ MVA-CHIKV were broad, with most of the responses being directed mainly against the E1 and E2 peptides (77%) and, to a lesser extent, against C (Fig. 8B), as with the results obtained in the adaptive phase.
  • MVA-CHIKV/MVA-CHIKV significantly enhanced the magnitudes of C-, E1 -, and E2- specific CD8 + T cell responses (PO.001 ) (Fig. 8C).
  • CHIKV-specific CD8 + T cells producing CD107a, IFN- ⁇ , or TNF-a are the populations most induced by the MVACHIKV/MVA-CHIKV immunization group (Fig. 8D), and their levels are also of a significantly higher magnitude than those induced by MVA-WT/MVA-WT (PO.001 ) (Fig. 8D), results similar to those obtained in the adaptive phase.
  • CHIKV-specific memory T cell immune responses was characterized by analyzing the simultaneous production of CD107a, IFN- ⁇ , TNF-a, and/or IL-2 from CHIKV specific CD8 + T cells (Fig. 8E), where 15 distinct CHIKV-specific CD8 + T cell populations could be identified.
  • MVA-CHIKV/MVA-CHIKV induced a polyfunctional profile, with 22% of CD8 + T cells exhibiting two, three, or four functions (Fig. 8E).
  • CD8 + T cells producing CD107a plus IFN- ⁇ plus TNF-a plus IL-2, CD107a plus IFN-y plus TNF-a, or CD107a were the most abundant populations elicited by MVACHIKV/ MVA-CHIKV, which also induced increases in the percentages of most of the populations that were significantly higher than those induced by MVA-WT/MVA-WT (P ⁇ 0.001 ) (Fig. 8E), results similar to those obtained in the adaptive phase.
  • CD127 and CD62L surface markers which allowed us to define the different memory subpopulations: central memory (TCM; CD127/CD62L), effector memory (TEM; CD1277CD62L “ ), and effector (TE;CD127 “ /CD62L “ ) T cells (Fig. 8F).
  • VACV-specific memory T cell immune responses elicited by both immunization groups were measured 52 days after the boost by ICS assay, which is similar to the protocol followed in the adaptive phase.
  • both immunization groups triggered overall VACV-specific memory immune responses mediated only by CD8 + T cells, determined as the sums of the individual levels of production of CD107a, IFN- ⁇ , TNF-a, and/or IL-2 obtained in MVA-infected EL4 cells (Fig.
  • VACV-specific CD8 + memory T cell immune responses are similar in the two immunization groups (Fig. 9A).
  • VACV-specific CD8 + T cells producing CD107a, IFN- ⁇ , or TNF-a are the most induced populations in both immunization groups, and these populations were also produced at similar magnitudes (Fig. 9B).
  • the quality of VACV-specific CD8 + memory T cell immune responses was characterized as the simultaneous production of CD107a, IFN- ⁇ , TNF- ⁇ , and/or IL-2 and was assessed by using a protocol like that followed in the adaptive phase, where 15 distinct VACV-specific CD8 + T cell populations could be identified (Fig.9C).
  • VACV-specific CD8 + T cell responses were similarly polyfunctional in the two immunization groups, with 95% of CD8 + T cells exhibiting two, three, or four functions.
  • CD8 + T cells producing CD107a plus IFN- ⁇ plus TNF-a plus IL-2, CD107a plus IFN- ⁇ plus TNF-a, IFN- ⁇ plus TNF-a, CD107a plus IFN- Y, or only CD107a were the most induced populations elicited by both immunization groups, and they were also induced at similar percentages (Fig. 9C).
  • MVA-CHIKV induces high titers of neutralizing antibodies against CHIKV.
  • Antibodies against CHIKV are crucial to control CHIKV infection.
  • MVA-CHIKV to study the ability of MVA-CHIKV to elicit humoral immune responses against CHIKV, we analyzed the levels of CHIKV envelope-specific antibodies present in the sera of C57BL/6 mice immunized with one or two doses of MVA-CHIKV (see Materials and Methods). Animals immunized with nonrecombinant MVA-WT were used as a control group. The results show that one immunization with MVA-CHIKV elicited high titers of IgG antibodies against CHIKV that were further enhanced by the second immunization (Fig. 10A).
  • mice were immunized with one or two doses of MVACHIKV or one dose of MVA-WT (as a control). Seven weeks after the last immunization, mice were challenged with a high dose of CHIKV in their feet (see Materials and Methods). Protection was evaluated by determining viremia and analyzing foot swelling in CHIKV-challenged mice during the days following challenge. The results showed that no CHIKV infection was developed in MVACHIKV- vaccinated mice, as no viremia was detected postchallenge (Fig. 10C).
  • MVA-WT-immunized mice developed CHIKV infection, with high levels of CHIKV (10 4 to 10 5 PFU/ml) detected in blood that peaked day 2 postchallenge (Fig. 10B). Moreover, MVA-CHIKV-vaccinated mice did not develop foot swelling after challenge, while MVA-WT-immunized mice developed severe foot swelling that peaked 6 days postchallenge (Fig. 10D).
  • MVA-CHIKV is a highly effective vaccine that protected mice against challenge with a high dose of CHIKV, with no viremia detected in their blood and no signs of inflammation. Remarkably, just a single dose of MVA- CHIKV protected all mice from challenge with CHIKV.
  • the DREP platform differs from conventional DNA plasmids in that it encodes the alphavirus (CHIKV) replicase, which drives the production of the subgenomic RNA and thus the expression of the encoded CHIKV antigen. Moreover, DREPs also possess intrinsic adjuvant properties as the replicase and RNA intermediates stimulate the production of type-1 interferons (IFNs) and apoptosis. Promising results have been reported for DNA replicons generated from other alphaviruses including Semliki Forest virus, Sindbis virus and Venezuelan equine encephalitis virus, when used for priming immunizations prior to boosting with other vaccine modalities.
  • IFNs type-1 interferons
  • the heterologous prime-boost approach takes advantage of the unique immune profile being induced by the different vaccine platforms. For example, both attenuated and genetic vaccines are being produced endogenously and can thus give rise to T cell- mediated immune responses. In contrast, protein antigens generally lack the ability to elicit cytotoxic T cell responses and are thus limited to the induction of humoral responses. Combining different vaccine strategies in heterologous prime-boost immunizations should induce a more balanced immune response in terms of cellular and humoral immune responses, and enhance the magnitude and quality of immune responses compared to homologous vaccination using a single vaccine modality alone.
  • CHIKV vaccine candidates All vaccine constructs were based on the CHIKV clone LR2006-OPY1 .
  • a DNA replicon vaccine encoding CHIKV envelope was constructed on the basis of cytomegalovirus (CMV) promoter launched infectious cDNA clone of CHIKV. Briefly, the region corresponding to the nucleotide residues 7565-8350 of CHIKV genome was replaced by sequence 5' CCTAGGCCACCATG 3' by PCR-based mutagenesis followed by multi-step subcloning procedure. The resulting deletion removed the CHIKV capsid coding sequence. In addition, the first amino acid residue of E3 protein (serine) was replaced by methionine.
  • A5nsP3 is an attenuated CHIKV that was formed by attenuating an infectious CHIKV by a large deletion in the nsP3 gene
  • MVA-CHIKV was constructed by inserting the cDNA encoding for the structural polyprotein of CHIKV (C, E3, E2, 6K, E1 ) into MVA
  • soluble recombinant p62-E1 CHIKV protein was formed by joining the ectodomains of CHIKV p62 and E1 proteins with a glycine serine linker.
  • Wild type (WT) CHIKV was used for challenge and as a reference for immunizations.
  • mice Female C57BL/6 mice, 5-6 weeks old, were immunized once or twice with the different CHIKV vaccine candidates DREP-Env, A5nsP3, MVA-CHIKV or CHIKV p62-E1 , using various homologous or heterologous prime-boost immunization protocols with a three weeks immunization interval between prime and boost.
  • a total of 10 ⁇ g of DREP-Env DNA diluted in 2x20 ⁇ of PBS was injected intradermal ⁇ followed by in vivo electroporation using the DermaVaxTM electroporation device as previously described.
  • 105 plaque-forming units (PFU) of A5nsP3 or WT CHIKV diluted in 2x50 ⁇ of PBS was injected subcutaneously and 10 7 PFU of recombinant MVA-CHIKV was diluted in 200 ⁇ of PBS and injected intraperitoneally.
  • 1 ⁇ g CHIKV p62-E1 protein was diluted in 2x50 ⁇ of PBS and administered intramuscularly.
  • CHIKV p62-E1 protein was injected alone or mixed with 25 ⁇ of the adjuvant AS03 (1 :10 human dose) (GlaxoSmithKline Biologicals S.A., Rixensart, Belgium) or with 5 ⁇ g of the adjuvant Matrix-M (Novavax, MD, USA). Some groups of mice were immunized with two vaccine candidates at the same occasion but at different injection sites. All experiments were performed in at least two separate iterations. B and T cell responses.
  • CHIKV-specific humoral and cellular immune responses induced prior to challenge by the different prime-boost immunization protocols were assessed by enzyme-linked immunosorbent assay (ELISA), neutralization assay and I FN- ⁇ ELISpot (Mabtech AB, Nacka Strand, Sweden) as previously described.
  • I FN- ⁇ ELISpot assay splenocytes from immunized mice were collected 8 days after the last immunization and 10 5 splenocytes were plated per well. Then, cells were stimulated with 2.5 ⁇ / ⁇ of a CD8 T cell-restricted CHIKV E1 peptide (HSMTNAVTI) or 10 ⁇ / ⁇ of CHIKV p62-E1 protein. Responses over 25 Spot Forming Units (SFU)/10 6 splenocytes and a minimum of four times above background were regarded as positive.
  • SFU Spot Forming Units
  • Peptides were first dissolved in dimethyl sulphoxide (DMSO) to a concentration of 15 ⁇ g/m ⁇ before being diluted 1 :1000 in 0.1 % PBST and subsequently coated onto the plates (100 ⁇ /well). Heat-inactivated pooled sera were diluted 1 :500 in 0.1 % sodium caseinate in 0.1 % PBST and 100 ⁇ were added into each well and incubated for 1 h at 37 °C. Horseradish peroxidase (HRP)-conjugated goat anti-mouse IgG Abs (Santa Cruz) diluted 1 :10000 in 0.1 % sodium caseinate in 0.1 % PBST were used to detect the bound Abs.
  • HRP horseradish peroxidase
  • mice Seven weeks after the last immunization mice were challenged with a total of 10 6 PFU of WT CHIKV in 2x20 ⁇ PBS subcutaneously in the dorsal side of the feet on both hind legs. Viremia and foot swelling post challenge was determined by plaque assay and by measuring the height and breadth of feet, respectively.
  • Prime-boost immunizations with different CHIKV vaccine candidates induce superior antibody responses.
  • Homologous and heterologous prime-boost immunizations using different CHIKV vaccines representing different CHIKV vaccine candidates (Table 1 ) were studied in C57BL/6 mice. A single immunization with 10 5 PFU of WT CHIKV was included for comparison. When combining different vaccine candidates they were either co-administered (at separate sites) in a single immunization (XY) or used in a prime-boost regimen (X,Y).
  • NT50 antibody titers Most vaccine candidates induced good neutralizing NT50 antibody titers in the range of 10 3 -10 5 .
  • the DREP-Env induced NT50 titers in the range of 10 2 -10 3 with some animals having no NT50 antibodies. These animals correspond to those that had binding (ELISA) lgG1 antibodies below or close to 10 4 (Fig. 1 1 A,B).
  • the lack of NT50 antibodies in some of the DREP-Env samples does not necessary mean that the animals were devoid of them but could be explained by the fact that the sensitivity of the assay is 1 :100 (first dilution). Another exception was p62-E1 protein alone given once that did not induce any detectable NT50 antibodies.
  • Prime boost immunization generally augmented NT50 titers with titers being the highest when priming was performed with DREP-Env followed by boosting with MVA (around 10 5 ).
  • boosting A5nsP3, DREP-Env or MVA-CHIK with p62-E1 protein did not significantly increase NT50 titers.
  • CHIKV vaccine candidates protect against CHIKV infection.
  • the efficacy of the different CHIKV vaccine candidates to protect from challenge with a high dose (10 6 PFU) WT CHIKV was studied in a challenge model using the established immunization schedules.
  • MVA-CHIKV, DREP, and p62-E1 vaccines used were as described in example 2. Immunisations and analysis were also as described.
  • Alphavirus replicon priming followed by heterologous booster immunizations induce different CD8+ T cell subpopulations.
  • C57BL/6 mice were primed with DREP-Env and then boosted either homologously with DREP-Env, or heterologously with either CHIKV p62-E1 protein, MVA-CHIKV or both protein and MVA-CHIKV at the same time.
  • the magnitude of the CHIKV-specific CD8+ T cell responses are greatly increased with heterologous prime-boost immunization.
  • the acute response is increased 9-fold with a CHIKV p62-E1 protein boost and 16-fold with an MVA-CHIKV boost.
  • Boosting with both CHIKV p62-E1 protein and MVA-CHIKV results in an 18-fold increase.
  • the phenotypes of CHIKV-specific CD8+ T cell responses were characterized 8 days after boost as described above.
  • the proportion of CD27+CD43+ CHIKV-specific CD8+ T cells was smaller whereas a higher proportion of CD27+CD43- cells was induced (Fig. 13, bottom pie chart).
  • mice given a heterologous boost had similar proportions of CD8+ Te cells in the range of 22-29%, whereas mice given a homologous DREP-Env boost triggered only 7% of CD8+ Te cells (Fig. 13, top pie charts).
  • Vijayakumar KP Nair Anish TS, George B, Lawrence T, Muthukkutty SC, Ramachandran R. 201 1 . Clinical Profile of Chikungunya Patients during the Epidemic of 2007 in India. J Glob Infect Dis 3:221 -226. Couturier E, Guillemin F, Mura M, Leon L, Virion JM, Letort MJ, De Valk H, Simon F, Vaillant V. 2012.
  • Pijlman GP. 201 1 Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells. Virol J 8:353.
  • a recombinant measles vaccine expressing chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus.
  • Sindbis virus DNA-based expression vectors utility for in vitro and in vivo gene transfer. Journal of virology 70:508-519.
  • 201 Inhibitors of alphavirus entry and replication identified with a stable Chikungunya replicon cell line and virus-based assays. PLoS One 6:e28923. Roos AK, Eriksson F, Timmons JA, Gerhardt J, Nyman U, Gudmundsdotter L, Brave A, Wahren B, Pisa P.
  • Gomez CE Perdiguero B, Garcia-Arriaza J, Esteban M. 2012. Poxvirus vectors as HIV/AIDS vaccines in humans. Hum. Vaccin. Immunother. 8:1 192-207. Gomez CE, Najera JL, Perdiguero B, Garcia-Arriaza J, Sorzano COS, Jimenez V, Gonzalez-Sanz R, Jimenez JL, Munoz-Fernandez MA, Lopez Bernaldo de Quiros JC, Guardo AC, Garcia F, Gatell JM, Plana M, Esteban M. 201 1.
  • the HIV/AIDS vaccine candidate MVA-B administered as a single immunogen in humans triggers robust, polyfunctional, and selective effector memory T cell responses to HIV-1 antigens. J. Virol. 85:1 1468-78.
  • Sallusto F Lanzavecchia A, Araki K, Ahmed R. 2010. From vaccines to memory and back. Immunity 33:451 -63. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. 1999. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401 :708-12.
  • CD80/CD86 in generation and maintenance of functional virus-specific CD8+ T cells in mice infected with lymphocytic choriomeningitis virus.
  • Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature 433:887-892.
  • PKR acts early in infection to suppress Semliki Forest virus production and strongly enhances the type I interferon response. J Gen Virol, 2009/03/07 ed. 90:1382-1391. Le Bon A, Durand V, Kamphuis E, Thompson C, Bulfone-Paus S, Rossmann C, Kalinke U, Tough DF.
  • Type I IFN negatively regulates CD8+ T cell responses through IL-10-producing CD4+ T regulatory 1 cells. J. Immunol. 174:99-

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

A composition for use as a vaccine to protect against Chikungunya virus (CHIKV) infection is disclosed. The composition comprises a nucleic acid comprising a nucleotide sequence encoding an attenuated poxvirus vector and one or more CHIKV structural proteins. In preferred embodiments, the nucleotide sequence encodes all of the C, E3, E2, 6K, and E1 structural proteins. Dosage schedules are also described.

