WO2023126882A1 - Denv ediii-ns1 consensus sequence-based dengue dna vaccine - Google Patents

Denv ediii-ns1 consensus sequence-based dengue dna vaccine Download PDF

Info

Publication number
WO2023126882A1
WO2023126882A1 PCT/IB2022/062891 IB2022062891W WO2023126882A1 WO 2023126882 A1 WO2023126882 A1 WO 2023126882A1 IB 2022062891 W IB2022062891 W IB 2022062891W WO 2023126882 A1 WO2023126882 A1 WO 2023126882A1
Authority
WO
WIPO (PCT)
Prior art keywords
denv
dna vaccine
vaccine construct
recombinant dna
dengue
Prior art date
Application number
PCT/IB2022/062891
Other languages
French (fr)
Inventor
Sudhir Krishna
Arun SANKARADOSS
Original Assignee
National Centre For Biological Sciences
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 National Centre For Biological Sciences filed Critical National Centre For Biological Sciences
Publication of WO2023126882A1 publication Critical patent/WO2023126882A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • 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
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24134Use 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

  • This disclosure relates to a DNA vaccine construct against Dengue virus.
  • the present disclosure relates to a DENV (dengue) DNA vaccine construct, comprising at least one antigenic regions of envelope domain and at least one protein-coding region.
  • the present disclosure also relates to a process for constructing a DNA vaccine construct and uses thereof.
  • the DENV ED III (envelope domain III) region of Dengue has emerged as a vaccine target, as it is known to stimulate serotype-specific antibodies, and antibodies generated against this region are either not involved or minimally involved in antibody-dependent enhancement (ADE) of the infection (Fahimi, et al., 2018. Dengue viruses and promising envelope protein domain Ill-based vaccines. Appl. Microbiol. Biotechnol. 2018 1027 102, 2977-2996)Recent studies have found that EDIII based DENV vaccines could circumvent ADE (ADE of infection) of infection in mice, whereas T cell response against EDIII DNA vaccine has shown to play a role in disease protection through effective viral clearance (Ramasamy, et al. 2018.
  • a tetravalent virus-like particle vaccine designed to display domain III of dengue envelope proteins induces multi- serotype neutralizing antibodies in mice and macaques which confer protection against antibody dependent enhancement in AG129 mice.
  • PLoS One 10, e0145717-e0145717 support the choice of EDIII for the next-generation vaccine development.
  • NS1 DENV non- structural protein 1
  • NS1 DENV non- structural protein 1
  • NS1 a highly conserved protein among flaviviruses, is involved in virus replication and immune evasion (Chen, et al., 2018. Dengue virus non- structural protein 1: A pathogenic factor, therapeutic target, and vaccine candidate. J. Biomed. Sci. 25).
  • it has also been reported to activate antibody Fc-mediated effector function and provide protection against flaviviruses (Wan, et al., 2014. Protection against dengue virus infection in mice by administration of antibodies against modified nonstructural protein 1.
  • PLoS One 9 NS1 has been demonstrated to trigger T-cell responses in both experimental animals and humans (Grubor-Bauk, et al. 2019.
  • NS1 DNA vaccination protects against Zika infection through T cell-mediated immunity in immunocompetent mice. Sci. Adv. 5, eaax2388). Previous studies have shown that NS1 induced T cell responses involved in protection against dengue infection. Furthermore, it is seen that vaccination with DENV-1, DENV-3, or DENV-4 NS1 offers protection against a heterologous DENV2 challenge (Beatty, et al., 2015. Dengue virus NS1 triggers endothelial permeability and vascular leak that is prevented by NS1 vaccination. Sci. Transl. Med. 7, 304ral41-304ral41; Lai, et al., 2017. Antibodies against Modified NS1 Wing Domain Peptide Protect Against Dengue Virus Infection. Sci. Rep. 7).
  • DENV vaccine development is complicated by four antigenically diverse serotypes (DENV 1-4) and their intra-serotype (genotype) diversity at both local and global scales (Bhatt, et al. 2013. The global distribution and burden of dengue. Nature 496, 504-507).
  • the genetic and antigenic differences between genotypes of the same serotype were not considered to impact long-term protective immunity and vaccine efficacy.
  • several recent studies have challenged this assumption (A, M. et al., 2013. Lineage shift in Indian strains of Dengue virus serotype-3 (Genotype III), evidenced by detection of lineage IV strains in clinical cases from Huawei. Virol. J. 10; BP, et al., 2013.
  • US20210188947 discloses compositions comprising structurally modified DNA encoded antibodies (DMAbs), methods of structurally modifying DMAbs, and methods of using structurally modified DMAbs.
  • DMAbs structurally modified DNA encoded antibodies
  • WO2018/217897 discloses an immunogenic comprising an antigen and optionally IL- 12 and a method for administering the vaccine through intradermal electroporation.
  • WO2016034974A1 provides a recombinant polypeptide comprising the EDIII domain of each of Dengue virus serotype DENV-1, DENV-2, DENV-3, and DENV-4 linked to the N-terminal of HBsAg.
  • WO2017142831 Al relates to specific Dengue virus glycoprotein subunit E Dill variants and their uses in preventing and treating Dengue virus infection.
  • T Cell Responses Induced by DNA Vaccines Based on the DENV2 E and NS1 Proteins in Mice: Importance in Protection and Immunodominant Epitope Identification” (https://pubmed.ncbi.nlm.nih.gov/31333657/) emphasizes the importance of the T cell response involved in protection against dengue induced by E and NS1 based DNA vaccines.
  • DNA based vaccines is predicted to be a real game changer in India, Africa and other tropical regions where cold chain (storage and distribution of vaccines to health services) is a main concern.
  • the low cost and thermostable aspect of the DNA vaccine should allow countries to drive vaccination programs on a larger scale than is currently possible, especially in middle- and low-income countries.
  • FIG. 1 illustrates the DENV DNA vaccine (DDV) expression cassette.
  • Figure 2 shows DENV Serotype distribution across various geographical sites.
  • Figure 3 illustrates DENV amino acid variation in clinical isolates for all four serotypes.
  • Figure 4 shows DENV DNA vaccine construction.
  • Figure 5 shows antibody response induced by DENV DNA vaccination in BALB/c and C57BL/6J mice.
  • Figure 6 shows that characterization of cellular immune response induced by DENV DNA vaccine in mice.
  • Figure 7 shows the Th 1 -biased immune responses elicited by DDV.
  • Figure 8 illustrates transcriptomic analysis of immune genes after vaccination with
  • Figure 9 illustrates the capacity of anti-DDV immune sera to confer protection against lethal DENV-2 challenge in AG129 mice.
  • Figure 10 illustrates the genetic relationship of Indo-Africa DENV2 strains.
  • Figure 11 illustrates DENV amino acid variation among Indian strains of all four serotypes isolated from 2000-2020. (A, D, G and J).
  • Figure 12 shows the structural validation of the modelled constructs.
  • Figure 13 shows in-vitro antigen expression using monoclonal EDIII and NS1 antibodies.
  • FIG 14 shows DENV DNA vaccine (DDV) expression cassette for a variant of Dengue DNA vaccine construct of present invention.
  • the present invention provides a DNA based vaccine for pan dengue viruses.
  • the invention discloses a DNA vaccine, wherein the antigenic regions DENV EDIII of dengue virus serotypes 1-4 and DENV-2 NS1 are incorporated in the vaccine construct, such that the DNA vaccine induces robust B cell and T cell immune response without causing ADE of the infection.
  • the present invention also provides a process for constructing a DNA vaccine by incorporating EDIII and NS1 in the vaccine design.
  • the present invention further provides a pharmaceutical composition; pharmaceutical combination and a kit comprising DNA based vaccine for pan dengue viruses.
  • the invention furthermore provides a method of treatment and/or prevention of one or more symptoms of dengue viruses in a subject in need thereof.
  • Vaccination of mice with DDV induces pan-serotype neutralizing antibodies and antigen-specific T cell responses.
  • Assaying of intracellular IFN-y staining, immunoglobulin IgG2(a/c)/IgGl ratios and immune gene profiling suggest a strong Th 1 -dominant immune response.
  • passive transfer of DDV immune serum protected AG129 mice challenged with a virulent, non-mouse adapted DENV-2 strain.
  • the surprising findings of preset invention collectively suggest an alternative strategy for dengue vaccine design; and offers a novel vaccine candidate with a possible broad spectrum protection and successful clinical translation either as a stand-alone or in a mix and match strategy.
  • the present disclosure describes a process for constructing a DNA vaccine by incorporating EDIII and NS1 in the vaccine design.
  • the present invention provides a recombinant DNA vaccine construct comprising at least one antigenic region of envelope domain and at least one protein-coding region of dengue virus.
  • the present invention provides that the protein-coding region in the recombinant DNA vaccine construct is a non- structural protein-coding region.
  • the present invention provides that the DNA vaccine construct comprises sequences selected from SEQ. ID NO 1 or SEQ. ID NO 3 or a variant thereof.
  • the present invention provides that the variant of DNA vaccine construct is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 99% identical to the SEQ. ID NO 1 or SEQ. ID NO 3.
  • the present invention provides that the antigenic region of envelope domain of dengue virus (DENV) of DNA vaccine construct comprises the envelope domain III of serotypes 1-4.
  • DEV dengue virus
  • the present invention provides that the protein-coding region of dengue virus in DNA vaccine construct is dengue-2 non- structural protein 1 (NS1).
  • NS1 dengue-2 non- structural protein 1
  • the present invention provides that recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non-structural protein 1 (NS1).
  • recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non-structural protein 1 (NS1).
  • NS1 DENV non-structural protein 1
  • Another embodiment of the present invention provides a method for preparing a recombinant DNA vaccine construct comprising steps of:
  • the present invention provides that the step of inserting the DENV expression cassette into a modified expression vector in the method for preparing a recombinant DNA vaccine construct comprises incorporating the DENV DNA expression cassette into a modified pVAXl expression vector between Nhel and Hindlll under the control of cytomegalovirus immediate early promoter to result in recombinant vector for the DENV DNA vaccine.
  • the present invention provides that the method for preparing a recombinant DNA vaccine construct further comprises the step of optimizing the DNA vaccine construct by codon optimisation for optimal protein expression.
  • the present invention provides that in the method for preparing a recombinant DNA vaccine construct the recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non- structural protein 1 (NS1).
  • the recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non- structural protein 1 (NS1).
  • the present invention provides that in the method for preparing a recombinant DNA vaccine construct the recombinant DNA vaccine construct comprises sequences selected from SEQ. ID NO 1 or SEQ. ID NO 3 or a variant thereof.
  • the present invention provides a method of preparation of DENV expression cassette comprises the steps of:
  • the present invention provides that in the method of preparation of DENV expression cassette the DENV serotype in step (i) is selected from serotypes DENV-1, DENV-2, DENV-3, and/or DENV-4.
  • the present invention provides that in the method of preparation of DENV expression cassette the DENV serotype in step (ii) is DENV-2.
  • the present invention provides that in the method of preparation of DENV expression cassette the proteolytic enzyme is furin.
  • the present invention provides that the method of preparation of DENV expression cassette further comprises the step of adding Kozak sequences and IgE leader sequence upstream to the DENV expression cassette.
  • the present invention also provides an isolated nucleic acid molecule encoding the recombinant DNA vaccine construct of the present invention.
  • the present invention provides an expression vector comprising the nucleic acid molecule of the present invention.
  • the present invention provides a host cell comprising the expression vector of the present invention.
  • the present invention provides a pharmaceutical composition or combination for treatment and/or prevention of one or more symptoms of dengue viruses in a subject in need thereof, said composition comprising about 1 pl to 1000 pl of recombinant DNA vaccine construct along with pharmaceutically acceptable excipients.
  • the present invention provides a pharmaceutical composition or combination for inducing immunogenicity against dengue viruses in a subject in need thereof, wherein said subject is administered with 1
  • the present invention provides a method of preparing pharmaceutical composition or combination for treating and/or preventing one or more symptoms of dengue virus in a subject in need thereof comprising the steps: a) adding about 1 pl to 1000 pl of recombinant DNA vaccine construct at suitable conditions to one or more pharmaceutically acceptable excipients to obtain a mixture; b) subjecting the mixture obtained in step ‘a’ to suitable conditions to obtain the formulation or combination in desired dosage form.
