WO2018215766A1 - Compositions and methods for inducing an immune response - Google Patents

Compositions and methods for inducing an immune response Download PDF

Info

Publication number
WO2018215766A1
WO2018215766A1 PCT/GB2018/051399 GB2018051399W WO2018215766A1 WO 2018215766 A1 WO2018215766 A1 WO 2018215766A1 GB 2018051399 W GB2018051399 W GB 2018051399W WO 2018215766 A1 WO2018215766 A1 WO 2018215766A1
Authority
WO
WIPO (PCT)
Prior art keywords
mers
chadoxi
cov
composition
vaccine
Prior art date
Application number
PCT/GB2018/051399
Other languages
French (fr)
Inventor
Sarah C. Gilbert
Adrian V. S. Hill
Susan Jane MORRIS
Original Assignee
Oxford University Innovation Limited
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 Oxford University Innovation Limited filed Critical Oxford University Innovation Limited
Publication of WO2018215766A1 publication Critical patent/WO2018215766A1/en

Links

Classifications

    • 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
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • 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
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10044Chimeric viral vector comprising heterologous viral elements for production of another viral vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the invention relates to induction of immune responses, suitably protective immune responses, against Middle East respiratory syndrome coronavirus (MERS-CoV).
  • MERS-CoV Middle East respiratory syndrome coronavirus
  • MERS-CoV Middle East respiratory syndrome coronavirus
  • Middle East respiratory syndrome is caused by a novel betacoronavirus (MERS-CoV) that was isolated in late 2012 in Saudi Arabia (1).
  • the syndrome (MERS) is described as a viral infection that causes fever, cough, and/ or shortness of breath and to a lesser extent gastrointestinal symptoms such as diarrhea (2).
  • Severe disease from MERS-CoV infection can cause respiratory failure and organ failure, and cases can be fatal, especially in patients with co-morbidities such as diabetes and cardiac
  • MERS-CoV MERS-CoV
  • the mechanism of camel to human transmission is still not clear, but several primary cases have been associated with camel contact, which is considered an important risk factor (14-16). Therefore, camels are being considered an intermediate host and one of the sources of MERS-CoV infection (8-13).
  • Other livestock animals such as sheep, goats, cows, chicken, and horses have proved seronegative in many studies (17-20).
  • Vaccination approaches using full-length S protein of MERS-CoV have proven problematic.
  • Hotez et al. 2014 (Microbes and Infection 2016, pages 529 - 531) discusses calls for rapid development of a safe and effective MERS vaccine. It is disclosed at page 530 that use of subunit vaccines comprising the full spike protein of the severe acute respiratory syndrome (SARS) coronavirus caused eosinophilic immune enhancement in the lungs - a form of hypersensitivity that is very undesirable.
  • SARS severe acute respiratory syndrome
  • the authors note efforts to develop a more restricted receptor binding domain (RBD) of the coronavirus spike protein as a recombinant vaccine against SARS.
  • RBD restricted receptor binding domain
  • Such an approach is said to elicit highly effective cross-neutralising antibody responses against SARS in vaccinated animals, but the authors note a general unwillingness to follow this route for generating a MERS vaccine on account of the time and cost involved in this
  • SARS vaccines comprising a UV-inactivated SARS virus together with Toll-like receptor (TLR) agonists including lipopolysaccharide, poly(U), and poly(I-C) (UV-V+TLR) to reduce excess eosinophilic infiltration in the lungs.
  • TLR Toll-like receptor
  • Haagmans et al. 2016 discloses an orthopoxvirus based vaccine which reduces MERS-CoV viral RNA excretion after MERS-CoV infection in dromedary camels.
  • the authors show that a modified vaccinia virus Ankara (MVA) vaccine expressing the MERS-CoV spike protein confers mucosal immunity in dromedary camels when it is administered in two doses following a prime-boost regimen, and no protective immunity is demonstrated.
  • MVA modified vaccinia virus Ankara
  • the present seeks to overcome problem(s) associated with the prior art.
  • a combination which comprises a simian adenoviral vector (such as ChAdOxi) delivering a MERS-CoV antigen (the spike protein).
  • a simian adenoviral vector such as ChAdOxi
  • MERS-CoV antigen the spike protein
  • sequence/secretory sequence such as the tissue plasminogen activator (tPA) amino acid sequence fused to the N-terminus of the MERS-CoV spike protein antigen.
  • tPA tissue plasminogen activator
  • the invention relates to a composition
  • a composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is an adenovirus based vector.
  • MERS-CoV middle eastern respiratory syndrome coronavirus
  • said adenovirus based vector is ChAdOx 1.
  • said spike protein is full length spike protein.
  • said spike protein is present as a fusion with the tissue plasminogen activator (tPA) sequence.
  • tPA tissue plasminogen activator
  • the fusion is in the order N-terminus - tPA - spike protein - C- terminus.
  • said spike protein has the amino acid sequence given in the MERS genome GenBank accession number KJ650098.1.
  • said spike protein has the amino acid sequence given in GenBank accession number AHX71946.1.
  • Middle East respiratory syndrome coronavirus may be isolate
  • said spike protein has the amino acid sequence of SEQ ID NO: 1.
  • the invention relates to a composition as described above for induction of an immune response against MERS-CoV.
  • the invention relates to a composition as described above for induction of an immune response against MERS-CoV, wherein a single dose of said composition is administered.
  • the invention relates to a composition as described above for induction of an immune response against MERS-CoV, wherein said composition is administered once.
  • said composition is administered once per 6 months.
  • said composition is administered once per 12 months.
  • the primary vaccination regimen is one dose, whereas all prior art MERS vaccines require two doses to provide protection.
  • the invention relates to a composition as described above for preventing MERS-CoV infection.
  • the invention relates to a composition as described above for preventing MERS-CoV infection, wherein a single dose of said composition is administered.
  • the invention relates to a composition as described above for preventing MERS-CoV infection, wherein said composition is administered once.
  • said composition is administered once per 6 months.
  • said composition is administered once per 12 months.
  • the invention relates to use of a composition as described above in medicine.
  • the invention relates to use of a composition as described above in the preparation of a medicament for prevention of MERS-CoV infection.
  • the invention relates to use of a composition as described above in inducing an immune response against middle eastern respiratory syndrome coronavirus (MERS-CoV).
  • the invention relates to use of a composition as described above in immunising a subject against MERS-CoV.
  • the invention relates to use of a composition as described above in prevention of MERS-CoV infection.
  • a method of inducing an immune response against middle eastern respiratory syndrome coronavirus (MERS-CoV) in a mammalian subject comprising administering a composition as described above to said subject.
  • MERS-CoV middle eastern respiratory syndrome coronavirus
  • a single dose of said composition is administered to said subject.
  • composition is administered once.
  • composition is administered once per 6 months.
  • composition is administered once per 12 months.
  • composition is administered by a route of administration selected from a group consisting of subcutaneous, intranasal, aerosol, nebuliser, intradermal and intramuscular.
  • administration is intramuscular.
  • the invention relates to an adeno-based viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV).
  • MERS-CoV middle eastern respiratory syndrome coronavirus
  • the adeno-based viral vector is ChAdOx 1.
  • the invention relates to a ChAdOx comprising a polynucleotide encoding glycoprotein S from the MERS virus.
  • adeno-based viral vector has the sequence and/or construction as described in one or more of the examples.
  • the invention relates to a method of raising an immune response by administering the adeno-based viral vector as described above.
  • the invention relates to the adeno-based viral vector as described above for use in preventing MERS infection.
  • the invention relates to the adeno-based viral vector as described above for use in raising an anti-MERS immune response.
  • ChAdOxi and MVA based vaccine candidates against MERS-CoV elicit neutralising antibodies and cellular immune responses in mice.
  • ChAdOxi MERS vaccines were produced with or without the leader sequence of the human tissue plasminogen activator gene (tPA) where MVA MERS vaccines were produced with tPA, and either the 1T1H5 or F11 promoter driving expression of the spike gene. All vaccine candidates were evaluated in a mouse model in prime only or prime-boost regimens. ChAdOxi MERS with tPA induced higher neutralising antibodies than ChAdOxi MERS without tPA.
  • ChAdOxi and MVA vectored vaccines have been safely evaluated in camels and humans and these MERS vaccine candidates are demonstrated to be promising and the data shown herein support their industrial application for example that they are expected to be effective in camels and in clinical trials in humans.
  • MERS-CoV vaccine candidates that are based on two different viral vectors: Chimpanzee Adenovirus, Oxford University #1 (ChAdOxi) (26) and Modified Vaccinia virus Ankara (MVA) (27, 28).
  • Each viral vector was developed by generating two alternative versions, resulting in four vaccine candidates that all encode the same complete MERS-CoV spike gene (S).
  • the two ChAdOxi based vaccines were produced with or without the signal peptide of the human tissue plasminogen activator gene (tPA) at the N terminus.
  • tPA tissue plasminogen activator gene
  • the two MVA based vaccines were produced with either the 1T1H5 or F11 poxviral promoter driving antigen expression, both including the tPA sequence at the N terminus of MERS-CoV Spike protein.
  • ChAdOxi based vaccines have been tested in different animal models, including camels (32), and in human clinical trials and proved safe and immunogenic (33). Therefore, based on our data, ChAdOxi MERS can be readily employed for use as a MERS vaccine in humans. Furthermore, utilising ChAdOxi MERS for camel vaccination can serve the one-health approach whereby blocking MERS-CoV transmission in camels is expected to prevent human infections.
  • the inventors identified the major surface antigen of MERS-CoV as the Spike (S protein) and demonstrated that ChAdOxi expressing this protein induces the production of anti-S antibodies, including neutralising antibodies, after a single intramuscular immunisation. This single dose induced a response equivalent to that provided by a prime-boost administration of the same antigen in another viral vector as demonstrated in mice. Studies in camels and humans are outlined below.
  • Hypersensitivity responses were not seen in vaccinated animals and human subjects.
  • the invention finds particular application in prevention or containment of outbreaks of MERS.
  • it is extremely advantageous to achieve protective immunity with only a single dose of vaccine.
  • emerging pathogens such as MERS
  • patients may need to walk from village to village in order to receive their dose and have many pressures on their time which can prevent attendance for a second dose.
  • they may have to travel from distance to receive a dose, or they may need to attend to their deaths which can prevent them from attending for more than a single dose.
  • the present invention also advantageously allows for avoidance of quarantine of patients in between doses which might otherwise be required since acquiring the infection in between doses would be potentially deleterious for the individual.
  • the invention finds particular application in the immunisation of camels.
  • Camels are believed to be the only, or at least the main, animal source of human infection with MERS.
  • MERS human infection with MERS.
  • the invention delivers protective immunity with only a single dose - this advantage extends to camels as well as to human or other subjects.
  • the subject is a camel.
  • the subject is a human.
  • Malczyk et al. 2015 disclose a measles virus vaccine comprising the MERS spike protein.
  • protective immunity is only demonstrated for this measles based vaccine after two doses.
  • the invention delivers benefits compared to Malczyk et al. 2015.
  • the method is a method of immunising.
  • the immune response comprises a humoral response.
  • the immune response comprises an antibody response.
  • the immune response comprises a neutralising antibody response.
  • the immune response comprises a cell mediated response.
  • the immune response comprises cell mediated immunity (CMI).
  • CMI cell mediated immunity
  • the immune response comprises induction of CD8+ T cells.
  • the immune response comprises induction of a CD8+ cytotoxic T cell (CTL) response.
  • CTL cytotoxic T cell
  • the immune response comprises both a humoral response and a cell mediated response.
  • the immune response comprises protective immunity.
  • the composition is an antigenic composition.
  • composition is an immunogenic composition.
  • composition is a vaccine composition.
  • composition is a pharmaceutical composition.
  • the composition is formulated for administration to mammals, suitably to primates or camelids.
  • Camelids are members of the biological family Cam elidae, the only currently living family in the suborder Tylopoda.
  • the extant members of this group are: dromedary camel, Bactrian camels, wild or feral camels, llamas, alpacas, vicunas, and guanacos.
  • the composition is formulated for administration to primates, camels, alpacas, llamas, vicunas, pigs, non-human primates or humans, more suitably to primates or camels, most suitably to humans
  • the composition is formulated taking into account its route of administration.
  • the composition is formulated to be suitable for the route of administration specified.
  • the composition is formulated to be suitable for the route of administration selected by the operator or physician.
  • Sequences deposited in databases can change over time.
  • the current version of sequence database(s) are relied upon.
  • the release in force at the date of filing is relied upon.
  • accession numbers may be version/dated accession numbers.
  • the citeable accession numbers for the current database entry are the same as above, but omitting the decimal point and any subsequent digits.
  • GenBank is the NIH genetic sequence database, an annotated collection of all publicly available DNA sequences (National Center for Biotechnology Information, U.S.).
  • GenBank database release referred to is 15 April 2017, NCBI-GenBank Release 219.0.
  • UniProt Universal Protein Resource
  • 'UniProt a hub for protein information' Nucleic Acids Res. 43: D204-D212 (2015).
  • UniProt Release 20i5_n is relied upon.
  • RVF River Valley Fever
  • Weingartl et al. 2004 ⁇ ibid discloses the use of a MVA vector to deliver antigen to ferrets.
  • Weingartl et al. observed pathology using this viral delivery system. Therefore, it would be incorrect to extrapolate from this paper as if merely using a viral vector would be the answer to achieving single dose immunity.
  • the invention delivers the advantage of avoiding immunopathology.
  • the invention provides the advantage of avoiding hypersensitivity.
  • the invention possesses the advantage of protective immunity after single dose (single administration).
  • protective immune response or “protective immunity” as used herein means that the composition is capable of generating a protective response in a host organism, such as a human or a non-human mammal (e.g. a camel), to whom it is administered according to the invention.
  • a protective immune response protects against subsequent infection or disease caused by MERS-CoV.
  • the antigen in the MERS spike protein Suitably the full length spike protein is used.
  • Suitably full length means each amino acid in the spike protein is included.
  • An exemplary spike protein is as disclosed in accession number AHX71946.1 - SEQ ID NO: 1 (above).
  • the full length spike protein is used.
  • a further advantage of using the full length spike protein is that it allows for better T- cell responses. Without wishing to be bound by theory, it is believed that the more amino acid sequences present, then the more potential targets there are for the T-cell responses. Thus, suitably every amino acid of the wild type spike protein is included in the antigen of the invention.
  • tPA tPA tissue plasminogen activator
  • tPA is fused to the MERS spike protein antigen of the invention.
  • tPA is fused to the N-terminus of the spike protein sequence.
  • tPA leader sequence means the tPA amino acid sequence of SEQ ID NO: 5 SEQ ID NO: 5
  • the C terminal 'RR' is not actually part of the tPA leader sequence. It comes from the fusion of two restriction sites.
  • the tPA leader sequence may be used with or without the C terminal 'RR' e.g. SEQ ID NO: 7 or SEQ ID NO: 8. Most suitably the sequence is used as shown in SEQ ID NO: 5.
  • the underlined A is P in the naturally occurring tPA leader sequence.
  • the P->A mutation has the advantage of improved antigen secretion.
  • the tPA leader sequence may be used with or without the P->A mutation, i.e. suitably the tPA leader sequence may be used as SEQ ID NO: 5 or SEQ ID NO: 6.
  • sequence is used with the P->A mutation (with or without the C terminal 'RR'). Most suitably the sequence is used as shown in SEQ ID NO: 5.
  • SEQ ID NO: 9 An exemplary nucleotide sequence encoding tPA, which has been codon optimised for human codon usage, is as shown in SEQ ID NO: 9 (this is the sequence encoding SEQ ID NO: 5):
  • tPA promotes secretion of proteins to which it is fused. It is believed that tPA increases expression of proteins to which it is fused. Notwithstanding the underlying mechanism, the advantage in the invention of fusing tPA to the N-terminus of the spike protein antigen is that improved immunogenicity is achieved.
  • the antigen of the invention is provided as a fusion with tPA.
  • the tPA is fused to the N-terminus of the spike protein antigen.
  • the antigen does not comprise any further sequence tags.
  • the antigen does not comprise any further linker sequences.
  • Adeno-based viral vectors Any suitable adeno-based viral vector may be used.
  • any replication-deficient viral vector for human use preferably derived from a non-human adenovirus may be used.
  • Ad5 may be used for veterinary use.
  • ChAd0x2 is an example of a suitable non-human adenovirus vector for human use.
  • the adeno-based viral vector is ChAdOxi.
  • ChAdOxi is described in Dicks MDJ, Spencer AJ, Edwards NJ, Wadell G, Bojang K, et al. (2012) A Novel Chimpanzee Adenovirus Vector with Low Human Seroprevalence: Improved Systems for Vector Derivation and Comparative Immunogenicity.
  • PLoS ONE 7(7): 640385 and in WO2012/172277. Both these documents are hereby incorporated herein by reference, in particular for the specific teachings of the ChAdOxi vector, including its construction and manufacture.
  • the El site may be used, suitably with the hCMV IE promoter.
  • the short or the long version may be used; most suitably the long version as described in WO2008/122811, which is specifically incorporated herein by reference for the teaching of the promoters, particularly the long promoter.
  • a clone of ChAdOxi containing GFP is deposited with the ECACC: a sample of E. coli strain SW1029 (a derivative of DH10B) containing bacterial artificial chromosomes (BACs) containing the cloned genome of AdChOXi (pBACe3.6 AdChOxi (E4 modified) TIPeGFP, cell line name "AdChOxi (E4 modified) TIPeGFP" was deposited by Isis Innovation Limited on 24 May 2012 with the European Collection of Cell Cultures (ECACC) at the Health Protection Agency Culture Collections, Health Protection Agency, Porton Down, Salisbury SP4 oJG, United Kingdom under the Budapest Treaty and designated by provisional accession no. 12052403. Isis
  • the nucleotide sequence of the ChAd0x2 vector (with a GatewayTM cassette in the El locus) is shown in SEQ ID NO. 2 This is a viral vector based on Chimpanzee adenovirus C68. (This is the sequence of SEQ ID NO: 10 in gb patent application number
  • ChAd0x2 containing GFP is deposited with the ECACC: deposit accession number 16061301 was deposited by Isis Innovation Limited on 13 June 2016 with the European Collection of Cell Cultures (ECACC) at the Health Protection Agency Culture Collections, Health Protection Agency, Porton Down, Salisbury SP4 oJG, United Kingdom under the Budapest Treaty. Isis Innovation Limited is the former name of the proprietor/applicant of this patent/application.
  • any suitable route of administration may be used.
  • the route of administration is selected from group consisting of subcutaneous, intranasal, aerosol, nebuliser, intradermal and intramuscular.
  • the route of administration is selected from a group consisting of intranasal, aerosol and intramuscular.
  • the route of administration is selected from a group consisting of intranasal and intramuscular.
  • the route of administration is intramuscular.
  • the route of administration maybe applied to camels or humans.
  • Viral particles - vp/mL This refers to the count of total viral particles administered.
  • Preferred doses according to the present invention are:
  • Humans preferred range is from io 9 to 10 11 viral particles
  • Pigs, io 8 to io 10 viral particles Pigs, io 8 to io 10 viral particles.
  • the viral vector of the invention is formulated with simple buffer.
  • An exemplary buffer may be as shown below under the heading 'Formulation'.
  • nucleic acid sequence is codon optimised for mammalian codon usag suitably for camel or human codon usage, most suitably for human codon usage.
  • a container containing a composition as described above is provided.
  • said container may be a vial.
  • said container may be a syringe.
  • a nebuliser containing a composition as described above is provided.
  • a nasal applicator containing a composition as described above is provided.
  • an inhaler containing a composition as described above is provided.
  • a pressurised canister containing a composition as described above is provided.
  • a method of making a composition as described above comprising preparing a nucleic acid encoding the MERS-CoV spike protein, optionally fused to the tPA protein, and incorporating said nucleic acid into an adeno-based viral vector, suitably a ChAdOxi vector.
  • the nucleic acid is operably linked to a promoter suitable for inducing expression of said MERS-CoV spike protein (or MERS- CoV spike protein-tPA fusion protein) when in a mammalian cell such as a human or camel cell.
  • ChAdOxi formulation buffer as used for the clinical product is: FORMULATION BUFFER COMPONENTS
  • Formulations for other administration routes such as aerosol will be adjusted accordingly by the skilled operator.
  • composition and/or formulation does not comprise adjuvant.
  • adjuvant is omitted from the composition and/or formulation of the invention.