Description

Vaccine Composition
FIELD OF THE INVENTION
The present invention relates to a vaccine composition for protection against Chikungunya virus infection. Aspects of the invention relate to methods for protecting subjects against Chikungunya virus infection, and to compositions for use in the preparation of a vaccine composition.
BACKGROUND TO THE INVENTION
Chikungunya is a disease caused by the Chikungunya virus (CHIKV) with a mortality rate of about 1 :1000. The disease is characterized by skin rash, high fever, headache, vomiting, myalgia, and, mainly, polyarthralgia. Most of the symptoms resolve after 10 days, but the polyarthralgia can persist for months or years, and severe symptoms, such as encephalitis, hemorrhagic disease, and mortality, have also been described. The disease is transmitted to humans by Aedes mosquito vectors. CHIKV infection has been reported from Africa, islands in the Indian Ocean, India, Southeast Asia, and southern Europe, and recently in the Americas where over 700,000 individuals are estimated to have encountered CHIKV. In 2014 several mortality cases related to CHIKV infection have been reported in Venezuela. CHIKV contains a positive, single-stranded RNA genome of around 1 1 .8 kb which encodes four nonstructural and five structural proteins. The nonstructural proteins (nsP1 , nsP2, nsP3, and nsP4) are required for virus replication. The structural proteins are cleaved by capsid (C) autoproteinase and signalases from a polyprotein precursor to generate the C and envelope (pE2, 6K, and E1 ) proteins. The pE2 precursor is cleaved to E3 and E2 by the furin protease in the trans-Golgi during transport of the envelope complex to the cell surface where virus assembly occurs. pE2 is also known as p62. Virions are 70-nm enveloped particles containing 240 heterodimers of E1/E2 glycoproteins on their surfaces. There is currently no specific treatment for Chikungunya, nor are there currently any approved vaccines, although a number of candidate vaccines are in development. It would be desirable to develop a vaccine to protect against CHIKV.
Ross River Virus (RRV) is closely related to CHIKV, and causes an illness displaying similar symptoms. It has been shown that antibodies against RRV and CHIKV cross react. Therefore it is possible that a CHIKV vaccine could also provide protection against RRV.
SUMMARY OF THE INVENTION
Described herein is a vaccine candidate against CHIKV, based on an attenuated poxvirus vector expressing the CHIKV structural genes. The vaccine was described for the first time in J. Virol. 2014, 88(6):3527, Garcia-Arriaza et al, "A Novel Poxvirus Based Vaccine, MVA-CHIKV, Is Highly Immunogenic and Protects Mice against Chikungunya Infection", published ahead of print 8 January 2014. The contents of this publication are incorporated herein by reference.
According to a first aspect of the invention, there is provided a composition for use in the production of a vaccine, the composition comprising a nucleic acid comprising a nucleotide sequence encoding an attenuated poxvirus vector and one or more CHIKV structural proteins.
Preferably the nucleotide sequence encodes two, three, four, or five CHIKV structural proteins; most preferably five structural proteins. The structural proteins may be selected from the group consisting of the C, E3, E2, 6K, and E1 proteins. Preferably the nucleotide sequence encodes at least the E3 and E2 proteins; most preferably the nucleotide sequence encodes all of the C, E3, E2, 6K, and E1 proteins. The CHIKV sequences can be derived from virus isolates which may differ in nucleotide sequence from one another and from neutralization resistant mutants. The nucleotide sequence may further encode one or more replicase proteins (that is, one or more of nsP1 , nsP2, nsP3, and nsP4). The sequence may encode two, three, or four of said replicase proteins. It is possible that host immune system responses against the replicase may further enhance protectivity of the vaccine, for example by reducing long term infection of macrophages by CHIKV.
The CHIKV nucleotide sequences can be derived from one or more virus isolates. The isolates may be selected such that they differ from one another to the extent that a neutralisation response by sera generated from one isolate does not neutralise a second isolate. Including sequences from a number of isolates improves the chances of an immune response generated by the vaccine being effective against multiple strains of CHIKV. Sequences from different isolates may be combined in a single poxvirus vector, or multiple vectors representing different CHIKV isolates may be combined in a single vaccine composition. The nucleic acid may be DNA, RNA, or a combination thereof, and can include any combination of naturally occurring, chemically modified or enzymatically modified nucleotides. The nucleic acid of the invention may take the form of an expression vector that is capable of expression in an organism or in a cell of the organism, in culture or in vivo. An organism or cell in which the coding sequence of the vector can be expressed can be eukaryotic or prokaryotic, and can be, without limitation, a bacterium, a yeast, an insect, a protozoan, or animals, such as a human, a bird, or a non-human mammal. The nucleotide sequence of the nucleic acid may be codon optimised for expression in a particular organism or cell; for example, humans. It should be understood that nucleic acids of the invention can be single-stranded or double-stranded, and further that a single-stranded nucleic acid of the invention includes a polynucleotide fragment having a nucleotide sequence that is complementary to a nucleotide sequence that encodes said poxvirus vector and structural proteins according to the invention. As used herein, the term "complementary" refers to the ability of two single stranded polynucleotide fragments to base pair with each other.
Further, a single-stranded nucleic acid of the invention also includes a polynucleotide fragment having a nucleotide sequence that is substantially complementary to a nucleotide sequence that encodes said poxvirus vector and structural proteins according to the invention, or to the complement of the nucleotide sequence that encodes said poxvirus vector and structural proteins. Substantially complementary polynucleotide fragments can include at least one base pair mismatch, such that at least one nucleotide present on a first polynucleotide fragment will not base pair to at least one nucleotide present on a second polynucleotide fragment, however the two polynucleotide fragments will still have the capacity to hybridize. The present invention therefore encompasses polynucleotide fragments that are substantially complementary. Two polynucleotide fragments are substantially complementary if they hybridize under hybridization conditions exemplified by 2xSSC (SSC: 150 mM NaCI, 15 mM trisodium citrate, pH 7.6) at 55eC. Substantially complementary polynucleotide fragments for purposes of the present invention preferably share at least about 85% nucleotide identity, preferably at least about 90%, 95% or 99% nucleotide identity. Locations and levels of nucleotide sequence identity between two nucleotide sequences can be readily determined using, for example, CLUSTALW multiple sequence alignment software.
The poxvirus vector may be a vaccinia virus; preferably a modified vaccinia virus Ankara (MVA). In preferred embodiments, the MVA vector is further modified by deletion of one or more vaccinia immunomodulatory genes; preferably one, two, or three of the genes C6L, K7R, and A46R. Most preferably, all three genes are deleted. Deletion of these genes has been shown to enhance the innate immune response triggered by the viral vector, and potentially also specific immune response. Alternative attenuated poxvirus vectors include NYVAC, an isolate of the Copenhagen vaccinia vaccine strain.
A further aspect of the invention provides an attenuated poxvirus vector expressing one or more CHIKV structural genes. Also provided is a vaccine composition comprising such an attenuated poxvirus vector. The vaccine composition may also be used in combination with other vectors expressing CHIKV genes and protein components, and/or as a combination vaccine against other infections. A combination vaccine may comprise one or more adjuvants. Preferred adjuvants include AS03, or Matrix - M.
A vaccine of the invention is conveniently administered to subjects using ingestion, topical administration, or direct injection, preferably subcutaneous or intramuscular injection. The subject is preferably mammalian, more preferably human, although alternative subjects may be treated, for example, in the course of vaccine or disease research. The dose of vaccine to be administered to a subject depends on the species and size of subject, the nature of the condition being treated, and can be readily determined by one of skill in the art.
The invention further provides an attenuated poxvirus vector expressing one or more CHIKV structural genes for use as a vaccine. Also provided is the use of an attenuated poxvirus vector expressing one or more CHIKV structural genes in the manufacture of a vaccine. The vaccine is intended for use in protecting a subject against CHIKV infection. The vaccine may also or instead be for use in protecting a subject against RRV infection.
A still further aspect of the invention provides a method of immunising a subject against CHIKV infection, the method comprising administering a vaccine comprising an attenuated poxvirus vector expressing one or more CHIKV structural genes to a subject. A single administration may be sufficient to provide suitable immunization, but in preferred embodiments, more than one dose of vaccine is administered. For example, a first dose may be followed by a booster dose after one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months or longer intervals.
In alternative embodiments, the first dose of the attenuated poxvirus vector vaccine may be followed by a second dose of an alternative vaccine; a so-called heterologous prime-boost protocol. Or the attenuated poxvirus vector vaccine may be administered second, after an initial dose of an alternative vaccine. The alternative vaccine may comprise a covalently linked p62-E1 heterodimer (E1 being the CHIKV E1 protein, and p62 being the CHIKV precursor protein which is cleaved during virus replication into the E2 and E3 proteins). Alternatively, the alternative vaccine may comprise a DNA- replicon (DREP) based vaccine; this may comprise a nucleotide sequence comprising CHIKV genomic sequences operably linked to a promoter sequence. The genomic sequences may comprise genes coding for the envelope proteins (E3, E2, 6K, and E1 ). Preferably the genomic sequences do not include genes coding for the capsid (C) protein; in the absence of this protein, the DREP-CHIKV is unable to form viral particles, and is only able to express envelope proteins on the host cell surface. In preferred embodiments of the invention the first dose is of DREP-CHIKV vaccine, and the second is of the attenuated poxvirus vector. The second dose may be combined with a dose of p62 - E1 vaccine. The invention may further provide a kit comprising first and second doses of CHIKV vaccine. The first dose is preferably a vaccine comprising an attenuated poxvirus vector expressing one or more CHIKV structural genes. The second dose may be the same as the first dose, or may be a vaccine comprising a covalently linked p62-E1 heterodimer, or a DREP-CHIKV vaccine. "First" and "second" here do not necessary refer to the order in which the vaccines are to be administered. The doses may be provided in a suitable pharmaceutical container; for example, sealed vials, or alternatively as preloaded syringes.
BRIEF DESCRIPTION OF THE DRAWINGS FIG 1 Generation and in vitro characterization of recombinant MVA-CHIKV. (A) Scheme of the MVA-CHIKV genome map. The different regions are indicated by capital letters. The right and left terminal regions are shown. Below the map, the deleted or fragmented genes are depicted as black boxes. The CHIKV C, E3, E2, 6K, and E1 structural genes (from CHIKV isolate LR2006-OPY1 ), driven by the synthetic early/late (sE/L) virus promoter and inserted within the VACV TK viral locus (J2R), are indicated. The deleted VACV C6L, K7R, and A46R genes are also indicated. TK-L, TK left; TK-R, TK right. (B) PCR analysis of the VACV TK locus. Viral DNA was extracted from chick DF-1 cells mock infected or infected at 5 PFU/cell with MVA-CHIKV, MVA-GFP, or MVA-WT. Primers spanning the TK locus-flanking regions were used for PCR analysis of the CHIKV genes inserted within the TK locus. DNA products with their corresponding sizes (base pairs) are indicated by an arrow on the right. A molecular size marker (1 -kb ladder) with the corresponding sizes (base pairs) is indicated on the left. (C) PCR analysis of the C6L, K7R, and A46R loci. Viral DNA was extracted from DF-1 cells mock infected or infected at 5 PFU/cell with MVA-CHIKV, MVA-GFP, or MVA-WT. Primers spanning C6L-, K7R-, and A46R-flanking regions were used for PCR analysis of the C6L, K7R, and A46R loci, respectively. DNA products with the corresponding size (base pairs) of the parental virus (wt) or the virus with the C6L, K7R, and A46R deletions are indicated by an arrow on the right. A molecular size marker (1 -kb ladder) with the corresponding sizes (base pairs) is indicated on the left. (C) Expression of CHIKV E1 and E2 proteins. DF-1 cells were mock infected or infected at 5 PFU/cell with MVA-CHIKV, MVA-GFP, or MVA-WT. At 24 h postinfection, cells were lysed in Laemmli buffer, fractionated by 10% SDS-PAGE, and analyzed by Western blotting using rabbit polyclonal antibody against E1 or mouse polyclonal antibody against E2. Arrows on the right indicate the positions of the CHIKV E1 and E2 proteins, with the corresponding sizes in kDa. The sizes of standards (in kDa) (Precision Plus protein standards; Bio-Rad Laboratories) are indicated on the left.
FIG 2 Viral growth kinetics and stability of MVA-CHIKV. (A) Viral growth kinetics. Monolayers of DF-1 cells were infected at 0.01 PFU/cell with MVA-WT or MVA-CHIKV. At different times postinfection (0, 24, 48, and 72 h), cells were collected and virus titers in cell lysates were quantified by a plaque immunostaining assay with anti-WR antibodies. The means of results from two independent experiments are shown. (B) Stability of MVA-CHIKV.MVA-CHIKV(P2 stock) was continuously grown to passage 9 in DF-1 cells. Then, DF-1 cells were mock infected or infected with MVA-CHIKV from the different passages or with MVA-WT. At 24h postinfection, cells were lysed in Laemmli buffer, fractionated by 10% SDS-PAGE, and analyzed by Western blotting using rabbit polyclonal antibody against E1 or mouse polyclonal antibody against E2. Rabbit anti-B-actin antibody was used as a protein loading control. Rabbit anti-VACV early E3 protein antibody was used as a VACV loading control. Arrows on the right indicate the positions of the CHIKV E1 and E2 proteins, B-actin, and the VACV E3 protein, with their corresponding sizes in kDa. The sizes of standards (in kDa) (Precision Plus protein standards; Bio-Rad Laboratories) are indicated on the left. (C) Genetic stability of MVA-CHIKV, as determined by plaque immunostaining. Virus titers of MVA-CHIKV (P2 stock, P3 stock, and passage 9) were quantified by a plaque immunostaining assay with anti-VACV and anti-CHIKV antibodies. The means and standard deviations of results from two independent determinations are shown. It should be noted that the P3 stock represents virus purified with two sucrose cushions, while P2 and passage 9 stocks are virus titers from crude cell lysates. FIG 3 Immunofluorescence analysis of CHIKV E2 protein produced in HeLa cells infected with MVA-CHIKV. Subconfluent HeLa cells mock infected or infected with MVA-CHIKV or MVA-WT were fixed at 6 and 16 hpi (MVA-WT- and mock-infected cells are represented at 16 hpi), labeled with the corresponding primary antibodies or probes, monitored for appropriate fluorescence from protein-conjugated secondary antibodies, and visualized by confocal microscopy. The antibodies or probes used were anti-E2 (D3-62; to detect CHIKV E2 protein, shown in green), phalloidin (to stain actin, shown in red), anti-calnexin (to detect the ER, shown in red), WGA(to detect Golgi apparatus membranes and the plasma membrane, shown in red), and DAPI (to mark DNA, shown in blue). Bar, 10 urn.
FIG 4 Architecture of HeLa cells following infection with MVA-CHIKV or MVA-WT. HeLa cells infected with MVA-WT or MVA-CHIKV were chemically fixed at 6 hpi and then processed for conventional embedding in an epoxy resin and observed by electron microscopy. (A) General overview of a cell infected with MVA-WT (left) and MVA-CHIKV (right); (B) two high-magnification panels of a cell infected with MVA- CHIKV. m, mitochondria; IV, immature virus; GM, Golgi apparatus membranes. Bars:
FIG 5 MVA-CHIKV triggers an innate immune response in human macrophages and dendritic cells, upregulating type I IFN, proinflammatory cytokines, and chemokine expression. Human THP-1 macrophages (A) and moDCs (B) were mock infected or infected with MVA-WT, MVA-GFP, or MVA-CHIKV at 5 PFU/cell (A) and 1 PFU/cell (B). At different times postinfection (3 h and 6 h in panel A, 6 h in panel B), RNA was extracted and the mRNA levels of IFN-, TNF-, MIP-1 , RANTES, IP-10, IFIT1 , IFIT2, RIG-I, MDA-5, and HPRT were analyzed by reverse transcription-PCR. Results are expressed as the ratio of the gene of interest to the hypoxanthine phosphoribosyltransferase (HPRT) mRNA level. A.U., arbitrary units. P values indicate significantly higher responses in comparisons of MVA-WT to MVA-GFP or MVA-CHIKV at the same hour (*, P < 0.05; **, P < 0.005; ***, P < 0.001 ). Data are means +/- standard deviations of results with duplicate samples from one experiment and are representative of two independent experiments.