  • the present invention provides a method of treating and/or preventing one or more symptoms of dengue viruses in a subject in need thereof, comprising administering to the subject a composition or combination comprising about 1 pl to 1000 pl of recombinant DNA vaccine construct of the present invention.
  • the present invention provides use of a composition or combination comprising recombinant DNA vaccine construct of the present invention for treating and/or preventing one or more symptoms of dengue viruses in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of said composition or combination.
  • the present invention provides a kit comprising a recombinant DNA vaccine construct of the present invention, pharmaceutical composition or combination of the present invention and instructions for administration of the pharmaceutical formulation and/or combination to a subject in need of treatment and/or prevention of dengue infection.
  • the present invention relates to a DNA based vaccine for pan dengue viruses.
  • the present disclosure relates to a DENV DNA vaccine, wherein the antigenic regions DENV EDIII of dengue virus serotypes 1-4 and DENV-2 non- structural protein 1 (NS1) are incorporated in the vaccine construct, such that the DNA vaccine induces robust B cell and T cell immune response without causing ADE of the infection.
  • DENV DNA vaccine wherein the antigenic regions DENV EDIII of dengue virus serotypes 1-4 and DENV-2 non- structural protein 1 (NS1) are incorporated in the vaccine construct, such that the DNA vaccine induces robust B cell and T cell immune response without causing ADE of the infection.
  • the DENV EDIII-based DNA vaccine (DDV) of the present disclosure was developed by integrating consensus sequence information from circulating genotypes of each serotype instead of a single strain genome sequence; incorporating NS1 protein coding region to induce broad T cell responses; and then optimizing DNA vaccine construct for optimal protein expression.
  • Consensus vaccine antigens were derived from strains which are circulating in India and Africa.
  • the present disclosure also describes a process for constructing a DNA vaccine by incorporating EDIII and NS1 in the vaccine design.
  • the DENV 1-4 EDIII and NS1 gene of DENV2 were linked together in a single construct with furin cleavage sites between the individual genes. Consensus sequences were optimized for EDIII and NS1 expression, including codon and RNA optimization. The Kozak sequences and IgE leader sequence were added upstream to the DNA sequences as were furin cleavage sites to facilitate EDIII and NS1 processing.
  • the synthetic DENV DNA expression cassette was inserted into the modified pVAXl expression vector (Source: Invitrogen, CA, synthesized commercially at Genscript) between Nhel and Hindlll under the control of the cytomegalovirus immediate early promoter (Source: Genscript Biotech, USA) to result in the recombinant vector for the DENV DNA vaccine.
  • Figure 1 illustrates the DENV DNA vaccine expression cassette (SEQ. ID NO 1 and SEQ. ID NO 2).
  • nucleotide sequence of the recombinant DENV DNA vaccine sequence is set forth as SEQ. ID NO 1 in Table A below.
  • the peptide sequence encoded by the DENV vaccine expression cassette is set forth as SEQ. ID NO 2 in Table B below.
  • Table B List of peptide sequence of present invention
  • the DENV DNA vaccine of the present disclosure could be alternative strategy for traditional vaccines.
  • the vaccine design enables fast tracked vaccine production and eliminates much of the logistics associated with vaccine distribution.
  • the low cost and thermostable aspect of the DNA vaccine should allow countries to drive vaccination programs on a larger scale than is currently possible, especially in India and Africa.
  • the recombinant DNA sequence of present invention can be a variant of SEQ. ID NO 1 or SEQ. ID NO 3.
  • the variant is a functionally active variant and may be obtained by changing sequence of SEQ. ID NO 1 or SEQ. ID NO 3 and is characterized by having a biological activity similar to that displayed by nucleotide sequence of SEQ. ID NO 1 or SEQ. ID NO 3 from which the variant is derived.
  • the variant includes ability of recombinant DNA sequence for treatment; prevention and/or amelioration of one or more symptoms of pan Dengue viruses in a subject in need thereof.
  • the functionally active variant of SEQ. ID NO 1 or SEQ. ID NO 3 may be obtained by sequence alterations in sequence of SEQ. ID NO 1 or SEQ.
  • the variant can comprise at least 70% of the sequence of SEQ. ID NO 1 or SEQ. ID NO 3, at least 75% of the sequence of SEQ. ID NO 1 or SEQ. ID NO 3, at least 80% of the sequence of SEQ. ID NO 1, preferably at least 85%, still more preferably at least 90%, even more preferably at least 95% and most preferably at least 97%, 98% or 99%.
  • the variant is derived from the SEQ. ID NO 1 or SEQ.
  • SEQ. ID NO 3 by at least one amino acid substitution and/or deletion, wherein the functionally active variant has a sequence identity to SEQ. ID NO 1 or SEQ. ID NO 3 of at least 80%, more preferably at least 85%, still more preferably at least 90%, even more preferably at least 95% and most preferably at least 97%, 98% or 99%.
  • the variant of SEQ. ID NO 1 or SEQ. ID NO 3 is functionally active in the context of the present invention, if the activity of the variant amounts to at least 10%, preferably at least 25%, more preferably at least 50%, even more preferably at least 70%, still more preferably at least 80%, especially at least 90%, particularly at least 95%, most preferably at least 99% of the activity of SEQ. ID NO 1 or SEQ. ID NO 3 without sequence alteration.
  • the activity of the variant may be determined or measured as described in the examples and then compared to that obtained for SEQ. ID NO 1 or SEQ. ID NO 3 of the present invention.
  • Example 1 Obtaining Samples, Ethical consideration, and DENY Viral DNA sequencing
  • DENY viral RNA sequencing The phylogenetic-based genotype analysis of the present study revealed that all Indian DENV 1-4 strains belong to a single genotype within the serotypes, except DENV1 ( Figure 2B to 2E).
  • DENV-1, genotype III is the most dominant, apart from a few sequences of genotype I.
  • DENV2 sequences belong to the cosmopolitan genotype, whereas DENV-3 and DENV-4 sequences belong to genotype III and I, respectively.
  • the data show that two different DENV-2 lineages within the cosmopolitan genotype are propagating simultaneously in all four sites in this study. Both the lineages emerged in the middle of the 1980s. Most of the neighboring sequences are from Asian countries.
  • Serum/ Plasma was tested for dengue virus -specific immunoglobulin M (IgM) and IgG antibodies using Panbio Dengue IgM and IgG capture ELISA kits, respectively. Dengue infection was classified as primary or secondary, and further samples were categorized as per WHO 2009 guidelines.
  • Viral RNA was extracted from about 150 pl of patient’s serum using QIAamp Viral RNA Mini Kit (Qiagen). Sequencing library preparation was performed with about 1.2 ng of normalized double stranded cDNA using the Illumina Nextera XT sequencing kit and sequenced on an Illumina Miseq platform, using 2 X 75 and 2 X 150 bp paired-end reads.
  • Raw sequence reads were inspected for quality using FASTQC (https://www.bioinformatics.babraham.ac.uk/projects/fastqc/), reads were filtered and trimmed based on the quality score. Reads were then mapped to the reference sequences obtained from the RefSeq database (NC_001477(DENV-l), NC_001474 (DENV-2), NC_001475 (DENV-3), NC_002640 (DENV-4) using Geneious assembler (3 iterations and medium-low sensitivity). Assembled reads were checked manually for errors.
  • Serotypes were assigned manually and coverage was assigned based on the following criteria: >10X coverage if the depth of the coverage at each base was >20 and breadth of the coverage >95%, 5X-10X coverage if depth ⁇ 20 and breadth>95, IX if depth ⁇ 10 and breadth>90, for less coverages serotype.
  • Consensus sequences were generated from the samples having >5X coverage based on the majority rule and were clipped to the length of reference sequences, iqtree v 1.6.10 was used to construct maximum likelihood trees with 1000 bootstrap replicates.
  • Sylvatic strains EF457905 (for DENV-1), EF105379 (for DENV-2), KT424097 (for DENV- 3) and JF262779-80 (for DENV-4) were used as outgroups to root the tree. Genotypes were assigned to our sequences based on their positions in the tree. Figtree vl.4 was used to visualize the tree.
  • Example 2 DENV EDIII genetic diversity
  • the E protein primarily Dill domain of the E protein, stimulates host immune responses by evoking protective and neutralizing antibodies. Mutations in the EDIII region could potentially impact the neutralization of DENV and the host-receptor interaction.
  • the EDIII protein diversity in global strains and how the present study’s sequences are divergent from reference strains was investigated. To assess antigenic differences between the EDIII of DENV 1-4 strains from distinct parts of the world, a neighbor-joining tree was generated and representative genotype variants were selected from each branch to comprehensively represent global diversity within the serotype. A sequence alignment of these proteins was performed and their EDIII genetic diversity relative to the respective reference strains was compared.
  • Variable sites were designated when at least one virus showed an amino acid change at any of the 103 amino acid positions in the alignment. While DENV genotypes are closely related, considerable genetic variation was observed across genotypes of the same serotype in the EDIII region with a range of 1.62-5.83%.
  • DENV-1 exhibited the highest EDIII diversity with a median of 4.85% (range, 2.91 %-5.83%), followed by DENV-4 and DENV-2 with 2.91% (range, 2.91%-3.88%) and 1.94% (range, 0.97 %-3.88%), respectively.
  • the percentage diversity of DENV-3 EDIII sequences was found to be limited, ranging from 0% to 0.97 %.
  • the frequency of amino acid variants was calculated across strains and expressed as a percentage ( Figure 3B, 3D, 3F and 3H). These mutations were also spotted on the EDIII PDB structure ( Figure 3C, 3E, 3G and 31). Additionally, several highly variable sites were identified across serotypes.
  • a site was considered highly variable when greater than 50% of the study isolates showed a mutation at that position.
  • the number of highly variable sites in EDIII was 5, 3, 2, and 1 in DENV-1, DENV-4, DENV-2, and DENV-3 strains, respectively.
  • EDIII mutations in all Indian DENV1-4 strain sequences deposited in the ViPR database were investigated.
  • Figure 11 shows amino acid variants and their frequency, implicating the EDIII amino acid residues as sites under immune pressure.
  • DNA based vaccines would be a real game changer in India, Africa and other tropical regions where cold chain is a main concern.
  • the DENV DNA vaccine candidate of the present disclosure is more adapted to the strains of dengue viruses found in India and Africa.
  • a new DNA vaccine candidate was generated by combining in tandem envelope protein domain III of dengue virus serotypes 1-4 and a DENV-2 NS1 protein coding region. Each domain was designed as a serotype -specific consensus coding sequence derived from different genotypes based on whole genome sequencing of clinical isolates in India (aforementioned study) and complemented with data from Africa that were retrieved from the ViPR database. Generated consensus sequences were codon and RNA optimized, synthesized commercially, cloned into pVAXl expression vector and the generated plasmid was designated as DDV (Dengue DNA Vaccine) ( Figure 4A and 4B).
  • Table. 3 Comparison of multiple EDIIIs within each serotype with the EDIII consensus sequences used to design DENV DNA vaccine.
  • DDV has 100% identity with African DENV-2 and DENV-3 strains, while African DENV-1 and DENV-4 strains exhibited >96% identity with their corresponding serotype DDV sequences. DDV also shares >95.15 identities with the EDIII of the top 1000 international dengue sequences of the cognate serotype in the ViPR database.
  • the polyvalent DENV DNA vaccine construct encodes EDIII of all four serotypes and NS1 sequence of DENV-2.
  • African DENV-2 and DENV-3 genotypes are identical to Indian genotypes, these sequences were pooled directly with Indian DENV2 and 3 strains for consensus generation. Africa DENV1 and 4 genotypes differ from those in India, hence consensus sequences were generated separately and compared to Indian DENV-1 and DENV-4 strains represented in the vaccine expression cassette. African DENV-1 and DENV-4 strains exhibited >96 % identity with their respective serotype DDV sequences.
  • the DENV DNA vaccine expression cassette was designed with the consensus sequences of the EDIII gene of all four serotypes and consensus sequences of the NS1 gene of DENV-2 linked together in a single construct with furin cleavage sites between the individual genes. Consensus sequences were optimized for EDIII and NS1 expression, including codon and RNA optimization. The Kozak sequences and IgE leader sequence were added upstream to the DNA sequences as were furin cleavage sites to facilitate EDIII and NS1 processing.
  • the synthetic DENV DNA expression cassette was inserted into the pVAXl expression vector between Nhel and Hindlll under the control of the cytomegalovirus immediate early promoter (Genscript Biotech, USA) (deposition to the Microbial Type Culture Collection is under process).