  • MVA-MERS spike protein vaccine has been described in the prior art, for example by Volz et al. 2015/Haagmans et al. 2016 ⁇ ibid).
  • MVA vector carrying the MERS spike protein which is described in more detail in the Examples section.
  • the MVA vector described herein features a 1T1H5/F11 promoter system in one embodiment, or relies on a standard F11 promoter in another embodiment.
  • these promoter systems are known in the art, for example in published patent US 9 > 273, 327B2 (Cottingham - granted 1 March 2016 - 'Poxvirus Expression System') - this document is hereby incorporated by reference, in particular for the specific teachings of promoter(s) for use herein.
  • MVA vector delivering MERS spike protein is taught as a useful optional boost in an immunisation regimen as described.
  • the first dose should preferably be ChAdOxi-MERS spike protein (most preferably comprising the tPA fusion to the N-terminus of the spike protein) and the optional second administration preferably comprises MVA-MERS spike protein.
  • the main focus of the invention is in provision of a single dose MERS vaccine.
  • the second (boosting) administration is in a different viral vector i.e. a heterologous "prime-boost" regime.
  • the second (boosting) administration comprises a MVA vector.
  • a "prime-boost" regimen comprising a first administration of an adenoviral vector-MERS composition such as a ChAdOx-MERS composition, followed by a second (boosting) administration of a viral vector comprising the MERS spike protein, such as a MVA vector expressing the MERS spike protein.
  • an adenoviral vector-MERS composition such as a ChAdOx-MERS composition
  • a second (boosting) administration of a viral vector comprising the MERS spike protein such as a MVA vector expressing the MERS spike protein.
  • MVA-MERS spike protein has limited use but may find particular application as a heterologous boost following a ChAdOx- MERS spike protein priming vaccination.
  • MVA-MERS vaccine may be administered first followed by the ChAdOx-MERS vaccine after an interval of typically 1 - 8 weeks.
  • MVA boosting for extended duration of immunity may also be useful for subjects who are occupationally exposed to camels, e.g. camel shepherds or
  • slaughterhouse workers may also become infected, experience only mild illness but transmit the infection to family or community members. There is a slightly higher rate of seropositivity to MERS in these workers than the general population.
  • this embodiment of the invention may be applied to such subjects who are occupationally exposed to camels, most suitably camel shepherds or slaughterhouse workers.
  • the invention provides a method of inducing an immune response against middle eastern respiratory syndrome coronavirus (MERS-CoV) in a mammalian subject, the method comprising
  • composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is an adenovirus based vector to said subject, and
  • MERS-CoV middle eastern respiratory syndrome coronavirus
  • composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is a MVA based vector to said subject.
  • MERS-CoV middle eastern respiratory syndrome coronavirus
  • step (i) is a priming composition.
  • step (ii) is a boosting composition.
  • step (ii) is carried out 1-8 weeks after the step (i), most suitably 4 weeks after step (i).
  • A schematic representation of ChAdOxi and MVA based vaccines, each encodes the same MERS-CoV spike gene (MERS-CoV S).
  • the S gene was inserted into the El region of ChAdOxi genome or into the F11L locus of MVA genome.
  • tPA Human tissue plasminogen activator (tPA) signal peptide sequence.
  • IE CMV The human
  • cytomegalovirus major immediate early promoter ⁇ 1 ⁇ 5 and F11: Poxviral promoters.
  • LHA left homology arm sequence.
  • RHA right homology arm sequence.
  • B The expression of spike transgene, cloned into a plasmid vector, was validated by transfection into an African green monkey kidney cell line (Vero cells) and confirmed by immunostaining.
  • C Untransfected cells control. Green colour represents detection of the spike protein. Blue colour represents nuclei by staining nucleic acid with DAPI.
  • Figure 2 Antibody responses to ChAdOxi MERS vaccine candidates.
  • a control group of mice were immunised with ChAdOxi expressing eGFP instead of MERS-CoV S gene. Serum samples were collected at 14 and 28 days post immunisation (d.p.i.). Si- binding antibodies were detected at both time points by ELISA (A) and neutralisation activity of the antibodies were confirmed by MERS-CoV pseudotyped viral particles (MERSpp) neutralisation assay (B) or neutralisation assay (C). Individual data points are shown with line as the median. Data are representative of two independent experiments. Statistical significance by Kruskal-Wallis test is shown.
  • FIG. 3 Cellular im m une responses to ChAdOxi MERS vaccine candidate .
  • Twenty eight days post- immunisation IFN- ⁇ ex vivo ELISpot (A) or Intracellular Cytokine Staining (ICS (B)), were performed to determine the percentage of splenic IFN- ⁇ secreting CD4 + and CD8 + after in vitro re-stimulation with a MERS-CoV S-specific peptide. Individual data points are shown with line as the median (A) or error bars as the SD (B). Data are representative of two independent experiments.
  • Figure 4 Hum oral and cellular im m unogenicity of heterologous ChAdOxi MERS and MVA MERS vaccination.
  • mice were boosted with MVA MERS at ixio A 6 pfu.
  • MVA MERS candidates either contain ⁇ 1 ⁇ 5 or F11 promoter for transgene expression.
  • Serum samples were collected at 28 (post-prime) and 42 (post- boost) d.p.i.
  • Si-binding antibodies were detected at both time points by ELISA (A) and neutralisation activity of serum antibodies at 42 d.p.i. were confirmed by MERSpp neutralisation assay (B).
  • Cytokine Staining were performed to determine the percentage of CD8 + IFN- Y + splenocytes after in vitro re-stimulation with a MERS-CoV S-specific peptide.
  • ICS of splenocytes re-stimulated with MVA- specific peptides was also performed (E and F). Individual data points are shown with line as the median. Data are representative of two independent experiments. Statistical significance by Kruskal- Wallis test is shown. Symbols are closed squares ( ⁇ ) for ChAdOxi prime responses, open circles (o) for 1T1H5-MVA boost responses, and closed circles ( ⁇ ) for F11-MVA boost responses.
  • Figure 5 Hum oral and cellular im m unogenicity of hom ologous MVA MERS vaccination.
  • MVA MERS candidates either contain ⁇ 1 ⁇ 5 or F11 promoter for transgene expression.
  • Serum samples were collected at 21 (post-prime) and 42 (post-boost) d.p.i.
  • Si-binding antibodies were detected at both time points by ELISA (A) and neutralisation activity of serum antibodies at 42 d.p.i. were confirmed by MERSpp neutralisation assay (B).
  • splenocytes were processed and re-stimulated with a MERS-CoV S-specific peptide (CD8 + T cell specific) for IFN- ⁇ ex vivo ELISpot (C).
  • ICS of splenocytes re- stimulated with MVA-specific peptides was also performed (D and E) as was performed in figure 4. Individual data points are shown with line as the median. Data are representative of two independent experiments. Statistical significance by Kruskal-Wallis test is shown. Symbols are open circles (o) for 1T1H5-MVA and closed circles ( ⁇ ) for Fii-MVA.
  • FIG. 6 Protective Efficacy of ChAdOxi MERS vaccine .
  • Groups of 10 mice were vaccinated with 10 8 TCID 50 ChAdOxi GFP or ChAdOxi MERS via the intranasal or intramuscular route, blood samples were collected before vaccination, and before challenge at 28 days post vaccination.
  • hDPP4 mice were challenged intranasally with 104 TCID 5 o MERS-CoV (strain HC0V-EMC2012). At three dpi, four animals were sacrificed and lungs collected for analyses. The remaining six animals per group were sacrificed 28 dpi, or when they reached the endpoint criteria.
  • hDPP4 mouse tissues were evaluated for pathology and the presence of viral antigen as described previously Briefly, tissues were fixed in 10% neutral-buffered formalin for 7 days and paraffin- embedded. Tissue sections were stained with hematoxylin and eosin (H&E). An in- house produced rabbit polyclonal antiserum against HCoV- EMC/2012 (1:1000) was used as a primary antibody for the detection of viral antigen. Grading of histopathology and immunohistochemistry was done blinded by a board-certified veterinary pathologist. Lung tissues are shown at 100X and 1000X (insert) magnification.
  • Lung tissue shows multifocal scattered positivity in the lungs.
  • the inserts display MERS-CoV antigen within the Type I & II pneumocytes.
  • Figure 8 shows a plot
  • Figure 9 shows a plot, a bar chart and a graph.
  • Figure 11 shows a plot of ELISPOT responses to ChAdOxi MERS vaccine in Group 1 (5 x 10 9 vp, low dose) volunteers.
  • Figure 12 shows a plot of evaluation of MERS-CoV specific antibodies in camels at different ages using the commercial ELISA from Euroimmun.
  • Figure 13 shows a plot of evaluation of MERS-CoV specific antibodies in camels at different ages using an in house endpoint titre ELISA.
  • Figure 14 shows a plot of virus neutralisation titres for control and vaccinated camels.
  • Figure 15 shows a plot of na ' ve camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using Euroimmun ELISA kit.
  • Figure 16 shows a plot of seropositive camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using Euroimmun ELISA kit.
  • Figure 17 shows na ' rve camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using an in house end point titre ELISA.
  • Figure 18 shows a plot of seropositive camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using an in house end point titre ELISA.
  • the spike (S) gene of MERS-CoV camel isolate (Genbank accession number:
  • KJ650098.1 was synthesised by GeneArt Gene Synthesis (Thermo Fisher Scientific). The S transgene was then cloned into four shuttle plasmid vectors following In-Fusion cloning (Clontech). Two plasmids contained the S transgene within the El homologous region of ChAdOxi, driven by the human cytomegalovirus major immediate early promoter (IE CMV) that includes intron A. One of the ChAdOxi shuttle plasmids was designed to include the tPA signal sequence upstream of the transgene sequence while the second plasmid did not contain the tPA. The ChAdOxi shuttle plasmids contained the S transgene within Gateway® recombination cassettes. To construct MVA MERS, one of the shuttle plasmids for MVA was designed to have the upstream and
  • mH5-MVA MERS contained the 1T1H5 promoter at the F11L locus, however, the endogenous F11 promoter is intact and located upstream of the mH5 promoter. The endogenous F11 promoter could not be replaced with the mH5 since it is part of the essential upstream ORF. Im m unostaining for Transgene Expression
  • the ChAdOxi shuttle plasmid was used to validate the expression of MERS-CoV spike protein in vitro.
  • An African green monkey kidney cell line (Vero cells) was seeded into 6-well plate to 80% confluence. Then the plasmid DNA was transfected into Vero cells using Lipofectamine® 2000 (Thermo Fisher Scientific) following manufacturer's instruction. Twenty four hours after transfection, cells were fixed, permeabilised, and immunostained using a rabbit polyclonal anti-MERS-CoV spike antibody, following standard protocols. DAPI stain was used to label nuclei.
  • ChAdOxi MERS vaccines were prepared by Gateway® recombination between the ChAdOxi destination DNA BAC vector (described in (26)) and entry plasmids containing the coding sequence for MERS-CoV spike gene (ChAdOxi shuttle vectors explained above), according to standard protocols. ChAdOxi MERS genomes were then derived in HEK293A cell lines (Invitrogen, Cat. R705-07), the resultant viruses were purified by CsCl gradient ultracentrifugation as previously described (34).
  • the titres were determined on HEK293A cells using anti-hexon immunostaining assay based on the QuickTiterTM Adenovirus Titer Immunoassay kit (Cell Biolabs Inc).
  • MVA MERS vaccines chicken embryo fibroblast cells (CEFs) were infected with MVA parental virus that encodes dsRed marker instead of the native F11L ORF and transfected with MVA shuttle plasmids containing MERS-CoV spike gene (explained above) to allow recombination with the MVA genome and deletion of dsRed marker whilst keeping the F11 promoter sequence.
  • Recombinant MVA expressing MERS-CoV S protein was purified by plaque-picking and fluorescent selection using the sorting function of CyCLONE robotic module of a MoFlo Flow cytometer (Dako Cytomation, Denmark) as previously described (31).
  • F11-MVA MERS and mH5-MVA MERS were confirmed to lack the native F11L ORF (and the dsRed marker), and contain MERS-CoV S by PCR (identity and purity PCR screening).
  • the sequence of the S transgene amplified from these vaccines was confirmed.
  • the recombinant viruses (vaccines) were amplified in 1500 cm 2 monolayers of CEFs cells, partially purified over sucrose cushions and titrated in CEFs cells according to standard practice, and purity and identity were again verified by PCR.
  • mice Female BALB/c mice (Harlan, UK) aged 6 to 8 weeks were immunised intramuscularly (i.m.) in the upper leg (total volume 50 ⁇ .) with a total of 10 8 IU of ChAdOxi MERS with or without tPA or with a total of 10 6 pfu of either F11-MVA MERS or mH5-MVA MERS.
  • animals were anaesthetised using vaporised IsoFloH.
  • mice were vaccinated with ChAdOxi with blood samples taken at 14 days post immunisation (d.p.i) or 28 d.p.i.
  • Splenocytes were harvested for analysis by IFN- ⁇ ELISpot or intracellular cytokine staining (ICS) and flow cytometry as previously described (35, 36), using re-stimulation with 2 g/mL S291 MERS-CoV S-specific peptide (VYDTIKYYSIIPHSI); for vaccine cellular immunogenicity (37)); or 1 g/mL E3 and F2(G) MVA vector-specific peptides (38) (for anti-MVA immune responses).
  • VYDTIKYYSIIPHSI 2 g/mL S291 MERS-CoV S-specific peptide
  • E3 and F2(G) MVA vector-specific peptides for anti-MVA immune responses.
  • the frequency of IFN-y + cells which was typically 0.1% by flow cytometry or less than 50 SFC by ELISpot, was subtracted from tested re-stimulated samples.
  • MERS pseudotyped viral particles were produced and titrated using Huh7.5 cell line as described previously (40).
  • MERSpp neutralization assay serum samples were serially diluted in 96- well white plates (Nunc). A standard concentration of the MERSpp were added to the wells and plates were incubated for 1 h at 37 °C. After incubation, Huh7.5 cells (10,000 cells per well) were added to the plate in duplicates. Following 48 h incubation, cells were lysed and luciferase activity was measured. IC90 neutralisation titres were calculated for each mouse serum sample using GraphPad Prism.
  • MERS-CoV vaccine candidates The spike gene from a camel isolate (Camel/Qatar_2_20i4 MERS-CoV isolate, GenBank accession number KJ650098.1) was cloned into four shuttle vectors that facilitate homologous recombination with the genome of ChAdOxi or MVA.
  • ChAdOxi based vaccine candidates were generated with or without the signal peptide of the human tissue plasminogen activator gene (tPA).
  • the spike transgene expression in ChAdOxi MERS vaccine candidates is under the control of the human cytomegalovirus major immediate early promoter (CMV IE) that includes intron A.
  • CMV IE human cytomegalovirus major immediate early promoter
  • the tPA was also inserted upstream of the spike transgene, which was under the control of either the ectopic 1T1H5 promoter or the endogenous F11 promoter ( Figure lA).
  • All of our MERS-CoV vaccine candidates contain the same codon-optimized spike transgene.
  • the expression of the newly synthesized transgene was first tested by transfection of an African green monkey kidney cell line (Vero cells) with the adenovirus shuttle vector, and immunofluorescence staining of the transfected cells ( Figure lB and lC). This was performed to confirm the expression of the codon optimized spike transgene in mammalian cells.
  • the level of transgene expression from the four vaccine candidates was not evaluated in vitro. We have previously reported that differences in MVA promoter activity detectable in vitro does not correlate with in vivo immunogenicity (31), and that only in vivo expression correlates with the in vivo immunogenicity.
  • ChAdOxi MERS ChAdOxi MERS in the rest of this report
  • spleens were collected at 28 d.p.i. from immunised BALB/c mice.
  • Splenocytes were processed to evaluate cellular immune responses to ChAdOxi MERS in ELISpot and Intracellular cytokine staining (ICS).
  • ICS Intracellular cytokine staining
  • ICS data confirmed the IFN- ⁇ secreting CD8 + splenocytes also secreted TNF-a and IL-17 ( Figure 3B).
  • mice were immunised with ChAdOxi MERS vaccine and boosted with one of two different MVA MERS vaccine candidates four weeks later.
  • the MVA based candidates differ in the promoters that controls the transgene expression: Fii-MVA MERS utilises the endogenous strong early F11 promoter and mH5-MVA MERS utilises the ectopic early/late 1T1H5 promoter. Serum samples from 28 d.p.i. (post-prime) or 42 d.p.i. (post-boost) were collected and evaluated by ELISA and MERSpp neutralisation assay. At 28 d.p.i.
  • ChAdOxi MERS induced similar levels of Si-specific antibodies and nAb as observed previously ( Figure 4A and B).
  • Figure 4A) with nAb also enhanced to a statistically significant level (mean titre IC 90 (Logi 0 ) 2.87 by ChAdOxi MERS boosted to 3.3 by mH5-MVA MERS or 3.5 by F11-MVA MERS; Figure 4B).
  • IC 90 (Logi 0 ) 2.87 by ChAdOxi MERS boosted to 3.3 by mH5-MVA MERS or 3.5 by F11-MVA MERS;
  • Figure 4B There was no difference in antibody levels induced using either the F11 or ⁇ 1 ⁇ 5 promoter in the MVA.
  • splenocytes were also processed to evaluate cellular immune responses to ChAdOxi MERS MVA MERS prime-boost vaccination in ELISpot and ICS as shown in Figure 3.
  • the T cell responses to MERS S were boosted by the MVA vaccinations; in the ICS experiments, Fii-MVA and 1T1H5-MVA boosted the percentage of IFN-Y + splenic CD8 + T cells to 7.3 and 5.2% respectively ( Figure 4D) whereas the percentage was 2.5% after ChAdOxi MERS prime in Figure 3B.
  • the percentage of TNF-a + splenic CD8 + T cells were also increased by MVA boost (comparing Figure 3B and 4D).
  • ChAdOxi MERS without tPA was still a potent vaccine candidate, inducing a high level of both Si-specific binding antibodies and MERS-CoV neutralising antibodies.
  • ChAdOxi MERS also induced cellular responses for MERS S, with polyfunctional CD8 + T cells detected in the spleen of immunized mice. This supports the potency of the ChAdOxi viral vector in inducing T cellular immunity, observed previously in animal models (26, 32, 47) as well as in humans (33).
  • ChAdOxi expressing the external glycoprotein of Rift Valley Fever Virus has been demonstrated in multiple species and it is already known that ChAdOxi RVF is highly immunogenic in camels (32). However, RVF is a completely different viral family to the coronaviruses (as discussed above).
  • MVA MERS as a heterologous boost
  • ChAdOxi/MVA would be more likely to provide complete protection than MVA/MVA.
  • ChAdOxi MERS is thus demonstrated to be a plausible and credible vaccine for both camels and humans.
  • Antrobus RD Coughlan L, Berthoud TK, Dicks MD, Hill AV, Lambe T, et al. Clinical assessment of a novel recombinant simian adenovirus ChAdOxl as a vectored vaccine expressing conserved Influenza A antigens. Molecular therapy : the journal of the American Society of Gene Therapy. 2014;22(3):668-74.
  • EXAMPLE 2 Protective Efficacy of a Novel Simian Adenovirus Vaccine Against Lethal MERS-CoV Challenge in a Transgenic Human DPP4 Mouse Model: A Simian
  • Middle East respiratory syndrome coronavirus is a novel zoonotic virus that causes severe respiratory disease in humans with case fatality rates close to 40%, but for which no vaccines are available.
  • ChAdOxi a promising replication-deficient simian adenovirus vaccine vector platform with an established safety profile in humans and dromedary camels, for MERS-CoV vaccine development.
  • Using a transgenic lethal BALB/c MERS-CoV mouse model we show that single dose intranasal or intramuscular immunization with ChAdOxi MERS, encoding full-length MERS-CoV Spike glycoprotein, is highly immunogenic and confers protection against lethal viral challenge. Immunogenicity and efficacy were comparable between immunisation routes. Together these data show credible utility of ChAdOxi MERS vaccine in humans and dromedary camels, the animal reservoir of infection.
  • ChAdOxi 6 - One ChAd vector, termed ChAdOxi 6 -, has undergone testing in dromedary camels, showing excellent safety and immunogenicity when encoding Rift Valley Fever viral glycoproteins
  • ChAdOxi MERS encoding the full-length MERS-CoV spike glycoprotein (GenBank accession number KJ650098.1) targeted by protective neutralising antibodies a .