FIG 6 Immunization with MVA-CHIKV induces strong, broad, and polyfunctional adaptive CHIKV-specific T cell immune responses. Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA-CHIKV 10 days after the last immunization. Next, adaptive, CHIKV-specific CD4 and CD8 T cell immune responses triggered by both immunization groups were measured by the ICS assay following stimulation of splenocytes with different CHIKV peptides (C, E1 , and E2). Values from unstimulated controls were subtracted in all cases. P values indicate significantly higher responses in comparisons of MVA-WT/MVA-WTto MVA- CHIKV/MVA-CHIKV (***, P<0.001 ). Data are from one experiment and are representative of data from two independent experiments. (A) Overall magnitudes of CHIKV-specific CD4 and CD8 T cells. The values represent the sums of the percentages of T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against the C, E1 , and E2 peptides. (B) Pattern of adaptive, CHIKV-specific CD8 T cell immune responses to C, E1 , and E2. Frequencies were calculated by reporting the number of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 and are specific for each peptide to the total number of CD8 T cells in the MVA- CHIKV/MVA-CHIKV immunization group. (C) Magnitudes of C, E1 , and E2 CHIKV- specific CD8 T cells. Frequencies represent the sums of the percentages of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against the C, E1 , or E2 peptide. (D) Magnitudes of CHIKV-specific CD8 T cells producing CD107a, IFN-γ, TNF-a, or IL-2. Frequencies represent percentages of CD8 T cells producing CD107a, IFN-γ, TNF-a, or IL-2 against the C, E1 , and E2 peptides. (E) Polyfunctional profile of adaptive, CHIKV-specific CD8 T cell immune responses. All the possible combinations of the responses are shown on the x axis (15 different T cell populations), while the percentages of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against the C, E1 , and E2 peptides are shown on the y axis. Responses are grouped and color coded on the basis of the number of functions (4, 3, 2, or 1 ). The pie charts summarize the data. Each slice corresponds to the proportion of CHIKV-specific CD8 T cells exhibiting one, two, three, or four functions within the total population of CHIKV- specific CD8 T cells.
FIG 7 Adaptive, VACV-specific T cell immune responses. Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA- CHIKV 10 days after the last immunization. Next, adaptive, VACV-specific CD4 and CD8 T cell immune responses triggered by both immunization groups were measured by ICS assay following stimulation of splenocytes with MVA-infected EL4 cells. Values from unstimulated controls were subtracted in all cases. Data are from one experiment that is representative of two independent experiments. (A) Overall magnitude of VACV- specific CD4 and CD8 T cells. The values represent the sums of the percentages of T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells. (B) Percentages of VACV-specific CD8 T cells producing CD107a, IFN-γ , TNF-a, or IL-2. Frequencies represent percentages of CD8 T cells producing CD107a, IFN-γ , TNF-a, or IL-2 against MVA-infected EL4 cells. (C) Functional profiles of adaptive, VACV-specific CD8 T cell immune responses. All the possible combinations of the responses are shown on the x axis (15 different T cell populations), while the percentages of CD8 T cells producing CD107a and/or IFN-γ and/or TNFa and/or IL-2 against MVA-infected EL4 cells are shown on the y axis. Responses are grouped and color coded on the basis of the number of functions (4, 3, 2, or 1 ). The pie charts summarize the data. Each slice corresponds to the proportion of VACV-specific CD8 T cells exhibiting one, two, three, or four functions within the total population of VACV- specific CD8 T cells. FIG 8 Immunization with MVA-CHIKV induces strong, broad, and polyfunctional CHIKV-specific memory T cell immune responses. Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA-CHIKV 52 days after the last immunization. CHIKV-specific CD4 and CD8 memory T cell immune responses triggered by both immunization groups were measured by ICS assay as described in the legend to Fig. 6. Values from unstimulated controls were subtracted in all cases. P values indicate significantly higher responses in comparisons of MVA- WT/MVA-WT to MVA-CHIKV/MVA-CHIKV (***, P<0.001 ). Data are from one experiment that is representative of two independent experiments. (A) Overall magnitudes of CHIKV-specific CD4 and CD8 T cells. The values represent the sums of the percentages of T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against C plus E1 plus E2. (B) Pattern of C, E1 , and E2 CHIKV-specific CD8 memory T cell immune responses. Frequencies were calculated by reporting the number of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 and are specific for each peptide to the total number of CD8 T cells in the MVA-CHIKV/MVA-CHIKV immunization group. (C) Magnitudes of C, E1 , and E2 CHIKV-specific CD8 T cells. Frequencies represent the sums of the percentages of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against the C, E1 , or E2 peptide. (D) Magnitudes of CHIKV-specific CD8 T cells producing CD107a, IFN-γ, TNF-a or IL-2. Frequencies represent percentages of CD8 T cells producing CD107a, IFN-γ, TNF-a, or IL-2 against C plus E1 plus E2. (E) Polyfunctional profile of CHIKV-specific CD8 memory T cell immune responses. All the possible combinations of the responses are shown on the x axis (15 different T cell populations), while the percentages of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against C plus E1 plus E2 are shown on the y axis. Responses are grouped and color coded on the basis of the number of functions (4, 3, 2, or 1 ). The pie charts summarize the data. Each slice corresponds to the proportion of CHIKV-specific CD8 T cells exhibiting one, two, three, or four functions within the total population of CHIKV-specific CD8 T cells. (F) Phenotypic profile of CHIKV-specific CD8 memory T cells. CD127 and CD62L expression was used to identify central memory (TCM, CD127 CD62), effector memory (TEM, CD127 CD62L), and effector (TE, CD127CD62L) subpopulations. Magnitudes of TCM, TEM, and TE CHIKV-specific CD8 T cells are represented. The values represent percentages of TCM, TEM, and TE populations producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against the C plus E1 plus E2 peptides. The pie charts summarize the data. Each slice corresponds to the proportion of the TCM, TEM, or TE population within the total CHIKV-specific CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2. P values indicate significantly higher responses in comparisons of MVA-WT/MVA-WT to MVA-CHIKV/MVA-CHIKV (***, P< 0.001 ). FIG 9 VACV-specific memory T cell immune responses. Splenocytes were collected from mice (5 per group) immunized with MVA-WT/MVA-WT or MVA-CHIKV/MVA- CHIKV 52 days after the last immunization. Next, VACV-specific CD4 and CD8 memory T cell immune responses triggered by both immunization groups were measured by ICS assay following stimulation of splenocytes with MVA-infected EL4 cells. Values from unstimulated controls were subtracted in all cases. P values indicate significantly higher responses in comparisons of MVA-WT/MVA-WT to MVA- CHIKV/MVA-CHIKV (***, P<0.001 ). Data are from one experiment that is representative of two independent experiments. (A) Overall magnitude of VACV- specific CD4 and CD8 memory T cells. The values represent the sums of the percentages of T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells. (B) Percentage of VACV-specific CD8 T memory cells producing CD107a, IFN-γ, TNF-a, or IL-2. Frequencies represent percentages of CD8 memory T cells producing CD107a, IFN-γ, TNF-a, or IL-2 against MVA-infected EL4 cells. (C) Functional profiles of VACV-specific CD8 memory T cell immune responses. Responses are grouped and color coded on the basis of the number of functions (4, 3, 2, or 1 ). All possible combinations of the responses are shown on the x axis (15 different T cell populations), while the percentages of CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells are shown on the y axis. The pie charts summarize the data. Each slice corresponds to the proportion of VACV-specific CD8 memory T cells exhibiting one, two, three, or four functions within the total population of VACV-specific CD8 T cells. (D) Phenotypic profile of VACV-specific CD4 and CD8 memory T cells. Fluorescence-activated cell sorter (FACS) plots of MVA-infected EL4 cells are represented. CD127 and CD62L expression was used to identify central memory (TCM, CD127 CD62), effector memory (TEM, CD127 CD62L), and effector (TE, CD127CD62L) subpopulations. The memory T cell subpopulations are depicted as density plots. Blue dots represent T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2. The graph represents the magnitudes of TCM, TEM, and TE VACV-specific CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2. Frequencies represent percentages of TCM, TEM, and TE populations producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2 against MVA-infected EL4 cells. The pie charts summarize the data. Each slice corresponds to the proportion of the TCM, TEM, or TE population within the total population of VACV-specific CD8 T cells producing CD107a and/or IFN-γ and/or TNF-a and/or IL-2.
FIG 10 Immunization with MVA-CHIKV induces neutralizing antibodies against CHIKV and protects mice against CHIKV infection. Five C57BL/6 mice per group were immunized as described in Materials and Methods. Data are from one experiment that is representative of two independent experiments. (A) Antibody titers against CHIKV E1 -p62 detected by ELISA at 3 weeks after the prime or 6 weeks after the boost in sera of animals immunized with one dose of MVA-WT or one or two doses of MVA-CHIKV. The detection limit is 100. (B) CHIKV-neutralizing antibody titers (NT50) detected at 6 weeks after the last immunization in sera of animals immunized with one dose of MVA- WT or one or two doses of MVA-CHIKV. The detection limit is 100. (C) Viremia detected in animals immunized with one dose of MVA-WT or one or two doses of MVA- CHIKV and challenged with a total of 106 PFU of CHIKV subcutaneously in both feet. Blood samples were collected at days 1 to 3 postchallenge from tail bleedings, and viremia was analyzed by plaque assay (PFU/ml). The detection limit is 250 PFU/ml. (D) Foot swelling in animals immunized with one dose of MVA-WT or one or two doses of MVA-CHIKV and challenged with a total of 106 PFU of CHIKV subcutaneously in both feet. Foot swelling was analyzed on the day of challenge and days 4 to 8 following challenge.
FIG 1 1 CHIKV specific antibody responses induced by different CHIKV vaccine candidates using various prime-boost immunization strategies. C57BL/6 mice (n=5) were immunized once or twice with three weeks immunization intervals with various combinations of the CHIKV vaccine candidates depicted in Table 1 . (A) Total IgG titers and (B) lgG1 (red) and lgG2c (blue) titers in serum collected six weeks post the last immunization. (C) 50% neutralization titers (NT50) on serum collected prior to challenge. Bars show mean values (n=5). A Kruskal-Wallis test followed by Dunn's posttest was used to compare responses from CHIKV (WT). *, ** and *** indicate statistical differences of p<0.05, p<0.01 and p<0.001 , respectively.
FIG 12 Viremia and foot swelling after CHIKV challenge. Mice were challenged seven weeks post the last immunization with 106 PFU of CHIKV in the feet. (A) Viremia in serum collected two days after challenge. (B) Peak foot swelling of each mouse (mean of height times breadth of both feet relative to day 0) day 4-9 after challenge. Bars show mean values (n=5). A Kruskal-Wallis test followed by Dunn's post-test was used to compare responses from nal've mice (PBS) and a Spearman rank test was used to examine correlation between the magnitude of antibody responses prior to challenge and the level of viremia and foot swelling post challenge. *, ** and *** indicate statistical differences of p<0.05, p<0.01 and p<0.001 , respectively. (C and D) Correlation between the magnitude of anti-CHI KV antibody responses prior to challenge and the level of viremia and foot swelling after challenge. A Spearman rank test was used to examine the correlations.
FIG 13 Induction of CD8+ T cells with different CHIKV vaccine candidates. Groups of 5 C57BL/6 mice were infected once with wild-type CHIKV, or primed with DREP-Env and boosted 3 weeks later with DREP-Env, p62-E1 protein antigen and/or MVA-CHIKV, as indicated. Mice were sacrificed, and spleens were collected 8 days after the last immunization. CHIKV E1 peptide (HSMTNAVTI)-specific CD8+ T cells were identified by pentamer staining and characterized for the expression of CD62L/CD127 and CD27/CD43. DETAILED DESCRIPTION OF THE INVENTION
EXAMPLE 1 - MVA-CHIKV is highly immunogenic and protects mice against Chikungunya infection MATERIALS AND METHODS
Viruses. The poxvirus strain used in this study as the parental virus for the generation of the recombinant MVA-CHIKV is a modified vaccinia virus Ankara (MVA) strain (obtained from the Ankara strain after 586 serial passages in CEFs; this strain is derived from clone F6 at passage 585, kindly provided by G. Sutter, Germany). We modified this virus by inserting the gene encoding the green fluorescent protein (GFP) into the thymidine kinase (TK) locus and by deleting the immunomodulatory vaccinia virus (VACV) genes C6L, K7R, and A46R. The GFP cassette in the TK locus was replaced by the CHIKV cassette to generate MVA-CHIKV. We also used in this study the parental attenuated MVA (referred to as wild-type MVA [MVA-WT]). All viruses were grown in primary CEFs, purified by centrifugation through two 36% (wt/vol) sucrose cushions in 10 mM Tris-HCI, pH 9, and titrated in DF-1 cells by a plaque immunostaining assay, as previously described. The titer determinations of the different viruses were performed at least three times. All viruses were free of contamination with mycoplasmas, fungi, or bacteria.
Construction of the infectious CHIKV clone LR2006-OPY1 has previously been described. Briefly, a plasmid for production of CHIKV was generated by cloning the CHIKV cDNA under the control of the SP6 RNA polymerase promoter. CHIKV was subsequently produced in BHK-21 cells as previously described.
Construction of the plasmid transfer vector pCyA20-ICRESl . The plasmid transfer vector pCyA-ICRES1 was generated and used for the construction of recombinant MVA-CHIKV, in which CHIKV structural genes (those for C, E3, E2, 6K, and E1 ) instead of the GFP gene were inserted into the TK locus of the parental MVA-GFP. In detail, a 3.7-kbp DNA fragment encoding the CHIKV structural proteins (C, E3, E2, 6K, and E1 ) was amplified with oligonucleotides ICRES1 upper (5'-ATATA GTTTAAACATGGAGTTCATCCCAACCCAAAC-3': Pmel site underlined) and ICRES1 lower (5'-TATATAGATCTTTAGTGCCTGCTGAACGACACG-3'; Bglll site underlined) from plasmid pSP6-ICRES1 , which contains the structural genes from the CHIKV genome (CHIKV isolate LR2006-OPY1 ). The amplification reaction was performed with Platinum Pfx DNA polymerase (Invitrogen) according to the manufacturer's recommendations. Then, this fragment was digested with Pmel and Bglll and cloned into the VACV insertion vector pCyA-20 (7,582 bp), previously digested with the same restriction enzymes and dephosphorylated by incubation with shrimp alkaline phosphatase, to generate the plasmid transfer vector pCyA-ICRES1 (1 1 ,324 bp). The plasmid pCyA-20, used for the generation of plasmid pCyA-ICRES1 , was previously described and contains a synthetic band containing the viral early/late promoter and a multiple-cloning site in the plasmid pLZAWI (provided by Sanofi-Pasteur). The correct generation of pCyA-ICRES1 was further confirmed by DNA sequence analysis. This plasmid was used in transient infection and transfection assays for the insertion of CHIKV structural genes (those for C, E3, E2, 6K, and E1 ) into the TK locus of the MVA genome under the transcriptional control of the viral synthetic early/late (sE/L) promoter. It contains a B-galactosidase (B-Gal) reporter gene sequence between two repetitions of the left TK-flanking arm, which allows the reporter gene to be deleted from the final recombinant virus by homologous recombination after successive passages.
Construction of recombinant MVA-CHIKV. DF-1 cells (3 x 106 cells) were infected with parental MVA-GFP at a multiplicity of infection (MOI) of 0.05 PFU/cell and transfected 1 h later with 10 ug of DNA of plasmid pCyA-ICRES1 , using Lipofectamine reagent according to the manufacturer's recommendations (Invitrogen). At 48 h postinfection (hpi), the cells were harvested, lysed by freeze-thaw cycling, sonicated, and used for recombinant-virus screening. Recombinant MVAs containing the 3.7-kb CHIKV DNA fragment (encoding the structural proteins of CHIKV, C, E3, E2, 6K, and E1 ), with the GFP gene deleted, and transiently coexpressing the B-Gal marker gene (MVA-CHIKV, X-Gal+) were selected by consecutive rounds of plaque purification in DF-1 cells stained with X-Gal (5-bromo-4-chloro-3-indolyl-B-D-galactopyranoside, 400 ug/ml, for three passages in total). In the following plaque purification steps, recombinant MVAs containing the 3.7-kb CHIKV DNA fragment and with the B-Gal gene deleted by homologous recombination from between the left TK arm and the short left TK arm repeat flanking the marker (MVA-CHIKV, X-Gal-) were isolated by three additional consecutive rounds of plaque purification screening for nonstaining viral foci in DF-1 cells in the presence of X-Gal (400 ug/ml). In each round of purification, the isolated plaques were expanded in DF-1 cells for 3 days, and the crude viruses obtained were used for the next plaque purification round. The resulting recombinant virus, MVA-CHIKV (passage 2 [P2] stock), was grown in CEFs, purified through two 36% (wt/vol) sucrose cushions, and titrated by plaque immunostaining assay.