  • a variant of Dengue DNA vaccine construct of present invention The inventors of present invention also optimized Dengue vaccine construct by changing the order of EDIII segments to DENV-3, DENV-4, DENV-1 and DENV-2, followed by DENV-2 NS1.
  • the EDIII of DENV 1-4 and NS1 of DENV-2 sequences have been further expanded to contain all global strains of dengue and have been constructed irrespective of genotype. Additionally, P2A cleavage sites from porcine echovirus were incorporated between DENV EDIII 1-4 and NS1 segments to promote equimolar production of these antigens.
  • the generated consensus sequences were codon- optimized and RNA-optimized, synthesized commercially, and cloned into nanoplasmid expression plasmids NTC9385R-eRNA41H-CpG and NTC 9385R by Nature Technology, USA ( Figure 14, SEQ. ID NO 3, SEQ. ID NO 4).
  • Example 5 In vitro antigen expression and Immunofluorescence assays
  • the encoded DENV EDIII and NS1 transgene expression was assessed at the RNA level in HEK293T cells [ATCC and NCOS, Pune] transfected with DDV. Using the total RNA isolated from the transfected 293T cells, the EDIII and NS1 mRNA expression was confirmed by qRT-PCR ( Figure 4C). In vitro, EDIII and NS1 protein expression in HEK- 293T cells was measured by western blot using anti-DDV immune sera on cell lysates.
  • HEK293T cells were transfected with DENV DNA Vaccine (DDV) using X-treme GENE HP DNA transfection reagent as per the manufacturer’s instructions.
  • the transfected cell lysate and supernatants were collected about 36 hours of post-transfection, and antigen expression was confirmed by western blot analysis.
  • Cells were washed with phosphate-buffered saline (PBS) and lysed with NP40 supplemented with protease inhibitor cocktail (Roche) and ImM PMSF(Sigma) was used to make cell lysates.
  • PBS phosphate-buffered saline
  • NP40 protease inhibitor cocktail
  • ImM PMSF(Sigma) was used to make cell lysates.
  • Protein lysates were separated on SDS-polyacrylamide gel, transferred onto a nitrocellulose membrane (Bio-Rad), and blocked for about 1 hour in about 5% skimmed milk. Subsequently, membranes were incubated in mouse antisera (1:200 dilution) against DDV. Secondary antibodies conjugated to horseradish peroxidase (HRP) were used at a dilution of about 1:1500. After washing with PBS/PBST the blots were developed using an enhanced chemiluminescence system (Thermo Fisher).
  • HRP horseradish peroxidase
  • TA Tibialis anterior
  • mice were bled at day 0 and two weeks after each vaccination to obtain sera, which were assayed for the presence of DENV antibodies by enzyme-linked immunosorbent assay (ELISA) against recombinant protein as a capture protein ( Figure 5A).
  • ELISA enzyme-linked immunosorbent assay
  • Figure 5A Binding antibody ELISA data revealed that the DDV induced DENV specific antibody responses.
  • the results showed that all mice developed anti-DENV antibodies after a single immunization.
  • the anti-dengue IgG responses were significantly increased after 1-2 booster immunizations, appearing to peak 2 weeks after the second booster in both BALB/c and C57BL/6J strains ( Figure 5B and 5D).
  • mice were immunized with about 50 pg of DNA in a total volume of about 50 pl of sterile water by a syringe into the anterior tibialis (TA) muscles then electroporated using BTX ECM 830 with 8 square 40-V electric pulses in alternating direction with a time constant of about 0.05 s and an inter-pulse interval of about Is. Each group received 2 booster doses at about 2-week intervals, and mice were euthanized about two weeks following the last immunization. All the experimental procedures were approved by the Institutional Animal Ethics Committee (IAEC) of National Centre for Biological Sciences (NCBS) and Theralndx Lifesciences Pvt. Ltd.
  • IAEC Institutional Animal Ethics Committee
  • NCBS National Centre for Biological Sciences
  • NCBS National Centre for Biological Sciences
  • Flow-based virus neutralization assay was performed in U937-DC-SIGN cells (U937-DC- SIGN cell line related assays were outsourced at the Bioassay Lab, THSTI, Faridabad) to assess the levels of anti-DDV immune sera-induced neutralizing antibody titers against laboratory DENV strains.
  • DDV-vaccinated mice immune sera showed a clear neutralizing antibody titer against all four serotypes simultaneously; the median FNT50 titers against DENV 1-4 ranged from 182-3500 ( Figure 5F and 5G).
  • the neutralizing potency of anti-DDV immune sera against DENV 1-4 recent clinical isolates was investigated.
  • DENV 1-4 serotypes were isolated from DENV NS1 positive patients in India and FNT was performed.
  • the data showed anti-DDV immune sera effectively neutralized clinical isolates as well and FNT50 titers against DENV 1- 4 ranged from 150-900 ( Figure 5H and 51). These data provide evidence that DDV-induced neutralizing antibody responses cover major currently circulating strains as well.
  • Method The flow cytometry-based neutralization assays were performed in triplicate in 96-well cell culture plates with flatbottom wells. Each well contained 5 X 10 4 DCSIGN- expressing U937 cells. Immune sera were serially diluted, and the virus was pre-incubated with the sera for about 1 hour at about 37°C. The cells were washed, and virus and serum mixture was added to the cells for about 1 hour at about 37°C. Next, the wells were filled with cell culture medium, and the plates were incubated for about 24 to 48 hours at about 37°C in about 5% CO2. The cells were prepared for flow cytometry analysis by washing them in phosphate-buffered saline and transferring them to 96-well plates with round-bottom wells.
  • the cells were fixed and permeabilized by using a Cytofix/ Cytoperm kit (BD-PharMingen, San Diego, CA) and stained with monoclonal antibody 4G2, a monoclonal antibody that recognizes the flavivirus E protein.
  • the cells were analyzed with a FACScan flow cytometer.
  • the serum dilution that neutralized 50% of the viruses was calculated by nonlinear, doseresponse regression analysis with Prism 4.0 software (GraphPad Software, Inc., San Diego, CA).
  • Example 8 IFN-y Enzyme-Linked Immunosorbent Spot (ELISPOT) Assay
  • DENV specific cellular responses were assessed in vaccinated animals by ELISPOT.
  • MHC class I binding predictions, peptide selection, and antigenicity of those peptides were analyzed by NetCTL 1.2 and Vaxijen 2.0.
  • a total of 15 CTL epitopes (9 mer peptides) were screened out of which 11 were found to possess antigenicity and were chosen for synthesis ( Figure 6B).
  • T cell response against dengue antigens via IFN-y ELISPOT was assayed. IFN-y has been described as a mediator of T cell responses and plays a distinctive role in antiviral activity against dengue viruses.
  • mice were immunized as before. Two weeks after the second booster DDV or pVAXl plasmid control immunized mice were euthanized and splenocytes were isolated (Figure 6A). Single-cell suspensions were stimulated with the peptide pools (Pool 1- EDIII peptide mixture; Pool 2- NS1 peptide mixture; Pool 3- EDIII and NS1 peptide mixture) number of IFN-y producing CD8+ T cells was analyzed.
  • Results show that both EDIII- and NS1 specific cellular responses (presented as IFN-y SFU/cells) were detected by ELISPOT in DDV vaccinated animals while negligible spots were detected in the plasmid vector-vaccinated animals ( Figure 6C to 6F).
  • PMA/IONOMYCIN was used as a non-specific positive control.
  • stimulation with PMA/IONO in all ELISPOT assays performed with cells from DENV vaccinated or control animals induced a high IFN-y response.
  • ELISpot was performed with the Mouse IFN-y ELISpot BASIC Kit (Mabtech).
  • VDF polyvinylidene fluoride
  • IO lonomycin
  • Example 9 Intracellular cytokine staining
  • Intracellular cytokine staining was performed using freshly isolated mouse splenocytes. The intracellular IFN-y in both CD8+ and CD4+ cells of mouse splenocytes in both vaccine and control groups of animals were analyzed. As a positive stimulus for T cell activation, PMA/Ionomycin were used. Exocytosis of cytokines was blocked by the addition of Brefeldin A (10 pg/ml) during stimulation. Cells were permeabilized, labeled, and fixed for flow cytometry.
  • IFN-y CD8+ T cells and IFN-y CD4+ T cells were proportionately higher, as found using intracellular staining with flow cytometry, in DDV as compared to pVAXl vaccinated animals ( Figure 61 and 6J).
  • DENV DNA vaccine candidate DDV induces robust T-cell response in mice.
  • Intracellular cytokine staining was performed by stimulating freshly isolated splenocytes with about 50 ng/mL PMA and about 500 ng/mL IO in the presence of Brefeldin for about 5-6 hours. After stimulation, surface staining of CD4, and CD8 was performed, followed by intracellular staining of IFN-y (Biolegend). Data acquisition was performed on a BD LSRFortessa and analyzed with FlowJo.
  • Example 10 DDV vaccination skewed a Thl-dominant response
  • Thl skewness has been shown to elicit a robust adaptive response in terms of cellular activation and antibody production.
  • the induction of polyfunctional Thl cells is an essential element of a protective vaccine response.
  • Respiratory disease viruses such as SARS-CoV and MERS-CoV vaccine development have highlighted the importance of Thl skewed response in mitigating the risk of vaccine-induced disease enhancement.
  • the Thl/Th2 balance elicited by vaccination with DENV DNA vaccine was investigated.
  • the IgG subclass fate of plasma cells is highly governed by Th cells.
  • Thl-associated IgG subclasses-IgG2a (BAlB/c) ( Figure 7B and 7C) and IgG2c (C57BL/6J) ( Figure 7D and 7E)- against the Th2 associated IgGl were measured.
  • C57BL/6J and BALB/c mice are prototypical Thl and Th2 animal strains, with C57BL/6J producing high IgG2c and BALB/c, mostly IgG2a.
  • evaluation of IgG2c and IFN-y is critical to correct interpretation of Thl immune responses.
  • DDV vaccination- induced Thl skewed IgG subclass responses In both strains of mice, DDV vaccination- induced Thl skewed IgG subclass responses.
  • DENV binding antibody titer was analyzed by indirect ELISA.
  • 96 well plates (Thermo Scientific) were coated with recombinant protein in a coating buffer (0.1M NaHCOs) and incubated overnight at about 4 °C. The following day, plates were blocked with about 3% BSA in PBS for about 2 hours at room temperature. Triplicate samples of serially diluted plasma ranging from 1:100 to 1:5,00,000 were added to the plate and incubated for about 2 hours at room temperature or overnight at about 4 °C.
  • Cut-off values for each dilution were set using the O.D of naive samples in the formula: naive O.D at a dilution + (2.5 * standard deviation). Starting from the lowest dilution, the sample dilution prior to the one which was exceeded by the cut-off was considered as the end titer value.
  • RNA was extracted with TRIzol LS.
  • RNA 50 ng
  • RNA was hybridized to the NanoString nCounter mouse inflammation and immunology v2 panels (NanoString Technologies), respectively.
  • RNA was hybridized with reconstituted CodeSet and ProbeSet. Reactions were incubated for about 24 hours at about 65°C and ramped down to about 4°C. Hybridized samples were then immobilized onto a nCounter cartridge and imaged on a nCounter SPRINT (NanoString Technologies).
  • DEGs differentially expressing genes
  • Graph pad Prism software was used to analyze variance with a cutoff of p ⁇ 0.05.
  • Log 2 fold changes generated were used for volcano plots constructed with Prism 5 software.
  • DEGs were identified by a fold change cutoff of 1.5.
  • Unsupervised PCA was performed to visualize variability between DDV and pVAXl control animals.
  • the DENV-inf ection-only group showed an initial increase in body weight; however, with a steep decrease from day 5 or 6.
  • the maintenance of body weight or percentage increase were observed to be better in 300 pl immunized mice compared to DENV-challenged group and pVAXl control group ( Figure 9B).
  • the mice started showing noticeable symptoms from 3rd day post-infection starting with ruffled fur which continued to become more aggressive and prominent with the disease progression. Despite the initial rise in symptoms, clinical scoring showed reduced manifestation of the disease in the 300 pl immunized group at later stages compared to the infection control and pVAXl control.
  • AG129 mice [deficient in interferon-a/p and y receptors] were obtained from B&K Universal (UK). The mice were bred and maintained under specific pathogen-free conditions in individually ventilated cages. 3-4 weeks old AG129 mice were injected intraperitoneal with about 100 pl and about 300 pl immune serum obtained from vaccinated BAUB/c mice. After about 2 hours of passive immunization, the mice were subjected to DENV challenge with 1 X 10 4 pfu of DENV-2 strain through subcutaneous route. In pVAXl control group, the AG129 mice were passively immunized with serum from pVAXl control group of BAEB/c mice. Mice with DENV-2 virus challenge alone were kept as infection control.
  • the recombinant DNA sequence for prevention and/or treatment of one or more symptoms of pan Dengue viruses as set forth in the present invention also describes the efficacy and utility of said sequence to restore healthy functioning in humans and treat the conditions and disorders in humans as identified and described in this patent application.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Mycology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to relates to a DNA vaccine construct against Dengue virus. In particular, the present disclosure relates to a DENV (dengue) DNA vaccine construct, comprising at least one antigenic regions of envelope domain and at least one protein-coding region. The present disclosure also relates to a process for constructing a DNA vaccine construct and uses thereof.