  • hDPP4 human dipeptidyl peptidase
  • ChAdOxi GFP enhanced green fluorescent protein
  • ChAdOxi MERS vaccine encoding enhanced green fluorescent protein
  • Sera were obtained before vaccination and 28 days post-vaccination.
  • ChAdOxi GFP vaccinees developed signs of disease, including loss of body weight, ruffled fur and lethargy (Fig. 6B).
  • MERS-CoV staining for MERS-CoV in lung tissue showed abundance of antigen in the ChAdOxi GFP vaccinated mice, but not the ChAdOxi-MERS vaccinees ( Figure 7). MERS-CoV staining was observed in the type I and type II pneumocytes ( Figure 7 inserts) but not in any of the other respiratory cells such as endothelial cells, bronchiolar epithelium or macrophages.
  • ChAdOxi MERS induces T cell responses against MERS Spike protein as well as neutralizing antibodies (example 1) . Identification of immune correlates of protection against MERS-CoV in humans and camels allows cost-effective disease surveillance and vaccine monitoring.
  • ChAdOxi platform for MERS-CoV vaccine development in a lethal mouse model.
  • the excellent immunogenicity and efficacy observed here show the value of ChAdOxi MERS in dromedary camels and humans.
  • MERS-CoV Middle East respiratory syndrome coronavirus
  • Ad m in is tr a tio n ChAdOxi MERS-S 2.5 x 10 10 vp
  • the dipeptidyl peptidase 4 (DPP4) receptor is used by the MERS-CoV virus during infection and is highly conserved between Camels and Humans.
  • the MERS-CoV spike (S) protein is a characteristic structural component of the virion membrane and its Si domain mediates binding to DDP4.
  • the spike protein has been chosen as the target antigen for use in the replication-deficient simian adenovirus developed by the University of Oxford, ChAdOxi vaccine vector. ChAdOxi has shown successful results in the development of Oxford lead vaccines which have gone onto enter phase I trials within the UK (7). In this study we propose the manufacture to GMP of the ChAdOXi MERS-S vaccine and its use in a small open labelled phase I trial in Oxford.
  • the Si protein subunit and RBD have also been the basis for several MERS-CoV vaccine candidates. Both constructs have elicited neutralizing antibodies of high potency across multiple viral strains. Despite their demonstrated immunogenicity in animal models and anticipated safety in humans, RBD or Si-subunit based vaccine candidates are limited in their epitope breadth. Vaccine candidates that elicit a more diverse antibody repertoire as well as a robust cellular immuneresponse may offer the advantage of broader and more durable protection (8).
  • Live attenuated viruses have historically been among the most immunogenic platforms available, as they have the capacity to present multiple antigens across the viral life cycle in their native conformations.
  • manufacturing live-attenuated viruses requires complex containment and biosafety measures.
  • live-attenuated viruses carry the risks of inadequate attenuation causing disseminated disease, particularly in immunocompromised hosts.
  • moderately immunocompromised adults with co-morbidities have suffered the most severe MERS-CoV disease
  • making a live-attenuated virus vaccine is a less viable option.
  • Replication competent viral vectors could pose a similar threat for disseminated disease in the immuno-suppressed.
  • Replication deficient vectors avoid that risk while maintaining the advantages of native antigen presentation, elicitation of T cell immunity and the ability to express multiple antigens (8).
  • the first MERS-CoV vaccine to be used in humans has recently entered a phase I dose ranging safety study in January 2016.
  • the GLS-5300 a DNA plasmid vaccine that expresses the MERS-CoV spike (S) glycoprotein, is being administered to 75 healthy adult volunteers in the USA, by the Walter Reed Army Institute of Research. Safety and immunogenicity data are expected to be reported by the end of 2017.
  • MERS spike protein as a vaccine antigen
  • Coronaviruses are spherical and enveloped viruses with large, unsegmented, single positive RNA genomes.
  • One-third of the genome is responsible for coding the structural proteins: spike (S) glycoprotein, small envelope protein (E), integral membrane protein (M), and genome-associated nucleocapsid protein (N).
  • S spike glycoprotein
  • E small envelope protein
  • M integral membrane protein
  • N genome-associated nucleocapsid protein
  • the proteins E, M, and N are mainly responsible for the assembly of the virions, while the S protein is involved in receptor binding and bears membrane fusion capabilities during CoVs infection.
  • the S protein has an essential role in virus entry and determines tissue and cell tropism, as well as host range (11).
  • S is a type I, trimeric, transmembrane protein located at the surface of the viral envelope, giving rise to spikeshaped protrusions from the virion.
  • S is 1353 amino acids in length, heavily glycosylated (with 21 predicted N-linked glycosylation sites), and consists of a large ectodomain and a short cytosolic tail.
  • the S proteins of CoVs can be divided into two functional subunits: the N-terminal Si subunit forms the globular head, and the membrane-embedded C-terminal S2 (11).
  • Si and S2 subunits are respectively, responsible for cellular receptor DPP4 binding via the receptor-binding domain (RBD), and fusion of virus and cell membranes, thereby mediating the entry of MERS-CoV into the target cells.
  • the MERS-CoV RBD consists of a core structure, which is homologous to that of the SARS-CoV S protein RBD, and a receptor-binding motif, which is unique to MERS-CoV, thus determining viral pathogenesis and receptor recognition (12).
  • ChAdOxi MERS-S expresses a codon-optimised coding sequence for Spike protein from the MERS-CoV isolate Camel/Qatar_2_20i4
  • Adenoviruses are attractive vectors for human vaccination. They possess a stable genome so that inserts of foreign genes are not deleted and they can infect large numbers of cells without any evidence of insertional mutagenesis.
  • Replication defective adenovirus can be engineered by deletion of genes from the El locus, which is required for viral replication, and these viruses can be propagated easily with good yields in cell lines expressing El from AdHu5 such as human embryonic kidney cells 293 (HEK 293 cells) (13).
  • Human adenoviruses are under development as vectors for malaria, HIV and hepatitis C vaccines, amongst others. They have been used extensively in human trials with excellent safety profile mainly as vectors for HIV vaccines.
  • a limiting factor to widespread use of human adenovirus as vaccine vectors has been the level of anti-vector immunity present in humans where adenovirus is a ubiquitous infection.
  • the prevalence of immunity to human adenoviruses prompted the consideration of simian adenoviruses as vectors, as they exhibit hexon structures homologous to human adenoviruses (15).
  • Simian adenoviruses are not known to cause pathological illness in humans and the prevalence of antibodies to chimpanzee origin adenoviruses is less than 5% in humans residing in the US.
  • ChAdOxi is a novel recombinant chimpanzee adenovirus designed as a vaccine vector, developed by The Jenner Institute at the University of Oxford. This viral vector has been used by researchers at the University of Oxford to produce a number of vaccines expressing a range of different antigens. Three phase I clinical trials have been completed in the UK using ChAdOxi with different inserts (two influenza trials and one TB trial).
  • ChAdOxi is produced from a replication-deficient (El and E3 deleted) simian adenovirus and it has been described by Dicks et al (16).
  • the vector was constructed in a bacterial artificial chromosome (BAC) to facilitate genetic manipulation of genomic clones with improved stability and flexibility.
  • BAC bacterial artificial chromosome
  • Cellular immunogenicity of recombinant Ei E3-deleted ChAdOxi was comparable to that of other species E derived chimpanzee adenovirus vectors including ChAd63, the first simian adenovirus vector to enter clinical trials in humans.
  • the El region is essential for viral replication, hence the ability to delete Ei renders the new vector immediately replication incompetent.
  • the deletion of the non-essential adenovirus E3 region increases the insert capacity of the new vector by approximately 5Mb. It is known that the proteins encoded by the E4 region of adenoviruses interact with El during viral replication, and the imperfect interaction between the gene products of the AdHu5 Ei gene produced by HEK293 cells and simian E4 gene products has been found to result in impaired viral replication in this cell line, and consequently lower virus yields. In ChAdOxi, Ad5 E40rf4 has been inserted to replace the homologous simian virus coding sequence, resulting in improved viral replication during vaccine production. Since no replication of the virus takes place after immunization, this replacement has no effect on immunogenicity of the viral vector. Insertion of recombinant antigens at the El locus is performed using Gateway® site specific recombination technology (Invitrogen).
  • ChAdOxi MERS-S encodes the Spike (S) surface glycoprotein of the coronavirus.
  • a genomic clone of ChAdOxi MERS-S was prepared by Gateway® recombination between an entry plasmid containing the codon-optimised coding sequence for Spike protein from the MERS-CoV isolate Camel/Qatar_2_20i4 (GenBank:KJ650098.i), and the Ei-and E3-deleted ChAdOxi destination vector.
  • mice (balb/c) were immunised with ChAdOxi or MVA vectored vaccines expressing
  • MERS-CoV Spike protein Serum samples were taken after 28 days and endpoint titres measured by ELISA. This study showed that a single dose of ChAdOxi results in equivalent immunogenicity to two doses of MVA.
  • Fig. 8 shows Immunogenicity of viral vectored vaccines MERS vaccines in mice.
  • mice transgenic for the hDPP4 receptor were immunised with a single dose of ChAdOxi MERS-S by either intranasal or intramuscular injection.
  • the control ChAdOxi vaccine expressed eGFP as the vaccine antigen.
  • Serum neutralising titres were measured 28 days after vaccination, when the mice were then challenged with MERS CoV by intranasal inoculation.
  • the results showed that mice immunised with the MERS vaccine by either route were completely protected against MERS CoV infection. No virus was detected in the lungs of the mice receiving the MERS vaccine and they all survived, whereas all of the sham- vaccinated mice succumbed to infection within 8 days (Vincent Munster,unpublished data).
  • Fig. 9 A. Virus neutralising titres in mice amongst ChAdOxi MERS-S and controls administered via intranasal or intramuscularly. B. Viral load after MERS-CoV challenge. C Survival amongst ChAdOxi MERS-S and control mice after intranasal MERS-CoV challenge.
  • ChAdOxi vectored vaccines expressing different inserts have previously been used in 126 healthy volunteers taking part in clinical trials conducted by the University of Oxford in the UK (table 1).
  • ChAdOxi encoding the influenza fusion protein NP+Mi has been safely administered to 84 healthy adult volunteers in the UK in two completed clinical trials conducted at The Jenner Institute (FLU004 and FLU005).
  • FLU004 was a phase I, open-label, non- randomised dose escalation study of ChAdOxi NP+Mi.
  • the vaccine was safe, well tolerated and immunogenic, inducing ELISpot responses at all doses.
  • the dose of 2.5 ⁇ io 10 vp was chosen for further studies of ChAdOxi NP+Mi (7).
  • FLU005 was a multicentre phase I, randomised study to determine the safety and immunogenicity of vaccination regimens employing the candidate influenza vaccines MVA-NP+Mi and ChAdOxi NP+Mi.
  • Sixty-nine (69) healthy adult volunteers have received ChAdOxi NP+Mi at a concentration of 2.5 x 10 10 vp.
  • Administrations of ChAdOxi NP+Mi and MVA-NP+Mi vaccines were found to be safe and well-tolerated, in agreement with our previous studies (7, 17-20).
  • the majority of adverse events were mild to moderate in nature and lasted for 1-2 days.
  • the most common local adverse event was arm pain at the site of injection and the most common systemic adverse event was mild fatigue and headache.
  • ChAdOxi 85A Mycobacterium tuberculosis antigen 85A
  • ChAdOxi 5T4 has been given in the VANCEoi study which is an ongoing first-in-man open label randomized phase I study to determine the safety and immunogenicity of heterologous prime boost ChAd-MVA vaccination against oncofetal antigen 5T4. To date, 34 participants have received the ChAdOxi 5T4 vaccine at a dose of 2.5 x 10 10 vp and only mild AEs related to the vaccination have been reported
  • VAC067 is an ongoing first-in-man study of the ChAdOxi viral vector expressing dual second generation liver-stage malaria antigens LSAi and LSAP2 (ChAdOxi LS2). No significant safety concerns have been reported until this date.
  • MERS-CoV is an emerging zoonotic viral disease considered a global threat and listed as a priority pathogen for urgent Research and Development.
  • Chimpanzee adenovirus vaccine vectors have been safely administered to over 1000 people using a wide range of infectious disease targets including malaria (21), HIV (22), tuberculosis, influenza (7), hepatitis C (23), RSV (24) and, most recently, Ebola (25).
  • ChAdOxi viral vectored vaccines have shown to be both safe and immunogenic in previous clinical trials in the UK (FLU004, FLU005 and TB034).
  • ChAdOxi MERS-S vaccine has shown to elicit high levels of neutralising antibody in animal model.
  • the vaccine could be licensed for use in camels in the Middle East and North Africa. If licensed, human vaccines could be deployed for occupationally exposed individuals such as camel workers and health care professionals, with stockpiles available for use in the case of an outbreak.
  • Volunteers will be recruited and vaccinated at the CCVTM, Oxford. There will be 3 study groups and a total of 24 volunteers will be enrolled (table 2). Staggered enrolment will apply for the first three volunteers within each group. Volunteers will be first recruited into Group 1 and subsequently into Groups 2 and 3 following interim clinical safety reviews (see section 8.4.2). Volunteers will be allocated to a study group by selecting eligible volunteers for enrolment in the order in which they were deemed eligible, following screening.
  • ChAdOxi adenovirus vector expressing different inserts and similar adenovirus vectored vaccines eg. ChAd63.
  • ChAd63 Another simian adenovirus vector (ChAd63) has been safely administered at doses up to 2 x 10 11 vp with an optimal dose of 5 x 10 10 vp, balancing immunogenicity and reactogenicity.
  • the first dose of ChAdOxi MERS-S proposed in this study (5 x 10 9 vp) is therefore at least 10 fold less than what this new insert is expected to be tolerated (5X 10 10 vp). Doses will be gradually increased aiming to provide an optimal dose of ChAdOxi MERS-S
  • Volunteers will be enrolled and doses will be escalated according to the plan outlined below.
  • the first volunteer in the study will receive 5 xio 9 vp of ChAdOxi MERS-S (group 1). This volunteer will be vaccinated ahead of any other volunteers and the profile of adverse events will be examined after 48h. Provided there are no safety concerns as assessed by the Chief Investigator (CI) and the Local Safety Monitor (LSM), another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following vaccination of the first volunteer and at least 1 hour apart from each other. An independent safety review will be conducted by the LSM after vaccination of the first 3 volunteers in group 1. This review will include the results of safety blood tests at day 7 post vaccination and an assessment of the profile of the adverse events reported.
  • CI Chief Investigator
  • LSM Local Safety Monitor
  • the CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 1 and the first volunteer to receive the next incremental dose in group 2. If there are no safety concerns, the remaining volunteers in Group 1 and the first volunteer in group 2 may be vaccinated.
  • the total duration of the study will be 26 weeks from the day of enrolment for all volunteers.
  • the start of the trial is defined as the date of the first vaccination of the first volunteer.
  • the end of the trial is the date of the last visit of the last volunteer.
  • the potential risk to participants is considered as low.
  • the potential risks are those associated with phlebotomy and vaccination.
  • recombinant adenoviral vectors are safe. Similar vaccines encoding different antigens have been given to several thousand volunteers (including children) with a good safety profile.
  • ChAdOxi MERS-S has not been used in humans before and therefore will be initially administered at the lower dose of 5 x 10 9 vp before progressing to the higher doses of 2.5 x 10 10 and 5 x 10 10 in Groups 2 and 3.
  • Potential expected risks from vaccination include local effects such as pain, redness, warmth, swelling, tenderness or itching.
  • Systemic reactions that could potentially occur following immunisation with a recombinant adenovirus vaccine include a flu-like illness with feverishness, fatigue, malaise, arthralgia, myalgia and headache.
  • Measures of immunogenicity to the ChAdOxi MERS-S vaccine may include:
  • ChAdOxi MERS-S vaccine consists of the replication-deficient simian adenovirus vector ChAdOxi, containing the structural surface glycoprotein (Spike protein) antigens of the MERS-CoV expressed from the strong CMV IE promoter.
  • ChAdOxi MERS-S is manufactured in formulation buffer at a nominal concentration of >i.i x 10 11 vp/mL.
  • the drug product is filled into 2mL glass vials with a 13 mm grey bromobutyl rubber freeze-dry stopper (CE Marked, supplied by Adelphi Tubes) and a 13 mm aluminium seal.
  • the nitrogen filled vials are supplied sterile.
  • the containers and closures are tested for compliance with defined specifications.
  • the vials are made from Ph Eur Type 1 glass.
  • ChAdOxi MERS-S has been formulated and vialed under Good Manufacturing Practice conditions at the Clinical Bio-Manufacturing Facility (CBF), University of Oxford.
  • CBF Clinical Bio-Manufacturing Facility
  • QP Qualified Person
  • the vaccine is stored at nominal -8o°C in a locked freezer, at the clinical site. All movements of the study vaccines will be documented in accordance with existing standard operating procedure (SOP). Vaccine accountability, storage, shipment and handling will be in accordance with relevant SOPs and forms.
  • SOP standard operating procedure
  • ChAdOxi MERS-S will be allowed to thaw to room temperature and will be administered within 1 hour of removal from the freezer.
  • the vaccine will be administered intramuscularly into the deltoid of the non-dominant arm (preferably). All volunteers will be observed in the unit for 1 hour ( ⁇ 10 minutes) after vaccination.
  • Advanced Life Support drugs and resuscitation equipment will be immediately available for the management of anaphylaxis. Vaccination will be performed and the IMPs handled according to the relevant SOPs.
  • GMO genetically m odified organism s
  • inoculation sites will be covered with a dressing after immunisation. This should absorb any virus that may leak out through the needle track.
  • the dressing will be removed from the injection site after 30 minutes (+15/- 5 minutes) and will be disposed as GMO waste by autoclaving.
  • the volunteer must satisfy all the following criteria to be eligible for the study:
  • the volunteer may not enter the study if any of the following apply:
  • Bleeding disorder eg. factor deficiency, coagulopathy or platelet disorder
  • Acute disease at the time of vaccination is defined as the presence of a moderate or severe illness with or without fever. All vaccines can be administered to persons with a minor illness such as diarrhoea, mild upper respiratory infection with or without low-grade febrile illness, i.e. temperature of ⁇ 37.5°C/99.5°F.
  • This section describes the clinical procedures for evaluating study participants and follow-up after administration of study vaccine.
  • ChAdOxi MERS-S vaccine ChAdOxi MERS-S vaccine, and undergo follow-up for a total of 26 weeks.
  • the total volume of blood donated during the study will be mL. Additional visits or procedures may be performed at the discretion of the investigators, e.g., further medical history and physical examination, urine microscopy in the event of positive urinalysis or additional blood tests if clinically relevant.
  • 8.2 Observations Pulse, blood pressure and temperature will be measured at the time-points indicated in the schedule of procedures and may also be measured as part of a physical examination if indicated at other time-points.
  • HLA Human Leukocyte Antigen
  • Additional safety blood tests may be performed if clinically relevant at the discretion of the medically qualified investigators. These generally include, but are not limited to, AST, GGT and a coagulation screen.
  • Immunogenicity will be assessed by a variety of immunological assays. This may include antibodies to MERS Spike protein, ex vivo ELISpot assays for interferon gamma and flow cytometry assays, functional antibody assays and B cell analyses. Other exploratory immunological assays including cytokine analysis and other antibody assays, DNA analysis of genetic polymorphisms potentially relevant to vaccine immunogenicity and gene expression studies amongst others may be performed at the discretion of the Investigators.
  • Urinalysis Urine will be tested for protein, blood and glucose at screening. For female volunteers only, urine will be tested for beta-human chorionic gonadotrophin ( ⁇ -HCG) at screening and immediately prior to each vaccination.
  • ⁇ -HCG beta-human chorionic gonadotrophin
  • Volunteers will not be considered enrolled in the study until they have received a vaccine. Before vaccination, the eligibility of the volunteer will be reviewed. Pulse, blood pressure and temperature will be observed and if necessary, a medical history and physical examination may be undertaken to determine need to postpone vaccination depending on criteria listed in section 7.3.5. Vaccinations will be administered as described below.
  • Diary cards will collect information on the timing and severity of the following solicited AEs:
  • the first volunteer in Group ⁇ will be vaccinated ahead of any other volunteers and the profile of adverse events will be reviewed after 48 hours post vaccination.
  • Another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following the first volunteer being vaccinated and at least 1 hour apart from each other.
  • An independent safety review will be conducted by the LSM after vaccination of the first three volunteers. This review will include an assessment of the profile of adverse events and the results of safety blood tests at day 7 post vaccination.