Expression of CHIKV proteins from recombinant MVA-CHIKV by Western blotting. To check the correct expression of CHIKV antigens by the recombinant MVA- CHIKV, monolayers of DF-1 cells were mock infected or infected at 5 PFU/cell with MVA-WT, MVA-GFP, or MVACHIKV. At 24 hpi, cells were lysed in Laemmli buffer, and cells extracts were fractionated in 10% SDS-PAGE and analyzed by Western blotting with mouse polyclonal antibody against E2 (antibody 3E4, kindly provided by Philippe Despres from the Pasteur Institute, Paris, France; diluted 1 :500), mouse monoclonal antibody against E1 -E2 (antibody D3-62; kindly provided by Marc Lecuit and Therese Couderc from the Pasteur Institute, Paris, France; diluted 1 :500), or rabbit polyclonal antibody against E1 (kindly provided by Lisa F. P. Ng from the Singapore Immunology Network, Agency for Science, Technology and Research, Biopolis, Singapore; diluted 1 :1 ,000) to evaluate the expression of the different CHIKV structural proteins. The anti- rabbit HRP-conjugated antibody (Sigma; diluted 1 :5,000), or anti-mouse-HRP- conjugated antibody (Sigma; diluted 1 :2,000), were used as secondary antibodies. The immunocomplexes were detected using an enhanced HRP-luminol chemiluminescence system (ECL Plus; GE Healthcare).
Analysis of MVA-CHIKV
Analysis of genetic stability of recombinant MVA-CHIKV, immunofluorescence studies, and electron microscopy analysis were carried out as previously described in Gomez CE, Perdiguero B, Cepeda MV, Mingorance L, Garcia-Arriaza J, Vandermeeren A, Sorzano CO, Esteban M. 2013. High, broad, polyfunctioned and durable T cell immune responses induced in mice by a novel hepatitis C virus (HCV) vaccine candidate (MVA- HCV) based on modified vaccinia virus Ankara expressing the nearly full-length HCV genome. J. Virol. 87:7282-7300. http://dx.doi.org/10.1 128/JVI.03246-12. (reference 58).
Peptides. CHIKV peptides representative of the capsid (C), E1 , and E2 CHIKV proteins were used in the immunological analysis. Peptides sequences are as follows: ACLVGDJVM (capsid), HSMTNAAVTI (E1 ), and IILYYYELY (E2).
C57BL/6 mouse immunogenicity study. Female C57BL/6 mice (6 to 8 weeks old) were purchased from Harlan Laboratories and stored in a pathogen-free barrier area of the Centra Nacional de Biotecnologia (Madrid, Spain) in accordance with recommendations of the Federation of European Laboratory Animal Science Associations. A homologous MVA prime-MVA boost immunization protocol was performed to assay the immunogenicity of the recombinant MVA-CHIKV. Groups of animals (n 10) were immunized with 1 x107 PFU of MVA-WT or MVA-CHIKV by the intraperitoneal (i.p.) route in 200 ul of PBS. Two weeks later, animals received 2 x 107 PFU of MVA-WT or MVA-CHIKV by the i.p. route in 200ul of PBS. At 10 and 52 days after the last immunization, five mice in each group were sacrificed using carbon dioxide (C02), and their spleens were processed to measure the adaptive and memory immune responses, respectively, to the CHIKV and VACV antigens by an intracellular cytokine staining (ICS) assay. Two independent experiments were performed. The ability of MVA-CHIKV to induce antibody responses was studied in mice at the AFL at Karolinska Institutet, Stockholm, Sweden. Groups of 5- to 6-week-old female C57BL/6 mice (n 5) were immunized with 1 x107 PFU of MVA-WT or MVA-CHIKV by the i.p. route in 200 ul PBS. MVA-WT was administered once, whereas MVA-CHIKV was administered once or twice with a 3-week immunization interval. Serum was collected by tail bleedings 3 weeks after the first immunization and 6 weeks after the second immunization, and antibody responses were analyzed by enzyme-linked immunosorbent assay (ELISA) and a neutralization assay.
CHIKV challenge study. A challenge study to evaluate the efficacy of the recombinant MVA-CHIKV was performed in a biosafety level 3 laboratory at the Astrid Fagraeus Laboratory at Karolinska Institutet, Stockholm, Sweden. Female C57BL/6 mice previously immunized with MVA-WT or MVA-CHIKV to study antibody responses were used (see "C57BL/6 mouse immunogenicity study" above [5 mice/group]). Seven weeks after the last immunization, mice were challenged with a total of 106 PFU of CHIKV (CHIKV isolate LR2006-OPY1 ) via the subcutaneous route in the dorsal side of each hind foot. Blood samples were collected at days 1 to 3 post challenge from tail bleedings, and viremia was analyzed by plaque assay (PFU/ml). Foot swelling (height by breadth) was measured using a digital caliper before challenge and days 4 to 8 after challenge. Two independent experiments were performed.
ICS assay. The magnitudes, polyfunctionality, and phenotypes of the VACV- and CHIKV-specific adaptive and memory T cell responses were analyzed by ICS, as previously described, with some modifications. After an overnight rest, 4 x 106 splenocytes (depleted of red blood cells) were seeded on M96 plates and stimulated for 6 h incomplete RPMI 1640 medium supplemented with 10% FCS containing 1 ul/ml GolgiPlug (BD Biosciences) (to inhibit cytokine secretion), anti-CD107a-Alexa Fluor 488 (BD Biosciences), Monensin (1 x; eBioscience), and 1 ug/ml of the different CHIKV peptides (C, E1 , or E2) or MVA-infected EL4 cells (4 x 105 EL4 cells in 1 x 106 splenocytes; the ratio of MVA infected EL4 cells to splenocytes was equal to 1 :2.5). Then, cells were washed, stained for the surface markers, fixed, permeabilized (Cytofix/ Cytoperm kit; BD Biosciences), and stained intracellular^ with the appropriate fluorochromes. Dead cells were excluded using the violet LIVE/ DEAD stain kit (Invitrogen). For functional analyses, the following fluorochrome-conjugated antibodies were used: CD3-phycoerythrin (PE)-CF594, CD4-allophycocyanin (APC)-Cy7, CD8- V500, IFN-Y-PE-Cy7, tumor necrosis factor alpha (TNF-a)-PE, and interleukin 2 (IL- 2)-APC. In addition, for phenotypic analyses, the following antibodies were used: CD62L-Alexa 700 and CD127-PerCP-Cy5.5. All antibodies were from BD Biosciences. Cells were acquired using a Gallios flow cytometer (Beckman Coulter). Analyses of the data were performed using FlowJo software version 8.5.3 (Tree Star, Ashland, OR). The number of lymphocyte- gated events was 106. After gating, Boolean combinations of single functional gates were then created using FlowJo software to determine the frequency of each response based on all possible combinations of cytokine.
RESULTS
Generation and in vitro characterization of an MVA recombinant expressing CHIKV structural genes (MVA-CHIKV). Fig. 1 A shows a diagram of the MVA-CHIKV genome. The correct insertion and purity of recombinant MVA-CHIKV was confirmed by PCR and DNA sequence analysis. PCR using primers annealing in the VACV TK gene-flanking regions confirmed the presence of the CHIKV structural genes in MVA- CHIKV, with no wild-type contamination in the preparation (Fig. 1 B); in the parental virus MVA-GFP, the GFP was present, and in the MVA-WT, the VACV TK locus was amplified (Fig. 1 B). Moreover, PCRs using primers annealing in the C6L-, K7R-, and A46R-flanking regions confirmed the deletion of the VACV genes C6L, K7R, and A46R, respectively, from MVA-CHIKV (Fig. 1 C), also with no wild-type contamination in the preparation. The proper insertion and correct sequence of the CHIKV structural genes in the TK locus and the correct deletions of the VACV genes C6L, K7R, and A46R were also confirmed by DNA sequencing (data not shown). Additionally, to confirm that MVA-CHIKV constitutively expresses and correctly processes the CHIKV structural polyprotein C-E3-E2-6K-E1 , we carried out a Western blot analysis of DF-1 cells infected with MVA-CHIKV, MVA-GFP, or MVA-WT using specific antibodies that recognize the CHIKV E1 and E2 proteins. The results demonstrated that the viral polyprotein was correctly processed and that MVA-CHIKV expressed the CHIKV E1 and E2 proteins (Fig. 1 D). Expression of the C, E3, and 6K proteins is not shown, since we did not have specific antibodies. However, the proper processing of the polyprotein indicates that the other proteins should be correctly expressed by MVA-CHIKV.
MVA-CHIKV replicates in cell culture. To further determine whether expression of CHIKV structural proteins affects MVA replication in cell culture, we next analyzed the growth of MVACHIKV and MVA-WT in DF-1 cells. The results showed that the kinetics of viral growth were similar between the two viruses (Fig. 2A), indicating that the constitutive expression of CHIKV structural proteins does not impair vector replication under permissive conditions.
MVA-CHIKV is stable in cell culture. To ensure that MVACHIKV can be maintained in cultured cells without the loss of the CHIKV transgene, a stability test was performed. Here, the recombinant MVA-CHIKV was further grown in DF-1 cells infected at a low multiplicity during 9 consecutive passages, and expression of the CHIKV E1 and E2 proteins during the different passages was determined by Western blotting (Fig. 2B). The results showed that MVA-CHIKV efficiently expresses the CHIKV E1 and E2 proteins after successive passages, indicating that the recombinant MVACHIKV is genetically stable. Moreover, we also analyzed the genetic stability of MVA-CHIKV at passage 9 by a plaque immunostaining assay (Fig. 2C). The results showed that MVA-CHIKV titers (in the P2 stock, in the P3 stock, and at passage 9) were similar when plaques expressing VACV antigens or plaques expressing CHIKV antigens were analyzed, confirming the high genetic stability of MVA-CHIKV.
MVA-CHIKV expresses the CHIKV E2 protein in the cytoplasm and in the cell membrane. The expression and intracellular localization of the structural CHIKV E2 protein was also analyzed by immunofluorescence in human HeLa cells infected with MVA-WT or MVA-CHIKV using antibodies specific to E2 and specific probes or antibodies for different proteins and compartments, such as actin (phalloidin), ER (anticalnexin), and the Golgi apparatus or Golgi apparatus-derived membranes, as well as for the plasma membrane (WGA). As shown in Fig. 3, the CHIKV E2 protein is expressed mainly at the plasma membrane and throughout the cytoplasm, where it is localized in discrete accumulations at the viral factories. Moreover, no colocalization between E2 and the ER was observed. Nevertheless, partial colocalization between E2 and actin was found in discrete areas and spots, and it was also detected between E2 and the plasma membrane and between E2 and the Golgi apparatus or Golgi apparatus-derived membranes.
MVA-CHIKV induces specific morphological cell alterations, with the formation and accumulation of Golgi apparatus derived membranes. We next examined the impact of CHIKV structural proteins constitutively expressed by MVA-CHIKV on HeLa cell architecture using transmission electron microscopy analysis. Thus, ultrathin sections of HeLa cells infected with MVA-WT or MVA-CHIKV were visualized by electron microscopy at low and high magnifications (Fig. 4). Both in MVA-WT and in MVA-CHIKV-infected cells, the assembly of immature virus (IV) forms of MVA was detected, and remarkably, in MVA-CHIKV- infected cells, an atypical growth and accumulation of Golgi apparatus-like membranes was observed.
MVA-CHIKV triggers an innate immune response in human macrophages and dendritic cells, inducing type I IFN, proinflammatory cytokines, and chemokine expression. Type I IFN innate immune responses play a critical role in controlling CHIKV viral replication. Thus, to evaluate whether the presence of the CHIKV structural genes in the MVA genome is able to impair the response of innate immune cells to MVA infection, we analyzed by real-time PCR the expression of type I IFN (IFN-β), proinflammatory cytokines (TNF-a), chemokines (MIP-1 a, IP-10, and RANTES), IFN-inducible genes (IFIT1 and IFIT2), and some key cytosolic sensors that lead to antiviral IFN production (RIG-I and MDA-5) by human THP-1 macrophages that had been mock infected or infected for 3 and 6 h with 5 PFU/cell of MVA-WT, MVA-GFP, and MVA-CHIKV (Fig. 5A). The results showed that, compared to MVA- WT, MVA-CHIKV significantly upregulated the mRNA levels of most of these genes (Fig. 5A). To verify these results, we infected human moDCs with 1 PFU/ cell of MVA- WT, MVA-GFP, and MVA-CHIKV and measured IFN-β, TNF-a, MIP-1 a, IP-10, RANTES, IFIT1 , IFIT2, RIG-I, and MDA-5 mRNA levels at 6 h postinfection (Fig. 5B). In the same way as in human THP-1 cells, in moDCs, MVA-CHIKV strongly increased the mRNA levels of most of these genes, compared to levels in MVA-WT-infected cells (Fig. 5B). In conclusion, MVA-CHIKV promotes a robust innate immune response in human macrophages and moDCs by inducing the expression of type I IFN (IFN-β), proinflammatory cytokines (TNF-a), chemokines (MIP-1 a, IP-10, and RANTES), IFN- in-ducible genes (IFIT1 and IFIT2), and some key cytosolic sensors that lead to antiviral IFN production (RIG-I and MDA-5).
MVA-CHIKV induces strong, broad, and polyfunctional adaptive CHIKV-specific T cell immune responses. The role of T cell responses in controlling CHIKV infection is not well known. Thus, to study in vivo the effect of the presence of the CHIKV structural genes in the MVA genome on cellular immunogenicity against CHIKV antigens, we next analyzed the CHIKV specific immune responses induced by MVA- CHIKV in C57BL/6 mice by using a homologous MVA-CHIKV prime (1 x107 PFU) and MVA-CHIKV boost (2x107 PFU) immunization protocol (see Materials and Methods). Animals primed with nonrecombinant MVA-WT and boosted with MVA-WT were used as a control group. Adaptive, CHIKV-specific T cell immune responses elicited by both immunization groups (MVA-WT/MVA-WT and MVA-CHIKV/MVA-CHIKV) were measured 10 days after the last immunization by a polychromatic intracellular cytokine staining (ICS) assay, after the stimulation of splenocytes with specific peptides representative of the C, E1 , and E2 CHIKV proteins. Immunization with MVA- CHIKV/MVA-CHIKV elicited robust adaptive CHIKV-specific CD8+ T cell immune responses (determined as the sum of the individual responses obtained for the C, E1 , and E2 CHIKV peptides, producing IFN-γ , TNF-a, and/or IL-2 cytokines, as well as the expression of CD107a on the surfaces of activated T cells as an indirect marker of cytotoxicity) (Fig. 6A). The MVA-CHIKV/MVA-CHIKV immunization group triggered an overall CHIKV-specific immune response mediated only by CD8+ T cells, with no CHIKV-specific CD4+ T cells detected, indicating the selectivity for CD8+ T cells of the peptides used in the ICS assay (Fig. 6A). The pattern of adaptive, CHIKV-specific T cell immune responses showed that CD8+ T cell responses induced by MVACHIKV/ MVA-CHIKV were broad, with most of the responses directed mainly against the E1 and E2 peptides (80%) and to a lesser extent against C (Fig. 6B). Furthermore, compared to MVAWT/MVA-WT, MVA-CHIKV/MVA-CHIKV significantly enhanced the magnitude of C-, E1 - and E2-specific CD8+ T cell responses (P< 0.001 ) (Fig. 6C). Moreover, CHIKV-specific CD8+ T cells producing CD107a, IFN-γ, or TNF-a are the populations most induced by the MVACHIKV/MVA-CHIKV immunization group (Fig. 6D); levels were also of a significantly higher magnitude than those induced by MVAWT/MVA-WT (P< 0.001 ) (Fig. 6D). The quality of the adaptive, CHIKV-specific T cell immune response was characterized in part by the pattern of cytokine production and its cytotoxic potential. Thus, on the basis of the production of CD107a, IFN-y, TNF-a, and IL-2 from CHIKV-specific CD8+ T cells, 15 different CHIKV-specific CD8+ T cell populations could be identified (Fig. 6E). MVA-CHIKV/MVACHIKV induced a high polyfunctional profile, with 97% of CD8+ T cells exhibiting two, three, or four functions (Fig.6E). Furthermore, CD8+ T cells producing CD107a plus IFN-y plus TNF-a plus IL- 2, CD107a plus IFN-y plus TNF-a, or IFN-y plus TNF-a were the most abundant populations elicited by MVA-CHIKV/MVA-CHIKV, which, compared to MVA-WT/MVA- WT, also induced significantly higher increases in the percentages of most of the populations (P< 0.001 ) (Fig. 6E). Collectively, these results demonstrate that MVACHIKV induced strong, broad, and polyfunctional adaptive CHIKV-specific CD8+ T cell immune responses. Similar findings were observed in two independent experiments. MVA-WT and MVA-CHIKV induce similar magnitudes and polyfunctionalities of adaptive, VACV-specific T cell immune responses. To further characterize the immune responses elicited in mice by both immunization groups (MVA-WT/MVA-WT and MVA-CHIV/MVA-CHIKV), it was of interest to analyze the responses to the MVA vector. Thus, we next measured adaptive, VACV-specific T cell immune responses induced by MVA-WT and MVA-CHIKV 10 days after the boost by an ICS assay (similar to the protocol followed in the analysis of adaptive CHIKV-specific T cell immune responses) after the stimulation of splenocytes with MVA-infected EL4 cells. Both immunization groups triggered an overall adaptive, VACV-specific immune response mediated only by CD8+ T cells (determined as the sum of the individual responses producing IFN-γ, TNF-a, and/or IL-2 cytokines, as well as the expression of CD107a on the surfaces of activated T cells as an indirect marker of cytotoxicity); this experiment was carried out with MVA-infected EL4 cells, and the magnitudes of adaptive, VACV-specific CD8+ T cell immune responses were similar in the two immunization groups (Fig. 7A). Moreover, VACV-specific CD8+ T cells producing CD107a, IFN-γ, or TNF a-were the most induced populations in both immunization groups, and they were induced at similar magnitudes (Fig. 7B). The quality of adaptive, VACV-specific CD8+ T cell immune responses was characterized by the production of CD107a, IFN-γ, TNF-a, and/or IL-2, and 15 distinct VACV-specific CD8+ T cell populations could be identified (Fig. 7C). VACV-specific CD8+ T cell responses were similarly polyfunctional in the two immunization groups, with 88 to 91 % of the CD8+ T cells exhibiting two, three, or four functions. CD8+ T cells producing CD107a plus IFN-Y plus TNF-a plus IL-2, CD107a plus IFN-γ plus TNF-a, IFN-γ plus TNF-a, CD107a plus IFN-γ, or only CD107a were the most induced populations elicited by both immunization groups (at similar percentages) (Fig. 7C). In summary, these results showed that MVA-WT and MVACHIKV elicited similar magnitudes and levels of quality of adaptive, VACV-specific CD8+ T cell immune responses. Similar findings were observed in two independent experiments.