Description

DENY EDIII-NS1 CONSENSUS SEQUENCE-BASED DENGUE DNA VACCINE
FIEED OF THE INVENTION
[0001] This disclosure relates to a DNA vaccine construct against Dengue virus. In particular, the present disclosure relates to a DENV (dengue) DNA vaccine construct, comprising at least one antigenic regions of envelope domain and at least one protein-coding region. The present disclosure also relates to a process for constructing a DNA vaccine construct and uses thereof.
BACKGROUND OF THE INVENTION
[0002] The DENV ED III (envelope domain III) region of Dengue has emerged as a vaccine target, as it is known to stimulate serotype-specific antibodies, and antibodies generated against this region are either not involved or minimally involved in antibody-dependent enhancement (ADE) of the infection (Fahimi, et al., 2018. Dengue viruses and promising envelope protein domain Ill-based vaccines. Appl. Microbiol. Biotechnol. 2018 1027 102, 2977-2996)Recent studies have found that EDIII based DENV vaccines could circumvent ADE (ADE of infection) of infection in mice, whereas T cell response against EDIII DNA vaccine has shown to play a role in disease protection through effective viral clearance (Ramasamy, et al. 2018. A tetravalent virus-like particle vaccine designed to display domain III of dengue envelope proteins induces multi- serotype neutralizing antibodies in mice and macaques which confer protection against antibody dependent enhancement in AG129 mice. PLoS Negl. Trop. Dis. 12; HW, et al. 2015. The Immunodominance Change and Protection of CD4+ T-Cell Responses Elicited by an Envelope Protein Domain Ill-Based Tetravalent Dengue Vaccine in Mice. PLoS One 10, e0145717-e0145717). These studies support the choice of EDIII for the next-generation vaccine development.
[0003] DENV non- structural protein 1 (NS1) has also emerged as an attractive vaccine target. NS1, a highly conserved protein among flaviviruses, is involved in virus replication and immune evasion (Chen, et al., 2018. Dengue virus non- structural protein 1: A pathogenic factor, therapeutic target, and vaccine candidate. J. Biomed. Sci. 25). However, it has also been reported to activate antibody Fc-mediated effector function and provide protection against flaviviruses (Wan, et al., 2014. Protection against dengue virus infection in mice by administration of antibodies against modified nonstructural protein 1. PLoS One 9). NS1 has been demonstrated to trigger T-cell responses in both experimental animals and humans (Grubor-Bauk, et al. 2019. NS1 DNA vaccination protects against Zika infection through T cell-mediated immunity in immunocompetent mice. Sci. Adv. 5, eaax2388). Previous studies have shown that NS1 induced T cell responses involved in protection against dengue infection. Furthermore, it is seen that vaccination with DENV-1, DENV-3, or DENV-4 NS1 offers protection against a heterologous DENV2 challenge (Beatty, et al., 2015. Dengue virus NS1 triggers endothelial permeability and vascular leak that is prevented by NS1 vaccination. Sci. Transl. Med. 7, 304ral41-304ral41; Lai, et al., 2017. Antibodies Against Modified NS1 Wing Domain Peptide Protect Against Dengue Virus Infection. Sci. Rep. 7).
[0004] DENV vaccine development is complicated by four antigenically diverse serotypes (DENV 1-4) and their intra-serotype (genotype) diversity at both local and global scales (Bhatt, et al. 2013. The global distribution and burden of dengue. Nature 496, 504-507). The genetic and antigenic differences between genotypes of the same serotype were not considered to impact long-term protective immunity and vaccine efficacy. However, several recent studies have challenged this assumption (A, M. et al., 2013. Lineage shift in Indian strains of Dengue virus serotype-3 (Genotype III), evidenced by detection of lineage IV strains in clinical cases from Kerala. Virol. J. 10; BP, et al., 2013. Circulation of different lineages of Dengue virus 2, genotype American/Asian in Brazil: dynamics and molecular and phylogenetic characterization. PLoS One 8)1. Studies have demonstrated that antigenic variation of genotypes can have a high impact on the breadth of antibody neutralization against different genotypes elicited by immune sera from people who have been exposed to natural infections or vaccination. Moreover, the limited efficacy of Dengvaxia against DENV2 points towards differences between the vaccine strain and disease-causing DENV in the clinical sites (Juraska, et al. 2018. Viral genetic diversity and protective efficacy of a tetravalent dengue vaccine in two phase 3 trials. Proc. Natl. Acad. Sci. U. S. A. 115, E8378- E8387). These findings suggest that while selecting vaccine strains, investigators and vaccine developers should consider circulating genotype variants.
[0005] US20210188947 discloses compositions comprising structurally modified DNA encoded antibodies (DMAbs), methods of structurally modifying DMAbs, and methods of using structurally modified DMAbs. [0006] WO2018/217897 discloses an immunogenic comprising an antigen and optionally IL- 12 and a method for administering the vaccine through intradermal electroporation.
[0007] WO2016034974A1 provides a recombinant polypeptide comprising the EDIII domain of each of Dengue virus serotype DENV-1, DENV-2, DENV-3, and DENV-4 linked to the N-terminal of HBsAg.
[0008] WO2017142831 Al relates to specific Dengue virus glycoprotein subunit E Dill variants and their uses in preventing and treating Dengue virus infection.
[0009] “Dengue Virus and Vaccines: How Can DNA Immunization Contribute to This Challenge? ” (https://www.frontiersin.org/articles/10.3389/fmedt.2021.640964/full) reports preclinical and clinical tests with DNA vaccines against the dengue virus.
[0010] “T Cell Responses Induced by DNA Vaccines Based on the DENV2 E and NS1 Proteins in Mice: Importance in Protection and Immunodominant Epitope Identification” (https://pubmed.ncbi.nlm.nih.gov/31333657/) emphasizes the importance of the T cell response involved in protection against dengue induced by E and NS1 based DNA vaccines.
[0011] “Recent Developments in Recombinant Protein-Based Dengue Vaccines ” (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6115509/) summarizes recent developments in recombinant protein based dengue vaccines that could lead to a good number of efficacious vaccine candidates for future human use and ultimately alternative dengue vaccine candidates.
[0012] The current DENV vaccine effort is also thought to be hampered by ADE, whereby cross-reactive antibodies against one serotype can enhance subsequent infection by heterologous serotype. To circumvent such issues, regions or motifs of the antigen responsible for causing ADE must be eliminated from the vaccine design. [0013] Hence, there is a need of a DENV vaccine candidate which is more adapted to the genotypes of dengue viruses found in India and Africa. An ideal DENV vaccine should be able to induce robust B cell and T cell immune response without causing ADE of the infection which are critical for vaccination, regardless of age and immune status.
[0014] DNA based vaccines is predicted to be a real game changer in India, Africa and other tropical regions where cold chain (storage and distribution of vaccines to health services) is a main concern. The low cost and thermostable aspect of the DNA vaccine should allow countries to drive vaccination programs on a larger scale than is currently possible, especially in middle- and low-income countries.
BRIEF DESCRIPTION OF THE ACCOMPANYING DRAWINGS
[0015] The invention herein will be better understood from the following detailed description with reference to the drawings, in which:
[0016] Figure 1 illustrates the DENV DNA vaccine (DDV) expression cassette.
[0017] Figure 2 shows DENV Serotype distribution across various geographical sites.
[0018] Figure 3 illustrates DENV amino acid variation in clinical isolates for all four serotypes.
[0019] Figure 4 shows DENV DNA vaccine construction.
[0020] Figure 5 shows antibody response induced by DENV DNA vaccination in BALB/c and C57BL/6J mice.
[0021] Figure 6 shows that characterization of cellular immune response induced by DENV DNA vaccine in mice.
[0022] Figure 7 shows the Th 1 -biased immune responses elicited by DDV. [0023] Figure 8 illustrates transcriptomic analysis of immune genes after vaccination with
DDV.
[0024] Figure 9 illustrates the capacity of anti-DDV immune sera to confer protection against lethal DENV-2 challenge in AG129 mice.
[0025] Figure 10 illustrates the genetic relationship of Indo-Africa DENV2 strains.
[0026] Figure 11 illustrates DENV amino acid variation among Indian strains of all four serotypes isolated from 2000-2020. (A, D, G and J).
[0027] Figure 12 shows the structural validation of the modelled constructs.
[0028] Figure 13 shows in-vitro antigen expression using monoclonal EDIII and NS1 antibodies.
[0029] Figure 14 shows DENV DNA vaccine (DDV) expression cassette for a variant of Dengue DNA vaccine construct of present invention.
SUMMARY OF THE INVENTION
[0030] The present invention provides a DNA based vaccine for pan dengue viruses. The invention discloses a DNA vaccine, wherein the antigenic regions DENV EDIII of dengue virus serotypes 1-4 and DENV-2 NS1 are incorporated in the vaccine construct, such that the DNA vaccine induces robust B cell and T cell immune response without causing ADE of the infection. The present invention also provides a process for constructing a DNA vaccine by incorporating EDIII and NS1 in the vaccine design. The present invention further provides a pharmaceutical composition; pharmaceutical combination and a kit comprising DNA based vaccine for pan dengue viruses. The invention furthermore provides a method of treatment and/or prevention of one or more symptoms of dengue viruses in a subject in need thereof.
[0031] Vaccination of mice with DDV induces pan-serotype neutralizing antibodies and antigen-specific T cell responses. Assaying of intracellular IFN-y staining, immunoglobulin IgG2(a/c)/IgGl ratios and immune gene profiling suggest a strong Th 1 -dominant immune response. Finally, passive transfer of DDV immune serum protected AG129 mice challenged with a virulent, non-mouse adapted DENV-2 strain. The surprising findings of preset invention collectively suggest an alternative strategy for dengue vaccine design; and offers a novel vaccine candidate with a possible broad spectrum protection and successful clinical translation either as a stand-alone or in a mix and match strategy. Further, the present disclosure describes a process for constructing a DNA vaccine by incorporating EDIII and NS1 in the vaccine design.
[0032] In one of the embodiments, the present invention provides a recombinant DNA vaccine construct comprising at least one antigenic region of envelope domain and at least one protein-coding region of dengue virus.
[0033] In another embodiment, the present invention provides that the protein-coding region in the recombinant DNA vaccine construct is a non- structural protein-coding region.
[0034] In yet another embodiment, the present invention provides that the DNA vaccine construct comprises sequences selected from SEQ. ID NO 1 or SEQ. ID NO 3 or a variant thereof.
[0035] In a further embodiment, the present invention provides that the variant of DNA vaccine construct is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 99% identical to the SEQ. ID NO 1 or SEQ. ID NO 3.
[0036] In still another embodiment, the present invention provides that the antigenic region of envelope domain of dengue virus (DENV) of DNA vaccine construct comprises the envelope domain III of serotypes 1-4.
[0037] In a further embodiment, the present invention provides that the protein-coding region of dengue virus in DNA vaccine construct is dengue-2 non- structural protein 1 (NS1).
[0038] In yet another embodiment, the present invention provides that recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non-structural protein 1 (NS1). [0039] Another embodiment of the present invention provides a method for preparing a recombinant DNA vaccine construct comprising steps of:
(a) preparing dengue (DENV) expression cassette comprising recombinant DNA vaccine construct wherein said vaccine construct comprises at least one antigenic region of envelope domain and at least one protein-coding region of dengue virus; and
(b) inserting the DENV expression cassette into a modified expression vector to obtain said DNA vaccine construct.
[0040] In yet another embodiment, the present invention provides that the step of inserting the DENV expression cassette into a modified expression vector in the method for preparing a recombinant DNA vaccine construct comprises incorporating the DENV DNA expression cassette into a modified pVAXl expression vector between Nhel and Hindlll under the control of cytomegalovirus immediate early promoter to result in recombinant vector for the DENV DNA vaccine.
[0041] In a further embodiment, the present invention provides that the method for preparing a recombinant DNA vaccine construct further comprises the step of optimizing the DNA vaccine construct by codon optimisation for optimal protein expression.
[0042] In still another embodiment, the present invention provides that in the method for preparing a recombinant DNA vaccine construct the recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non- structural protein 1 (NS1).
[0043] In another embodiment, the present invention provides that in the method for preparing a recombinant DNA vaccine construct the recombinant DNA vaccine construct comprises sequences selected from SEQ. ID NO 1 or SEQ. ID NO 3 or a variant thereof.
[0044] In a further embodiment, the present invention provides a method of preparation of DENV expression cassette comprises the steps of:
(i) integrating EDIII consensus sequence information from circulating genotypes of each DENV serotype in a single construct with at least one cleavage site for proteolytic enzyme between said EDIII consensus sequences; (ii) linking NS1 consensus sequence information from one of the DENV serotype by at least one cleavage site for proteolytic enzyme; and
(iii) optimizing consensus sequences for ED III and NS1 expression obtained in steps (i) and (ii) to obtain said DENV expression cassette.
[0045] In yet another embodiment, the present invention provides that in the method of preparation of DENV expression cassette the DENV serotype in step (i) is selected from serotypes DENV-1, DENV-2, DENV-3, and/or DENV-4.
[0046] In still another embodiment, the present invention provides that in the method of preparation of DENV expression cassette the DENV serotype in step (ii) is DENV-2.
[0047] In another embodiment, the present invention provides that in the method of preparation of DENV expression cassette the proteolytic enzyme is furin.
[0048] In a further embodiment, the present invention provides that the method of preparation of DENV expression cassette further comprises the step of adding Kozak sequences and IgE leader sequence upstream to the DENV expression cassette.
[0049] In one of the embodiments, the present invention also provides an isolated nucleic acid molecule encoding the recombinant DNA vaccine construct of the present invention.
[0050] In another embodiment, the present invention provides an expression vector comprising the nucleic acid molecule of the present invention.
[0051] In yet another embodiment, the present invention provides a host cell comprising the expression vector of the present invention.
[0052] In still another embodiment, the present invention provides a pharmaceutical composition or combination for treatment and/or prevention of one or more symptoms of dengue viruses in a subject in need thereof, said composition comprising about 1 pl to 1000 pl of recombinant DNA vaccine construct along with pharmaceutically acceptable excipients.