  • the CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 1 and the first volunteer to receive the next incremental dose in group 2. If there are no safety concerns, the remaining volunteers in Group 1 and the first volunteer in group 2 may be vaccinated.
  • Enrolment of the first volunteer in Group 2 will only proceed if the CI and LSM assess the data from the first three vaccinees in Group 1 as indicating that it is safe to do so.
  • the first subject in Group 2 will be vaccinated alone, and a 48 hour gap allowed before vaccinating further subjects in this group.
  • Another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following the first volunteer being vaccinated and at least 1 hour apart from each other.
  • An independent safety review will be conducted by the LSM after vaccination of the first three volunteers. This review will include an assessment of the profile of adverse events and the results of safety blood tests at day 7 post vaccination.
  • the CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 2 and the first volunteer to receive the next incremental dose in group 3. If there are no safety concerns, the remaining volunteers in Group 2 and the first volunteer in group 3 may be vaccinated.
  • Enrolment of the first volunteer in Group 3 will only proceed if the CI and LSM assess the data from the first three vaccinees in Group 2 as indicating that it is safe to do so.
  • the first subject in Group 3 will be vaccinated alone, and a 48 hour gap allowed before vaccinating further subjects in this group.
  • Another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following the first volunteer being vaccinated and at least 1 hour apart from each other.
  • An independent safety review will be conducted by the LSM after vaccination of the first three volunteers. This review will include an assessment of the profile of adverse events and the results of safety blood tests at day 7 post vaccination.
  • the CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 3. If there are no safety concerns, the remaining volunteers in Group 3 may be vaccinated. 8.4.3 Subsequent visits : days 2, 7, 14, 28 , 56 and 182.
  • the volunteer may be admitted to an NHS hospital for observation and further medical management under the care of the Consultant on call.
  • ChAdOxi MERS-S The foreseeable ARs following vaccination with ChAdOxi MERS-S include injection site pain, erythema, warmth, swelling, pruritus, myalgia, arthralgia, headache, fatigue, fever, feverishness, malaise and nausea. 9.3 Expected Serious Adverse Events
  • immunogenicity outcome (ELISpot) data A Wilcoxon signed rank test will permit testing of differences in responses between time points within a group and a Mann- Whitney U test will permit testing of differences in responses between different groups.
  • GCP Good Clinical Practice
  • A-B Hematoxylin & Eosin
  • A Vaccinated with unrelated Adenovirus
  • Lung Normal - 100X
  • This example presents safety and immunogenicity data for the MERS-CoV vaccine ChAdOxi MERS in both human volunteers (MERSooi phase I clinical trial) and camels, the principal animal reservoir for MERS coronavirus (MERS-CoV).
  • MERSooi is a phase I clinical trial to determine the safety and immunogenicity of the candidate MERS-CoV vaccine ChAdOxi MERS in UK healthy adult volunteers.
  • the study groups are shown in the table below.
  • MERS-00101006 14/03/20 18 16/03/20 18 21/03/20 18 Group 1 MERS-00101004 20/03/2018 22/03/2018 27/03/2018
  • Volunteer MER-00101006 reported mild (grade 1) 'Blocked nose', 'Running nose' and 'Cold symptoms' from Dl until Dio post vaccination. They also reported a mild and self-limited 'Loose bowel movement' episode on D5. Volunteer MER-00101011 reported mild and short-lived 'Back pain' on Dl. They also reported mild 'Swollen glands / soreness when swallowing' symptoms from D2 until D6.
  • Volunteers MER-00101016 reported mild and short-lived warmth at injection site at their 30 min observation in clinic which completely resolved by their 60 min review. All volunteers reported at least one systemic AE after vaccination. Systemic AEs were mild or moderate in nature and resolved within 5 days.
  • Volunteer MER-00101012 reported mild (grade 1) Photophobia and Earache on Di which resolved within 24.I1. Volunteer MER-00101010 reported mild Lack of appetite on Di and moderate 'Diarrhoea' on D2. They remained asymptomatic on D3, but then reported gastroenteritis symptoms starting with mild Nausea, Malaise & Vomiting on D4 which got progressively worse until D7 when they experienced severe (grade 3) Nausea, Abdominal Pain, Vomiting and Diarrhoea. Volunteer called their GP who prescribed anti-emetics and advised increased fluids intake.
  • the first Group 3 volunteer is scheduled to be vaccinated with the highest dose of ChAdOxi MERS (5 x 10 10 vp) on the 14th May 2018, pending the review of the safety report data.
  • the Enzyme-Linked ImmunoSpot (ELISPOT) assay is a widely used method for monitoring cellular immune responses. Herein it is used to measure interferon gamma (IFNy produced by T cells (activated in volunteers in response to vaccination) responding to MERS CoV peptides in culture. Ex vivo (18-h stimulation) ELISPOT assays were performed using Multiscreen IP ELISPOT plates (Millipore), human IFN SA-ALP antibody kits (Mabtech) and BCIP NBT-plus chromogenic substrate (Moss Inc). Cells were cultured in RPMI (Sigma) containing 10% heat-inactivated, sterile- filtered fetal calf serum, previously screened for low reactivity (Labtech International).
  • Antigens were tested in duplicate with 250,000 PBMC added to each well of the ex vivo ELISPOT plate.
  • Peptides were 20 amino acids in length, overlapping by 10 amino acids (Neopeptide), assayed in 13 pools of peptides at 10 mg/ml (tables below).
  • ELISPOT responses as spot forming cells (IFN/ producing activated T cells) per million PBMCs were determined for each volunteer pre vaccination, and at 14 and 28 days post vaccination. Results are displayed in figure 11.
  • Figure 11 shows spot forming cells (T cells activated in response to MERS CoV peptides) per million peripheral blood mononuclear cells (the rest of the cells in a given volume of blood).
  • ChAdOxl MERS vaccine All volunteers showed a significant increase in response at day 14 after vaccination (p ⁇ o.05, Kruskall-Wallis test).
  • Antibody responses in vaccinated camels were evaluated using a commercial camel MERS-CoV Si specific ELISA kit from Euroimmun (1).
  • the results of this kit are presented as ratios of optical density (OD) of each serum sample over the OD of a provided positive calibrator, the positive cut-off is 1.1 following manufacturers recommendation.
  • Enzyme-linked immunosorbent assay is an assay technique designed for detecting and quantifying substances such as peptides, proteins, antibodies and hormones. Herein it is used to measure serum antibody responses to the MERS CoV Si protein.
  • ELISA plates were coated with 2 ⁇ g/ml capturing antigen (Si recombinant protein from ATGen Co. Ltd., CA, USA, provided as part of a collaboration with the International Vaccine Institute of Seoul National University, Seoul, Korea), and standard endpoint ELISA protocol was followed, as previously described (2).
  • Sera were prepared in a 10-fold serial dilution in PBS/T and then 50 ⁇ were plated in duplicate wells.
  • Anti-camel IgG conjugated to alkaline phosphatase (Sigma) and PNPP tablet (20 mg p-nitrophenylphosphate, SIGMA) substrate were used in the assay.
  • Camels were purchased from rural markets in the north of Saudi Arabia and maintained at a camel research farm, managed by the Saudi Ministry of Water, Environment, and Agriculture (MEWA). Animal maintenance and procedures were in accordance with King Abdullah International Medical Research Centre (KAIMRC, Riyadh, Kingdom of Saudi Arabia) Institutional Animal Care and Use Committee (IACUC) recommendations, under the regulation of MEWA. Project designation is KAIMRC research project RC16-093.
  • the age information for the camels in the main study is very young.
  • the seropositive camels vary from 1 to 3 years. See the table below - the top set is G01, lower set G02.
  • IU Infectious Units
  • virus neutralisation titre was calculated for camel serum samples collected at o and 28 d.p.i. Serum samples were tested for their ability to neutralise MERS CoV infection in tissue culture by using 100 50% tissue culture infective doses (TCID 50 ) in in Huh-7 cells. Virus neutralisation titre is calculated as the reciprocal of the serum dilution that neutralizes the virus infection. Data points represent individual camels with the mean shown as a horizontal line.

Abstract

The invention relates to a composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is an adenovirus based vector. Suitably said adenovirus based vector is ChAdOx 1, and said spike protein is full length spike protein. More suitably said spike protein is present as a fusion with the tissue plasminogen activator (tPA) sequence in the order N-terminus - tPA - spike protein - C-terminus. The invention also relates to uses and methods.

Description

Com positions and Methods for inducing an im m une response
FIELD OF THE INVENTION The invention relates to induction of immune responses, suitably protective immune responses, against Middle East respiratory syndrome coronavirus (MERS-CoV).
BACKGROUND TO THE INVENTION The Middle East respiratory syndrome coronavirus (MERS-CoV) has infected more than 1900 humans since 2012. The syndrome ranges from asymptomatic and mild cases to severe pneumonia and death. The virus is believed to be circulating in dromedary camels without notable symptoms since the 1980s. Therefore, dromedary camels are considered the only animal source of infection. Neither antiviral drugs nor vaccines are approved for veterinary or medical use despite active research on this area, which is a problem in the art.
Middle East respiratory syndrome (MERS) is caused by a novel betacoronavirus (MERS-CoV) that was isolated in late 2012 in Saudi Arabia (1). The syndrome (MERS) is described as a viral infection that causes fever, cough, and/ or shortness of breath and to a lesser extent gastrointestinal symptoms such as diarrhea (2). Severe disease from MERS-CoV infection can cause respiratory failure and organ failure, and cases can be fatal, especially in patients with co-morbidities such as diabetes and cardiac
complications. However, the infection can be asymptomatic or mild in many cases (3- 7). MERS-CoV has spread to 27 countries and infected more than 1900 humans with a mortality rate of 40% (2). Dromedary camels, especially juveniles, contract the infection and shed the virus, without notable symptoms of disease; this is now known to have been occurring since the early 1980s (8-13). The mechanism of camel to human transmission is still not clear, but several primary cases have been associated with camel contact, which is considered an important risk factor (14-16). Therefore, camels are being considered an intermediate host and one of the sources of MERS-CoV infection (8-13). Other livestock animals such as sheep, goats, cows, chicken, and horses have proved seronegative in many studies (17-20). Further, these animals did not productively contract MERS-CoV when they were inoculated experimentally (21, 22). Therefore, to date, dromedary camels are the only confirmed animal reservoir. There is currently no approved vaccine against MERS-CoV for camels or humans despite active vaccine research and development. A number of vaccine candidates have been developed using various platforms and regimens and have been tested in several animal models (23). These all require multiple doses (administrations) to achieve any effect. Moreover, these can also induce immunopathology such as hypersensitivity. No licensed vaccines or treatments are currently available for MERS-CoV infections. Ongoing disease control strategies have so far relied on minimising contact with dromedary camels, observing standard infection control measures to limit nosocomial transmission, contact tracing and quarantine. Addressing this unmet need for MERS- CoV interventions has been prioritised by the WHO for urgent action 4. A major gap remains in the understanding of key immune mechanisms responsible for protection from disease; whilst MERS-CoV infections elicit high titre neutralising antibody, these do not appear sufficient to provide long-term protection against re-infection 11-13.
Vaccination approaches using full-length S protein of MERS-CoV have proven problematic. Hotez et al. 2014 (Microbes and Infection 2016, pages 529 - 531) discusses calls for rapid development of a safe and effective MERS vaccine. It is disclosed at page 530 that use of subunit vaccines comprising the full spike protein of the severe acute respiratory syndrome (SARS) coronavirus caused eosinophilic immune enhancement in the lungs - a form of hypersensitivity that is very undesirable. The authors note efforts to develop a more restricted receptor binding domain (RBD) of the coronavirus spike protein as a recombinant vaccine against SARS. Such an approach is said to elicit highly effective cross-neutralising antibody responses against SARS in vaccinated animals, but the authors note a general unwillingness to follow this route for generating a MERS vaccine on account of the time and cost involved in this
development and the accepted existence of uncertain risk factors (see page 530, left column, first full paragraph).
The unwillingness around development of a MERS vaccine as reported in Hotez et al is amply supported by the reported studies in related coronaviruses such as that responsible for severe acute respiratory syndrome (SARS-CoV). Jaume et al. 2012
(Hong Kong Med J, Volume 18, page 31) disclose enhancement of SARS-CoV infection by subunit vaccines comprising a recombinant native full length spike protein trimer (triSpike). This study raises very serious safety concerns over the use of full-length coronavirus spike proteins as antigens in vaccine compositions. Such concerns are supported by the findings reported by many research groups including Tseng et al.
2012 (PLoS ONE, Volume 7, document 635421) who discloses that immunisation with a SARS coronavirus vaccine leads to pulmonary immunopathology on challenge with the SARS virus and advises caution in proceeding to application of a coronavirus vaccine in humans.
Efforts to avoid hypersensitivity reactions when immunising with proteins from other coronavirus pathogens have relied on new antigen-adjuvant combinations. Honda- Okubo et al. 2015 (Journal of Virology, Volume 89, page 2995) disclose SARS vaccines comprising inactivated virus or recombinant viral protein formulated with delta inulin adjuvants to ameliorate lung eosinophilic immunopathology.
Iwata-Yoshikawa et al. 2014 (Journal of Virology, Volume 88, page 8597) disclose SARS vaccines comprising a UV-inactivated SARS virus together with Toll-like receptor (TLR) agonists including lipopolysaccharide, poly(U), and poly(I-C) (UV-V+TLR) to reduce excess eosinophilic infiltration in the lungs.
Other investigators have attempted to develop vaccines against coronaviruses using viral vectors. Weingartl et al. 2004 (Journal of Virology, Volume 78, page 12672) disclose that immunisation with modified vaccinia virus Ankara expressing the SARS- CoV spike protein is associated with enhanced hepatitis in ferrets.
Haagmans et al. 2016 (Science, Volume 351, page 77) discloses an orthopoxvirus based vaccine which reduces MERS-CoV viral RNA excretion after MERS-CoV infection in dromedary camels. The authors show that a modified vaccinia virus Ankara (MVA) vaccine expressing the MERS-CoV spike protein confers mucosal immunity in dromedary camels when it is administered in two doses following a prime-boost regimen, and no protective immunity is demonstrated.
Volz et al. 2015 (Journal of Virology, Volume 89, page 8651) replicates and extends the findings of Haagmans et al. in mice. This reference discloses "protective efficacy" of a recombinant MVA vaccine expressing the MERS-CoV spike protein delivered in two doses as a prime-boost regimen. Notably, the first or prime dose produced weak immune responses that only became protective after the second or boost vaccination ("prime-boost" vaccination i.m. in Figure 2).
Malczyk et al. 2015 (Journal of Virology, Volume 89, page 11654) disclose a MERS-CoV vaccine based on a recombinant measles virus vaccine platform that induced a protective immune response only after administration of two doses. No evidence of a protective effect after only a single dose is provided. Thus, there exists no vaccine in the prior art capable of delivering protection against the MERS coronavirus after only a single dose of vaccine.
The present seeks to overcome problem(s) associated with the prior art.
SUMMARY OF THE INVENTION
We describe a combination which comprises a simian adenoviral vector (such as ChAdOxi) delivering a MERS-CoV antigen (the spike protein). This combination has surprising technical benefits including the absence of hypersensitivity type reactions after immunisation. A key benefit delivered by this new combination is the induction of protective immune response after only a single vaccine administration.
In addition, the inventors describe the optional incorporation of a leader
sequence/secretory sequence such as the tissue plasminogen activator (tPA) amino acid sequence fused to the N-terminus of the MERS-CoV spike protein antigen. This triple combination (ChAdOxi + tPA + MERS spike protein) delivers enhanced
immunogenicity. The inventors provide data demonstrating that a single dose of this combined construct delivers 5-log increase in the relevant immune responses - data demonstrating these advantages are provided in the Examples section below.
Thus, in one aspect the invention relates to a composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is an adenovirus based vector.
Suitably said adenovirus based vector is ChAdOx 1. Suitably said spike protein is full length spike protein.
Suitably said spike protein is present as a fusion with the tissue plasminogen activator (tPA) sequence. Suitably the fusion is in the order N-terminus - tPA - spike protein - C- terminus.
Suitably said spike protein has the amino acid sequence given in the MERS genome GenBank accession number KJ650098.1. Suitably said spike protein has the amino acid sequence given in GenBank accession number AHX71946.1.
Suitably the Middle East respiratory syndrome coronavirus may be isolate
Camel/Qatar_2_20i4.
Suitably said spike protein has the amino acid sequence of SEQ ID NO: 1.