MVA-CHIKV induces strong, broad, polyfunctional, and durable CHIKV-specific memory T cell immune responses. The durability of a vaccine-induced T cell response is an important feature, since long-term protection is a requirement for prophylactic vaccination. Thus, we analyzed CHIKV-specific memory T cell immune responses elicited by both immunization groups 52 days after the last immunization by the ICS assay in a manner similar to that of the protocol followed in the adaptive phase. Immunization with MVA-CHIKV/MVA-CHIKV elicited robust CHIKV-specific CD8+ memory T cell immune responses (determined as the sum of the individual responses to the CHIKV C, E1 , and E2 peptides in splenocytes producing IFN-γ , TNF-a, and/or IL-2 cytokines, and the expression of CD107a on the surfaces of activated T cells was used as an indirect marker of cytotoxicity) (Fig. 8A). As with the results obtained in the adaptive phase, the MVA-CHIKV/MVA-CHIKV immunization group triggered an overall CHIKV-specific memory immune response mediated only by CD8+ T cells, with no CHIKV-specific CD4+ T cells detected, indicating the selectivity for CD8+ T cells of the peptides used in the ICS assay (Fig. 8A). The pattern of CHIKV-specific memory T cell immune responses showed that CD8 T cell responses induced by MVACHIKV/ MVA-CHIKV were broad, with most of the responses being directed mainly against the E1 and E2 peptides (77%) and, to a lesser extent, against C (Fig. 8B), as with the results obtained in the adaptive phase. Furthermore, compared to MVA-WT/MVAWT, MVA-CHIKV/MVA-CHIKV significantly enhanced the magnitudes of C-, E1 -, and E2- specific CD8+ T cell responses (PO.001 ) (Fig. 8C). Furthermore, CHIKV-specific CD8+ T cells producing CD107a, IFN-γ, or TNF-a are the populations most induced by the MVACHIKV/MVA-CHIKV immunization group (Fig. 8D), and their levels are also of a significantly higher magnitude than those induced by MVA-WT/MVA-WT (PO.001 ) (Fig. 8D), results similar to those obtained in the adaptive phase. The quality of the CHIKV-specific memory T cell immune responses was characterized by analyzing the simultaneous production of CD107a, IFN-γ , TNF-a, and/or IL-2 from CHIKV specific CD8+ T cells (Fig. 8E), where 15 distinct CHIKV-specific CD8+ T cell populations could be identified. MVA-CHIKV/MVA-CHIKV induced a polyfunctional profile, with 22% of CD8+ T cells exhibiting two, three, or four functions (Fig. 8E). Furthermore, again, CD8+ T cells producing CD107a plus IFN-γ plus TNF-a plus IL-2, CD107a plus IFN-y plus TNF-a, or CD107a were the most abundant populations elicited by MVACHIKV/ MVA-CHIKV, which also induced increases in the percentages of most of the populations that were significantly higher than those induced by MVA-WT/MVA-WT (P<0.001 ) (Fig. 8E), results similar to those obtained in the adaptive phase.
Moreover, we also determined the phenotype of the C-, E1 -, and E2-specific memory T cells by measuring the expression of CD127 and CD62L surface markers, which allowed us to define the different memory subpopulations: central memory (TCM; CD127/CD62L), effector memory (TEM; CD1277CD62L"), and effector (TE;CD127" /CD62L") T cells (Fig. 8F). The results showed that MVA-CHIKV/MVA-CHIKV induced CHIKV-specific CD8+ memory T cells (determined as the sum of the individual responses, i.e., the levels of CD107a, IFN-γ , TNF-a, and/or IL-2 produced against the C plus E1 plus E2 peptides), mainly of the TEM phenotype (78%) (Fig.8F, right). Furthermore, immunization with MVA-CHIKV/MVA-CHIKV induced significantly higher increases in the percentages of CHIKV-specific CD8 TCM,TEM,and TE cells than immunization with MVA-WT (PO.001 ) (Fig. 8F, left). In summary, these results demonstrate that MVA-CHIKV induced strong, broad, polyfunctional, and durable CHIKV-specific CD8+ memory T cell immune responses. Similar findings were observed in two independent experiments.
MVA-WT and MVA-CHIKV induce similar magnitudes and polyfunctionalities of VACV-specific memory T cell immune responses. VACV-specific memory T cell immune responses elicited by both immunization groups (MVA-WT/MVA-WT and MVA- CHIV/MVA-CHIKV) were measured 52 days after the boost by ICS assay, which is similar to the protocol followed in the adaptive phase. Similarly to the results obtained in the adaptive phase, both immunization groups triggered overall VACV-specific memory immune responses mediated only by CD8+ T cells, determined as the sums of the individual levels of production of CD107a, IFN-γ, TNF-a, and/or IL-2 obtained in MVA-infected EL4 cells (Fig. 9A), with the magnitudes of VACV-specific CD8+ memory T cell immune responses being similar in the two immunization groups (Fig. 9A). Moreover, VACV-specific CD8+ T cells producing CD107a, IFN-γ, or TNF-a are the most induced populations in both immunization groups, and these populations were also produced at similar magnitudes (Fig. 9B). The quality of VACV-specific CD8+ memory T cell immune responses was characterized as the simultaneous production of CD107a, IFN- γ , TNF-α, and/or IL-2 and was assessed by using a protocol like that followed in the adaptive phase, where 15 distinct VACV-specific CD8+ T cell populations could be identified (Fig.9C). VACV-specific CD8+ T cell responses were similarly polyfunctional in the two immunization groups, with 95% of CD8+ T cells exhibiting two, three, or four functions. CD8+ T cells producing CD107a plus IFN-γ plus TNF-a plus IL-2, CD107a plus IFN- γ plus TNF-a, IFN-γ plus TNF-a, CD107a plus IFN- Y, or only CD107a were the most induced populations elicited by both immunization groups, and they were also induced at similar percentages (Fig. 9C). Additionally, the analysis of the memory phenotype of VACV specific CD8+ memory T cells by measurement of the expression of CD127 and CD62L surface markers showed that both immunization groups elicited mainly VACV-specific CD8+ memory T cells of the TEM phenotype and did so at similar magnitudes (Fig. 9D). In summary, these results showed that MVA-WT and MVACHIKV elicited similar magnitudes and levels of quality of VACV specific CD8+ memory T cell immune responses. Similar findings were observed in two independent experiments.
MVA-CHIKV induces high titers of neutralizing antibodies against CHIKV.
Antibodies against CHIKV are crucial to control CHIKV infection. Thus, to study the ability of MVA-CHIKV to elicit humoral immune responses against CHIKV, we analyzed the levels of CHIKV envelope-specific antibodies present in the sera of C57BL/6 mice immunized with one or two doses of MVA-CHIKV (see Materials and Methods). Animals immunized with nonrecombinant MVA-WT were used as a control group. The results show that one immunization with MVA-CHIKV elicited high titers of IgG antibodies against CHIKV that were further enhanced by the second immunization (Fig. 10A). Nevertheless, a single immunization with MVA-CHIKV was enough to induce high levels of IgG antibody responses to CHIKV. Next, we analyzed the titers of neutralizing antibodies against CHIKV present in the sera of immunized mice at 6 weeks postboost. The results showed that in contrast to infection with MVA-WT, immunization with one or two doses of MVA-CHIKV resulted in the production of high titers of CHIKV-neutralizing antibodies (Fig. 10B). In comparison to neutralizing antibody titers after a single immunization, titers after two doses were slightly increased. MVA-CHIKV protects mice against CHIKV infection. To study whether MVA-CHIKV protected mice against a CHIKV challenge, we immunized mice with one or two doses of MVACHIKV or one dose of MVA-WT (as a control). Seven weeks after the last immunization, mice were challenged with a high dose of CHIKV in their feet (see Materials and Methods). Protection was evaluated by determining viremia and analyzing foot swelling in CHIKV-challenged mice during the days following challenge. The results showed that no CHIKV infection was developed in MVACHIKV- vaccinated mice, as no viremia was detected postchallenge (Fig. 10C). However, MVA-WT-immunized mice developed CHIKV infection, with high levels of CHIKV (104 to 105 PFU/ml) detected in blood that peaked day 2 postchallenge (Fig. 10B). Moreover, MVA-CHIKV-vaccinated mice did not develop foot swelling after challenge, while MVA-WT-immunized mice developed severe foot swelling that peaked 6 days postchallenge (Fig. 10D). In conclusion, MVA-CHIKV is a highly effective vaccine that protected mice against challenge with a high dose of CHIKV, with no viremia detected in their blood and no signs of inflammation. Remarkably, just a single dose of MVA- CHIKV protected all mice from challenge with CHIKV.
EXAMPLE 2 - PRIME-BOOST IMMUNIZATION STRATEGIES AGAINST CHIKUNGUNYA VIRUS
In this study, in addition to the previously described attenuated A5nsp3 (Hallengard et al, 2013, J Virol epub ahead of print 26 December 2013, doi: 10.1 128/JVI.03453-13) and recombinant MVA-CHIKV vaccine candidates, novel p62- E1 protein and DNA replicon (DREP) based CHIKV vaccines were compared. We evaluated the immunogenicity and efficacy in mice immunized with several homologous and heterologous prime-boost immunization protocols using distinct CHIKV vaccine candidates representing different antigens and vaccine modalities. The DREP platform differs from conventional DNA plasmids in that it encodes the alphavirus (CHIKV) replicase, which drives the production of the subgenomic RNA and thus the expression of the encoded CHIKV antigen. Moreover, DREPs also possess intrinsic adjuvant properties as the replicase and RNA intermediates stimulate the production of type-1 interferons (IFNs) and apoptosis. Promising results have been reported for DNA replicons generated from other alphaviruses including Semliki Forest virus, Sindbis virus and Venezuelan equine encephalitis virus, when used for priming immunizations prior to boosting with other vaccine modalities.
The heterologous prime-boost approach takes advantage of the unique immune profile being induced by the different vaccine platforms. For example, both attenuated and genetic vaccines are being produced endogenously and can thus give rise to T cell- mediated immune responses. In contrast, protein antigens generally lack the ability to elicit cytotoxic T cell responses and are thus limited to the induction of humoral responses. Combining different vaccine strategies in heterologous prime-boost immunizations should induce a more balanced immune response in terms of cellular and humoral immune responses, and enhance the magnitude and quality of immune responses compared to homologous vaccination using a single vaccine modality alone.
The novel DREP and protein CHIKV vaccine candidates described in this study were both immunogenic and efficient when used in priming and boosting immunizations. Furthermore, we have identified several homologous and heterologous prime-boost immunization strategies that were able to elicit protective immune responses in the CHIKV mouse model. This important information will form the basis for immunogen selection in further preclinical and clinical testing of these CHIKV vaccine candidates. MATERIALS AND METHODS
CHIKV vaccine candidates. All vaccine constructs were based on the CHIKV clone LR2006-OPY1 . A DNA replicon vaccine encoding CHIKV envelope (termed DREP- Env) was constructed on the basis of cytomegalovirus (CMV) promoter launched infectious cDNA clone of CHIKV. Briefly, the region corresponding to the nucleotide residues 7565-8350 of CHIKV genome was replaced by sequence 5' CCTAGGCCACCATG 3' by PCR-based mutagenesis followed by multi-step subcloning procedure. The resulting deletion removed the CHIKV capsid coding sequence. In addition, the first amino acid residue of E3 protein (serine) was replaced by methionine.
Construction of the A5nsP3, MVA-CHIKV and p62-E1 CHIKV vaccine candidates have been described previously. Briefly, A5nsP3 is an attenuated CHIKV that was formed by attenuating an infectious CHIKV by a large deletion in the nsP3 gene, MVA-CHIKV was constructed by inserting the cDNA encoding for the structural polyprotein of CHIKV (C, E3, E2, 6K, E1 ) into MVA, and soluble recombinant p62-E1 CHIKV protein was formed by joining the ectodomains of CHIKV p62 and E1 proteins with a glycine serine linker. Wild type (WT) CHIKV was used for challenge and as a reference for immunizations.
Immunizations. Female C57BL/6 mice, 5-6 weeks old, were immunized once or twice with the different CHIKV vaccine candidates DREP-Env, A5nsP3, MVA-CHIKV or CHIKV p62-E1 , using various homologous or heterologous prime-boost immunization protocols with a three weeks immunization interval between prime and boost. A total of 10 μg of DREP-Env DNA diluted in 2x20 μΙ of PBS was injected intradermal^ followed by in vivo electroporation using the DermaVax™ electroporation device as previously described. 105 plaque-forming units (PFU) of A5nsP3 or WT CHIKV diluted in 2x50 μΙ of PBS was injected subcutaneously and 107 PFU of recombinant MVA-CHIKV was diluted in 200 μΙ of PBS and injected intraperitoneally. 1 μg CHIKV p62-E1 protein was diluted in 2x50 μΙ of PBS and administered intramuscularly. CHIKV p62-E1 protein was injected alone or mixed with 25 μΙ of the adjuvant AS03 (1 :10 human dose) (GlaxoSmithKline Biologicals S.A., Rixensart, Belgium) or with 5 μg of the adjuvant Matrix-M (Novavax, MD, USA). Some groups of mice were immunized with two vaccine candidates at the same occasion but at different injection sites. All experiments were performed in at least two separate iterations. B and T cell responses. CHIKV-specific humoral and cellular immune responses induced prior to challenge by the different prime-boost immunization protocols were assessed by enzyme-linked immunosorbent assay (ELISA), neutralization assay and I FN-γ ELISpot (Mabtech AB, Nacka Strand, Sweden) as previously described. In the I FN-γ ELISpot assay, splenocytes from immunized mice were collected 8 days after the last immunization and 105 splenocytes were plated per well. Then, cells were stimulated with 2.5 μς/ιτιΙ of a CD8 T cell-restricted CHIKV E1 peptide (HSMTNAVTI) or 10 μς/ιτιΙ of CHIKV p62-E1 protein. Responses over 25 Spot Forming Units (SFU)/106 splenocytes and a minimum of four times above background were regarded as positive.
Epitope determination and structural localization. Specific linear B cell epitopes on CHIKV E2 glycoprotein were identified via peptide-based ELISA assays. Briefly, heat- inactivated pooled sera from vaccinated mice were screened using overlapping synthetic 18-mer biotinylated-peptides synthesized based on consensus E2 glycoprotein sequence (Mimotopes). Streptavidin-coated 96-wells plates (Nunc) were first blocked with 1 % w/v sodium caseinate (Sigma-Aldrich) in 0.1 % PBST (0.1 % Tween-20 in PBS) for 1 h at room temperature. Peptides were first dissolved in dimethyl sulphoxide (DMSO) to a concentration of 15 \g/m\ before being diluted 1 :1000 in 0.1 % PBST and subsequently coated onto the plates (100 μΙ/well). Heat-inactivated pooled sera were diluted 1 :500 in 0.1 % sodium caseinate in 0.1 % PBST and 100 μΙ were added into each well and incubated for 1 h at 37 °C. Horseradish peroxidase (HRP)-conjugated goat anti-mouse IgG Abs (Santa Cruz) diluted 1 :10000 in 0.1 % sodium caseinate in 0.1 % PBST were used to detect the bound Abs. Reactions were developed with 3,3',5,5'-Tetramethylbenzidine substrate (Sigma-Aldrich) and terminated by Stop reagent (Sigma-Aldrich). Absorbance at 450 nm was measured using TECAN Infinite® M200 microplate reader and analyzed using Magellan™ software. Peptides were considered positive when the absorbance value is greater than mean + 6 SD that of the non-vaccinated mice. Positive peptides were plotted as signal relative to a positive peptide E2EP3 obtained with WT-CHIKV infected mice sera. Structural data of the E2 glycoprotein were retrieved from the Protein Data Bank (PDB) (ID: 3N42) and visualized using the UCSF CHIMERA software.
Challenge. Seven weeks after the last immunization mice were challenged with a total of 106 PFU of WT CHIKV in 2x20 μΙ PBS subcutaneously in the dorsal side of the feet on both hind legs. Viremia and foot swelling post challenge was determined by plaque assay and by measuring the height and breadth of feet, respectively.
Prime-boost immunizations with different CHIKV vaccine candidates induce superior antibody responses. Homologous and heterologous prime-boost immunizations using different CHIKV vaccines representing different CHIKV vaccine candidates (Table 1 ) were studied in C57BL/6 mice. A single immunization with 105 PFU of WT CHIKV was included for comparison. When combining different vaccine candidates they were either co-administered (at separate sites) in a single immunization (XY) or used in a prime-boost regimen (X,Y). The results showed that all vaccine candidates except p62-E1 protein generated high CHIKV-specific IgG titers (105-106) after a single immunization, and titers were further enhanced upon a booster immunization (titers of 106-107) (Fig.1 1 A). Simultaneous immunization with the p62-E1 protein antigen and either DREP-Env, A5nsP3 and MVA-CHIKV seemed to slightly augment IgG titers. However, using p62-E1 protein as a boost post prime immunization with the other CHIKV vaccine candidates induced even higher titers. Finally, most prime-boost immunized mice induced stronger antibody responses than mice inoculated once with WT CHIKV.
Vaccine Description Dose and route
D DREP-Env DNA replicwi (DIEP) encoding 10 u£ i.d.+EP
nsPl-nsP4 and E1-E3 (Ertv)
V A5nsP3 CHIKV attenuated by a large 10s PFU s.c.
deletion in nsP3
Figure imgf000030_0001
M MVA-CHIKV Recombinant MVA encoding 107 PFU «,p.
caps id and E1-E3
Table 1 Similarly to DREP-Env, the majority of CHIKV-specific antibody responses induced by the different prime-boost immunizations were of lgG2c rather than lgG1 isotype (Fig. 1 1 B) with the exception of mice immunized with p62-E1 protein, which had a balanced lgG1/lgG2c response, or even a slightly pronounced lgG1 response post homologous prime-boost immunizations. Furthermore, when DREP-Env was co-immunized with p62-E1 protein a balanced Th1/Th2 response was obtained. In contrast, when DREP- Env was first used as prime and then later boosted with p62-E1 protein a Th1 -type response was induced. Similar results were obtained when using AnsP3 plus p62-E1 vs AnsP3 prime followed by AnsP3 and p62-E1 boost.