[0053] In a further embodiment, the present invention provides a pharmaceutical composition or combination for inducing immunogenicity against dengue viruses in a subject in need thereof, wherein said subject is administered with 1 |jg to 100 |ag of recombinant dengue DNA vaccine construct of the present invention.
[0054] In an embodiment, the present invention provides a method of preparing pharmaceutical composition or combination for treating and/or preventing one or more symptoms of dengue virus in a subject in need thereof comprising the steps: a) adding about 1 pl to 1000 pl of recombinant DNA vaccine construct at suitable conditions to one or more pharmaceutically acceptable excipients to obtain a mixture; b) subjecting the mixture obtained in step ‘a’ to suitable conditions to obtain the formulation or combination in desired dosage form.
[0055] In another embodiment, the present invention provides a method of treating and/or preventing one or more symptoms of dengue viruses in a subject in need thereof, comprising administering to the subject a composition or combination comprising about 1 pl to 1000 pl of recombinant DNA vaccine construct of the present invention.
[0056] In still embodiment, the present invention provides use of a composition or combination comprising recombinant DNA vaccine construct of the present invention for treating and/or preventing one or more symptoms of dengue viruses in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of said composition or combination.
[0057] In yet another embodiment the present invention provides a kit comprising a recombinant DNA vaccine construct of the present invention, pharmaceutical composition or combination of the present invention and instructions for administration of the pharmaceutical formulation and/or combination to a subject in need of treatment and/or prevention of dengue infection.
[0058] The foregoing description and following detailed description are exemplary and explanatory and are intended to provide further explanation of the invention. Other embodiments, aspects, advantages, and features will be readily apparent to those skilled in the art from the following detailed description of the invention. DETAILED DESCRIPTION OF THE INVENTION
[0059] The present invention relates to a DNA based vaccine for pan dengue viruses. In particular, the present disclosure relates to a DENV DNA vaccine, wherein the antigenic regions DENV EDIII of dengue virus serotypes 1-4 and DENV-2 non- structural protein 1 (NS1) are incorporated in the vaccine construct, such that the DNA vaccine induces robust B cell and T cell immune response without causing ADE of the infection.
[0060] This DENV DNA vaccine addresses the following four major dengue vaccine issues:
[0061] Selecting circulating dengue genotypes for vaccine design following large scale sequencing in India and published data from east Africa.
[0062] Consensus sequence selection to cover high frequency variants in the given region.
[0063] Targeting EDIII region of all four serotypes to avoid ADE of the infection and incorporating NS1 region to the vaccine as an additional target for cellular immunity to broaden the immune response generated by this vaccine.
[0064] Optimized DNA for delivery.
[0065] The DENV EDIII-based DNA vaccine (DDV) of the present disclosure was developed by integrating consensus sequence information from circulating genotypes of each serotype instead of a single strain genome sequence; incorporating NS1 protein coding region to induce broad T cell responses; and then optimizing DNA vaccine construct for optimal protein expression.
[0066] Consensus vaccine antigens were derived from strains which are circulating in India and Africa. The consensus gene sequences were constructed using the predicted consensus sequences from sequences obtained from the study (DENV1 n =40, DENV2 n =48, DENV3 n =22, DENV4 n =9, where n is the number of sequences) and Indo-Africa specific sequences available in the Virus Pathogen database and analysis resource (ViPR). Further, Aliview and Geneious tools were used to align and select the consensus amino acid sequence. A consensus was generated from the most frequent residues at each site. Total sequences employed in the generation of the vaccine construct are; DENV1: 182, DENV2: 406, DENV3: 131, DENV4: 61.
[0067] The present disclosure also describes a process for constructing a DNA vaccine by incorporating EDIII and NS1 in the vaccine design.
[0068] In an embodiment, the DENV 1-4 EDIII and NS1 gene of DENV2 were linked together in a single construct with furin cleavage sites between the individual genes. Consensus sequences were optimized for EDIII and NS1 expression, including codon and RNA optimization. The Kozak sequences and IgE leader sequence were added upstream to the DNA sequences as were furin cleavage sites to facilitate EDIII and NS1 processing. Finally, the synthetic DENV DNA expression cassette was inserted into the modified pVAXl expression vector (Source: Invitrogen, CA, synthesized commercially at Genscript) between Nhel and Hindlll under the control of the cytomegalovirus immediate early promoter (Source: Genscript Biotech, USA) to result in the recombinant vector for the DENV DNA vaccine. Figure 1 illustrates the DENV DNA vaccine expression cassette (SEQ. ID NO 1 and SEQ. ID NO 2).
[0069] The nucleotide sequence of the recombinant DENV DNA vaccine sequence is set forth as SEQ. ID NO 1 in Table A below.
Table A: List of nucleotide sequence of present invention
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
[0070] The peptide sequence encoded by the DENV vaccine expression cassette is set forth as SEQ. ID NO 2 in Table B below. Table B: List of peptide sequence of present invention
Figure imgf000016_0002
Figure imgf000017_0001
[0071] The DENV DNA vaccine of the present disclosure could be alternative strategy for traditional vaccines. The vaccine design enables fast tracked vaccine production and eliminates much of the logistics associated with vaccine distribution. Moreover, the low cost and thermostable aspect of the DNA vaccine should allow countries to drive vaccination programs on a larger scale than is currently possible, especially in India and Africa.
[0072] It has been demonstrated that immunization with DENV DNA vaccine resulted in robust neutralizing antibody responses against all four serotypes simultaneously and passive transfer of vaccinated immune sera confer protection against lethal DENV challenge. In addition, the DNA vaccine induces multifunctional antigen- specific T cell responses in murine models.
[0073] The recombinant DNA sequence of present invention can be a variant of SEQ. ID NO 1 or SEQ. ID NO 3. The variant is a functionally active variant and may be obtained by changing sequence of SEQ. ID NO 1 or SEQ. ID NO 3 and is characterized by having a biological activity similar to that displayed by nucleotide sequence of SEQ. ID NO 1 or SEQ. ID NO 3 from which the variant is derived. The variant includes ability of recombinant DNA sequence for treatment; prevention and/or amelioration of one or more symptoms of pan Dengue viruses in a subject in need thereof. The functionally active variant of SEQ. ID NO 1 or SEQ. ID NO 3 may be obtained by sequence alterations in sequence of SEQ. ID NO 1 or SEQ. ID NO 3, wherein the peptide with the sequence alterations retains function of the unaltered peptide. Such sequence alterations can include, but are not limited to, (conservative) substitutions, deletions, mutations and insertions. The variant can comprise at least 70% of the sequence of SEQ. ID NO 1 or SEQ. ID NO 3, at least 75% of the sequence of SEQ. ID NO 1 or SEQ. ID NO 3, at least 80% of the sequence of SEQ. ID NO 1, preferably at least 85%, still more preferably at least 90%, even more preferably at least 95% and most preferably at least 97%, 98% or 99%. The variant is derived from the SEQ. ID NO 1 or SEQ. ID NO 3 by at least one amino acid substitution and/or deletion, wherein the functionally active variant has a sequence identity to SEQ. ID NO 1 or SEQ. ID NO 3 of at least 80%, more preferably at least 85%, still more preferably at least 90%, even more preferably at least 95% and most preferably at least 97%, 98% or 99%. The variant of SEQ. ID NO 1 or SEQ. ID NO 3 is functionally active in the context of the present invention, if the activity of the variant amounts to at least 10%, preferably at least 25%, more preferably at least 50%, even more preferably at least 70%, still more preferably at least 80%, especially at least 90%, particularly at least 95%, most preferably at least 99% of the activity of SEQ. ID NO 1 or SEQ. ID NO 3 without sequence alteration. The activity of the variant may be determined or measured as described in the examples and then compared to that obtained for SEQ. ID NO 1 or SEQ. ID NO 3 of the present invention.
[0074] In the detailed description, reference is made to the accompanying drawings that form a part hereof, and in which is shown by way of illustration specific embodiments, which may be practiced. These embodiments are described in sufficient detail to enable those skilled in the art to practice the embodiments, and it is to be understood that other embodiments may be utilized without departing from the scope of the embodiments. The invention is described in connection with such embodiments, but the invention is not limited to any embodiment. While only certain features of the invention have been illustrated and described herein, many modifications and changes will occur to those skilled in the art.
[0075] The foregoing description of the specific examples will so fully reveal the general nature of the examples herein that others can, by applying current knowledge, readily modify and/or adapt for various applications without departing from the generic concept, and, therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed examples. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not for limitation. Therefore, while the examples herein have been described in terms of preferred examples, those skilled in the art will recognize that the examples herein can be practiced with modification within the spirit and scope of the embodiments.
EXAMPLES:
[0076] Example 1: Obtaining Samples, Ethical consideration, and DENY Viral DNA sequencing
In the present study, the genetic diversity of circulating DENV strains in India was investigated, and a DNA vaccine was designed based on circulating DENV genotypes and evaluated the immunogenicity of this vaccine candidate in two murine models. A retrospective analysis of 1000 archived serum/plasma samples from NS1 positive patients was performed, with dengue clinical presentation collected from 2012-2018 from 4 hospitals in India. Whole-genome sequencing of dengue viruses was carried out from the serum of 319 patients.
[0077] Out of 319 samples sequenced, 120 whole-genome sequences as well as infected serotype infection of 217 patients were obtained. Among the 217 patients, 174 patients were infected with a single DENV serotype (mono-infection), and 43 patients had concurrent DENV co-inf ections. From the 174 patients, 68 (39.1%) of them typed as DENV1, 63 (36.2%) of them as DENV2, 29 (16.7%) of them as DENV3, and 14 (8%) of them as DENV4. The distribution of serotypes across various geographical sites/locations in the country, during the sample collection period is shown in (Figure 2A). This dengue surveillance data collectively suggest that India is endemic to all four serotypes therefore; an effective vaccine must be tetravalent.
[0078] Method: Serum/Plasma samples of DENV- NS1 positive patients were obtained from four hospitals in India viz. St. John’s medical college and hospital (SJRI), Bangalore, All India Institute of Medical Sciences (AIIMS), Jodhpur, Kasturba hospital for Infectious diseases (KHI), Mumbai, and All India Institute of Medical Sciences (AIIMS), Delhi during the years 2012-2018. The Institutional Ethical Clearance Review Boards approved the study of all institutions participating in this study. Informed consent was obtained from enrolled patients. Clinical information of patients whose samples were included in this study was recorded in a preformed proforma which included the patient's demographic details, signs, and symptoms, of illness, laboratory parameters, and treatment details.
[0079] DENY viral RNA sequencing: The phylogenetic-based genotype analysis of the present study revealed that all Indian DENV 1-4 strains belong to a single genotype within the serotypes, except DENV1 (Figure 2B to 2E). DENV-1, genotype III is the most dominant, apart from a few sequences of genotype I. DENV2 sequences belong to the cosmopolitan genotype, whereas DENV-3 and DENV-4 sequences belong to genotype III and I, respectively. The data show that two different DENV-2 lineages within the cosmopolitan genotype are propagating simultaneously in all four sites in this study. Both the lineages emerged in the middle of the 1980s. Most of the neighboring sequences are from Asian countries. However, one cluster in the cosmopolitan-b lineage that is found in Kenya (Figure 2C and Figure 10) and one sequence of a traveler from Ethiopia and Djibouti. All the Indian sequences of DENV3 genotype III cluster with other Asian countries. The only genotype I of DENV-4 is observed in India. All Indian DENV4-I sequences along with one sequence from Pakistan cluster separately within genotype I from other Southeast Asian sequences. These data highlight that even though there is an enormous intra-serotype variation recorded all over the world, India has distinct genotypes for all four DENV serotypes with prominent intermixing between neighboring countries. Based on the implications of the findings on region- specific genotypes, a vaccine design approach was proposed against the circulating strains.
[0080] Method: Serum/ Plasma was tested for dengue virus -specific immunoglobulin M (IgM) and IgG antibodies using Panbio Dengue IgM and IgG capture ELISA kits, respectively. Dengue infection was classified as primary or secondary, and further samples were categorized as per WHO 2009 guidelines. Viral RNA was extracted from about 150 pl of patient’s serum using QIAamp Viral RNA Mini Kit (Qiagen). Sequencing library preparation was performed with about 1.2 ng of normalized double stranded cDNA using the Illumina Nextera XT sequencing kit and sequenced on an Illumina Miseq platform, using 2 X 75 and 2 X 150 bp paired-end reads. Raw sequence reads were inspected for quality using FASTQC (https://www.bioinformatics.babraham.ac.uk/projects/fastqc/), reads were filtered and trimmed based on the quality score. Reads were then mapped to the reference sequences obtained from the RefSeq database (NC_001477(DENV-l), NC_001474 (DENV-2), NC_001475 (DENV-3), NC_002640 (DENV-4) using Geneious assembler (3 iterations and medium-low sensitivity). Assembled reads were checked manually for errors. Serotypes were assigned manually and coverage was assigned based on the following criteria: >10X coverage if the depth of the coverage at each base was >20 and breadth of the coverage >95%, 5X-10X coverage if depth <20 and breadth>95, IX if depth<10 and breadth>90, for less coverages serotype. Consensus sequences were generated from the samples having >5X coverage based on the majority rule and were clipped to the length of reference sequences, iqtree v 1.6.10 was used to construct maximum likelihood trees with 1000 bootstrap replicates. All available DENV complete coding nucleotide sequences (DENV-1 n = 1800, DENV-2 n = 1395, DENV-3 n = 823, DENV-4 n = 220) from human host, were used for tree construction. Sylvatic strains EF457905 (for DENV-1), EF105379 (for DENV-2), KT424097 (for DENV- 3) and JF262779-80 (for DENV-4) were used as outgroups to root the tree. Genotypes were assigned to our sequences based on their positions in the tree. Figtree vl.4 was used to visualize the tree.
[0081] Example 2: DENV EDIII genetic diversity