SEQ ID NO: 1 - GenBank accession number AHX71946.1
MIHSVFLLMFLLTPTESYVDVGPDSVKSACIEVDIQQTFFDKTW
PRPIDVSKADGI IYPQGRTYSNITITYQGLFPYQGDHGDMYVYSAGHATGTTPQKLFV ANYSQDVKQFANGFVVRIGAAANSTGTVI ISPSTSATIRKIYPAFMLGSSVGNFSDGK MGRFFNHTLVLLPDGCGTLLRAFYCILEPRSGNHCPAGNSHTSFATYHTPATDCSDGN YNRNASLNSFKEYFNLRNCTFMYTYNITEDEILEWFGITQTAQGVHLFSSRYVDLYGG NMFQFATLPVYDTIKYYSI IPHS IRSIQSDRKAWAAFYVYKLQPLTFLLDFSVDGYIR RAIDCGFNDLSQLHCSYESFDVESGVYSVSSFEAKPSGSWEQAEGVECDFSPLLSGT PPQVYNFKRLVFTNCNYNLTKLLSLFSVNDFTCSQISPAAIASNCYSSLILDYFSYPL SMKSDLSVSSAGPISQFNYKQSFSNPTCLILATVPHNLTTITKPLKYSYINKCSRLLS DDRTEVPQLVNANQYSPCVSIVPSTVWEDGDYYRKQLSPLEGGGWLVASGSTVAMTEQ LQMGFGITVQYGTDTNSVCPKLEFANDTKIASQLGNCVEYSLYGVSGRGVFQNCTAVG VRQQRFVYDAYQNLVGYYSDDGNYYCLRACVSVPVSVIYDKETKTHATLFGSVACEHI SSTMSQYSRSTRSMLKRRDSTYGPLQTPVGCVLGLVNSSLFVEDCKLPLGQSLCALPD TPSTLTPRSVRSVPGEMRLAS IAFNHPIQVDQLNSSYFKLS IPTNFSFGVTQEYIQTT IQKVTVDCKQYVCNGFQKCEQLLREYGQFCSKINQALHGANLRQDDSVRNLFASVKSS QSSPI IPGFGGDFNLTLLEPVSI STGSRSARSAIEDLLFDKVTIADPGYMQGYDDCMQ QGPASARDLICAQYVAGYKVLPPLMDVNMEAAYTSSLLGSIAGVGWTAGLSSFAAIPF AQSIFYRLNGVGITQQVLSENQKLIANKFNQALGAMQTGFTTTNEAFRKVQDAVNNNA QALSKLASELSNTFGAI SASIGDI IQRLDVLEQDAQIDRLI GRLTTLNAFVAQQLVR SESAALSAQLAKDKVNECVKAQSKRSGFCGQGTHIVSFVVNAPNGLYFMHVGYYPSNH IEVVSAYGLCDAANPTNCIAPVNGYFIKTNNTRIVDEWSYTGSSFYAPEPITSLNTKY VAPQVTYQNISTNLPPPLLGNSTGIDFQDELDEFFKNVSTS IPNFGSLTQINTTLLDL TYEMLSLQQWKALNESYIDLKELGNYTYYNKWPWYIWLGFIAGLVALALCVFFILCC TGCGTNCMGKLKCNRCCDRYEEYDLEPHKVHVH Suitably the nucleic acid encoding the spike protein antigen, and/ or encoding the tPA- spike protein antigen fusion, is codon optimised for humans. Suitably said polynucleotide sequence comprises the sequence of SEQ ID NO: 3 SEQ ID NO: 3 - Without tPA leader
ATGATCCACTCCGTGTTTCTGCTGATGTTCCTGCTGACCCCCACCGAGAGCTACGTGG ACGTGGGCCCCGATAGCGTGAAGTCCGCCTGTATCGAAGTGGACATCCAGCAGACCT TTTTCGACAAGACCTGGCCCAGACCCATCGACGTGTCCAAGGCCGACGGCATCATCTA TCCACAAGGCCGGACCTACAGCAACATCACCATTACCTACCAGGGCCTGTTCCCATAT CAAGGCGACCACGGCGATATGTACGTGTACTCTGCCGGCCACGCCACCGGCACCACA CCCCAGAAACTGTTCGTGGCCAACTACAGCCAGGACGTGAAGCAGTTCGCCAACGGC TTCGTCGTGCGGATTGGCGCCGCTGCCAATAGCACCGGCACAGTGATCATCAGCCCCA GCACCAGCGCCACCATCCGGAAGATCTACCCCGCCTTCATGCTGGGCAGCAGCGTGG GCAATTTCAGCGACGGCAAGATGGGCCGGTTCTTCAACCACACCCTGGTGCTGCTGCC TGACGGCTGTGGCACACTGCTGAGAGCCTTCTACTGCATCCTGGAACCCAGAAGCGG CAACCACTGCCCTGCCGGCAATAGCCACACCAGCTTCGCCACCTACCACACACCCGCC ACCGATTGCTCCGACGGCAACTACAACCGGAACGCCAGCCTGAACAGCTTCAAAGAG TACTTCAACCTGCGGAACTGCACCTTCATGTACACCTACAATATCACCGAGGACGAGA TCCTGGAATGGTTCGGCATCACCCAGACCGCCCAGGGCGTGCACCTGTTCAGCAGCA GATACGTGGACCTGTACGGCGGCAACATGTTCCAGTTTGCCACCCTGCCCGTGTACGA CACCATCAAGTACTACAGCATCATCCCCCACAGCATCCGGTCCATCCAGAGCGACAGA AAAGCCTGGGCCGCCTTCTACGTGTACAAGCTGCAGCCCCTGACCTTCCTGCTGGACT TCAGCGTGGACGGCTACATCAGACGGGCCATCGACTGCGGCTTCAACGACCTGAGCC AGCTGCACTGCTCCTACGAGAGCTTCGACGTGGAAAGCGGCGTGTACAGCGTGTCCA GCTTCGAGGCCAAGCCTAGCGGCAGCGTGGTGGAACAGGCTGAGGGCGTGGAATGC GACTTCAGCCCTCTGCTGAGCGGCACCCCTCCCCAGGTGTACAACTTCAAGCGGCTGG TGTTCACCAACTGCAATTACAACCTGACCAAGCTGCTGAGCCTGTTCTCCGTGAACGA CTTCACCTGTAGCCAGATCAGCCCTGCCGCCATTGCCAGCAACTGCTACAGCAGCCTG ATCCTGGACTACTTCAGCTACCCCCTGAGCATGAAGTCCGATCTGAGCGTGTCCTCCG CCGGACCCATCAGCCAGTTCAACTACAAGCAGAGCTTCAGCAACCCTACCTGCCTGAT TCTGGCCACCGTGCCCCACAATCTGACCACCATCACCAAGCCCCTGAAGTACAGCTAC ATCAACAAGTGCAGCAGACTGCTGTCCGACGACCGGACCGAAGTGCCCCAGCTCGTG AACGCCAACCAGTACAGCCCCTGCGTGTCCATCGTGCCCAGCACCGTGTGGGAGGAC GGCGACTACTACAGAAAGCAGCTGAGCCCCCTGGAAGGCGGCGGATGGCTGGTGGCT TCTGGAAGCACAGTGGCCATGACCGAGCAGCTGCAGATGGGCTTTGGCATCACCGTG CAGTACGGCACCGACACCAACAGCGTGTGCCCCAAGCTGGAATTCGCCAATGACACCA AGATCGCCAGTCAGCTGGGCAACTGCGTGGAATACTCCCTGTATGGCGTGTCCGGAC GGGGCGTGTTCCAGAATTGCACAGCAGTGGGAGTGCGGCAGCAGAGATTCGTGTACG ATGCCTACCAGAACCTCGTGGGCTACTACAGCGACGACGGCAATTACTACTGCCTGCG GGCCTGTGTGTCCGTGCCCGTGTCCGTGATCTACGACAAAGAGACAAAGACCCACGC CACACTGTTCGGCTCCGTGGCCTGCGAGCACATCAGCTCCACCATGAGCCAGTACTCC CGCTCCACCCGGTCCATGCTGAAGCGGAGAGATAGCACCTACGGCCCCCTGCAGACA CCTGTGGGATGTGTGCTGGGCCTCGTGAACAGCTCCCTGTTTGTGGAAGATTGCAAG CTGCCCCTGGGCCAGAGCCTGTGTGCCCTGCCAGATACCCCTAGCACCCTGACCCCTA GAAGCGTGCGCTCTGTGCCCGGCGAAATGCGGCTGGCCTCTATCGCCTTCAATCACCC CATCCAGGTGGACCAGCTGAACTCCAGCTACTTCAAGCTGAGCATCCCCACCAACTTC AGCTTCGGCGTGACCCAGGAGTACATCCAGACCACAATCCAGAAAGTGACCGTGGAC TGCAAGCAGTACGTGTGCAACGGCTTTCAGAAGTGCGAACAGCTGCTGCGCGAGTAC GGCCAGTTCTGCAGCAAGATCAACCAGGCCCTGCACGGGGCCAACCTGAGACAGGAT GACAGCGTGCGGAACCTGTTCGCCAGCGTGAAAAGCAGCCAGTCCAGCCCCATCATC CCTGGCTTCGGCGGCGACTTTAACCTGACCCTGCTGGAACCTGTGTCCATCAGCACCG GCTCCAGAAGCGCCAGATCCGCCATCGAGGACCTGCTGTTCGACAAAGTGACCATTG CCGACCCCGGCTACATGCAGGGCTACGACGATTGCATGCAGCAGGGCCCAGCCAGCG CCAGGGATCTGATCTGTGCCCAGTATGTGGCCGGCTACAAGGTGCTGCCCCCCCTGAT GGACGTGAACATGGAAGCCGCCTACACCTCCAGCCTGCTGGGATCTATTGCTGGCGT GGGCTGGACAGCCGGCCTGTCTAGCTTTGCCGCCATCCCTTTCGCCCAGAGCATCTTC TACCGGCTGAACGGCGTGGGAATCACCCAGCAGGTGCTGAGCGAGAACCAGAAGCTG ATCGCCAACAAGTTTAACCAGGCACTGGGCGCCATGCAGACCGGCTTCACCACCACCA ACGAGGCCTTCAGAAAGGTGCAGGACGCCGTGAACAACAACGCCCAGGCTCTGAGCA AGCTGGCCTCCGAGCTGAGCAATACCTTCGGCGCCATCAGCGCCTCCATCGGCGACAT CATCCAGCGGCTGGACGTGCTGGAACAGGACGCCCAGATCGACCGGCTGATCAACGG CAGACTGACCACCCTGAACGCCTTCGTGGCACAGCAGCTCGTGCGGAGCGAATCTGC CGCTCTGTCTGCTCAGCTGGCCAAGGACAAAGTGAACGAGTGCGTGAAGGCCCAGTC CAAGCGGAGCGGCTTTTGTGGCCAGGGCACCCACATCGTGTCCTTCGTCGTGAATGC CCCCAACGGCCTGTACTTTATGCACGTGGGCTATTACCCCAGCAACCACATCGAGGTG GTGTCCGCCTATGGCCTGTGCGACGCCGCCAATCCTACCAACTGTATCGCCCCCGTGA ACGGCTACTTCATCAAGACCAACAACACCCGGATCGTGGACGAGTGGTCCTACACCG GCAGCAGCTTCTACGCCCCCGAGCCTATCACCTCCCTGAACACCAAATACGTGGCCCC CCAAGTGACATACCAGAACATCTCCACCAACCTGCCCCCACCCCTGCTGGGCAATAGC ACAGGGATCGACTTCCAGGACGAGCTGGACGAGTTCTTCAAGAACGTGTCCACCTCC ATCCCCAACTTCGGCAGCCTGACCCAGATCAACACCACTCTGCTGGACCTGACCTACG AGATGCTGTCCCTGCAGCAGGTCGTGAAAGCCCTGAACGAGTCCTACATCGACCTGA AAGAGCTGGGGAACTACACCTACTACAACAAGTGGCCTTGGTACATTTGGCTGGGCT TTATCGCCGGCCTGGTGGCCCTGGCCCTGTGCGTGTTCTTCATCCTGTGCTGTACCGG CTGCGGCACCAATTGCATGGGCAAGCTGAAATGCAACCGGTGCTGCGACAGATACGA GGAATACGACCTGGAACCTCACAAAGTGCATGTGCACTGA More suitably said polynucleotide sequence comprises the sequence of SEQ ID NO: 4 SEQ ID NO: 4 - With tPA leader
ATGGACGCCATGAAGAGGGGCCTGTGCTGCGTGCTGCTGCTGTGTGGCGCCGTGTTT GTGTCCGCCAGCCAGGAAATCCACGCCCGGTTCAGACGGATCCACTCCGTGTTTCTGC TGATGTTCCTGCTGACCCCCACCGAGAGCTACGTGGACGTGGGCCCCGATAGCGTGA AGTCCGCCTGTATCGAAGTGGACATCCAGCAGACCTTTTTCGACAAGACCTGGCCCAG ACCCATCGACGTGTCCAAGGCCGACGGCATCATCTATCCACAAGGCCGGACCTACAGC AACATCACCATTACCTACCAGGGCCTGTTCCCATATCAAGGCGACCACGGCGATATGT ACGTGTACTCTGCCGGCCACGCCACCGGCACCACACCCCAGAAACTGTTCGTGGCCAA CTACAGCCAGGACGTGAAGCAGTTCGCCAACGGCTTCGTCGTGCGGATTGGCGCCGC TGCCAATAGCACCGGCACAGTGATCATCAGCCCCAGCACCAGCGCCACCATCCGGAAG ATCTACCCCGCCTTCATGCTGGGCAGCAGCGTGGGCAATTTCAGCGACGGCAAGATG GGCCGGTTCTTCAACCACACCCTGGTGCTGCTGCCTGACGGCTGTGGCACACTGCTGA GAGCCTTCTACTGCATCCTGGAACCCAGAAGCGGCAACCACTGCCCTGCCGGCAATAG CCACACCAGCTTCGCCACCTACCACACACCCGCCACCGATTGCTCCGACGGCAACTAC AACCGGAACGCCAGCCTGAACAGCTTCAAAGAGTACTTCAACCTGCGGAACTGCACCT TCATGTACACCTACAATATCACCGAGGACGAGATCCTGGAATGGTTCGGCATCACCCA GACCGCCCAGGGCGTGCACCTGTTCAGCAGCAGATACGTGGACCTGTACGGCGGCAA CATGTTCCAGTTTGCCACCCTGCCCGTGTACGACACCATCAAGTACTACAGCATCATC CCCCACAGCATCCGGTCCATCCAGAGCGACAGAAAAGCCTGGGCCGCCTTCTACGTGT ACAAGCTGCAGCCCCTGACCTTCCTGCTGGACTTCAGCGTGGACGGCTACATCAGACG GGCCATCGACTGCGGCTTCAACGACCTGAGCCAGCTGCACTGCTCCTACGAGAGCTTC GACGTGGAAAGCGGCGTGTACAGCGTGTCCAGCTTCGAGGCCAAGCCTAGCGGCAGC GTGGTGGAACAGGCTGAGGGCGTGGAATGCGACTTCAGCCCTCTGCTGAGCGGCACC CCTCCCCAGGTGTACAACTTCAAGCGGCTGGTGTTCACCAACTGCAATTACAACCTGA CCAAGCTGCTGAGCCTGTTCTCCGTGAACGACTTCACCTGTAGCCAGATCAGCCCTGC CGCCATTGCCAGCAACTGCTACAGCAGCCTGATCCTGGACTACTTCAGCTACCCCCTG AGCATGAAGTCCGATCTGAGCGTGTCCTCCGCCGGACCCATCAGCCAGTTCAACTACA AGCAGAGCTTCAGCAACCCTACCTGCCTGATTCTGGCCACCGTGCCCCACAATCTGAC CACCATCACCAAGCCCCTGAAGTACAGCTACATCAACAAGTGCAGCAGACTGCTGTCC GACGACCGGACCGAAGTGCCCCAGCTCGTGAACGCCAACCAGTACAGCCCCTGCGTG TCCATCGTGCCCAGCACCGTGTGGGAGGACGGCGACTACTACAGAAAGCAGCTGAGC CCCCTGGAAGGCGGCGGATGGCTGGTGGCTTCTGGAAGCACAGTGGCCATGACCGAG CAGCTGCAGATGGGCTTTGGCATCACCGTGCAGTACGGCACCGACACCAACAGCGTG TGCCCCAAGCTGGAATTCGCCAATGACACCAAGATCGCCAGTCAGCTGGGCAACTGC GTGGAATACTCCCTGTATGGCGTGTCCGGACGGGGCGTGTTCCAGAATTGCACAGCA GTGGGAGTGCGGCAGCAGAGATTCGTGTACGATGCCTACCAGAACCTCGTGGGCTAC TACAGCGACGACGGCAATTACTACTGCCTGCGGGCCTGTGTGTCCGTGCCCGTGTCC GTGATCTACGACAAAGAGACAAAGACCCACGCCACACTGTTCGGCTCCGTGGCCTGC GAGCACATCAGCTCCACCATGAGCCAGTACTCCCGCTCCACCCGGTCCATGCTGAAGC GGAGAGATAGCACCTACGGCCCCCTGCAGACACCTGTGGGATGTGTGCTGGGCCTCG TGAACAGCTCCCTGTTTGTGGAAGATTGCAAGCTGCCCCTGGGCCAGAGCCTGTGTG CCCTGCCAGATACCCCTAGCACCCTGACCCCTAGAAGCGTGCGCTCTGTGCCCGGCGA AATGCGGCTGGCCTCTATCGCCTTCAATCACCCCATCCAGGTGGACCAGCTGAACTCC AGCTACTTCAAGCTGAGCATCCCCACCAACTTCAGCTTCGGCGTGACCCAGGAGTACA TCCAGACCACAATCCAGAAAGTGACCGTGGACTGCAAGCAGTACGTGTGCAACGGCT TTCAGAAGTGCGAACAGCTGCTGCGCGAGTACGGCCAGTTCTGCAGCAAGATCAACC AGGCCCTGCACGGGGCCAACCTGAGACAGGATGACAGCGTGCGGAACCTGTTCGCCA GCGTGAAAAGCAGCCAGTCCAGCCCCATCATCCCTGGCTTCGGCGGCGACTTTAACCT GACCCTGCTGGAACCTGTGTCCATCAGCACCGGCTCCAGAAGCGCCAGATCCGCCATC GAGGACCTGCTGTTCGACAAAGTGACCATTGCCGACCCCGGCTACATGCAGGGCTAC GACGATTGCATGCAGCAGGGCCCAGCCAGCGCCAGGGATCTGATCTGTGCCCAGTAT GTGGCCGGCTACAAGGTGCTGCCCCCCCTGATGGACGTGAACATGGAAGCCGCCTAC ACCTCCAGCCTGCTGGGATCTATTGCTGGCGTGGGCTGGACAGCCGGCCTGTCTAGC TTTGCCGCCATCCCTTTCGCCCAGAGCATCTTCTACCGGCTGAACGGCGTGGGAATCA CCCAGCAGGTGCTGAGCGAGAACCAGAAGCTGATCGCCAACAAGTTTAACCAGGCAC TGGGCGCCATGCAGACCGGCTTCACCACCACCAACGAGGCCTTCAGAAAGGTGCAGG ACGCCGTGAACAACAACGCCCAGGCTCTGAGCAAGCTGGCCTCCGAGCTGAGCAATA CCTTCGGCGCCATCAGCGCCTCCATCGGCGACATCATCCAGCGGCTGGACGTGCTGG AACAGGACGCCCAGATCGACCGGCTGATCAACGGCAGACTGACCACCCTGAACGCCT TCGTGGCACAGCAGCTCGTGCGGAGCGAATCTGCCGCTCTGTCTGCTCAGCTGGCCA AGGACAAAGTGAACGAGTGCGTGAAGGCCCAGTCCAAGCGGAGCGGCTTTTGTGGCC AGGGCACCCACATCGTGTCCTTCGTCGTGAATGCCCCCAACGGCCTGTACTTTATGCA CGTGGGCTATTACCCCAGCAACCACATCGAGGTGGTGTCCGCCTATGGCCTGTGCGA CGCCGCCAATCCTACCAACTGTATCGCCCCCGTGAACGGCTACTTCATCAAGACCAAC AACACCCGGATCGTGGACGAGTGGTCCTACACCGGCAGCAGCTTCTACGCCCCCGAG CCTATCACCTCCCTGAACACCAAATACGTGGCCCCCCAAGTGACATACCAGAACATCT CCACCAACCTGCCCCCACCCCTGCTGGGCAATAGCACAGGGATCGACTTCCAGGACGA GCTGGACGAGTTCTTCAAGAACGTGTCCACCTCCATCCCCAACTTCGGCAGCCTGACC CAGATCAACACCACTCTGCTGGACCTGACCTACGAGATGCTGTCCCTGCAGCAGGTCG TGAAAGCCCTGAACGAGTCCTACATCGACCTGAAAGAGCTGGGGAACTACACCTACTA CAACAAGTGGCCTTGGTACATTTGGCTGGGCTTTATCGCCGGCCTGGTGGCCCTGGC CCTGTGCGTGTTCTTCATCCTGTGCTGTACCGGCTGCGGCACCAATTGCATGGGCAAG CTGAAATGCAACCGGTGCTGCGACAGATACGAGGAATACGACCTGGAACCTCACAAA GTGCATGTGCACTGA
Suitably administration of a single dose of said composition to a mammalian subject induces protective immunity in said subject.
In one aspect, the invention relates to a composition as described above for induction of an immune response against MERS-CoV.
In one aspect, the invention relates to a composition as described above for induction of an immune response against MERS-CoV, wherein a single dose of said composition is administered.
In one aspect, the invention relates to a composition as described above for induction of an immune response against MERS-CoV, wherein said composition is administered once. Suitably said composition is administered once per 6 months. Suitably said composition is administered once per 12 months.
Suitably the primary vaccination regimen is one dose, whereas all prior art MERS vaccines require two doses to provide protection. In some embodiments it may be desired to re-administer at a later date, not less than 6 months after the first immunisation.
In one aspect, the invention relates to a composition as described above for preventing MERS-CoV infection.
In one aspect, the invention relates to a composition as described above for preventing MERS-CoV infection, wherein a single dose of said composition is administered.
In one aspect, the invention relates to a composition as described above for preventing MERS-CoV infection, wherein said composition is administered once. Suitably said composition is administered once per 6 months. Suitably said composition is administered once per 12 months.
In one aspect, the invention relates to use of a composition as described above in medicine.
In one aspect, the invention relates to use of a composition as described above in the preparation of a medicament for prevention of MERS-CoV infection. In another aspect, the invention relates to use of a composition as described above in inducing an immune response against middle eastern respiratory syndrome coronavirus (MERS-CoV). In another aspect, the invention relates to use of a composition as described above in immunising a subject against MERS-CoV. In another aspect, the invention relates to use of a composition as described above in prevention of MERS-CoV infection.
A method of inducing an immune response against middle eastern respiratory syndrome coronavirus (MERS-CoV) in a mammalian subject, the method comprising administering a composition as described above to said subject.
Suitably a single dose of said composition is administered to said subject.
Suitably said composition is administered once.
Suitably said composition is administered once per 6 months.
Suitably said composition is administered once per 12 months.
Suitably said composition is administered by a route of administration selected from a group consisting of subcutaneous, intranasal, aerosol, nebuliser, intradermal and intramuscular.
Most suitably said administration is intramuscular.
In one aspect the invention relates to an adeno-based viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV). Suitably the adeno-based viral vector is ChAdOx 1.
In one aspect, the invention relates to a ChAdOx comprising a polynucleotide encoding glycoprotein S from the MERS virus. Suitably said adeno-based viral vector has the sequence and/or construction as described in one or more of the examples.
In one aspect, the invention relates to a method of raising an immune response by administering the adeno-based viral vector as described above.
In one aspect, the invention relates to the adeno-based viral vector as described above for use in preventing MERS infection.
In one aspect, the invention relates to the adeno-based viral vector as described above for use in raising an anti-MERS immune response.
DETAILED DESCRIPTION It is believed that the combination of ChAdOxi with the MERS antigen used in this work has not been disclosed previously and is therefore novel. It is believed that the prior art suggests a prejudice against the use of MERS antigens to develop a vaccine - Hotez (ibid. ) has identified a risk of MERS vaccines based on S protein as possibly being affected by the risk of inducing eosinophilic hypersensitivity which was seen with S protein based SARS vaccine. Hypersensitivity in mice with MERS vaccines from the same laboratory has been noted. It is an advantage of the invention that no such hypersensitivity is seen.