Most vaccine candidates induced good neutralizing NT50 antibody titers in the range of 103-105. The DREP-Env induced NT50 titers in the range of 102-103 with some animals having no NT50 antibodies. These animals correspond to those that had binding (ELISA) lgG1 antibodies below or close to 104 (Fig. 1 1 A,B). The lack of NT50 antibodies in some of the DREP-Env samples does not necessary mean that the animals were devoid of them but could be explained by the fact that the sensitivity of the assay is 1 :100 (first dilution). Another exception was p62-E1 protein alone given once that did not induce any detectable NT50 antibodies. Prime boost immunization generally augmented NT50 titers with titers being the highest when priming was performed with DREP-Env followed by boosting with MVA (around 105). In contrast, boosting A5nsP3, DREP-Env or MVA-CHIK with p62-E1 protein did not significantly increase NT50 titers. CHIKV vaccine candidates protect against CHIKV infection. The efficacy of the different CHIKV vaccine candidates to protect from challenge with a high dose (106 PFU) WT CHIKV was studied in a challenge model using the established immunization schedules. The results showed that all the immunized animals were protected from CHIKV and, with the exception of nal've mice, only one out of five mice immunized with DREP-Env once and 80% mice immunized once with p62-E1 protein that induced detectable viremia post challenge, all other animals were completely protected (Fig. 12A). Similar to viremia, all the immunized animals developed low foot swelling, except mice immunized once with CHIKV p62-E1 protein that had pronounced foot swelling (Fig. 12B). Interestingly, there were clear correlations between both anti-CHIKV IgG / NT50 titers and vi rem ia / foot swelling (P < 0.001 ) (Fig. 12C and D), and an IgG titer of >104 seemed to be protective.
EXAMPLE 3 - Kinetic and phenotypic analysis of CD8+ T cell responses after priming with alphavirus replicons and homologous or heterologous booster immunizations
In the present study, we demonstrate the induction of different subsets of CD8+ T cells by prime-boost immunization of various replication-proficient attenuated alphavirus (CHIKV) and booster immunizations with poxvirus vectors and / or protein antigen vaccine candidates. This may have implications for the design of efficient vaccination regimens in the clinic.
Materials and Methods
MVA-CHIKV, DREP, and p62-E1 vaccines used were as described in example 2. Immunisations and analysis were also as described.
We wanted to study whether immunization with a WT alphavirus that was capable of productive infection would result in the induction of memory T cells. We used a replication-competent CHIKV. Infection of adult immunocompetent mice by CHIKV has pathogenic consequences, but is not associated with mortality.
For this purpose, we infected C57BL/6 mice with CHIKV and characterized the phenotype of the CD8+ T cell response 8 days after the inoculation using an MHC class I pentamer loaded with a CHIKV E1 Env-derived peptide to identify antigen- specific CD8+ T cells. The phenotype of the CHIKV-specific CD8+ T cell response after wild-type CHIKV infection (Fig. 13) showed that most of the CD8+ T cells displayed a Te or Tern phenotype, and 12% were of the Tern phenotype (Fig. 13, top pie chart). Furthermore, half of the CD8+ T cells were CD27+CD43+, whereas CD27+CD43- T cells constituted one quarter of the CD8+ T cell response (Fig. 13, bottom pie chart).
Alphavirus replicon priming followed by heterologous booster immunizations induce different CD8+ T cell subpopulations.
Given the fact that there was very little difference between VREP and WT CHIKV infection, we wanted to analyze whether we would obtain different subpopulations of CD8+ T cells when boosting with different vaccine candidates following alphavirus immunization. Different vectors encoding a common immunogen can be combined in heterologous prime-boost regimens in order to increase immunogenicity and avoid buildup of antivector immunity. For this purpose, we studied the response induced by different booster vaccinations given after a DREP-Env prime. The DREP-Env vaccine candidate contains all the genomic sequence of CHIKV, but the gene encoding for the capsid protein. Hence, transfection with DREP-Env does not result in production of new virions. C57BL/6 mice were primed with DREP-Env and then boosted either homologously with DREP-Env, or heterologously with either CHIKV p62-E1 protein, MVA-CHIKV or both protein and MVA-CHIKV at the same time. The magnitude of the CHIKV-specific CD8+ T cell responses are greatly increased with heterologous prime-boost immunization. Compared to a homologous DREP-Env boost, the acute response is increased 9-fold with a CHIKV p62-E1 protein boost and 16-fold with an MVA-CHIKV boost. Boosting with both CHIKV p62-E1 protein and MVA-CHIKV results in an 18-fold increase. The phenotypes of CHIKV-specific CD8+ T cell responses were characterized 8 days after boost as described above. The results demonstrated that administering DREP-Env twice induced a CHIKV- specific CD8+ T cell response with a smaller proportion of the Te phenotype and instead a higher proportion of the Tern phenotype (Fig. 13, top pie chart). As the infection rate with viral particles is higher than the transfection rate with DNA (our observations in vitro), these results are in line with current knowledge that weaker signal strength induces a higher degree of Tern. Furthermore, the proportion of CD27+CD43+ CHIKV-specific CD8+ T cells was smaller whereas a higher proportion of CD27+CD43- cells was induced (Fig. 13, bottom pie chart).
Administering a heterologous boost revealed that the phenotype of the CHIKV-specific CD8+ T cell response was close to identical in groups boosted with MVA-CHIKV, either alone or together with CHIKV p62-E1 protein, with the CD8+ T cells mostly of the Tern phenotype, and only a small proportion of the Tern phenotype (Fig. 13, top pie charts). On the other hand, mice given a CHIKV p62-E1 protein boost, without MVA-CHIKV, developed a larger proportion of CD8+ T cells of the Tern phenotype and a smaller proportion of the Tern phenotype (Fig. 13, top pie charts), again in line with the hypothesis that a stronger signal favors Tern development . The proportion of Tern after a CHIKV p62-E1 protein boost was similar to that after a DREP-Env boost, about one third. Moreover, mice given a heterologous boost had similar proportions of CD8+ Te cells in the range of 22-29%, whereas mice given a homologous DREP-Env boost triggered only 7% of CD8+ Te cells (Fig. 13, top pie charts).
Furthermore, analysis of the CD27/CD43 staining revealed the induction of four distinct CHIKV-specific CD8+ T cell subpopulations that were of similar size after MVA-CHIKV or MVA-CHIKV + CHIKV p62-E1 protein boost (Fig. 13, bottom pie charts). However, boosting with CHIKV p62-E1 protein only also induced four CD8+ T cell subpopulations, although with a slightly larger proportion of the CD27+CD43- subset, similar to what was observed after a homologous DREP-Env boost (Fig. 13, bottom pie charts).
References
Note: See also references cited in J. Virol. 2014, 88(6):3527, Garcia-Arriaza et al, "A Novel Poxvirus Based Vaccine, MVA-CHIKV, Is Highly Immunogenic and Protects Mice against Chikungunya Infection", published ahead of print 8 January 2014.
Her Z, Kam YW, Lin RT, Ng LF. 2009. Chikungunya: a bending reality. Microbes and infection / Institut Pasteur 1 1 :1 165-1 176.
Burt FJ, Rolph MS, Rulli NE, Mahalingam S, Heise MT. 2012. Chikungunya: a re-emerging virus. Lancet 379:662-671.
Van Bortel W, Dorleans F, Rosine J, Blateau A, Rousset D, Matheus S, Leparc-Goffart I, Flusin O, Prat C, Cesaire R, Najioullah F, Ardillon V, Balleydier E, Carvalho L, Lemaitre A, Noel H, Servas V, Six C, Zurbaran M, Leon L, Guinard A, van den Kerkhof J, Henry M, Fanoy E, Braks M, Reimerink J, Swaan C, Georges R, Brooks L, Freedman J, Sudre B, Zeller H. 2014. Chikungunya outbreak in the Caribbean region, December 2013 to March 2014, and the significance for Europe. Euro Surveill 19.
Schwartz O, Albert ML. 2010. Biology and pathogenesis of chikungunya virus. Nature reviews. Microbiology 8:491 -500.
Vijayakumar KP, Nair Anish TS, George B, Lawrence T, Muthukkutty SC, Ramachandran R. 201 1 . Clinical Profile of Chikungunya Patients during the Epidemic of 2007 in Kerala, India. J Glob Infect Dis 3:221 -226. Couturier E, Guillemin F, Mura M, Leon L, Virion JM, Letort MJ, De Valk H, Simon F, Vaillant V. 2012.
Impaired quality of life after chikungunya virus infection: a 2-year follow-up study. Rheumatology (Oxford) 51 :1315-1322.
Essackjee K, Goorah S, Ramchurn SK, Cheeneebash J, Walker-Bone K. 2013. Prevalence of and risk factors for chronic arthralgia and rheumatoid-like polyarthritis more than 2 years after infection with chikungunya virus. Postgrad Med J 89:440-447.
Mavalankar D, Shastri P, Bandyopadhyay T, Parmar J, Ramani KV. 2008. Increased mortality rate associated with chikungunya epidemic, Ahmedabad, India. Emerg Infect Dis 14:412-415.
Weaver SC, Osorio JE, Livengood JA, Chen R, Stinchcomb DT. 2012. Chikungunya virus and prospects for a vaccine. Expert Rev Vaccines 1 1 :1087-1 101. Chu H, Das SC, Fuchs JF, Suresh M, Weaver SC, Stinchcomb DT, Partidos CD, Osorio JE. 2013.
Deciphering the protective role of adaptive immunity to CHIKV/IRES a novel candidate vaccine against Chikungunya in the A129 mouse model. Vaccine 31 :3353-3360. Edelman R, Tacket CO, Wasserman SS, Bodison SA, Perry JG, Mangiafico JA. 2000. Phase II safety and immunogenicity study of live chikungunya virus vaccine TSI-GSD-218. Am J Trop Med Hyg 62:681 -685.
Hallengard D, Kakoulidou M, Lulla A, Kummerer BM, Johansson DX, Mutso M, Lulla V, Fazakerley JK, Roques P, Le Grand R, Merits A, Liljestrom P. 2013. Novel attenuated Chikungunya vaccine candidates elicit protective immunity in C57BL/6 mice. Journal of virology 88:2858-2866.
Levitt NH, Ramsburg HH, Hasty SE, Repik PM, Cole FE, Jr., Lupton HW. 1986. Development of an attenuated strain of chikungunya virus for use in vaccine production. Vaccine 4:157-162.
Partidos CD, Paykel J, Weger J, Borland EM, Powers AM, Seymour R, Weaver SC, Stinchcomb DT, Osorio JE. 2012. Cross-protective immunity against o'nyong-nyong virus afforded by a novel recombinant chikungunya vaccine. Vaccine 30:4638-4643.
Piper A, Ribeiro M, Smith KM, Briggs CM, Huitt E, Nanda K, Spears CJ, Quiles M, Cullen J, Thomas ME, Brown DT, Hernandez R. 2013. Chikungunya Virus Host Range E2 Transmembrane Deletion Mutants Induce Protective Immunity against Challenge in C57BL/6J Mice. Journal of virology 87:6748-6757.
Plante K, Wang E, Partidos CD, Weger J, Gorchakov R, Tsetsarkin K, Borland EM, Powers AM, Seymour R, Stinchcomb DT, Osorio JE, Frolov I, Weaver SC. 201 1. Novel chikungunya vaccine candidate with an
IRES-based attenuation and host range alteration mechanism. PLoS Pathog 7:e1002142.
Harrison VR, Eckels KH, Bartelloni PJ, Hampton C. 1971. Production and evaluation of a formalin-killed Chikungunya vaccine. J Immunol 107:643-647.
Tiwari M, Parida M, Santhosh SR, Khan M, Dash PK, Rao PV. 2009. Assessment of immunogenic potential of Vero adapted formalin inactivated vaccine derived from novel ECSA genotype of Chikungunya virus. Vaccine 27:2513-2522.
White A, Berman S, Lowenthal JP. 1972. Comparative immunogenicities of Chikungunya vaccines propagated in monkey kidney monolayers and chick embryo suspension cultures. Appl Microbiol 23:951 - 952. Chattopadhyay A, Wang E, Seymour R, Weaver SC, Rose JK. 2013. A chimeric vesiculo/alphavirus is an effective alphavirus vaccine. Journal of virology 87:395-402.
Wang D, Suhrbier A, Penn-Nicholson A, Woraratanadharm J, Gardner J, Luo M, Le TT, Anraku I, Sakalian M, Einfeld D, Dong JY. 201 1. A complex adenovirus vaccine against chikungunya virus provides complete protection against viraemia and arthritis. Vaccine 29:2803-2809.
Wang E, Volkova E, Adams AP, Forrester N, Xiao SY, Frolov I, Weaver SC. 2008. Chimeric alphavirus vaccine candidates for chikungunya. Vaccine 26:5030-5039.
Akahata W, Yang ZY, Andersen H, Sun S, Holdaway HA, Kong WP, Lewis MG, Higgs S, Rossmann MG, Rao S, Nabel GJ. 2010. A virus-like particle vaccine for epidemic Chikungunya virus protects nonhuman primates against infection. Nat Med 16:334-338. Kumar M, Sudeep AB, Arankalle VA. 2012. Evaluation of recombinant E2 protein-based and whole-virus inactivated candidate vaccines against chikungunya virus. Vaccine 30:6142-6149.
Metz SW, Gardner J, Geertsema C, Le TT, Goh L, Vlak JM, Suhrbier A, Pijlman GP. 2013. Effective chikungunya virus-like particle vaccine produced in insect cells. PLoS Negl Trop Dis 7:e2124. Metz SW, Geertsema C, Martina BE, Andrade P, Heldens JG, van Oers MM, Goldbach RW, Vlak JM,
Pijlman GP. 201 1 . Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells. Virol J 8:353.
Metz SW, Martina BE, van den Doel P, Geertsema C, Osterhaus AD, Vlak JM, Pijlman GP. 2013. Chikungunya virus-like particles are more immunogenic in a lethal AG129 mouse model compared to glycoprotein E1 or E2 subunits. Vaccine 31 :6092-6096.
Brandler S, Ruffie C, Combredet C, Brault JB, Najburg V, Prevost MC, Habel A, Tauber E, Despres P, Tangy F. 2013. A recombinant measles vaccine expressing chikungunya virus-like particles is strongly immunogenic and protects mice from lethal challenge with chikungunya virus. Vaccine 31 :3718-3725.
Garcia-Arriaza J, Cepeda V, Hallengard D, Sorzano CO, Kummerer BM, Liljestrom P, Esteban M. 2014. A Novel Poxvirus-Based Vaccine, MVA-CHIKV, Is Highly Immunogenic and Protects Mice against
Chikungunya Infection. Journal of virology 88:3527-3547.
Mallilankaraman K, Shedlock DJ, Bao H, Kawalekar OU, Fagone P, Ramanathan AA, Ferraro B, Stabenow J, Vijayachari P, Sundaram SG, Muruganandam N, Sarangan G, Srikanth P, Khan AS, Lewis MG, Kim JJ, Sardesai NY, Muthumani K, Weiner DB. 201 1 . A DNA vaccine against chikungunya virus is protective in mice and induces neutralizing antibodies in mice and nonhuman primates. PLoS Negl Trop
Dis 5:e928.
Muthumani K, Lankaraman KM, Laddy DJ, Sundaram SG, Chung CW, Sako E, Wu L, Khan A, Sardesai N, Kim JJ, Vijayachari P, Weiner DB. 2008. Immunogenicity of novel consensus-based DNA vaccines against Chikungunya virus. Vaccine 26:5128-5134. Barry G, Fragkoudis R, Ferguson MC, Lulla A, Merits A, Kohl A, Fazakerley JK. 2010. Semliki forest virus- induced endoplasmic reticulum stress accelerates apoptotic death of mammalian cells. Journal of virology 84:7369-7377.
Glasgow GM, McGee MM, Sheahan BJ, Atkins GJ. 1997. Death mechanisms in cultured cells infected by Semliki Forest virus. The Journal of general virology 78 ( Pt 7):1559-1563. Hidmark AS, Mclnerney GM, Nordstrom EK, Douagi I, Werner KM, Liljestrom P, Karlsson Hedestam GB.
2005. Early alpha/beta interferon production by myeloid dendritic cells in response to UV-inactivated virus requires viral entry and interferon regulatory factor 3 but not MyD88. Journal of virology 79:10376-10385.
Berglund P, Smerdou C, Fleeton MN, Tubulekas I, Liljestrom P. 1998. Enhancing immune responses using suicidal DNA vaccines. Nat Biotechnol 16:562-565. Nordstrom EK, Forsell MN, Barnfield C, Bonin E, Hanke T, Sundstrom M, Karlsson GB, Liljestrom P. 2005. Enhanced immunogenicity using an alphavirus replicon DNA vaccine against human immunodeficiency virus type 1. The Journal of general virology 86:349-354.
Knudsen ML, Mbewe-Mvula A, Rosario M, Johansson DX, Kakoulidou M, Bridgeman A, Reyes-Sandoval A, Nicosia A, Ljungberg K, Hanke T, Liljestrom P. 2012. Superior induction of T cell responses to conserved HIV-1 regions by electroporated alphavirus replicon DNA compared to that with conventional plasmid DNA vaccine. Journal of virology 86:4082-4090.
Dubensky TW, Jr., Driver DA, Polo JM, Belli BA, Latham EM, Ibanez CE, Chada S, Brumm D, Banks TA, Mento SJ, Jolly DJ, Chang SM. 1996. Sindbis virus DNA-based expression vectors: utility for in vitro and in vivo gene transfer. Journal of virology 70:508-519.
Hariharan MJ, Driver DA, Townsend K, Brumm D, Polo JM, Belli BA, Catton DJ, Hsu D, Mittelstaedt D, McCormack JE, Karavodin L, Dubensky TW, Jr., Chang SM, Banks TA. 1998. DNA immunization against herpes simplex virus: enhanced efficacy using a Sindbis virus-based vector. Journal of virology 72:950- 958. Ljungberg K, Whitmore AC, Fluet ME, Moran TP, Shabman RS, Collier ML, Kraus AA, Thompson JM,
Montefiori DC, Beard C, Johnston RE. 2007. Increased immunogenicity of a DNA-launched Venezuelan equine encephalitis virus-based replicon DNA vaccine. Journal of virology 81 : 13412- 13423.
Lu S. 2009. Heterologous prime-boost vaccination. Curr Opin Immunol 21 :346-351.
Voss JE, Vaney MC, Duquerroy S, Vonrhein C, Girard-Blanc C, Crublet E, Thompson A, Bricogne G, Rey FA. 2010. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography.
Nature 468:709-712.
Pohjala L, Utt A, Varjak M, Lulla A, Merits A, Ahola T, Tammela P. 201 1. Inhibitors of alphavirus entry and replication identified with a stable Chikungunya replicon cell line and virus-based assays. PLoS One 6:e28923. Roos AK, Eriksson F, Timmons JA, Gerhardt J, Nyman U, Gudmundsdotter L, Brave A, Wahren B, Pisa P.
2009. Skin electroporation: effects on transgene expression, DNA persistence and local tissue environment. PLoS One 4:e7226.
Glasker S, Lulla A, Lulla V, Couderc T, Drexler JF, Liljestrom P, Lecuit M, Drosten C, Merits A, KCimmerer BM. 2013. Virus replicon particle based Chikungunya virus neutralization assay using Gaussia luciferase as readout. Virol J 10:235.
Kam YW, Lee WW, Simarmata D, Harjanto S, Teng TS, Tolou H, Chow A, Lin RT, Leo YS, Renia L, Ng LF. 2012. Longitudinal analysis of the human antibody response to Chikungunya virus infection: implications for serodiagnosis and vaccine development. Journal of virology 86:13005-13015. Kam YW, Lee WW, Simarmata D, Le Grand R, Tolou H, Merits A, Roques P, Ng LF. 2014. Unique epitopes recognized by antibodies induced in chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLoS One 9:e95647.
Kam YW, Lum FM, Teo TH, Lee WW, Simarmata D, Harjanto S, Chua CL, Chan YF, Wee JK, Chow A, Lin RT, Leo YS, Le Grand R, Sam IC, Tong JC, Roques P, Wiesmuller KH, Renia L, Rotzschke O, Ng LF.