The E protein, primarily Dill domain of the E protein, stimulates host immune responses by evoking protective and neutralizing antibodies. Mutations in the EDIII region could potentially impact the neutralization of DENV and the host-receptor interaction. Hence, the EDIII protein diversity in global strains and how the present study’s sequences are divergent from reference strains, was investigated. To assess antigenic differences between the EDIII of DENV 1-4 strains from distinct parts of the world, a neighbor-joining tree was generated and representative genotype variants were selected from each branch to comprehensively represent global diversity within the serotype. A sequence alignment of these proteins was performed and their EDIII genetic diversity relative to the respective reference strains was compared. Variable sites were designated when at least one virus showed an amino acid change at any of the 103 amino acid positions in the alignment. While DENV genotypes are closely related, considerable genetic variation was observed across genotypes of the same serotype in the EDIII region with a range of 1.62-5.83%.
[0082] Table 2: EDIII diversity in DENV 1-4 genotype variants
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000022_0002
[0083] Further, EDIII amino acid sequences of DENV1-4 clinical isolates from the study were compared for their similarity with the wild-type DENV strains. Even though India has its unique genotypes, considerable genetic variations were found in the EDIII region across genotypes of the same serotype. Alignment of the EDIII amino acid sequences revealed 9, 7, 2, and 8 sites of variation in DENV-1, DENV-2, DENV-3, and DENV-4 among Indian genotypes, respectively (Figure 3 A). Among all four strains, DENV-1 exhibited the highest EDIII diversity with a median of 4.85% (range, 2.91 %-5.83%), followed by DENV-4 and DENV-2 with 2.91% (range, 2.91%-3.88%) and 1.94% (range, 0.97 %-3.88%), respectively. The percentage diversity of DENV-3 EDIII sequences was found to be limited, ranging from 0% to 0.97 %. The frequency of amino acid variants was calculated across strains and expressed as a percentage (Figure 3B, 3D, 3F and 3H). These mutations were also spotted on the EDIII PDB structure (Figure 3C, 3E, 3G and 31). Additionally, several highly variable sites were identified across serotypes. A site was considered highly variable when greater than 50% of the study isolates showed a mutation at that position. The number of highly variable sites in EDIII was 5, 3, 2, and 1 in DENV-1, DENV-4, DENV-2, and DENV-3 strains, respectively. In addition to the study sequences, EDIII mutations in all Indian DENV1-4 strain sequences deposited in the ViPR database were investigated. Figure 11 shows amino acid variants and their frequency, implicating the EDIII amino acid residues as sites under immune pressure.
[0084] Method: Indian DENV sequences specific to the serotypes, including those sequenced during the study, were retrieved from the ViPR (Virus Pathogen Database and Analysis Resource) database. Sequence alignment was performed using MUSCLE and visualized in AliView. EDIII domain sequences were retained and used for downstream diversity analysis. EDIII sequences were compared pairwise against respective DENV reference strains and analyzed for Percentage diversity (Diversity %) and Percentage identity (Identity%). Diversity% was given by: (no. of variable sites/ EDIII length)* 100 while Identity % was calculated as (100 - Diversity%).
[0085] Example 3: Dengue DNA vaccine construction
DNA based vaccines would be a real game changer in India, Africa and other tropical regions where cold chain is a main concern. The DENV DNA vaccine candidate of the present disclosure is more adapted to the strains of dengue viruses found in India and Africa. In the present study, a new DNA vaccine candidate was generated by combining in tandem envelope protein domain III of dengue virus serotypes 1-4 and a DENV-2 NS1 protein coding region. Each domain was designed as a serotype -specific consensus coding sequence derived from different genotypes based on whole genome sequencing of clinical isolates in India (aforementioned study) and complemented with data from Africa that were retrieved from the ViPR database. Generated consensus sequences were codon and RNA optimized, synthesized commercially, cloned into pVAXl expression vector and the generated plasmid was designated as DDV (Dengue DNA Vaccine) (Figure 4A and 4B).
[0086] It is also noteworthy that approximately 26-50% of Indian DENV 1-4 strains exhibited 100% identity with EDIII sequences represented in DDV. The remaining Indian sequences, for all the serotypes, exhibited greater than 93% identity (Table 3).
[0087] Table. 3: Comparison of multiple EDIIIs within each serotype with the EDIII consensus sequences used to design DENV DNA vaccine.
Figure imgf000023_0001
Figure imgf000024_0001
[0088] DENV EDIII sequences used in designing DDV were aligned with the global dengue sequences for each serotype (n=1000) in the VIPR database to obtain the range of percent identity within members of the given serotype.
[0089] Furthermore, DDV has 100% identity with African DENV-2 and DENV-3 strains, while African DENV-1 and DENV-4 strains exhibited >96% identity with their corresponding serotype DDV sequences. DDV also shares >95.15 identities with the EDIII of the top 1000 international dengue sequences of the cognate serotype in the ViPR database.
[0090] Method: The polyvalent DENV DNA vaccine construct encodes EDIII of all four serotypes and NS1 sequence of DENV-2. The consensus gene sequences were constructed using the predicted consensus sequences from sequences obtained from our study (DENV-1 n =40, DENV-2 n =48, DENV-3 n =22, DENV-4 n =9) and Indo-Africa specific sequences available in the ViPR. The Aliview and Geneious tools were used to align and select the consensus amino acid sequence. A consensus was generated from the most frequent residues at each site. Total sequences employed in generation of the vaccine construct are; DENV-1: 182, DENV-2: 406, DENV-3: 131, DENV-4: 61. Due to the fact that African DENV-2 and DENV-3 genotypes are identical to Indian genotypes, these sequences were pooled directly with Indian DENV2 and 3 strains for consensus generation. Africa DENV1 and 4 genotypes differ from those in India, hence consensus sequences were generated separately and compared to Indian DENV-1 and DENV-4 strains represented in the vaccine expression cassette. African DENV-1 and DENV-4 strains exhibited >96 % identity with their respective serotype DDV sequences.
[0091] The DENV DNA vaccine expression cassette was designed with the consensus sequences of the EDIII gene of all four serotypes and consensus sequences of the NS1 gene of DENV-2 linked together in a single construct with furin cleavage sites between the individual genes. Consensus sequences were optimized for EDIII and NS1 expression, including codon and RNA optimization. The Kozak sequences and IgE leader sequence were added upstream to the DNA sequences as were furin cleavage sites to facilitate EDIII and NS1 processing. Finally, the synthetic DENV DNA expression cassette was inserted into the pVAXl expression vector between Nhel and Hindlll under the control of the cytomegalovirus immediate early promoter (Genscript Biotech, USA) (deposition to the Microbial Type Culture Collection is under process).
[0092] Example 4: Construct optimization by EDIII rearrangements
A variant of Dengue DNA vaccine construct of present invention: The inventors of present invention also optimized Dengue vaccine construct by changing the order of EDIII segments to DENV-3, DENV-4, DENV-1 and DENV-2, followed by DENV-2 NS1. The EDIII of DENV 1-4 and NS1 of DENV-2 sequences have been further expanded to contain all global strains of dengue and have been constructed irrespective of genotype. Additionally, P2A cleavage sites from porcine echovirus were incorporated between DENV EDIII 1-4 and NS1 segments to promote equimolar production of these antigens. The generated consensus sequences were codon- optimized and RNA-optimized, synthesized commercially, and cloned into nanoplasmid expression plasmids NTC9385R-eRNA41H-CpG and NTC 9385R by Nature Technology, USA (Figure 14, SEQ. ID NO 3, SEQ. ID NO 4).
[0093] Example 5: In vitro antigen expression and Immunofluorescence assays
The encoded DENV EDIII and NS1 transgene expression was assessed at the RNA level in HEK293T cells [ATCC and NCOS, Pune] transfected with DDV. Using the total RNA isolated from the transfected 293T cells, the EDIII and NS1 mRNA expression was confirmed by qRT-PCR (Figure 4C). In vitro, EDIII and NS1 protein expression in HEK- 293T cells was measured by western blot using anti-DDV immune sera on cell lysates. Western blots of the lysates of HEK-293T cells transfected with DDV construct revealed bands near predicted molecular weights of ~11 kDa (Figure 4D) and ~48kDa for EDIII and NS1 (Figure 4E), respectively. The secreted DENV NS1 was detected in the culture supernatants of transfected HEK-293T cells. EDIII and NS1 protein expression were further validated using EDIII and NS1 monoclonal antibodies (Figure 13). In immunofluorescence studies, the EDIII and NS1 protein was detected in HEK-293T cells transfected with DDV and exhibit antigen staining of the expressed proteins mainly in the cytoplasm which suggested the immune reactivity of the encoded protein (Figure 4G). In summary, in vitro studies revealed the expression of antigens at both the RNA and protein levels after transfection of cell lines with the candidate vaccine construct DDV.
[0094] Method: HEK293T cells were transfected with DENV DNA Vaccine (DDV) using X-treme GENE HP DNA transfection reagent as per the manufacturer’s instructions. The transfected cell lysate and supernatants were collected about 36 hours of post-transfection, and antigen expression was confirmed by western blot analysis. Cells were washed with phosphate-buffered saline (PBS) and lysed with NP40 supplemented with protease inhibitor cocktail (Roche) and ImM PMSF(Sigma) was used to make cell lysates. Protein lysates were separated on SDS-polyacrylamide gel, transferred onto a nitrocellulose membrane (Bio-Rad), and blocked for about 1 hour in about 5% skimmed milk. Subsequently, membranes were incubated in mouse antisera (1:200 dilution) against DDV. Secondary antibodies conjugated to horseradish peroxidase (HRP) were used at a dilution of about 1:1500. After washing with PBS/PBST the blots were developed using an enhanced chemiluminescence system (Thermo Fisher).
[0095] For Immunofluorescence, about 105 cells were plated on a coverslip. The next day, cells were fixed in ice-cold methanol, permeabilized with 0.1% Triton X-100 for 10 min, followed by blocking with about 1% BSA for about 30 minutes at room temperature (RT). Permeabilized cells were then incubated with antisera (1:100) for about 1 hour at RT, washed about three times with IX PBS, followed by incubated with goat anti-mouse IgG-AF488 at about 37°C for about 30 minutes. Cells were again washed about three times with IX PBS, mounted (Prolong gold antifade Mountant with DAPI, Invitrogen), air-dried, and visualized using an FV1000 confocal microscope.
[0096] Example 6: Immunization of mice (BALB/c and C57BL/6J with DENV DNA vaccine construct with electroporation and analysis of DENV binding antibody titer DENV specific humoral responses following vaccination were characterized in two different murine strains, BALB/c and C57BL/6J (The Jackson Laboratory, USA)). Animals were obtained from The Jackson Laboratory, USA. The mice were bred and maintained under specific pathogen-free conditions in individually ventilated cages at NCBS. Mice (n=6) were vaccinated three times, two weeks apart, with about 50 pg of the DNA vaccines or control pVAXl l plasmid vector using Tibialis anterior (TA) muscle delivery. Vaccinated mice were bled at day 0 and two weeks after each vaccination to obtain sera, which were assayed for the presence of DENV antibodies by enzyme-linked immunosorbent assay (ELISA) against recombinant protein as a capture protein (Figure 5A). Binding antibody ELISA data revealed that the DDV induced DENV specific antibody responses. The results showed that all mice developed anti-DENV antibodies after a single immunization. The anti-dengue IgG responses were significantly increased after 1-2 booster immunizations, appearing to peak 2 weeks after the second booster in both BALB/c and C57BL/6J strains (Figure 5B and 5D). Comparison between the serum IgG endpoint titers of DDV and plasmid control groups in both murine models showed robust titers elicited by DDV immunization. The endpoint tiers ranged from 1:1000 to 1:500000 in individual animals (Figure 5C and 5E). These findings demonstrated the ability of the DENV DNA vaccine construct to express in mammalian cells potentially and the antibodies induced by these constructs were able to react with the Dengue vaccine antigens.
[0097] Method: To determine the immunogenicity of the DENV DNA vaccine constructs, mice were immunized with about 50 pg of DNA in a total volume of about 50 pl of sterile water by a syringe into the anterior tibialis (TA) muscles then electroporated using BTX ECM 830 with 8 square 40-V electric pulses in alternating direction with a time constant of about 0.05 s and an inter-pulse interval of about Is. Each group received 2 booster doses at about 2-week intervals, and mice were euthanized about two weeks following the last immunization. All the experimental procedures were approved by the Institutional Animal Ethics Committee (IAEC) of National Centre for Biological Sciences (NCBS) and Theralndx Lifesciences Pvt. Ltd.
[0098] Example 7: Flow cytometry-based neutralization test (FNT)
Flow-based virus neutralization assay was performed in U937-DC-SIGN cells (U937-DC- SIGN cell line related assays were outsourced at the Bioassay Lab, THSTI, Faridabad) to assess the levels of anti-DDV immune sera-induced neutralizing antibody titers against laboratory DENV strains. DDV-vaccinated mice immune sera showed a clear neutralizing antibody titer against all four serotypes simultaneously; the median FNT50 titers against DENV 1-4 ranged from 182-3500 (Figure 5F and 5G). As per WHO recommendation for DENV vaccines, the neutralizing potency of anti-DDV immune sera against DENV 1-4 recent clinical isolates was investigated. DENV 1-4 serotypes were isolated from DENV NS1 positive patients in India and FNT was performed. The data showed anti-DDV immune sera effectively neutralized clinical isolates as well and FNT50 titers against DENV 1- 4 ranged from 150-900 (Figure 5H and 51). These data provide evidence that DDV-induced neutralizing antibody responses cover major currently circulating strains as well.
[0099] Method: The flow cytometry-based neutralization assays were performed in triplicate in 96-well cell culture plates with flatbottom wells. Each well contained 5 X 104 DCSIGN- expressing U937 cells. Immune sera were serially diluted, and the virus was pre-incubated with the sera for about 1 hour at about 37°C. The cells were washed, and virus and serum mixture was added to the cells for about 1 hour at about 37°C. Next, the wells were filled with cell culture medium, and the plates were incubated for about 24 to 48 hours at about 37°C in about 5% CO2. The cells were prepared for flow cytometry analysis by washing them in phosphate-buffered saline and transferring them to 96-well plates with round-bottom wells. The cells were fixed and permeabilized by using a Cytofix/ Cytoperm kit (BD-PharMingen, San Diego, CA) and stained with monoclonal antibody 4G2, a monoclonal antibody that recognizes the flavivirus E protein. The cells were analyzed with a FACScan flow cytometer. The serum dilution that neutralized 50% of the viruses was calculated by nonlinear, doseresponse regression analysis with Prism 4.0 software (GraphPad Software, Inc., San Diego, CA).
[00100] Example 8: IFN-y Enzyme-Linked Immunosorbent Spot (ELISPOT) Assay