Prior art prime-boost using MVA based vaccine candidates produces a very robust immune response as has been demonstrated repeatedly with a large number of different antigens in various indications. It is an advantage of the invention that one administration of ChAdOx-MERS raised an immune response equivalent to that of a homologous MVA prime-boost regimen against the same antigen. This comparable response from a single dose of ChAdOx-MERS was unexpected and has a number of benefits including quicker, simpler treatment and cheaper manufacturing and treatment.
We disclose that ChAdOxi and MVA based vaccine candidates against MERS-CoV elicit neutralising antibodies and cellular immune responses in mice.
We describe four vaccines against MERS-CoV based on ChAdOxi and MVA viral vectors, two vaccines per vector. All vaccines contained the full-length spike gene of MERS-CoV; ChAdOxi MERS vaccines were produced with or without the leader sequence of the human tissue plasminogen activator gene (tPA) where MVA MERS vaccines were produced with tPA, and either the 1T1H5 or F11 promoter driving expression of the spike gene. All vaccine candidates were evaluated in a mouse model in prime only or prime-boost regimens. ChAdOxi MERS with tPA induced higher neutralising antibodies than ChAdOxi MERS without tPA. A single dose of ChAdOxi MERS with tPA elicited cellular immune responses as well as neutralising antibodies - these were boosted to a significantly higher level by MVA MERS. The humoral immunogenicity of a single dose of ChAdOxi MERS with tPA was equivalent to two doses of MVA MERS (also with tPA). MVA MERS with Π1Η5 or F11 promoter induced similar antibody levels. The F11 promoter enhanced the cellular immunogenicity of MVA MERS to significantly higher magnitudes. In conclusion, our study showed that MERS-CoV vaccines could be optimised by utilising different viral vectors, and/or various genetic designs of the vectors, and/ or different regimens to increase
immunogenicity. ChAdOxi and MVA vectored vaccines have been safely evaluated in camels and humans and these MERS vaccine candidates are demonstrated to be promising and the data shown herein support their industrial application for example that they are expected to be effective in camels and in clinical trials in humans.
We show development of MERS-CoV vaccine candidates that are based on two different viral vectors: Chimpanzee Adenovirus, Oxford University #1 (ChAdOxi) (26) and Modified Vaccinia virus Ankara (MVA) (27, 28). Each viral vector was developed by generating two alternative versions, resulting in four vaccine candidates that all encode the same complete MERS-CoV spike gene (S). The two ChAdOxi based vaccines were produced with or without the signal peptide of the human tissue plasminogen activator gene (tPA) at the N terminus. Previous studies have shown that encoding tPA upstream of recombinant antigens enhanced immunogencity, although results differed depending on the antigens employed. The tPA encoded upstream of influenza A virus nucleoprotein, in a DNA vector, enhanced both cellular and humoral immune responses in mice (29, 30), whereas the same leader sequence resulted in increased humoral sequences but decreased cellular responses to fflV Gag (30). The two MVA based vaccines were produced with either the 1T1H5 or F11 poxviral promoter driving antigen expression, both including the tPA sequence at the N terminus of MERS-CoV Spike protein. Previously, we reported the ability of the strong early F11 promoter to enhance cellular immunogenicity of vaccine antigen candidates for malaria and influenza, as compared to utilising P7.5 or Π1Η5 early/late promoters which resulted in a lower level of gene expression immediately after virus infection of target cells, but higher levels at a later stage (31). Here, we continue to assess the F11 promoter in enhancing cellular immunogenicity, and to investigate its ability to impact on humoral immune responses. The four vaccine candidates were evaluated in a number of different regimens in mouse models that showed a single dose of ChAdOxi MERS inducing higher cellular and humoral immunogenicity than a single dose of MVA MERS, or equivalent to two doses of MVA MERS. ChAdOxi based vaccines have been tested in different animal models, including camels (32), and in human clinical trials and proved safe and immunogenic (33). Therefore, based on our data, ChAdOxi MERS can be readily employed for use as a MERS vaccine in humans. Furthermore, utilising ChAdOxi MERS for camel vaccination can serve the one-health approach whereby blocking MERS-CoV transmission in camels is expected to prevent human infections. The inventors identified the major surface antigen of MERS-CoV as the Spike (S protein) and demonstrated that ChAdOxi expressing this protein induces the production of anti-S antibodies, including neutralising antibodies, after a single intramuscular immunisation. This single dose induced a response equivalent to that provided by a prime-boost administration of the same antigen in another viral vector as demonstrated in mice. Studies in camels and humans are outlined below.
Hypersensitivity responses were not seen in vaccinated animals and human subjects.
APPLICATIONS
The invention finds particular application in prevention or containment of outbreaks of MERS. In this scenario, it is extremely advantageous to achieve protective immunity with only a single dose of vaccine. In the special considerations which apply to emerging pathogens such as MERS, there is typically not time to give two doses. It is also exceptionally difficult to recall patients for their second dose. For example, patients may need to walk from village to village in order to receive their dose and have many pressures on their time which can prevent attendance for a second dose. For example, they may have to travel from distance to receive a dose, or they may need to attend to their livelihoods which can prevent them from attending for more than a single dose. Thus, there is a need for a rapid onset of protection, which need is met by the present invention. The present invention also advantageously allows for avoidance of quarantine of patients in between doses which might otherwise be required since acquiring the infection in between doses would be potentially deleterious for the individual.
The invention finds particular application in the immunisation of camels. Camels are believed to be the only, or at least the main, animal source of human infection with MERS. For similar reasons as outlined above, it is a technical benefit that the invention delivers protective immunity with only a single dose - this advantage extends to camels as well as to human or other subjects.
Suitably the subject is a camel.
Suitably the subject is a human.
As acknowledged in the background to the invention, Malczyk et al. 2015 disclose a measles virus vaccine comprising the MERS spike protein. However, protective immunity is only demonstrated for this measles based vaccine after two doses. Thus, the invention delivers benefits compared to Malczyk et al. 2015. Suitably the method is a method of immunising.
Suitably the immune response comprises a humoral response. Suitably the immune response comprises an antibody response. Suitably the immune response comprises a neutralising antibody response.
Suitably the immune response comprises a cell mediated response. Suitably the immune response comprises cell mediated immunity (CMI). Suitably the immune response comprises induction of CD8+ T cells. Suitably the immune response comprises induction of a CD8+ cytotoxic T cell (CTL) response.
Suitably the immune response comprises both a humoral response and a cell mediated response.
Suitably the immune response comprises protective immunity. Suitably the composition is an antigenic composition.
Suitably the composition is an immunogenic composition. Suitably the composition is a vaccine composition.
Suitably the composition is a pharmaceutical composition.
Suitably the composition is formulated for administration to mammals, suitably to primates or camelids. Camelids are members of the biological family Cam elidae, the only currently living family in the suborder Tylopoda. The extant members of this group are: dromedary camel, Bactrian camels, wild or feral camels, llamas, alpacas, vicunas, and guanacos. More suitably the composition is formulated for administration to primates, camels, alpacas, llamas, vicunas, pigs, non-human primates or humans, more suitably to primates or camels, most suitably to humans
It maybe useful to protect pigs against MERS. The virus does productively infect pigs (does not apply to sheep, cattle, chickens). Although there are few farmed pigs in the middle east, there are wild boars which may get infected and form another zoonotic reservoir which may then spread, or if MERS gets into farmed pigs somewhere else in the world, such as US or China, it would be a major problem. Thus the invention finds utility in this application. Suitably the composition is formulated taking into account its route of administration. Suitably the composition is formulated to be suitable for the route of administration specified. Suitably the composition is formulated to be suitable for the route of administration selected by the operator or physician.
DATABASE RELEASE
Sequences deposited in databases can change over time. Suitably the current version of sequence database(s) are relied upon. Alternatively, the release in force at the date of filing is relied upon.
As the skilled person knows, the accession numbers may be version/dated accession numbers. The citeable accession numbers for the current database entry are the same as above, but omitting the decimal point and any subsequent digits.
GenBank is the NIH genetic sequence database, an annotated collection of all publicly available DNA sequences (National Center for Biotechnology Information, U.S.
National Library of Medicine 86oo Rockville Pike, Bethesda MD, 20894 USA; Nucleic Acids Research, 2013 Jan;4i(Di):D36-42) and accession numbers provided relate to this unless otherwise apparent. Suitably the GenBank database release referred to is 15 April 2017, NCBI-GenBank Release 219.0.
UniProt (Universal Protein Resource) is a comprehensive catalogue of information on proteins ('UniProt: a hub for protein information' Nucleic Acids Res. 43: D204-D212 (2015).). For the avoidance of doubt, UniProt Release 20i5_n is relied upon.
In more detail, the UniProt consortium European Bioinformatics Institute (EBI), SIB Swiss Institute of Bioinformatics and Protein Information Resource (PIR)'s UniProt Knowledgebase (UniProtKB) Release 20i7_05, (io-May-2017) is relied upon. ADVANTAGES
The inventors have previously published a paper immunising against River Valley Fever (RVF). In this paper, protective immunity is achieved after only one dose of ChAdOxi containing the RVF antigen. However, RVF is not a coronavirus. RVF is a completely different type of virus. Thus, it would not be correct to extrapolate from the RVF study as if it could be expected that a single dose might produce protective immunity in other viral families. It is an advantage of the invention that a single dose delivers protective immunity against MERS.
Weingartl et al. 2004 {ibid) discloses the use of a MVA vector to deliver antigen to ferrets. However, Weingartl et al. observed pathology using this viral delivery system. Therefore, it would be incorrect to extrapolate from this paper as if merely using a viral vector would be the answer to achieving single dose immunity.
It should be noted that numerous publications (as explained in the background to the invention above) have attempted to use diverse viral vectors to deliver the MERS spike protein as an antigen for vaccination, but those prior art approaches have each suffered from immunopathology (sometimes described as hypersensitivity) effects such as eosinophil infiltration of the lungs. These very serious medical complications are advantageously overcome by the present invention.
This view in the art is further established by the Hotez review {ibid) which noted the numerous problems causing immunopathology in attempting to immunise against the SARS coronavirus. The Hotez review clearly identifies this as a problem extending to MERS vaccines. Advantageously, the present invention delivers a solution to this problem.
Thus, the invention delivers the advantage of avoiding immunopathology. The invention provides the advantage of avoiding hypersensitivity.
The invention possesses the advantage of protective immunity after single dose (single administration).
The phrase "protective immune response" or "protective immunity" as used herein means that the composition is capable of generating a protective response in a host organism, such as a human or a non-human mammal (e.g. a camel), to whom it is administered according to the invention. Suitably a protective immune response protects against subsequent infection or disease caused by MERS-CoV. These advantages flow from the particular combination of features as set out in the claims. SPIKE PROTEIN
Suitably the antigen in the MERS spike protein. Suitably the full length spike protein is used.
Suitably full length means each amino acid in the spike protein is included.
An exemplary spike protein is as disclosed in accession number AHX71946.1 - SEQ ID NO: 1 (above).
It may be possible to use only the Si domain of the spike protein, or only the soluble part of the spike protein, or only the receptor binding domain of the spike protein. However, most suitably according to the present invention the full length spike protein is used.
By choosing the full length spike protein, advantageously the correct confirmation of the protein in assured. Truncated proteins can assume unnatural conformations. This drawback is avoided by using the full length protein.
A further advantage of using the full length spike protein is that it allows for better T- cell responses. Without wishing to be bound by theory, it is believed that the more amino acid sequences present, then the more potential targets there are for the T-cell responses. Thus, suitably every amino acid of the wild type spike protein is included in the antigen of the invention. tPA tPA (tissue plasminogen activator) - more specifically the tPA leader sequence - is suitably fused to the MERS spike protein antigen of the invention. Suitably tPA is fused to the N-terminus of the spike protein sequence.
Suitably tPA leader sequence means the tPA amino acid sequence of SEQ ID NO: 5 SEQ ID NO: 5
MDAMKRGLCCVLLLCGAVFVSASQEIHARFRR In the above SEQ ID NO: 5 the C terminal 'RR' is not actually part of the tPA leader sequence. It comes from the fusion of two restriction sites. Suitably the tPA leader sequence may be used with or without the C terminal 'RR' e.g. SEQ ID NO: 7 or SEQ ID NO: 8. Most suitably the sequence is used as shown in SEQ ID NO: 5.
The underlined A is P in the naturally occurring tPA leader sequence. The P->A mutation has the advantage of improved antigen secretion.
Suitably the tPA leader sequence may be used with or without the P->A mutation, i.e. suitably the tPA leader sequence may be used as SEQ ID NO: 5 or SEQ ID NO: 6.
SEQ ID NO: 6
MDAMKRGLCCVLLLCGAVFVSPSQEIHARFRR
SEQ ID NO: 7 (=SEQ ID NO: 5 without C-terminal 'RR')
MDAMKRGLCCVLLLCGAVFVSASQEIHARF SEQ ID NO: 8 (=SEQ ID NO: 6 without C-terminal 'RR')
MDAMKRGLCCVLLLCGAVFVSPSQEIHARF
More suitably the sequence is used with the P->A mutation (with or without the C terminal 'RR'). Most suitably the sequence is used as shown in SEQ ID NO: 5. An exemplary nucleotide sequence encoding tPA, which has been codon optimised for human codon usage, is as shown in SEQ ID NO: 9 (this is the sequence encoding SEQ ID NO: 5):
ATGGACGCCATGAAGAGGGGCCTGTGCTGCGTGCTGCTGCTGTGTGGCGCCGTGTTT GTGTCCGCCAGCCAGGAAATCCACGCCCGGTTCAGACGG
It is believed that tPA promotes secretion of proteins to which it is fused. It is believed that tPA increases expression of proteins to which it is fused. Notwithstanding the underlying mechanism, the advantage in the invention of fusing tPA to the N-terminus of the spike protein antigen is that improved immunogenicity is achieved. Thus, most suitably the antigen of the invention is provided as a fusion with tPA. Most suitably the tPA is fused to the N-terminus of the spike protein antigen.
Suitably the antigen does not comprise any further sequence tags. Suitably the antigen does not comprise any further linker sequences.
Adeno-based viral vectors Any suitable adeno-based viral vector may be used.
In more detail, any replication-deficient viral vector, for human use preferably derived from a non-human adenovirus may be used. For veterinary use Ad5 may be used.
ChAd0x2 is an example of a suitable non-human adenovirus vector for human use.
Most suitably the adeno-based viral vector is ChAdOxi.
ChAdOxi
ChAdOxi is described in Dicks MDJ, Spencer AJ, Edwards NJ, Wadell G, Bojang K, et al. (2012) A Novel Chimpanzee Adenovirus Vector with Low Human Seroprevalence: Improved Systems for Vector Derivation and Comparative Immunogenicity. PLoS ONE 7(7): 640385, and in WO2012/172277. Both these documents are hereby incorporated herein by reference, in particular for the specific teachings of the ChAdOxi vector, including its construction and manufacture. For insertion of the nucleotide sequence encoding spike protein, suitably the El site may be used, suitably with the hCMV IE promoter. Suitably the short or the long version may be used; most suitably the long version as described in WO2008/122811, which is specifically incorporated herein by reference for the teaching of the promoters, particularly the long promoter.
It is also possible to insert antigens at the E3 site, or close to the inverted terminal repeat sequences, if desired.
In addition, a clone of ChAdOxi containing GFP is deposited with the ECACC: a sample of E. coli strain SW1029 (a derivative of DH10B) containing bacterial artificial chromosomes (BACs) containing the cloned genome of AdChOXi (pBACe3.6 AdChOxi (E4 modified) TIPeGFP, cell line name "AdChOxi (E4 modified) TIPeGFP") was deposited by Isis Innovation Limited on 24 May 2012 with the European Collection of Cell Cultures (ECACC) at the Health Protection Agency Culture Collections, Health Protection Agency, Porton Down, Salisbury SP4 oJG, United Kingdom under the Budapest Treaty and designated by provisional accession no. 12052403. Isis
Innovation Limited is the former name of the proprietor/ applicant of this
patent/ application. ChAd0x2
The nucleotide sequence of the ChAd0x2 vector (with a Gateway™ cassette in the El locus) is shown in SEQ ID NO. 2 This is a viral vector based on Chimpanzee adenovirus C68. (This is the sequence of SEQ ID NO: 10 in gb patent application number
1610967.0).
In addition, a clone of ChAd0x2 containing GFP is deposited with the ECACC: deposit accession number 16061301 was deposited by Isis Innovation Limited on 13 June 2016 with the European Collection of Cell Cultures (ECACC) at the Health Protection Agency Culture Collections, Health Protection Agency, Porton Down, Salisbury SP4 oJG, United Kingdom under the Budapest Treaty. Isis Innovation Limited is the former name of the proprietor/applicant of this patent/application.
ADMINISTRATION ROUTE
In principle any suitable route of administration may be used.
Suitably the route of administration is selected from group consisting of subcutaneous, intranasal, aerosol, nebuliser, intradermal and intramuscular.
Suitably the route of administration is selected from a group consisting of intranasal, aerosol and intramuscular.
Suitably the route of administration is selected from a group consisting of intranasal and intramuscular.
Most suitably the route of administration is intramuscular.
The route of administration maybe applied to camels or humans.
DOSE
It should be noted that there are alternate ways of describing the dose for adenoviral vectors. • Viral particles - vp/mL. This refers to the count of total viral particles administered.
• Infectious units - i.u./ mL. This refers to the number of infectious units
administered, and can be correlated more accurately with immunogenicity.
By convention, clinical trials in the UK tend to provide the dose in terms of viral particles.
Preferred doses according to the present invention are:
Humans, preferred range is from io9 to 1011 viral particles
Camels, io? to io12 viral particles
Pigs, io8 to io10 viral particles.
Suitably no adjuvant is administered with the viral vector of the invention.
Suitably the viral vector of the invention is formulated with simple buffer. An exemplary buffer may be as shown below under the heading 'Formulation'.
FURTHER FEATURES
Suitably the nucleic acid sequence is codon optimised for mammalian codon usag suitably for camel or human codon usage, most suitably for human codon usage.
Suitably a container containing a composition as described above is provided. Suitably said container may be a vial. Suitably said container may be a syringe.
Suitably a nebuliser containing a composition as described above is provided.
Suitably a nasal applicator containing a composition as described above is provided. Suitably an inhaler containing a composition as described above is provided.
Suitably a pressurised canister containing a composition as described above is provided.
A method of making a composition as described above is provided, said method comprising preparing a nucleic acid encoding the MERS-CoV spike protein, optionally fused to the tPA protein, and incorporating said nucleic acid into an adeno-based viral vector, suitably a ChAdOxi vector. Suitably the nucleic acid is operably linked to a promoter suitable for inducing expression of said MERS-CoV spike protein (or MERS- CoV spike protein-tPA fusion protein) when in a mammalian cell such as a human or camel cell.
FORMULATION
The ChAdOxi formulation buffer, as used for the clinical product is: FORMULATION BUFFER COMPONENTS
1. 10 mM Histidine
2. 7.5 % Sucrose
3· 35 mM Sodium chloride
4· 1 mM Magnesium chloride
5· 0.1 % Polysorbate 80
6. O.l mM EDTA
7- 0.5% Ethanol
8. Hydrochloric acid (for pH adjustment to ~pH 6.6)
Formulated in Water for Injection Ph Eur. Formulations for other administration routes such as aerosol will be adjusted accordingly by the skilled operator.
Suitably the composition and/or formulation does not comprise adjuvant. Suitably adjuvant is omitted from the composition and/or formulation of the invention.
MVA - MERS SPIKE PROTEIN
An MVA-MERS spike protein vaccine has been described in the prior art, for example by Volz et al. 2015/Haagmans et al. 2016 {ibid). We also disclose an MVA vector carrying the MERS spike protein, which is described in more detail in the Examples section. The MVA vector described herein features a 1T1H5/F11 promoter system in one embodiment, or relies on a standard F11 promoter in another embodiment. In any case, these promoter systems are known in the art, for example in published patent US 9> 273, 327B2 (Cottingham - granted 1 March 2016 - 'Poxvirus Expression System') - this document is hereby incorporated by reference, in particular for the specific teachings of promoter(s) for use herein. In the context of the present invention, MVA vector delivering MERS spike protein is taught as a useful optional boost in an immunisation regimen as described. The first dose should preferably be ChAdOxi-MERS spike protein (most preferably comprising the tPA fusion to the N-terminus of the spike protein) and the optional second administration preferably comprises MVA-MERS spike protein.