2012. Early neutralizing IgG response to Chikungunya virus in infected patients targets a dominant linear epitope on the E2 glycoprotein. EMBO Mol Med 4:330-343.
Lum FM, Teo TH, Lee WW, Kam YW, Renia L, Ng LF. 2013. An essential role of antibodies in the control of chikungunya virus infection. J Immunol 190:6295-6302. Kam YW, Simarmata D, Chow A, Her Z, Teng TS, Ong EK, Renia L, Leo YS, Ng LF. 2012. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with chikungunya virus clearance and long-term clinical protection. The Journal of infectious diseases 205:1 147-1 154.
Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE. 2004. UCSF Chimera-a visualization system for exploratory research and analysis. J Comput Chem 25:1605-1612. Magnusson SE, Reimer JM, Karlsson KH, Lilja L, Bengtsson KL, Stertman L. 2013. Immune enhancing properties of the novel Matrix-M adjuvant leads to potentiated immune responses to an influenza vaccine in mice. Vaccine 31 :1725-1733.
Gilca V, De Serres G, Hamelin ME, Boivin G, Ouakki M, Boulianne N, Sauvageau C, Dionne M, Gilca R, Skowronski D. 201 1. Antibody persistence and response to 2010-201 1 trivalent influenza vaccine one year after a single dose of 2009 AS03-adjuvanted pandemic H1 N1 vaccine in children. Vaccine 30:35-41.
Akahata W, Nabel GJ. 2012. A specific domain of the Chikungunya virus E2 protein regulates particle formation in human cells: implications for alphavirus vaccine design. Journal of virology 86:8879-8883.
Gardner CL, Hritz J, Sun C, Vanlandingham DL, Song TY, Ghedin E, Higgs S, Klimstra WB, Ryman KD. 2014. Deliberate attenuation of chikungunya virus by adaptation to heparan sulfate-dependent infectivity: a model for rational arboviral vaccine design. PLoS Negl Trop Dis 8:e2719.
McKay PF, Cope AV, Mann JF, Joseph S, Esteban M, Tatoud R, Carter D, Reed SG, Weber J, Shattock RJ. 2014. Glucopyranosyl Lipid A Adjuvant Significantly Enhances HIV Specific T and B Cell Responses Elicited by a DNA-MVA-Protein Vaccine Regimen. PLoS One 9:e84707.
Gardner J, Anraku I, Le TT, Larcher T, Major L, Roques P, Schroder WA, Higgs S, Suhrbier A. 2010. Chikungunya virus arthritis in adult wild-type mice. Journal of virology 84:8021 -8032.
Coffey LL, Vignuzzi M. 201 1 . Host alternation of chikungunya virus increases fitness while restricting population diversity and adaptability to novel selective pressures. Journal of virology 85:1025-1035.
Hughes HR, Crill WD, Chang GJ. 2012. Manipulation of immunodominant dengue virus E protein epitopes reduces potential antibody-dependent enhancement. Virol J 9:1 15. Swain SL, McKinstry KK, Strutt TM. 2012. Expanding roles for CD4(+) T cells in immunity to viruses. Nat Rev Immunol 12:136-148.
Plotkin SA. 2013. Complex correlates of protection after vaccination. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 56:1458-1465. Holzer GW, Coulibaly S, Aichinger G, Savidis-Dacho H, Mayrhofer J, Brunner S, Schmid K, Kistner O,
Aaskov JG, Falkner FG, Ehrlich H, Barrett PN, Kreil TR. 201 1. Evaluation of an inactivated Ross River virus vaccine in active and passive mouse immunization models and establishment of a correlate of protection. Vaccine 29:4132-4141.
Suhrbier A, Jaffar-Bandjee MC, Gasque P. 2012. Arthritogenic alphaviruses-an overview. Nature reviews. Rheumatology 8:420-429.
Kreil TR, Eibl MM. 1997. Pre- and postexposure protection by passive immunoglobulin but no enhancement of infection with a flavivirus in a mouse model. Journal of virology 71 :2921 -2927.
Lidbury BA, Mahalingam S. 2000. Specific ablation of antiviral gene expression in macrophages by antibody-dependent enhancement of Ross River virus infection. Journal of virology 74:8376-8381. Lobigs M, Larena M, Alsharifi M, Lee E, Pavy M. 2009. Live chimeric and inactivated Japanese encephalitis virus vaccines differ in their cross-protective values against Murray Valley encephalitis virus. Journal of virology 83:2436-2445.
Wallace MJ, Smith DW, Broom AK, Mackenzie JS, Hall RA, Shellam GR, McMinn PC. 2003. Antibody- dependent enhancement of Murray Valley encephalitis virus virulence in mice. The Journal of general virology 84:1723-1728.
Plotkin SA, Gilbert PB. 2012. Nomenclature for immune correlates of protection after vaccination. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 54:1615-1617.
Burchill M a, Tamburini B a, Pennock ND, White JT, Kurche JS, Kedl RM. 2013. T cell vaccinology: exploring the known unknowns. Vaccine 31 :297-305. Hansen SG, Piatak M, Ventura AB, Hughes CM, Gilbride RM, Ford JC, Oswald K, Shoemaker R, Li Y,
Lewis MS, Gilliam AN, Xu G, Whizin N, Burwitz BJ, Planer SL, Turner JM, Legasse AW, Axthelm MK, Nelson J a, Friih K, Sacha JB, Estes JD, Keele BF, Edlefsen PT, Lifson JD, Picker LJ. 2013. Immune clearance of highly pathogenic SIV infection. Nature 502:100—4.
Bassett JD, Swift SL, Bramson JL. 201 1. Optimizing vaccine-induced CD8(+) T-cell immunity: focus on recombinant adenovirus vectors. Expert Rev. Vaccines 10:1307-19.
Gomez CE, Perdiguero B, Garcia-Arriaza J, Esteban M. 2012. Poxvirus vectors as HIV/AIDS vaccines in humans. Hum. Vaccin. Immunother. 8:1 192-207. Gomez CE, Najera JL, Perdiguero B, Garcia-Arriaza J, Sorzano COS, Jimenez V, Gonzalez-Sanz R, Jimenez JL, Munoz-Fernandez MA, Lopez Bernaldo de Quiros JC, Guardo AC, Garcia F, Gatell JM, Plana M, Esteban M. 201 1. The HIV/AIDS vaccine candidate MVA-B administered as a single immunogen in humans triggers robust, polyfunctional, and selective effector memory T cell responses to HIV-1 antigens. J. Virol. 85:1 1468-78.
Kaech SM, Cui W. 2012. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12:749-61 .
Sallusto F, Lanzavecchia A, Araki K, Ahmed R. 2010. From vaccines to memory and back. Immunity 33:451 -63. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. 1999. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401 :708-12.
Masopust D, Vezys V, Marzo a L, Lefrancois L. 2001. Preferential localization of effector memory cells in nonlymphoid tissue. Science 291 :2413-7.
Bachmann MF, Wolint P, Schwarz K, Jager P, Oxenius A. 2005. Functional properties and lineage relationship of CD8+ T cell subsets identified by expression of IL-7 receptor alpha and CD62L. J. Immunol.
175:4686-96.
Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, Whizin N, Oswald K, Shoemaker R, Swanson T, Legasse AW, Chiuchiolo MJ, Parks CL, Axthelm MK, Nelson J a, Jarvis M a, Piatak M, Lifson JD, Picker LJ. 201 1. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473:523-7.
Bachmann MF, Wolint P, Schwarz K, Oxenius A. 2005. Recall proliferation potential of memory CD8+ T cells and antiviral protection. J. Immunol. 175:4677-4685.
Nolz JC, Harty JT. 201 1. Protective capacity of memory CD8+ T cells is dictated by antigen exposure history and nature of the infection. Immunity 34:781 -93. Wherry EJ, Teichgraber V, Becker TC, Masopust D, Kaech SM, Antia R, von Andrian UH, Ahmed R. 2003.
Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat. Immunol. 4:225-34.
Hikono H, Kohlmeier JE, Takamura S, Wittmer ST, Roberts AD, Woodland DL. 2007. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8+ T cells. J. Exp. Med. 204:1625-36. Grujic M, Bartholdy C, Remy M, Pinschewer DD, Christensen JP, Thomsen AR. 2010. The role of
CD80/CD86 in generation and maintenance of functional virus-specific CD8+ T cells in mice infected with lymphocytic choriomeningitis virus. J. Immunol. 185:1730—43. Mikkelsen M, Hoist PJ, Bukh J, Thomsen AR, Christensen JP. 201 1. Enhanced and sustained CD8+ T cell responses with an adenoviral vector-based hepatitis C virus vaccine encoding NS3 linked to the MHC class II chaperone protein invariant chain. J. Immunol. 186:2355-64.
Naslund Tl, Uyttenhove C, Nordstrom EKL, Colau D, Warnier G, Jondal M, Van den Eynde BJ, Liljestrom P. 2007. Comparative prime-boost vaccinations using Semliki Forest virus, adenovirus, and ALVAC vectors demonstrate differences in the generation of a protective central memory CTL response against the P815 tumor. J. Immunol. 178:6761 -6769.
Ahola T, Courderc T, Ng LFP, Hallengard D, Powers A, Lecuit M, Esteban M, Merits A, Liljestrom P. 2014. Therapeutics and vaccines against chikungunya virus. Vector-Borne Zoonotic Dis. In press. Smerdou C, Liljestrom P. 1999. Two-helper RNA system for production of recombinant Semliki forest virus particles. J. Virol. 73:1092-1098.
Karlsson GB, Liljestrom P. 2003. Live viral vectors: Semliki Forest virus. Methods Mol Med 87:69-82.
Sjoberg EM, Suomalainen M, Garoff H. 1994. A significantly improved Semliki Forest virus expression system based on translation enhancer segments from the viral capsid gene. Biotechnology. (N. Y). 12:1 127-1 131 .
Ryan MD, Drew J. 1994. Foot-and-mouth disease virus 2A oligopeptide mediated cleavage of an artificial polyprotein. EMBO J. 13:928-33.
Donnelly ML, Luke G, Mehrotra A, Li X, Hughes LE, Gani D, Ryan MD. 2001. Analysis of the aphthovirus 2A/2B polyprotein "cleavage" mechanism indicates not a proteolytic reaction, but a novel translational effect: a putative ribosomal "skip". J Gen Virol, 2001/04/12 ed. 82:1013-1025.
Knudsen ML, Ljungberg K, Liljestrom P, Johansson DX. 2014. Intradermal electroporation of RNA. Methods Mol. Biol. 1 121 :147-154.
Badovinac VP, Messingham K a N, Hamilton SE, Harty JT. 2003. Regulation of CD8+ T cells undergoing primary and secondary responses to infection in the same host. J. Immunol. 170:4933—42. Masopust D, Jiang J, Shen H, Lefrancois L. 2001 . Direct analysis of the dynamics of the intestinal mucosa
CD8 T cell response to systemic virus infection. J. Immunol. 166:2348-56.
Masopust D, Ha S-J, Vezys V, Ahmed R. 2006. Stimulation history dictates memory CD8 T cell phenotype: implications for prime-boost vaccination. J. Immunol. 177:831-9.
Grayson JM, Harrington LE, Lanier JG, Wherry EJ, Ahmed R. 2002. Differential sensitivity of naive and memory CD8+ T cells to apoptosis in vivo. J. Immunol. 169:3760-70.
Jabbari A, Harty JT. 2006. Secondary memory CD8+ T cells are more protective but slower to acquire a central-memory phenotype. J. Exp. Med. 203:919-32. Sellins K, Fradkin L, Liggitt D, Dow S. 2005. Type I interferons potently suppress gene expression following gene delivery using liposome(-)DNA complexes. Mol. Ther. 12:451 -9.
Naslund Tl, Kostic L, Nordstrom EK, Chen M, Liljestrom P. 201 1. Role of innate signalling pathways in the immunogenicity of alphaviral replicon-based vaccines. Virol. J., 201 1/01 /26 ed. 8:36. Betts MR, Nason MC, West SM, De Rosa SC, Migueles S a, Abraham J, Lederman MM, Benito JM,
Goepfert P a, Connors M, Roederer M, Koup R a. 2006. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood 107:4781 -9.
Harari A, Dutoit V, Cellerai C, Bart PA, Du Pasquier RA, Pantaleo G. 2006. Functional signatures of protective antiviral T-cell immunity in human virus infections. Immunol Rev, 2006/07/1 1 ed. 21 1 :236-254. Tarbatt CJ, Glasgow GM, Mooney D a, Sheahan BJ, Atkins GJ. 1997. Sequence analysis of the avirulent, demyelinating A7 strain of Semliki Forest virus. J. Gen. Virol. 78 ( Pt 7):1551 -7.
Liu MA. 2010. Immunologic basis of vaccine vectors. Immunity, 2010/10/30 ed. 33:504-515.
Schneider J, Gilbert SC, Blanchard TJ, Hanke T, Robson KJ, Hannan CM, Becker M, Sinden R, Smith GL, Hill A V. 1998. Enhanced immunogenicity for CD8+ T cell induction and complete protective efficacy of malaria DNA vaccination by boosting with modified vaccinia virus Ankara. Nat. Med. 4:397-402.
Fraser K a, Schenkel JM, Jameson SC, Vezys V, Masopust D. 2013. Preexisting high frequencies of memory CD8+ T cells favor rapid memory differentiation and preservation of proliferative potential upon boosting. Immunity 39:171 -83.
Miller JD, van der Most RG, Akondy RS, Glidewell JT, Albott S, Masopust D, Murali-Krishna K, Mahar PL, Edupuganti S, Lalor S, Germon S, Del Rio C, Mulligan MJ, Staprans SI, Altman JD, Feinberg MB, Ahmed
R. 2008. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity 28:710-22.
Blom K, Braun M, Ivarsson M a, Gonzalez VD, Falconer K, Moll M, Ljunggren H-G, Michaelsson J, Sandberg JK. 2013. Temporal dynamics of the primary human T cell response to yellow fever virus 17D as it matures from an effector- to a memory-type response. J. Immunol. 190:2150-8.
Tan WG, Jin H-T, West EE, Penaloza-MacMaster P, Wieland A, Zilliox MJ, McElrath MJ, Barouch DH, Ahmed R. 2013. Comparative analysis of simian immunodeficiency virus gag-specific effector and memory CD8+ T cells induced by different adenovirus vectors. J. Virol. 87:1359-72.
Quinn KM, Da Costa A, Yamamoto A, Berry D, Lindsay RWB, Darrah P a, Wang L, Cheng C, Kong W-P, Gall JGD, Nicosia A, Folgori A, Colloca S, Cortese R, Gostick E, Price D a, Gomez CE, Esteban M, Wyatt
LS, Moss B, Morgan C, Roederer M, Bailer RT, Nabel GJ, Koup R a, Seder R a. 2013. Comparative analysis of the magnitude, quality, phenotype, and protective capacity of simian immunodeficiency virus gag-specific CD8+ T cells following human-, simian-, and chimpanzee-derived recombinant adenoviral vector immunization. J. Immunol. 190:2720-35. Pillai VKB, Kannanganat S, Penaloza-Macmaster P, Chennareddi L, Robinson HL, Blackwell J, Amara RR. 201 1. Different patterns of expansion, contraction and memory differentiation of HIV-1 Gag-specific CD8 T cells elicited by adenovirus type 5 and modified vaccinia Ankara vaccines. Vaccine 29:5399-406.
Bruna-Romero O, Gonzalez-Aseguinolaza G, Hafalla JC, Tsuji M, Nussenzweig RS. 2001. Complete, long-lasting protection against malaria of mice primed and boosted with two distinct viral vectors expressing the same plasmodial antigen. Proc. Natl. Acad. Sci. U. S. A. 98:1 1491-6.
Ledgerwood JE, Zephir K, Hu Z, Wei C-J, Chang L, Enama ME, Hendel CS, Sitar S, Bailer RT, Koup R a, Mascola JR, Nabel GJ, Graham BS. 2013. Prime-boost interval matters: a randomized phase 1 study to identify the minimum interval necessary to observe the H5 DNA influenza vaccine priming effect. J. Infect. Dis. 208:418-22.
Berglund P, Fleeton MN, Smerdou C, Liljestrom P. 1999. Immunization with recombinant Semliki Forest virus induces protection against influenza challenge in mice. Vaccine 17:497-507.
Uematsu Y, Vajdy M, Lian Y, Perri S, Greer CE, Legg HS, Galli G, Saletti G, Often GR, Rappuoli R, Barnett SW, Polo JM. 2012. Lack of interference with immunogenicity of a chimeric alphavirus replicon particle-based influenza vaccine by preexisting antivector immunity. Clin. Vaccine Immunol. 19:991 -8.
Pushko P, Parker M, Ludwig G V, Davis NL, Johnston RE, Smith JF. 1997. Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology 239:389-401.
White LJ, Sariol C a, Mattocks MD, Wahala M P B W, Yingsiwaphat V, Collier ML, Whitley J, Mikkelsen R, Rodriguez I V, Martinez Ml, de Silva A, Johnston RE. 2013. An alphavirus vector-based tetravalent dengue vaccine induces a rapid and protective immune response in macaques that differs qualitatively from immunity induced by live virus infection. J. Virol. 87:3409-24.
Schulz O, Diebold SS, Chen M, Naslund Tl, Nolte MA, Alexopoulou L, Azuma Y-T, Flavell RA, Liljestrom P, Reis e Sousa C. 2005. Toll-like receptor 3 promotes cross-priming to virus-infected cells. Nature 433:887-892.
Pichlmair A, Reis e Sousa C. 2007. Innate recognition of viruses. Immunity, 2007/09/26 ed. 27:370-383.
Barry G, Breakwell L, Fragkoudis R, Attarzadeh-Yazdi G, Rodriguez-Andres J, Kohl A, Fazakerley JK. 2009. PKR acts early in infection to suppress Semliki Forest virus production and strongly enhances the type I interferon response. J Gen Virol, 2009/03/07 ed. 90:1382-1391. Le Bon A, Durand V, Kamphuis E, Thompson C, Bulfone-Paus S, Rossmann C, Kalinke U, Tough DF.
2006. Direct stimulation of T cells by type I IFN enhances the CD8+ T cell response during cross-priming. J. Immunol. 176:4682-9.
Le Bon A, Etchart N, Rossmann C, Ashton M, Hou S, Gewert D, Borrow P, Tough DF. 2003. Cross- priming of CD8+ T cells stimulated by virus-induced type I interferon. Nat. Immunol. 4:1009-15. Ramos HJ, Davis AM, Cole AG, Schatzle JD, Forman J, Farrar JD. 2009. Reciprocal responsiveness to interleukin-12 and interferon-alpha specifies human CD8+ effector versus central memory T-cell fates. Blood 1 13:5516-25.
Dikopoulos N, Bertoletti A, Kroger A, Hauser H, Schirmbeck R, Reimann J. 2005. Type I IFN negatively regulates CD8+ T cell responses through IL-10-producing CD4+ T regulatory 1 cells. J. Immunol. 174:99-
109.
Sanchez PJ, Kedl RM. 2012. An alternative signal 3: CD8+ T cell memory independent of IL-12 and type I IFN is dependent on CD27/OX40 signaling. Vaccine 30:1 154-61.
Xiao Z, Casey K a, Jameson SC, Curtsinger JM, Mescher MF. 2009. Programming for CD8 T cell memory development requires IL-12 or type I IFN. J. Immunol. 182:2786-94.
Cousens LP, Peterson R, Hsu S, Dorner A, Altman JD, Ahmed R, Biron CA. 1999. Two roads diverged: interferon alpha/beta- and interleukin 12-mediated pathways in promoting T cell interferon gamma responses during viral infection. J. Exp. Med. 189:1315-1328.
Robinson HL, Amara RR. 2005. T cell vaccines for microbial infections. Nat. Med. 1 1 :S25-32. Patel V, Valentin A, Kulkarni V, Rosati M, Bergamaschi C, Jalah R, Alicea C, Minang JT, Trivett MT, Ohlen
C, Zhao J, Robert-Guroff M, Khan AS, Draghia-Akli R, Felber BK, Pavlakis GN. 2010. Long-lasting humoral and cellular immune responses and mucosal dissemination after intramuscular DNA immunization. Vaccine 28:4827-36.
Valentin A, von Gegerfelt A, Rosati M, Miteloudis G, Alicea C, Bergamaschi C, Jalah R, Patel V, Khan AS, Draghia-Akli R, Pavlakis GN, Felber BK. 2010. Repeated DNA therapeutic vaccination of chronically SIV- infected macaques provides additional virological benefit. Vaccine 28:1962-74.
Gomez CE, Perdiguero B, Garcia-Arriaza J, Esteban M. 2013. Clinical applications of attenuated MVA poxvirus strain. Expert Rev. Vaccines 12:1395-416.
Reyes-Sandoval A, Wyllie DH, Bauza K, Milicic A, Forbes EK, Rollier CS, Hill AVS. 201 1. CD8+ T effector memory cells protect against liver-stage malaria. J. Immunol. 187:1347-57.
Masopust D, Picker LJ. 2012. Hidden memories: frontline memory T cells and early pathogen interception. J. Immunol. 188:581 1-7.
Schenkel JM, Fraser K a, Masopust D. 2014. Cutting Edge: Resident Memory CD8 T Cells Occupy Frontline Niches in Secondary Lymphoid Organs. J. Immunol.