DENV specific cellular responses were assessed in vaccinated animals by ELISPOT. A total of 95 and 344 EDIII and NS1 specific T cell peptides, respectively, were identified in the DENV DNA vaccine construct. MHC class I binding predictions, peptide selection, and antigenicity of those peptides were analyzed by NetCTL 1.2 and Vaxijen 2.0. A total of 15 CTL epitopes (9 mer peptides) were screened out of which 11 were found to possess antigenicity and were chosen for synthesis (Figure 6B). T cell response against dengue antigens via IFN-y ELISPOT was assayed. IFN-y has been described as a mediator of T cell responses and plays a distinctive role in antiviral activity against dengue viruses. Mice were immunized as before. Two weeks after the second booster DDV or pVAXl plasmid control immunized mice were euthanized and splenocytes were isolated (Figure 6A). Single-cell suspensions were stimulated with the peptide pools (Pool 1- EDIII peptide mixture; Pool 2- NS1 peptide mixture; Pool 3- EDIII and NS1 peptide mixture) number of IFN-y producing CD8+ T cells was analyzed. Results show that both EDIII- and NS1 specific cellular responses (presented as IFN-y SFU/cells) were detected by ELISPOT in DDV vaccinated animals while negligible spots were detected in the plasmid vector-vaccinated animals (Figure 6C to 6F). PMA/IONOMYCIN was used as a non-specific positive control. As expected, stimulation with PMA/IONO in all ELISPOT assays performed with cells from DENV vaccinated or control animals induced a high IFN-y response.
[00101] Method: ELISpot was performed with the Mouse IFN-y ELISpot BASIC Kit (Mabtech). In brief, freshly isolated 0.5M splenocytes/animal were plated into polyvinylidene fluoride (PVDF)-coated 96-well plates containing IFN-y capture antibodies. Cells were stimulated with immunogenic 9-mer T cell peptides predicted from DENV1-4 ED III and NS1 antigens. Negative control wells contained no peptide. Phorbol myristate acetate (PMA) / lonomycin (IO) used as a positive control. After overnight stimulation, plates were washed and sequentially incubated with biotinylated IFN-y detection antibody (R4-6A2), streptavidin-ALP, and finally BCIP/NBT. Plates were imaged with ImmunoSpot Analyzer and quantified with ImmunoSpot software.
[00102] Example 9: Intracellular cytokine staining
Intracellular cytokine staining was performed using freshly isolated mouse splenocytes. The intracellular IFN-y in both CD8+ and CD4+ cells of mouse splenocytes in both vaccine and control groups of animals were analyzed. As a positive stimulus for T cell activation, PMA/Ionomycin were used. Exocytosis of cytokines was blocked by the addition of Brefeldin A (10 pg/ml) during stimulation. Cells were permeabilized, labeled, and fixed for flow cytometry. IFN-y CD8+ T cells and IFN-y CD4+ T cells were proportionately higher, as found using intracellular staining with flow cytometry, in DDV as compared to pVAXl vaccinated animals (Figure 61 and 6J). In summary, DENV DNA vaccine candidate DDV induces robust T-cell response in mice.
[00103] Method: Intracellular cytokine staining was performed by stimulating freshly isolated splenocytes with about 50 ng/mL PMA and about 500 ng/mL IO in the presence of Brefeldin for about 5-6 hours. After stimulation, surface staining of CD4, and CD8 was performed, followed by intracellular staining of IFN-y (Biolegend). Data acquisition was performed on a BD LSRFortessa and analyzed with FlowJo.
[00104] Example 10: DDV vaccination skewed a Thl-dominant response
DDV vaccination skewed a Thl-dominant response. Thl skewness has been shown to elicit a robust adaptive response in terms of cellular activation and antibody production. The induction of polyfunctional Thl cells is an essential element of a protective vaccine response. Respiratory disease viruses such as SARS-CoV and MERS-CoV vaccine development have highlighted the importance of Thl skewed response in mitigating the risk of vaccine-induced disease enhancement. Thus, the Thl/Th2 balance elicited by vaccination with DENV DNA vaccine was investigated. The IgG subclass fate of plasma cells is highly governed by Th cells. To determine whether DDV showed skewing of Thl over Th2 responses, the Thl- associated IgG subclasses-IgG2a (BAlB/c) (Figure 7B and 7C) and IgG2c (C57BL/6J) (Figure 7D and 7E)- against the Th2 associated IgGl were measured. C57BL/6J and BALB/c mice are prototypical Thl and Th2 animal strains, with C57BL/6J producing high IgG2c and BALB/c, mostly IgG2a. Hence, in C57BL/6J mice, evaluation of IgG2c and IFN-y is critical to correct interpretation of Thl immune responses. In both strains of mice, DDV vaccination- induced Thl skewed IgG subclass responses. These findings indicate that the DDV elicits Thl dominant immune responses.
[00105] Method: IgG and IgG subtype binding antibody titers by indirect ELISA:
DENV binding antibody titer was analyzed by indirect ELISA. 96 well plates (Thermo Scientific) were coated with recombinant protein in a coating buffer (0.1M NaHCOs) and incubated overnight at about 4 °C. The following day, plates were blocked with about 3% BSA in PBS for about 2 hours at room temperature. Triplicate samples of serially diluted plasma ranging from 1:100 to 1:5,00,000 were added to the plate and incubated for about 2 hours at room temperature or overnight at about 4 °C. After washing, secondary anti -IgG (Sigma), anti-IgGl(Invitrogen), and anti-IgG2a (Invitrogen) or IgG2c (Abeam) antibodies conjugated with horseradish peroxidase was added at 1:2000 (IgG, IgGl, IgG2a) and 1:5000 (IgG2c) dilution for about 1 hour at room temperature. The plates were then developed with TMB substrate (Sigma) for about 15-20 minutes. The reaction was stopped with a stop buffer (Invitrogen) and the optical density (O.D) measured at about 450 nm. Cut-off values for each dilution were set using the O.D of naive samples in the formula: naive O.D at a dilution + (2.5 * standard deviation). Starting from the lowest dilution, the sample dilution prior to the one which was exceeded by the cut-off was considered as the end titer value.
[00106] Example 11: Immune gene expression following DDV vaccination
To determine how the DDV exerts its immunogenicity, C57BL/6J mice were vaccinated thrice, two weeks apart, with about 50 ug of the DDV and compared these to equivalent doses of pVAXl vector control. Since immune responses develop in germinal centers in draining lymph nodes, the inguinal lymph nodes of mice at two weeks after 2nd booster were isolated and analyzed with the Nanostring V2 immunology panel (Figure 8A). Principal-component analysis (PCA) of immune gene expression showed clustering of responses to DDV distinct from pVAXl plasmid controls, indicating clear differences in immune gene expression following DDV vaccination (Figure 8B).
[00107] Differentially expressed genes were examined in the lymph nodes of mice injected with DDV compared with those administered a similar dose of pVAXl plasmid control (Figure 8D). Volcano plot analysis identified significant enrichment of various innate and adaptive immune responses, lymphocyte activation, cytokine, interferon signaling, and Class I antigen presentation genes in DNA vaccine immunized animals (Figure 8C). Some of the most highly expressed genes included CxcllO, Socs3, Ccl7, Plaur and Defbl which play roles in directing Thl and Th2 effector responses. These genes have also been linked to antigen presentation, innate and adaptive immune cell recruitment, T cell stimulation, and dendritic cell maturation in the context of host immunity. Furthermore, there are several antiviral defense and IFN-Thl responsive genes that were also activated upon DENV DNA vaccination. These include Stat2 (IFN signaling), Irf7 (IFN inducible genes expressed on Thl cells), Bst2 (development of antiviral T cell distribution and function in addition to augmenting DC activation), and CD 99 (Thl type cytokine response) (Figure 8E to 8J). In addition, several inflammatory signalling genes such as TGFBR1, VCAM1 and CD40L were downregulated in mice after the DDV vaccination (Figure 8K to 8L). These genes have been shown to contribute to inflammatory and autoimmune diseases and their downregulation following vaccination indicates the controlled immune response evoked by the DDV. These findings collectively suggest the development of the immune response in the inguinal lymph nodes of mice immunized with DDV.
[00108] Method: Mice were sacrificed about 2 weeks post-second immunization and lymph nodes were collected for analysis of genes involved in immune response. Lymph nodes were homogenized, and RNA was extracted with TRIzol LS. RNA (50 ng) from whole blood cells and lymph nodes were hybridized to the NanoString nCounter mouse inflammation and immunology v2 panels (NanoString Technologies), respectively. RNA was hybridized with reconstituted CodeSet and ProbeSet. Reactions were incubated for about 24 hours at about 65°C and ramped down to about 4°C. Hybridized samples were then immobilized onto a nCounter cartridge and imaged on a nCounter SPRINT (NanoString Technologies). Data were analyzed with nSolver Analysis software and PRISM. For normalization, samples were excluded when percentage field of vision registration was <75, binding density outside the range 0.1-1.8, positive control R2 value was <0.95, and 0.5 fM positive control was %2 SD above the mean of the negative controls. Background subtraction was performed by subtracting estimated background from the geometric means of the raw counts of negative control probes. Probe counts less than the background were floored to a value of 1. The geometric mean of positive controls was used to compute positive control normalization parameters. Samples with normalization factors outside 0.3-3.0 were excluded. The geometric mean of housekeeping genes was used to compute the reference normalization factor. Samples with reference factors outside the 0.10-10.0 range were also excluded. To identify differentially expressing genes (DEGs) between groups, Graph pad Prism software was used to analyze variance with a cutoff of p < 0.05. Log 2 fold changes generated were used for volcano plots constructed with Prism 5 software. DEGs were identified by a fold change cutoff of 1.5. Unsupervised PCA was performed to visualize variability between DDV and pVAXl control animals.
[00109] Example 12: Protective efficacy in AG129 mice
To assess the role of humoral immune response in mediating protection from DENV challenge, serum from BALB/c mice immunized with either plasmid control or DDV was passively transferred into AG129 mice. Groups of AG129 mice received anti-pVAXl sera at 300 pl per mouse or anti-DDV immune sera, at two dosage levels, 100 and 300 pl per mouse. All these mice were challenged about 2 hours after-passive transfer with a lethal dose of DENV-2 (105 FlU/mouse). The control group did not receive immune sera but were subjected to the lethal challenge dose. All groups were tracked for body weight changes, clinical signs, and survival for up to 14 day’s post-challenge (Figure 9A).
[00110] The DENV-inf ection-only group showed an initial increase in body weight; however, with a steep decrease from day 5 or 6. The maintenance of body weight or percentage increase were observed to be better in 300 pl immunized mice compared to DENV-challenged group and pVAXl control group (Figure 9B). The mice started showing noticeable symptoms from 3rd day post-infection starting with ruffled fur which continued to become more aggressive and prominent with the disease progression. Despite the initial rise in symptoms, clinical scoring showed reduced manifestation of the disease in the 300 pl immunized group at later stages compared to the infection control and pVAXl control. However, the 100 pl sera immunized group did not show any significant difference in clinical symptoms from both the DENV-challenge and pVAXl controls (Figure 9C). In the infection control group, the mortality starts on 6th day post-infection and a 100 % mortality was observed by 8th day post-infection (Figure 9D). The pVAXl control group showed a similar mortality pattern as the infection control group. On the other hand, in the 300pl sera injected group, three mice survived for at least 12-days post-infection out of the total six. The data from the survival curve indicate about a 50% better survival for the 300 pl sera vaccinated group compared to the controls. Immunization with 100 pl immune sera failed to provide any advantage in surviv ability compared to the infection and pVAXl control group.
[00111] Method: AG129 mice [deficient in interferon-a/p and y receptors] were obtained from B&K Universal (UK). The mice were bred and maintained under specific pathogen-free conditions in individually ventilated cages. 3-4 weeks old AG129 mice were injected intraperitoneal with about 100 pl and about 300 pl immune serum obtained from vaccinated BAUB/c mice. After about 2 hours of passive immunization, the mice were subjected to DENV challenge with 1 X 104 pfu of DENV-2 strain through subcutaneous route. In pVAXl control group, the AG129 mice were passively immunized with serum from pVAXl control group of BAEB/c mice. Mice with DENV-2 virus challenge alone were kept as infection control. After infection, each mouse was observed for development of clinical symptoms and body weight changes till mortality or 14th day post-infection. The clinical symptom scoring was done based on the following criteria: Score 1: ruffled fur; Score 2: 1+ hunched back; Score 3: 2+ slow movements or lethargy; Score 4: 3+ facial edema; Score 5: 4+ facial edema with closed eyes; Score 6: 5+hemorrhage (bloody stool) or limb paralysis; Score 7: death. All the experimental procedures were approved by the Institutional Animal Ethics Committee (IAEC) of Rajiv Gandhi Centre for Biotechnology (RGCB).
[00112] Statistical analysis: Immunological and virologic data analysis was performed using GraphPad Prism. Kruskal-Wallis or Chi-square was used for the clinical data analysis, with *P < 0.05 considered significant. Vaccination of mice with DDV induces pan-serotype neutralizing antibodies and antigen- specific T cell responses. Assaying of intracellular IFN-y staining, immunoglobulin IgG2(a/c)/IgGl ratios and immune gene profiling suggest a strong Th 1 -dominant immune response. Finally, passive transfer of DDV immune serum protected AG129 mice challenged with a virulent, non-mouse adapted DENV-2 strain. The findings of present invention collectively suggest an alternative strategy for dengue vaccine design; and offers a novel vaccine candidate with a possible broad-spectrum protection and successful clinical translation either as a stand-alone or in a mix and match strategy.
[00113] The recombinant DNA sequence for prevention and/or treatment of one or more symptoms of pan Dengue viruses as set forth in the present invention also describes the efficacy and utility of said sequence to restore healthy functioning in humans and treat the conditions and disorders in humans as identified and described in this patent application.
[00114] Although the subject matter has been described herein with reference to certain preferred embodiments thereof, other embodiments are possible. For illustrative purpose, the vaccine candidate specified in description has been employed for treatment and/or prevention of specific strains or serotypes of DENV. However, those skilled in the art would appreciate that scope of the invention would extend to use of the vaccine candidate to treat and/or prevent one or more strains or serotypes of the dengue virus, including DENV-1, DENV-2, DENV-3 and DENV-4 or other mosquito-bome viral infections known in the field of art. It will be obvious to those skilled in the art to make various changes, modifications and alterations to the invention described herein. To the extent that these various changes, modifications and alteration do not depart from scope of the present invention, they are intended to be encompassed therein.