As will be apparent, the main focus of the invention is in provision of a single dose MERS vaccine. However, in this embodiment featuring a second (boosting) administration, preferably the second (boosting) administration is in a different viral vector i.e. a heterologous "prime-boost" regime. Suitably the second (boosting) administration comprises a MVA vector. This finds particular application for example in inducing immunity in subjects such as healthcare workers. It is a particular problem that healthcare workers can contract a MERS infection. Since they are typically in good health themselves, this has very little effect, if any, on their general health. However, when they are infected they can of course excrete virus, which can go on to infect immune compromised patients in their care with disastrous consequences. Therefore, there is a special and particular problem in the immunisation of healthcare workers. A durable and long lasting immunity is desired for these professionals. Therefore, whilst it is a core tenet of the invention that a single dose of vaccine provides protection against MERS infection, in the special case of healthcare workers the protective immunity is desired to last as far as possible into the future. For most applications, a temporary immunity ('temporary' contrasted with a lifelong immunity) is entirely adequate to protect the individual and/ or to halt the spread of the infection. However, in the special case of healthcare workers any way of extending their immunity in time is itself additionally advantageous. In this scenario, we teach a "prime-boost" regimen comprising a first administration of an adenoviral vector-MERS composition such as a ChAdOx-MERS composition, followed by a second (boosting) administration of a viral vector comprising the MERS spike protein, such as a MVA vector expressing the MERS spike protein. Thus, in the inventors' opinion, MVA-MERS spike protein has limited use but may find particular application as a heterologous boost following a ChAdOx- MERS spike protein priming vaccination. In one embodiment the order of
immunisations may be reversed so that the MVA-MERS vaccine is administered first followed by the ChAdOx-MERS vaccine after an interval of typically 1 - 8 weeks. Similarly, MVA boosting for extended duration of immunity may also be useful for subjects who are occupationally exposed to camels, e.g. camel shepherds or
slaughterhouse workers. They may also become infected, experience only mild illness but transmit the infection to family or community members. There is a slightly higher rate of seropositivity to MERS in these workers than the general population. Thus this embodiment of the invention may be applied to such subjects who are occupationally exposed to camels, most suitably camel shepherds or slaughterhouse workers.
Thus in one aspect the invention provides a method of inducing an immune response against middle eastern respiratory syndrome coronavirus (MERS-CoV) in a mammalian subject, the method comprising
(i) administering a composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is an adenovirus based vector to said subject, and
(ii) administering a composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is a MVA based vector to said subject.
Suitably step (i) is a priming composition.
Suitably step (ii) is a boosting composition.
Suitably step (ii) is carried out 1-8 weeks after the step (i), most suitably 4 weeks after step (i).
Further particular and preferred aspects are set out in the accompanying independent and dependent claims. Features of the dependent claims may be combined with features of the independent claims as appropriate, and in combinations other than those explicitly set out in the claims.
Where an apparatus feature is described as being operable to provide a function, it will be appreciated that this includes an apparatus feature which provides that function or which is adapted or configured to provide that function.
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention will now be described by way of example, with reference to the accompanying drawings, in which:
Figure 1: Construction of MERS-CoV vaccine candidates
A: schematic representation of ChAdOxi and MVA based vaccines, each encodes the same MERS-CoV spike gene (MERS-CoV S). The S gene was inserted into the El region of ChAdOxi genome or into the F11L locus of MVA genome. tPA: Human tissue plasminogen activator (tPA) signal peptide sequence. IE CMV: The human
cytomegalovirus major immediate early promoter. Π1Η5 and F11: Poxviral promoters. LHA: left homology arm sequence. RHA: right homology arm sequence. B : The expression of spike transgene, cloned into a plasmid vector, was validated by transfection into an African green monkey kidney cell line (Vero cells) and confirmed by immunostaining. C: Untransfected cells control. Green colour represents detection of the spike protein. Blue colour represents nuclei by staining nucleic acid with DAPI. Figure 2 : Antibody responses to ChAdOxi MERS vaccine candidates.
BALB/c mice (n = 6) were immunised with a single injection of ChAdOxi MERS that either encodes or lacks tPA signal peptide, intramuscularly at ixioA8 IU. A control group of mice were immunised with ChAdOxi expressing eGFP instead of MERS-CoV S gene. Serum samples were collected at 14 and 28 days post immunisation (d.p.i.). Si- binding antibodies were detected at both time points by ELISA (A) and neutralisation activity of the antibodies were confirmed by MERS-CoV pseudotyped viral particles (MERSpp) neutralisation assay (B) or neutralisation assay (C). Individual data points are shown with line as the median. Data are representative of two independent experiments. Statistical significance by Kruskal-Wallis test is shown.
Figure 3 : Cellular im m une responses to ChAdOxi MERS vaccine candidate . BALB/c mice (n = 6) were immunised with a single injection of ChAdOxi MERS that encodes tPA signal peptide intramuscularly at ixioA8 IU. Twenty eight days post- immunisation, IFN-γ ex vivo ELISpot (A) or Intracellular Cytokine Staining (ICS (B)), were performed to determine the percentage of splenic IFN-γ secreting CD4+ and CD8+ after in vitro re-stimulation with a MERS-CoV S-specific peptide. Individual data points are shown with line as the median (A) or error bars as the SD (B). Data are representative of two independent experiments.
Figure 4 : Hum oral and cellular im m unogenicity of heterologous ChAdOxi MERS and MVA MERS vaccination.
BALB/c mice (n = 6) were immunised with ChAdOxi MERS that encodes tPA signal peptide, intramuscularly at ixioA8 IU. At 28 d.p.i. mice were boosted with MVA MERS at ixioA6 pfu. MVA MERS candidates either contain Π1Η5 or F11 promoter for transgene expression. Serum samples were collected at 28 (post-prime) and 42 (post- boost) d.p.i. Si-binding antibodies were detected at both time points by ELISA (A) and neutralisation activity of serum antibodies at 42 d.p.i. were confirmed by MERSpp neutralisation assay (B). At 42 d.p.i, IFN-γ ex vivo ELISpot (C) or Intracellular
Cytokine Staining (ICS (D)) were performed to determine the percentage of CD8+ IFN- Y+ splenocytes after in vitro re-stimulation with a MERS-CoV S-specific peptide. ICS of splenocytes re-stimulated with MVA- specific peptides (F(G)2 and E3) was also performed (E and F). Individual data points are shown with line as the median. Data are representative of two independent experiments. Statistical significance by Kruskal- Wallis test is shown. Symbols are closed squares () for ChAdOxi prime responses, open circles (o) for 1T1H5-MVA boost responses, and closed circles (·) for F11-MVA boost responses.
Figure 5 : Hum oral and cellular im m unogenicity of hom ologous MVA MERS vaccination.
BALB/c mice (n = 6) were immunised with MVA MERS at ixioA6 pfu, intramuscularly, in a homologous prime-boost vaccination with three-weeks interval. MVA MERS candidates either contain Π1Η5 or F11 promoter for transgene expression. Serum samples were collected at 21 (post-prime) and 42 (post-boost) d.p.i. Si-binding antibodies were detected at both time points by ELISA (A) and neutralisation activity of serum antibodies at 42 d.p.i. were confirmed by MERSpp neutralisation assay (B). At 42 d.p.i splenocytes were processed and re-stimulated with a MERS-CoV S-specific peptide (CD8+ T cell specific) for IFN-γ ex vivo ELISpot (C). ICS of splenocytes re- stimulated with MVA-specific peptides (F(G)2 and E3) was also performed (D and E) as was performed in figure 4. Individual data points are shown with line as the median. Data are representative of two independent experiments. Statistical significance by Kruskal-Wallis test is shown. Symbols are open circles (o) for 1T1H5-MVA and closed circles (·) for Fii-MVA.
Figure 6. Protective Efficacy of ChAdOxi MERS vaccine . Groups of 10 mice were vaccinated with 108 TCID50 ChAdOxi GFP or ChAdOxi MERS via the intranasal or intramuscular route, blood samples were collected before vaccination, and before challenge at 28 days post vaccination. hDPP4 mice were challenged intranasally with 104 TCID5o MERS-CoV (strain HC0V-EMC2012). At three dpi, four animals were sacrificed and lungs collected for analyses. The remaining six animals per group were sacrificed 28 dpi, or when they reached the endpoint criteria. A) Neutralizing antibody titers of hDPP4 mouse serum samples against MERS-CoV strain HCoV-EMC/2012 after vaccination. B) Weight loss after intranasal challenge with 104 TCID50 MERS- CoV. After challenge mice were weighed daily and percent body weight per group was calculated compared to body weight at the time of challenge. C) Survival curves of the vaccinated groups. After challenge hDPP4 mice were sacrificed due to the severity of disease signs or at 28 dpi. D) MERS-CoV viral loads in the lower respiratory tract of vaccinated hDPP4 mice. Viral load in the lungs of hDPP4 mice at 3 dpi. Mean values ± SD were calculated. Statistical significance was calculated using the Mann-Whitney U test; p-values: * < 0.05. (D). All experimental procedures were performed as previously described. Red = ChAdOxi GFP intranasally vaccinated animals; Grey = ChAdOxi MERS intranasally vaccinated animals; Blue = ChAdOxi GFP intramuscularly vaccinated animals; Purple = ChAdOxi MERS intramuscularly vaccinated animals. Figure 7. Im m unohistochem istry staining for MERS-CoV antigen in respiratory tract of vaccinated hDPP4 m ice. hDPP4 mouse tissues were evaluated for pathology and the presence of viral antigen as described previously Briefly, tissues were fixed in 10% neutral-buffered formalin for 7 days and paraffin- embedded. Tissue sections were stained with hematoxylin and eosin (H&E). An in- house produced rabbit polyclonal antiserum against HCoV- EMC/2012 (1:1000) was used as a primary antibody for the detection of viral antigen. Grading of histopathology and immunohistochemistry was done blinded by a board-certified veterinary pathologist. Lung tissues are shown at 100X and 1000X (insert) magnification.
ChAdOxi GFP intranasally (A) and intramuscularly (B) vaccinated animals. Lung tissue shows multifocal scattered positivity in the lungs. The inserts display MERS-CoV antigen within the Type I & II pneumocytes. ChAdOxi MERS intranasally (C) and intramuscularly (D) vaccinated animals. No MERS-CoV antigen positivity observed.
Figure 8 shows a plot.
Figure 9 shows a plot, a bar chart and a graph.
Figure 10 shows photographs.
Figure 11 shows a plot of ELISPOT responses to ChAdOxi MERS vaccine in Group 1 (5 x 109 vp, low dose) volunteers.
Figure 12 shows a plot of evaluation of MERS-CoV specific antibodies in camels at different ages using the commercial ELISA from Euroimmun.
Figure 13 shows a plot of evaluation of MERS-CoV specific antibodies in camels at different ages using an in house endpoint titre ELISA.
Figure 14 shows a plot of virus neutralisation titres for control and vaccinated camels.
Figure 15 shows a plot of na've camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using Euroimmun ELISA kit.
Figure 16 shows a plot of seropositive camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using Euroimmun ELISA kit.
Figure 17 shows na'rve camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using an in house end point titre ELISA.
Figure 18 shows a plot of seropositive camels: assessment of MERS-CoV specific antibodies in vaccinated camel sera using an in house end point titre ELISA. EXAMPLES
Materials and m ethods
Transgene and shuttle vector cloning
The spike (S) gene of MERS-CoV camel isolate (Genbank accession number:
KJ650098.1) was synthesised by GeneArt Gene Synthesis (Thermo Fisher Scientific). The S transgene was then cloned into four shuttle plasmid vectors following In-Fusion cloning (Clontech). Two plasmids contained the S transgene within the El homologous region of ChAdOxi, driven by the human cytomegalovirus major immediate early promoter (IE CMV) that includes intron A. One of the ChAdOxi shuttle plasmids was designed to include the tPA signal sequence upstream of the transgene sequence while the second plasmid did not contain the tPA. The ChAdOxi shuttle plasmids contained the S transgene within Gateway® recombination cassettes. To construct MVA MERS, one of the shuttle plasmids for MVA was designed to have the upstream and
downstream (flanks) of the F11L ORF as homologous sequence arms. Inserting the S transgene within these arms enabled the utilisation of the endogenous F11 promoter, which is part of the right homologous arm, while deleting the native F11L ORF. This resulted in the shuttle vector for generation of Fii-MVA MERS (F11 shuttle vector). The Π1Η5 promoter sequence was subcloned upstream of the S transgene; and this mH5-S transgene was then subcloned into the F11 shuttle vector. This resulted in the shuttle vector for generation of mH5-MVA MERS (Fn/mH5 shuttle vector). mH5-MVA MERS contained the 1T1H5 promoter at the F11L locus, however, the endogenous F11 promoter is intact and located upstream of the mH5 promoter. The endogenous F11 promoter could not be replaced with the mH5 since it is part of the essential upstream ORF. Im m unostaining for Transgene Expression
The ChAdOxi shuttle plasmid, described above, was used to validate the expression of MERS-CoV spike protein in vitro. An African green monkey kidney cell line (Vero cells) was seeded into 6-well plate to 80% confluence. Then the plasmid DNA was transfected into Vero cells using Lipofectamine® 2000 (Thermo Fisher Scientific) following manufacturer's instruction. Twenty four hours after transfection, cells were fixed, permeabilised, and immunostained using a rabbit polyclonal anti-MERS-CoV spike antibody, following standard protocols. DAPI stain was used to label nuclei.
Construction of recom binant ChAdOxi and MVA encoding MERS-CoV S antigens
The ChAdOxi MERS vaccines were prepared by Gateway® recombination between the ChAdOxi destination DNA BAC vector (described in (26)) and entry plasmids containing the coding sequence for MERS-CoV spike gene (ChAdOxi shuttle vectors explained above), according to standard protocols. ChAdOxi MERS genomes were then derived in HEK293A cell lines (Invitrogen, Cat. R705-07), the resultant viruses were purified by CsCl gradient ultracentrifugation as previously described (34). The titres were determined on HEK293A cells using anti-hexon immunostaining assay based on the QuickTiter™ Adenovirus Titer Immunoassay kit (Cell Biolabs Inc). For MVA MERS vaccines chicken embryo fibroblast cells (CEFs) were infected with MVA parental virus that encodes dsRed marker instead of the native F11L ORF and transfected with MVA shuttle plasmids containing MERS-CoV spike gene (explained above) to allow recombination with the MVA genome and deletion of dsRed marker whilst keeping the F11 promoter sequence. Recombinant MVA expressing MERS-CoV S protein was purified by plaque-picking and fluorescent selection using the sorting function of CyCLONE robotic module of a MoFlo Flow cytometer (Dako Cytomation, Denmark) as previously described (31). F11-MVA MERS and mH5-MVA MERS were confirmed to lack the native F11L ORF (and the dsRed marker), and contain MERS-CoV S by PCR (identity and purity PCR screening). The sequence of the S transgene amplified from these vaccines was confirmed. The recombinant viruses (vaccines) were amplified in 1500 cm2 monolayers of CEFs cells, partially purified over sucrose cushions and titrated in CEFs cells according to standard practice, and purity and identity were again verified by PCR.
Mouse im m unogenicity
Female BALB/c mice (Harlan, UK) aged 6 to 8 weeks were immunised intramuscularly (i.m.) in the upper leg (total volume 50 μΐ.) with a total of 108 IU of ChAdOxi MERS with or without tPA or with a total of 106 pfu of either F11-MVA MERS or mH5-MVA MERS. For induction of short-term anaesthesia, animals were anaesthetised using vaporised IsoFloH. In prime only regimens, mice were vaccinated with ChAdOxi with blood samples taken at 14 days post immunisation (d.p.i) or 28 d.p.i. for serum isolation; and spleens were collected at 28 d.p.i. In heterologous prime-boost regimens, mice were vaccinated with ChAdOxi MERS and boosted with MVA MERS at 28 d.p.i; mice were bled at 28 d.p.i. (post-prime) or 42 d.p.i (14 days post-boost) for serum isolation, and spleens were collected at 42 d.p.i. In homologous regimens, mice were vaccinated with MVA MERS and boosted with MVA MERS at 21 d.p.i; mice were bled on 21 d.p.i. (post-prime) or 42 d.p.i (post-boost) for serum isolation and spleens were collected at 42 d.p.i.
ELISpot, ICS, and flow cytom etry
Splenocytes were harvested for analysis by IFN-γ ELISpot or intracellular cytokine staining (ICS) and flow cytometry as previously described (35, 36), using re-stimulation with 2 g/mL S291 MERS-CoV S-specific peptide (VYDTIKYYSIIPHSI); for vaccine cellular immunogenicity (37)); or 1 g/mL E3 and F2(G) MVA vector-specific peptides (38) (for anti-MVA immune responses). In the absence of peptide re-stimulation, the frequency of IFN-y+ cells, which was typically 0.1% by flow cytometry or less than 50 SFC by ELISpot, was subtracted from tested re-stimulated samples.
ELISA
2 μg/ml with capturing antigen (Si recombinant protein from MyBioSource, CA, USA) were used to coat ELISA plates, and standard endpoint ELISA protocol was followed, as previously described (39). Sera were prepared in a 10-fold serial dilution in PBS/T and then 50 μΐ were plated in duplicate wells. Serum from a na'ive BALB/c mouse was included as a negative control. Goat anti-mouse total IgG conjugated to alkaline phosphatase (Sigma) and PNPP tablet (20 mg p-nitrophenylphosphate, SIGMA) substrate were used in the assay.
MERSpp Neutralisation assay
MERS pseudotyped viral particles (MERSpp) were produced and titrated using Huh7.5 cell line as described previously (40). For the MERSpp neutralization assay, serum samples were serially diluted in 96- well white plates (Nunc). A standard concentration of the MERSpp were added to the wells and plates were incubated for 1 h at 37 °C. After incubation, Huh7.5 cells (10,000 cells per well) were added to the plate in duplicates. Following 48 h incubation, cells were lysed and luciferase activity was measured. IC90 neutralisation titres were calculated for each mouse serum sample using GraphPad Prism.
Virus neutralisation assay
Induction of virus-neutralising antibodies was confirmed according to previously published protocols (37, 41). Briefly, mouse serum samples were tested for their capacity to neutralise MERS-CoV (EMC isolate) infections in vitro with 100 50% tissue culture infective doses (TCID50) in Huh-7 cells. Sera of non-immunised mice served as negative control.
Statistical analysis
GraphPad Prism (GraphPad software) was used for statistical analysis and to plot data. Ethics statem ent
All animal procedures were performed in accordance with the terms of the UK Animals (Scientific Procedures) Act (ASPA) for the project licenses 30/2414 or 30/2889 and were approved by the University of Oxford Animal Care and Ethical Review Committee. All mice were housed for at least 7 days for settlement prior to any procedure in the University animal facility, Oxford, UK under Specific Pathogen Free (SPF) conditions. Results
Construction and antigen expression of MERS-CoV vaccine candidates The spike gene from a camel isolate (Camel/Qatar_2_20i4 MERS-CoV isolate, GenBank accession number KJ650098.1) was cloned into four shuttle vectors that facilitate homologous recombination with the genome of ChAdOxi or MVA. Four recombinant viral vectors, two ChAdOxi and two MVA, were derived as described in the materials and methods. ChAdOxi based vaccine candidates were generated with or without the signal peptide of the human tissue plasminogen activator gene (tPA). The spike transgene expression in ChAdOxi MERS vaccine candidates is under the control of the human cytomegalovirus major immediate early promoter (CMV IE) that includes intron A. In MVA MERS vaccine candidates, the tPA was also inserted upstream of the spike transgene, which was under the control of either the ectopic 1T1H5 promoter or the endogenous F11 promoter (Figure lA). All of our MERS-CoV vaccine candidates contain the same codon-optimized spike transgene.The expression of the newly synthesized transgene was first tested by transfection of an African green monkey kidney cell line (Vero cells) with the adenovirus shuttle vector, and immunofluorescence staining of the transfected cells (Figure lB and lC). This was performed to confirm the expression of the codon optimized spike transgene in mammalian cells. The level of transgene expression from the four vaccine candidates was not evaluated in vitro. We have previously reported that differences in MVA promoter activity detectable in vitro does not correlate with in vivo immunogenicity (31), and that only in vivo expression correlates with the in vivo immunogenicity.