Claims

CLAIMS:
1. A composition comprising a nucleic acid comprising a nucleotide sequence encoding an attenuated poxvirus vector and one or more CHIKV structural proteins.
2. The composition of claim 1 , wherein the nucleotide sequence encodes at least the E3 and E2 CHIKV structural proteins.
3. The composition of claim 1 or 2 wherein the nucleotide sequence encodes all of the C, E3, E2, 6K, and E1 CHIKV structural proteins.
4. The composition of any preceding claim wherein the nucleotide sequence further encodes one or more CHIKV replicase proteins.
5. The composition of any preceding claim wherein the poxvirus vector is a modified vaccinia virus Ankara (MVA) vector.
6. The composition of claim 5 wherein the MVA vector is further modified by deletion of one or more vaccinia immunomodulatory genes.
7. The composition of claim 6 wherein the MVA vector is modified by deletion of all of the immunomodulatory genes C6L, K7R, and A46R.
8. An attenuated poxvirus vector expressing one or more CHIKV structural genes.
9. The vector of claim 8, wherein the vector expresses at least the E3 and E2 CHIKV structural genes.
10 The vector of claim 8 or 9 wherein the vector expresses all of the C, E3, E2, 6K, and E1 CHIKV structural genes.
1 1 . The vector of any of claims 8 to 10 wherein the vector expresses one or more CHIKV replicase genes.
12. The vector of any of claims 8 to 1 1 wherein the poxvirus vector is a modified vaccinia virus Ankara (MVA) vector.
13. The vector of claim 12 wherein the MVA vector is further modified by deletion of one or more vaccinia immunomodulatory genes.
14. The vector of claim 13 wherein the MVA vector is modified by deletion of all of the immunomodulatory genes C6L, K7R, and A46R.
15. A vaccine composition comprising an attenuated poxvirus vector according to any of claims 8 to 14.
16. The vaccine composition of claim 15, further comprising a heterologous vaccine.
17. The vaccine composition of claim 16 wherein the heterologous vaccine is selected from a vaccine comprising a covalently linked p62-E1 heterodimer; or a DNA-replicon (DREP) vaccine comprising a nucleic acid comprising a nucleotide sequence comprising CHIKV genomic sequences operably linked to a promoter sequence.
18. A vector according to any of claims 8 to 14 for use as a vaccine.
19. The use of an attenuated poxvirus vector according to any of claims 8 to 14 in the manufacture of a vaccine.
20. A method of immunising a subject against CHIKV infection, the method comprising administering a vaccine comprising an attenuated poxvirus vector according to any of claims 8 to 14, or administering a vaccine composition according to any of claims 15 to
17, to a subject.
21 . The method of claim 20 further comprising administering a subsequent dose of a CHIKV vaccine to the subject.
22. The method of claim 21 wherein the subsequent dose is of a vaccine comprising an attenuated poxvirus vector according to any of claims 8 to 14, or a vaccine composition according to any of claims 15 to 17.
23. The method of claim 20 wherein the subsequent dose is of a heterologous vaccine.
24. The method of claim 23 wherein the heterologous vaccine is selected from a vaccine comprising a covalently linked p62-E1 heterodimer; or a DNA-replicon (DREP) vaccine comprising a nucleic acid comprising a nucleotide sequence comprising CHIKV genomic sequences operably linked to a promoter sequence.
25. The method of claim 24 wherein the vaccine is a DREP vaccine, and the CHIKV genomic sequences comprise genes coding for the envelope proteins (E3, E2, 6K, and
E1 ), but do not include genes coding for the capsid (C) protein.
26. A kit comprising first and second doses of CHIKV vaccine, wherein the first dose is of an attenuated poxvirus vector according to any of claims 8 to 14, or a vaccine composition according to any of claims 15 to 17.
27. The kit of claim 26 wherein the second dose is of an attenuated poxvirus vector according to any of claims 8 to 14, or a vaccine composition according to any of claims 15 to 17.
28. The kit of claim 26 wherein the second dose is of a vaccine comprising a covalently linked p62-E1 heterodimer; or a DNA-replicon (DREP) vaccine comprising a nucleic acid comprising a nucleotide sequence comprising CHIKV genomic sequences operably linked to a promoter sequence.
29. The kit of any of claims 26 to 28 wherein the doses are provided in sealed vials, or in preloaded syringes.
PCT/EP2014/076310 2014-12-02 2014-12-02 Vaccine composition WO2016086980A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/EP2014/076310 WO2016086980A1 (en) 2014-12-02 2014-12-02 Vaccine composition

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2014/076310 WO2016086980A1 (en) 2014-12-02 2014-12-02 Vaccine composition

Publications (1)

Publication Number Publication Date
WO2016086980A1 true WO2016086980A1 (en) 2016-06-09

Family

ID=52423677

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/076310 WO2016086980A1 (en) 2014-12-02 2014-12-02 Vaccine composition

Country Status (1)

Country Link
WO (1) WO2016086980A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018032057A1 (en) * 2016-08-19 2018-02-22 Sementis Limited Viral vaccines
JP2019521148A (en) * 2016-07-15 2019-07-25 エトゥビクス コーポレーション Compositions and methods for alphavirus vaccination
EP3928789A1 (en) 2020-06-24 2021-12-29 Consejo Superior de Investigaciones Científicas (CSIC) Mva-based vaccine against covid-19 expressing sars-cov-2 antigens
WO2021260065A1 (en) 2020-06-24 2021-12-30 Consejo Superior De Investigaciones Científicas (Csic) Mva-based vaccine against covid-19 expressing sars-cov-2 antigens
RU2778312C2 (en) * 2016-08-19 2022-08-17 Сементис Лимитед Viral vaccines
EP4108257A1 (en) 2021-06-23 2022-12-28 Consejo Superior De Investigaciones Científicas Mva-based vaccine against covid-19 expressing a prefusion-stabilized sars-cov-2 s protein
WO2022269003A1 (en) 2021-06-23 2022-12-29 Consejo Superior De Investigaciones Cientificas MVA-BASED VACCINE EXPRESSING A PREFUSION-STABILIZED SARS-CoV-2 S PROTEIN

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013011179A2 (en) * 2011-07-19 2013-01-24 Consejo Superior De Investigaciones Científicas (Csic) Recombinant vectors based on the modified vaccinia ankara (mva) virus, with deletion in the c6l gene, as vaccines against hiv/aids and other diseases

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013011179A2 (en) * 2011-07-19 2013-01-24 Consejo Superior De Investigaciones Científicas (Csic) Recombinant vectors based on the modified vaccinia ankara (mva) virus, with deletion in the c6l gene, as vaccines against hiv/aids and other diseases

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
D. HALLENGARD ET AL: "Novel Attenuated Chikungunya Vaccine Candidates Elicit Protective Immunity in C57BL/6 mice", JOURNAL OF VIROLOGY, vol. 88, no. 5, 1 March 2014 (2014-03-01), pages 2858 - 2866, XP055204816, ISSN: 0022-538X, DOI: 10.1128/JVI.03453-13 *
D. HALLENGARD ET AL: "Prime-Boost Immunization Strategies against Chikungunya Virus", JOURNAL OF VIROLOGY, vol. 88, no. 22, 15 November 2014 (2014-11-15), pages 13333 - 13343, XP055204781, ISSN: 0022-538X, DOI: 10.1128/JVI.01926-14 *
J. GARCIA-ARRIAZA ET AL: "A Novel Poxvirus-Based Vaccine, MVA-CHIKV, Is Highly Immunogenic and Protects Mice against Chikungunya Infection", JOURNAL OF VIROLOGY, vol. 88, no. 6, 15 March 2014 (2014-03-15), pages 3527 - 3547, XP055204747, ISSN: 0022-538X, DOI: 10.1128/JVI.03418-13 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019521148A (en) * 2016-07-15 2019-07-25 エトゥビクス コーポレーション Compositions and methods for alphavirus vaccination
US11104916B2 (en) 2016-07-15 2021-08-31 Etubics Corporation Compositions and methods for alphavirus vaccination
US20200009245A1 (en) * 2016-08-19 2020-01-09 Sementis Limited Viral vaccines
AU2017313450B2 (en) * 2016-08-19 2024-02-15 Sementis Limited Viral vaccines
WO2018032057A1 (en) * 2016-08-19 2018-02-22 Sementis Limited Viral vaccines
EP3500280A4 (en) * 2016-08-19 2020-05-13 Sementis Limited Viral vaccines
US10905759B2 (en) * 2016-08-19 2021-02-02 Sementis Limited Vector-based attenuated poxvirus vaccines
TWI728173B (en) * 2016-08-19 2021-05-21 澳大利亞商賽門堤斯有限公司 Viral vaccines
CN109862909A (en) * 2016-08-19 2019-06-07 赛门蒂斯有限公司 Viral vaccine
JP2019528271A (en) * 2016-08-19 2019-10-10 セメンティス リミテッド Virus vaccine
EP4331676A3 (en) * 2016-08-19 2024-05-15 Sementis Limited Viral vaccines
RU2778312C2 (en) * 2016-08-19 2022-08-17 Сементис Лимитед Viral vaccines
EP3928789A1 (en) 2020-06-24 2021-12-29 Consejo Superior de Investigaciones Científicas (CSIC) Mva-based vaccine against covid-19 expressing sars-cov-2 antigens
WO2021260065A1 (en) 2020-06-24 2021-12-30 Consejo Superior De Investigaciones Científicas (Csic) Mva-based vaccine against covid-19 expressing sars-cov-2 antigens
WO2022269003A1 (en) 2021-06-23 2022-12-29 Consejo Superior De Investigaciones Cientificas MVA-BASED VACCINE EXPRESSING A PREFUSION-STABILIZED SARS-CoV-2 S PROTEIN
EP4108257A1 (en) 2021-06-23 2022-12-28 Consejo Superior De Investigaciones Científicas Mva-based vaccine against covid-19 expressing a prefusion-stabilized sars-cov-2 s protein

Similar Documents

Publication Publication Date Title
Volz et al. Modified vaccinia virus Ankara: history, value in basic research, and current perspectives for vaccine development
García-Arriaza et al. COVID-19 vaccine candidates based on modified vaccinia virus Ankara expressing the SARS-CoV-2 spike protein induce robust T-and B-cell immune responses and full efficacy in mice
Enjuanes et al. Molecular basis of coronavirus virulence and vaccine development
García-Arriaza et al. A novel poxvirus-based vaccine, MVA-CHIKV, is highly immunogenic and protects mice against chikungunya infection
Hallengärd et al. Prime-boost immunization strategies against Chikungunya virus
García-Arriaza et al. Improving adaptive and memory immune responses of an HIV/AIDS vaccine candidate MVA-B by deletion of vaccinia virus genes (C6L and K7R) blocking interferon signaling pathways
Weger-Lucarelli et al. A novel MVA vectored Chikungunya virus vaccine elicits protective immunity in mice
Van Den Doel et al. Recombinant modified vaccinia virus Ankara expressing glycoprotein E2 of Chikungunya virus protects AG129 mice against lethal challenge
WO2016086980A1 (en) Vaccine composition
Indran et al. Novel approaches to develop Rift Valley fever vaccines
Knudsen et al. Kinetic and phenotypic analysis of CD8+ T cell responses after priming with alphavirus replicons and homologous or heterologous booster immunizations
Boshra et al. A lumpy skin disease virus deficient of an IL-10 gene homologue provides protective immunity against virulent capripoxvirus challenge in sheep and goats
EP3868398A1 (en) Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
García-Arriaza et al. Deletion of the vaccinia virus N2L gene encoding an inhibitor of IRF3 improves the immunogenicity of modified vaccinia virus Ankara expressing HIV-1 antigens
Hooper et al. Molecular smallpox vaccine delivered by alphavirus replicons elicits protective immunity in mice and non-human primates
Schäfer et al. Pre-clinical efficacy and safety of experimental vaccines based on non-replicating vaccinia vectors against yellow fever
US20230241201A1 (en) Coronavirus vaccines
DeFilippis Chikungunya virus vaccines: platforms, progress, and challenges
Volz et al. Immunogenicity and protective efficacy of recombinant Modified Vaccinia virus Ankara candidate vaccines delivering West Nile virus envelope antigens
WO2018152526A1 (en) Zika virus vaccines
Perdiguero et al. Highly attenuated poxvirus-based vaccines against emerging viral diseases
US20070036827A1 (en) West nile virus vaccine
Seregin et al. Immunogenicity of West Nile virus infectious DNA and its noninfectious derivatives
JP2006526393A (en) Flavivirus replicon packaging system
George et al. Distinct humoral and cellular immunity induced by alternating prime-boost vaccination using plasmid DNA and live viral vector vaccines expressing the E protein of dengue virus type 2

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14830800

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14830800

Country of ref document: EP

Kind code of ref document: A1