Claims

We Claim:
1. A recombinant DNA vaccine construct comprising at least one antigenic region of envelope domain and at least one protein-coding region of dengue virus.
2. The recombinant DNA vaccine construct as claimed in claim 1, wherein the proteincoding region is a non- structural protein-coding region.
3. The recombinant DNA vaccine construct as claimed in claim 1 or 2, wherein said DNA vaccine construct comprises sequences selected from SEQ. ID NO 1 or SEQ. ID NO 3 or a variant thereof.
4. The recombinant DNA vaccine construct as claimed in claim 3, wherein said variant is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 99% identical to the SEQ. ID NO 1 or SEQ. ID NO 3.
5. The recombinant DNA vaccine construct as claimed in any one of claims 1 to 4, wherein said antigenic region of envelope domain of dengue virus (DENV) comprises the envelope domain III of serotypes 1-4.
6. The recombinant DNA vaccine construct as claimed in any one of claim 1 to 5, wherein said protein-coding region of dengue virus is dengue-2 non-structural protein 1 (NS1).
7. The recombinant DNA vaccine construct as claimed in any one of claim 1 to 6, wherein said recombinant DNA vaccine construct comprises at least one antigenic region of DENV ED III and at least one DENV non-structural protein 1 (NS1).
8. A method for preparing a recombinant DNA vaccine construct comprising the steps of:
(a) preparing dengue (DENV) expression cassette comprising recombinant DNA vaccine construct wherein said vaccine construct comprises at least one antigenic region of envelope domain and at least one protein-coding region of dengue virus; and
(b) inserting the DENV expression cassette into a modified expression vector to obtain said DNA vaccine construct.
33
9. The method as claimed in claim 8, wherein step (b) optionally comprises: incorporating the DENV DNA expression cassette into a modified pVAXl expression vector between Nhel and Hindlll under the control of cytomegalovirus immediate early promoter to result in recombinant vector for the DENV DNA vaccine.
10. The method as claimed in claim 8 or 9, further comprising the step of: optimizing the DNA vaccine construct by codon optimisation for optimal protein expression.
11. The method as claimed in any one of claim 8 to 10, wherein said recombinant DNA vaccine construct comprises at least one antigenic region of DENV EDIII and at least one DENV non-structural protein 1 (NS1).
12. The method as claimed in any one of claim 8 to 11, wherein said recombinant DNA vaccine construct comprises sequences selected from SEQ. ID NO 1 or SEQ. ID NO 3 or a variant thereof.
13. The method as claimed in any one of claims 8 to 12, wherein preparation of DENV expression cassette comprises the steps of:
(i) integrating EDIII consensus sequence information from circulating genotypes of each DENV serotype in a single construct with at least one cleavage site for proteolytic enzyme between said EDIII consensus sequences;
(ii) linking NS1 consensus sequence information from one of the DENV serotype by at least one cleavage site for proteolytic enzyme; and
(iii) optimizing consensus sequences for EDIII and NS1 expression obtained in steps (i) and (ii) to obtain said DENV expression cassette.
14. The method as claimed in claim 13, wherein said DENV serotype in step (i) is selected from serotypes DENV-1, DENV-2, DENV-3, and/or DENV-4.
15. The method as claimed in claim 13 or 14, wherein said DENV serotype in step (ii) is DENV-2.
16. The method as claimed in any one of claim 13 to 15, wherein said proteolytic/ enzyme is
34 furin. The method as claimed in any one of claim 13 to 16, further comprising the step of adding Kozak sequences and IgE leader sequence upstream to the DENV expression cassette. An isolated nucleic acid molecule encoding the recombinant DNA vaccine construct as claimed in any one of claims 1 to 7. An expression vector comprising the nucleic acid molecule as claimed in claim 18. A host cell comprising the expression vector as claimed in claim 19. A pharmaceutical composition or combination for treatment and/or prevention of one or more symptoms of dengue viruses in a subject in need thereof, said composition comprising about 1 pl to 1000 pl of recombinant DNA vaccine construct as claimed in any one of claims 1 to 7 along with pharmaceutically acceptable excipients. A pharmaceutical composition or combination for inducing immunogenicity against dengue viruses in a subject in need thereof, wherein said subject is administered with 1 pg to 100 pg of recombinant dengue DNA vaccine construct as claimed in any one of claims 1 to 7. A method of preparing pharmaceutical composition or combination for treating and/or preventing one or more symptoms of dengue virus in a subject in need thereof comprising the steps: a) adding about 1 pl to 1000 pl of recombinant DNA vaccine construct at suitable conditions to one or more pharmaceutically acceptable excipients to obtain a mixture; b) subjecting the mixture obtained in step ‘a’ to suitable conditions to obtain the formulation or combination in desired dosage form. A method of treating and/or preventing one or more symptoms of dengue viruses in a subject in need thereof, comprising administering to the subject a composition or combination comprising about 1 pl to 1000 pl of recombinant DNA vaccine construct as claimed in any one of claims 1 to 7. Use of a composition or combination comprising recombinant DNA vaccine construct as claimed in any one of claims 1 to 7 for treating and/or preventing one or more symptoms of dengue viruses in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of said composition or combination. A kit comprising a recombinant DNA vaccine construct as claimed in any one of claims 1 to 7, pharmaceutical composition or combination as claimed in any one of claims 21 and 22 and instructions for administration of the pharmaceutical formulation and/or combination to a subject in need of treatment and/or prevention of dengue infection.
PCT/IB2022/062891 2021-12-30 2022-12-29 Denv ediii-ns1 consensus sequence-based dengue dna vaccine WO2023126882A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN202141061753 2021-12-30
IN202141061753 2021-12-30

Publications (1)

Publication Number Publication Date
WO2023126882A1 true WO2023126882A1 (en) 2023-07-06

Family

ID=86998379

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2022/062891 WO2023126882A1 (en) 2021-12-30 2022-12-29 Denv ediii-ns1 consensus sequence-based dengue dna vaccine

Country Status (1)

Country Link
WO (1) WO2023126882A1 (en)

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
COSTA ET AL.: "Protection against dengue type 2 virus induced in mice immunized with a DNA plasmid encoding the non-structural 1 (NS1) gene fused to the tissue plasminogen activator signal sequence", VACCINE, vol. 24, no. 2, 12 January 2006 (2006-01-12), pages 195 - 205, XP028010432, DOI: 10.1016/j.vaccine.2005.07.059 *
MELLADO-SANCHEZ ET AL.: "A plasmid encoding parts of the dengue virus E and NS1 proteins induces an immune response in a mouse model", ARCH VIROL, vol. 155, 2010, pages 847 - 856, XP019853321 *
RAMANATHAN ET AL.: "Development of a novel DNA SynCon tetravalent dengue vaccine that elicits immune responses against four serotypes", VACCINE, vol. 27, no. 46, 30 October 2009 (2009-10-30), pages 6444 - 53, XP055571496, DOI: 10.1016/j.vaccine.2009.06.061 *
SANKARADOSS ARUN, JAGTAP SURAJ, NAZIR JUNAID, MOULA SHEFTA E., MODAK AYAN, FIALHO JOSHUAH, IYER MEENAKSHI, SHASTRI JAYANTHI S., DI: "Immune profile and responses of a novel dengue DNA vaccine encoding an EDIII-NS1 consensus design based on Indo-African sequences", MOLECULAR THERAPY, ELSEVIER INC., US, vol. 30, no. 5, 1 May 2022 (2022-05-01), US , pages 2058 - 2077, XP093078055, ISSN: 1525-0016, DOI: 10.1016/j.ymthe.2022.01.013 *

Similar Documents

Publication Publication Date Title
CN109152826B (en) Novel vaccine against Zika virus
JP2023524054A (en) Betacoronavirus prevention and treatment
EP4153730A1 (en) Sars-cov-2 vaccines
US11801299B2 (en) Compositions and methods for generating an immune response to hepatitis B virus
US20220054625A1 (en) Immunogenic composition
CN110234346A (en) The vaccine of new confrontation zika virus and the combination of the DNA antibody construct for fighting zika virus
WO2022071513A1 (en) IMPROVED DNA VACCINE FOR SARS-CoV-2
Cecílio et al. Vaccines for human leishmaniasis: where do we stand and what is still missing
JP2010506926A (en) Efficacy of Ii-Key enhancement vaccine
Sankaradoss et al. Immune profile and responses of a novel dengue DNA vaccine encoding an EDIII-NS1 consensus design based on Indo-African sequences
US20200172585A1 (en) Vaccine and methods for detecting and preventing filariasis
Hu et al. Heterologous prime-boost vaccination against tuberculosis with recombinant Sendai virus and DNA vaccines
AU2018365296B2 (en) A Zika virus chimeric polyepitope comprising non-structural proteins and its use in an immunogenic composition
Cabrera-Mora et al. Induction of multifunctional broadly reactive T cell responses by a Plasmodium vivax circumsporozoite protein recombinant chimera
CN115894708B (en) Human cytomegalovirus epitope chimeric peptide and application thereof
WO2023126882A1 (en) Denv ediii-ns1 consensus sequence-based dengue dna vaccine
Rigato et al. Immunization of neonatal mice with LAMP/p55 HIV gag DNA elicits robust immune responses that last to adulthood
JP5844403B2 (en) Plasmodium falciparum sporozoites and liver stage antigens
KR102401682B1 (en) Recombinant mycobacterium strain expressing sars-cov-2 antigen and vaccine composition comprising same
US20220332772A1 (en) Vaccine and methods for detecting and preventing filariasis
Perdiguero et al. Immunogenicity and efficacy of a novel multi-patch SARS-CoV-2/COVID-19 vaccine candidate
CA2846486C (en) Novel clostridium difficile dna vaccine
US20230089516A1 (en) Vaccine and Methods for Detecting and Preventing Filariasis
US20220023412A1 (en) Compositions Useful in Both Homologous And Heterologous Vaccine Regimens
Shchelkunov et al. Enhancing the Immunogenicity of Vaccinia Virus. Viruses 2022, 14, 1453

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: 22915349

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)