Hum oral Im m unogenicity of ChAdOxi based MERS-CoV vaccine
candidates
To evaluate humoral immune responses to ChAdOxi MERS with or without tPA, BALB/c mice were vaccinated with lxio8 IU of ChAdOxi intramuscularly. Serum samples from 14 and 28 d.p.i. were collected and evaluated by ELISA. Both vaccine candidates induced a high level of Si-specific antibodies (mean endpoint titre (Logi0) = 4.8 with tPA, 4.7 without tPA) , unlike the control vaccine, ChAdOxi encoding enhanced green fluorescent protein (ChAdOxi- eGFP, mean endpoint titre (Logi0) = 1). These antibody levels were similar between the two candidates (with or without tPA) at day 14. However, at 28 d.p.i. ChAdOxi MERS with tPA induced significantly higher Si- specific antibodies than ChAdOxi MERS without tPA (mean endpoint titre (Logi0) = 5.13 with tPA, 4.6 without tPA, Figure 2A). Serum samples from day 28 were selected for MERSpp neutralisation assay. Serum antibodies induced by ChAdOxi MERS with tPA showed significantly higher neutralisation activity than without tPA (mean titre IC90 (Logio) = 2.8 with tPA, 2.2 without tPA; Figure 2B). In order to confirm that the psuedotyped virus neutralisation assay was producing biologically relevant results, serum samples from mice immunised with ChAdOxi MERS with tPA were also tested in a neutralisation assay utilising wildtype MERS virus. This assay confirmed the neutralisation activity of mouse antibodies (nAb) with a median of 360 VNT (Virus Neutralization Test antibody titre; Figure 2C). We therefore continued to evaluate ChAdOxi MERS with tPA in addition to generating MVA MERS vaccine candidates with tPA.
Cellular Im m unogenicity of ChAdOxi based MERS-CoV vaccine candidates
Having established the utility of tPA in ChAdOxi MERS vaccines (referred to as ChAdOxi MERS in the rest of this report) at increasing humoral responses, spleens were collected at 28 d.p.i. from immunised BALB/c mice. Splenocytes were processed to evaluate cellular immune responses to ChAdOxi MERS in ELISpot and Intracellular cytokine staining (ICS). Peptide S291, described by others (37), was used to re- stimulate the cells in both assays and ELISpot data showed a high level of IFN-γ secreting splenocytes (Median = 1300 SFU/106 splenocytes; Figure 3A). ICS data confirmed the IFN-γ secreting CD8+ splenocytes also secreted TNF-a and IL-17 (Figure 3B).
Im m unogenicity of Heterologous ChAdOxi and MVA vaccination against MERS-CoV
To evaluate humoral immune responses to heterologous prime-boost vaccination, BALB/c mice were immunised with ChAdOxi MERS vaccine and boosted with one of two different MVA MERS vaccine candidates four weeks later. The MVA based candidates differ in the promoters that controls the transgene expression: Fii-MVA MERS utilises the endogenous strong early F11 promoter and mH5-MVA MERS utilises the ectopic early/late 1T1H5 promoter. Serum samples from 28 d.p.i. (post-prime) or 42 d.p.i. (post-boost) were collected and evaluated by ELISA and MERSpp neutralisation assay. At 28 d.p.i. ChAdOxi MERS induced similar levels of Si-specific antibodies and nAb as observed previously (Figure 4A and B). At 42 d.p.i. Si-specific antibodies were boosted to a higher level (mean endpoint titre (Logi0) = 5 by ChAdOxi MERS boosted to 5.8 by mH5-MVA MERS or 5.9 by F11-MVA MERS); Figure 4A) with nAb also enhanced to a statistically significant level (mean titre IC90 (Logi0) = 2.87 by ChAdOxi MERS boosted to 3.3 by mH5-MVA MERS or 3.5 by F11-MVA MERS; Figure 4B). There was no difference in antibody levels induced using either the F11 or Π1Η5 promoter in the MVA.
At 42 d.p.i. splenocytes were also processed to evaluate cellular immune responses to ChAdOxi MERS MVA MERS prime-boost vaccination in ELISpot and ICS as shown in Figure 3. The T cell responses to MERS S were boosted by the MVA vaccinations; in the ICS experiments, Fii-MVA and 1T1H5-MVA boosted the percentage of IFN-Y+ splenic CD8+ T cells to 7.3 and 5.2% respectively (Figure 4D) whereas the percentage was 2.5% after ChAdOxi MERS prime in Figure 3B. The percentage of TNF-a+ splenic CD8+ T cells were also increased by MVA boost (comparing Figure 3B and 4D). Utilising the F11 promoter resulted in a trend towards greater cell-mediated immunogenicity (Figure 4C and D). Splenocytes were also re-stimulated with MVA backbone-specific E3 and F(G)2 peptides and evaluated in ICS. Both MVA based vaccines induced similar responses to E3 or to F(G)2 peptides, 2 weeks after MVA vaccination (Figure 4E and F). This similarity confirmed the efficiency of vaccine titration, vaccination, and sample processing because responses to each of those peptides are not expected to be different unless there is variation in the doses administered or sample preparation. Overall, MVA MERS vaccines were able to boost the humoral and cellular immune responses to ChAdOxi MERS prime vaccination. There was no difference between the F11 and 1T1H5 promoter in the resulting antibody titres after ChAdOxi prime/MVA boost, but there was a trend towards increased cellular immunogenicity when the F11 promoter was used.
Im m unogenicity of Hom ologous MVA vaccination against MERS-CoV
To evaluate humoral immune responses to a homologous MVA MERS prime-boost vaccination, two groups of BALB/c mice were immunised with Fii-MVA MERS or mH5"MVA MERS and boosted with the same vaccine after three weeks. Serum samples from 21 d.p.i. (post-prime) or 42 d.p.i. (post-boost) were collected and evaluated in ELISA and MERSpp neutralisation assays. At 21 d.p.i. Fii-MVA MERS and 1TLH5-MVA induced similar levels of Si-specific antibodies (mean endpoint titre (Logi0) = 3.2 and 2.8 respectively; Figure 5A). At 42 d.p.i Si-specific antibody levels had increased to 4.7 and 4.8 respectively (Figure 5A). The titres of nAb (MERS pp assay) were also similar for both vaccines (mean titre IC90 (Logio) = 2.71 (Fii-MVA MERS) and 2.76
respectively; Figure 5B). Utilising different promoters in MVA vectors did not result in differences in the induced antibody levels. However, at 42 d.p.i. IFN-γ secreting splenocytes induced by Fii-MVA MERS were statistically significantly higher than those of mH5-MVA MERS ((Median = 525 and 249 SFU/106 splenocytes, respectively, Figure 5C). Both MVA vaccines induced similar vector-specific immune responses as expected (Figure 5D and E).
Sum m ary
Prior art vaccines against MERS-CoV have been developed and tested in a number of animal models (including non-human primates (42-44) and camels (45)) as well as in human clinical trials (46). All vaccine candidates focused on the spike antigen because it contains the receptor-binding domain used for cell entry by the virus, against which neutralising antibodies may be induced, and it is conserved. Therefore, our approach to the improvement of MERS-CoV vaccines focusses on platform and vaccination regimens rather than antigen selection and optimisation. Here, we focused on using the same antigen (transgene) to develop a vaccine against MERS-CoV, and to assess different vectors, different versions of each vector, and different vaccination regimens. We generated a number of MERS-CoV vaccine candidates based on the same codon optimized spike transgene and ensured its expression in vitro before we evaluated the humoral and cellular immunogenicity in a pre-clinical BALB/c mouse model. ChAdOxi based vaccine candidates were produced with or without tPA. The tPA signal peptide was predicted to enhance the humoral immunogenicity of encoded vaccine antigens. Our data support this hypothesis and show a significant increase in the Si-specific antibody levels at 28 d.p.i. The level of neutralising antibodies was also increased when tPA was utilised. Thus it is clear that we have demonstrated a credible use as a human vaccine.
ChAdOxi MERS without tPA was still a potent vaccine candidate, inducing a high level of both Si-specific binding antibodies and MERS-CoV neutralising antibodies.
Neutralisation activity of mouse serum antibodies was assayed by using MERS-CoV pseudotyped viral particles (MERSpp), an approach used by a number of researchers for other human pathogens such as ΗΓν, Influenza, and HCV to overcome the necessity of handling BSL-3 viruses (40). Additionally, we confirmed the ability of serum samples from vaccinated mice to neutralise live MERS virus. We therefore selected ChAdOxi MERS with tPA (simply referred to ChAdOxi MERS) for further evaluation.
ChAdOxi MERS also induced cellular responses for MERS S, with polyfunctional CD8+ T cells detected in the spleen of immunized mice. This supports the potency of the ChAdOxi viral vector in inducing T cellular immunity, observed previously in animal models (26, 32, 47) as well as in humans (33).
Following ChAdOxi prime/MVA boost, MVA significantly boosted the neutralizing antibody titres to higher levels. No difference in humoral immunity was found when either the F11 or Π1Η5 promoter was used. Regarding the promoter effect on MVA cellular immunogenicity, we have previously reported that utilising the F11 promoter enhanced malaria and influenza antigens in MVA (31). Here, we again report that F11- MVA MERS induced higher T cell responses than 1TLH5-MVA MERS in a homologous prime-boost MVA MERS vaccination.
All of our vaccine candidates induced humoral (with nAb) and cellular immune (with polyfunctional CD8+ T cell) responses against MERS-CoV spike antigen. Effects on immunogenicity of different versions of the vaccines were noted, with the use of the tPA leader sequence in ChAdOxi, and the use of the F11 promoter in MVA producing increases in immunogenicity compared to no leader sequence, or the 1T1H5 promoter. The protective level of either antibodies or cellular immunity required to counter MERS-CoV infection in humans or in animal models is not yet defined, despite some efforts (48-51). The ideal vaccine would provide rapid onset of immunity and complete protective efficacy after a single dose, with a long duration of immunity. Complete protective efficacy of one dose of ChAdOxi expressing the external glycoprotein of Rift Valley Fever Virus has been demonstrated in multiple species and it is already known that ChAdOxi RVF is highly immunogenic in camels (32). However, RVF is a completely different viral family to the coronaviruses (as discussed above).
To date, the only vaccine against MERS to be tested in camels is an MVA vectored vaccine (41) which was protective in hDPP4 transgenic mice immunized with a homologous prime/boost regimen (37) but in camels required two doses given both intranasally and intramuscularly to provide partial protection and reduction of virus shedding (45). Here we show that a single dose of ChAdOxi MERS is as immunogenic as two doses of MVA MERS, demonstrating that this regimen should be tested for protective efficacy in camels. However if this is not completely protective,
administration of MVA MERS as a heterologous boost may be employed.
In our hands one dose of MVA resulted in an endpoint titre of 3 logs, two doses of MVA produced 4.7 logs, one dose of ChAdOxi produced 5 logs, and ChAdOxi/MVA prime boost produced 5.9 logs, showing that the invention delivers technical benefits.
If a two dose regimen is required, e.g. for particular applications such as sustained immunity in healthcare workers, ChAdOxi/MVA would be more likely to provide complete protection than MVA/MVA.
ChAdOxi MERS is thus demonstrated to be a plausible and credible vaccine for both camels and humans.
References
1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier A. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. The New England journal of medicine. 2012;367(19):1814-20.
2. World Health Organisation. Middle East respiratory syndrome coronavirus (MERS- CoV) 2016. Available from: http://www.who .int/emergencies/mers- ov/en/. [accessed 20.02.2017].
3. Lessler J, Salje H, Van Kerkhove MD, Ferguson NM, Cauchemez S, Rodriquez-Barraquer I, et al. Estimating the Severity and Subclinical Burden of Middle East Respiratory Syndrome
Coronavirus Infection in the Kingdom of Saudi Arabia. American journal of epidemiology. 2016;183(7):657-63.
4. Al Hammadi ZM, Chu DK, Eltahir YM, Al Hosani F, Al Mulla M, Tarnini W, et al.
Asymptomatic MERS-CoV Infection in Humans Possibly Linked to Infected Dromedaries Imported from Oman to United Arab Emirates, May 2015. Emerging infectious diseases.
2015;21(12):2197-200. 5. Oboho IK, Tomczyk SM, Al-Asmari AM, Banjar AA, Al-Mugti H, Aloraini MS, et al. 2014 MERS-CoV outbreak in Jeddah-a link to health care facilities. N Engl J Med. 2015;372(9):846- 54.
6. Who Mers-Cov Research G. State of Knowledge and Data Gaps of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) in Humans. PLoS currents. 2013;5.
7. Memish ZA, Zumla Al, Assiri A. Middle East respiratory syndrome coronavirus infections in health care workers. The New England journal of medicine. 2013;369(9):884-6.
8. Muller MA, Corman VM, Jores J, Meyer B, Younan M, Liljander A, et al. MERS coronavirus neutralizing antibodies in camels, Eastern Africa, 1983-1997. Emerging infectious diseases. 2014;20(12):2093-5.
9. Perera RA, Wang P, Gomaa MR, El-Shesheny R, Kandeil A, Bagato O, et al.
Seroepidemiology for MERS coronavirus using microneutralisation and pseudoparticle virus neutralisation assays reveal a high prevalence of antibody in dromedary camels in Egypt, June 2013. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2013;18(36):pii=20574.
10. Hemida MG, Perera RA, Al Jassim RA, Kayali G, Siu LY, Wang P, et al. Seroepidemiology of Middle East respiratory syndrome (MERS) coronavirus in Saudi Arabia (1993) and Australia (2014) and characterisation of assay specificity. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2014;19(23).
11. Gutierrez C, Tejedor-Junco MT, Gonzalez M, Lattwein E, Renneker S. Presence of antibodies but no evidence for circulation of MERS-CoV in dromedaries on the Canary Islands, 2015. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2015;20(37).
12. Ali MA, Shehata M M, Gomaa M R, Kandeil A, El-Shesheny R, Kayed AS, et al.
Systematic, active surveillance for Middle East respiratory syndrome coronavirus in camels in Egypt. Emerging microbes & infections. 2017;6(l):el.
13. van Doremalen N, Hijazeen ZS, Holloway P, Al Omari B, McDowell C, Adney D, et al. High Prevalence of Middle East Respiratory Coronavirus in Young Dromedary Camels in Jordan. Vector borne and zoonotic diseases. 2017;17(2):155-9.
14. Alraddadi BM, Watson JT, Almarashi A, Abedi GR, Turkistani A, Sadran M, et al. Risk Factors for Primary Middle East Respiratory Syndrome Coronavirus Illness in Humans, Saudi Arabia, 2014. Emerging infectious diseases. 2016;22(l):49-55.
15. Gossner C, Danielson N, Gervelmeyer A, Berthe F, Faye B, Kaasik Aaslav K, et al.
Human-Dromedary Camel Interactions and the Risk of Acquiring Zoonotic Middle East Respiratory Syndrome Coronavirus Infection. Zoonoses and public health. 2016;63(l):l-9.
16. Funk AL, Goutard FL, Miguel E, Bourgarel M, Chevalier V, Faye B, et al. M ERS-CoV at the Animal-Human Interface: Inputs on Exposure Pathways from an Expert-Opinion Elicitation. Frontiers in veterinary science. 2016;3:88.
17. Reusken CB, Ababneh M, Raj VS, Meyer B, Eljarah A, Abutarbush S, et al. Middle East Respiratory Syndrome coronavirus (MERS-CoV) serology in major livestock species in an affected region in Jordan, June to September 2013. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2013;18(50):20662.
18. Hemida MG, Perera RA, Wang P, Alhammadi MA, Siu LY, Li M, et al. Middle East Respiratory Syndrome (MERS) coronavirus seroprevalence in domestic livestock in Saudi Arabia, 2010 to 2013. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2013;18(50):20659.
19. Alagaili AN, Briese T, Mishra N, Kapoor V, Sameroff SC, Burbelo PD, et al. Middle East respiratory syndrome coronavirus infection in dromedary camels in Saudi Arabia. mBio.
2014;5(2):e00884-14.
20. Reusken CB, Haagmans BL, Muller MA, Gutierrez C, Godeke GJ, Meyer B, et al. Middle East respiratory syndrome coronavirus neutralising serum antibodies in dromedary camels: a comparative serological study. The Lancet Infectious diseases. 2013;13(10):859-66. 21. Vergara-Alert J, van den Brand JM, Widagdo W, Munoz Mt, Raj S, Schipper D, et al. Livestock Susceptibility to Infection with Middle East Respiratory Syndrome Coronavirus. Emerging infectious diseases. 2017;23(2):232-40.
22. Adney DR, Brown VR, Porter SM, Bielefeldt-Ohmann H, Hartwig AE, Bowen RA.
Inoculation of Goats, Sheep, and Horses with MERS-CoV Does Not Result in Productive Viral Shedding. Viruses. 2016;8(8).
23. Alharbi NK. Vaccines against Middle East respiratory syndrome coronavirus for humans and camels. Reviews in medical virology. 2016.
24. Draper SJ, Heeney JL. Viruses as vaccine vectors for infectious diseases and cancer. Nat Rev Microbiol. 2010;8(l):62-73.
25. Tao D, Barba-Spaeth G, Rai U, Nussenzweig V, Rice CM, Nussenzweig RS. Yellow fever 17D as a vaccine vector for microbial CTL epitopes: protection in a rodent malaria model. The Journal of experimental medicine. 2005;201(2):201-9.
26. Dicks MD, Spencer AJ, Edwards NJ, Wadell G, Bojang K, Gilbert SC, et al. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PloS one. 2012;7(7):e40385.
27. Gomez CE, Perdiguero B, Garcia-Arriaza J, Esteban M. Clinical applications of attenuated MVA poxvirus strain. Expert review of vaccines. 2013;12(12):1395-416.
28. Gilbert SC. Clinical development of Modified Vaccinia virus Ankara vaccines. Vaccine. 2013;31(39):4241-6.
29. Luo M, Tao P, Li J, Zhou S, Guo D, Pan Z. Immunization with plasmid DNA encoding influenza A virus nucleoprotein fused to a tissue plasminogen activator signal sequence elicits strong immune responses and protection against H5N1 challenge in mice. Journal of virological methods. 2008;154(l-2):121-7.
30. Wallace A, West K, Rothman AL, Ennis FA, Lu S, Wang S. Post-translational intracellular trafficking determines the type of immune response elicited by DNA vaccines expressing Gag antigen of Human Immunodeficiency Virus Type 1 (HIV-1). Hum Vaccin Immunother.
2013;9(10):2095-102.
31. Alharbi NK, Spencer AJ, Salman AM, Tully CM, Chinnakannan SK, Lambe T, et al.
Enhancing cellular immunogenicity of MVA-vectored vaccines by utilizing the FllL endogenous promoter. Vaccine. 2016;34(l):49-55.
32. Warimwe GM, Gesharisha J, Carr BV, Otieno S, Otingah K, Wright D, et al. Chimpanzee Adenovirus Vaccine Provides Multispecies Protection against Rift Valley Fever. Sci Rep.
2016;6:20617.
33. Antrobus RD, Coughlan L, Berthoud TK, Dicks MD, Hill AV, Lambe T, et al. Clinical assessment of a novel recombinant simian adenovirus ChAdOxl as a vectored vaccine expressing conserved Influenza A antigens. Molecular therapy : the journal of the American Society of Gene Therapy. 2014;22(3):668-74.
34. Cottingham MG, Carroll F, Morris SJ, Turner AV, Vaughan AM, Kapulu MC, et al.
Preventing spontaneous genetic rearrangements in the transgene cassettes of adenovirus vectors. Biotechnol Bioeng. 2012;109(3):719-28.
35. Sridhar S, Reyes-Sandoval A, Draper SJ, Moore AC, Gilbert SC, Gao GP, et al. Single- dose protection against Plasmodium berghei by a simian adenovirus vector using a human cytomegalovirus promoter containing intron A. J Virol. 2008;82(8):3822-33.
36. Cottingham MG, Andersen RF, Spencer AJ, Saurya S, Furze J, Hill AV, et al.
Recombination-mediated genetic engineering of a bacterial artificial chromosome clone of modified vaccinia virus Ankara (MVA). PloS one. 2008;3(2):el638.
37. Volz A, Kupke A, Song F, Jany S, Fux R, Shams-Eldin H, et al. Protective efficacy of recombinant Modified Vaccinia virus Ankara (MVA) delivering Middle East Respiratory Syndrome coronavirus spike glycoprotein. Journal of virology. 2015.
38. Tscharke DC, Woo WP, Sakala IG, Sidney J, Sette A, Moss DJ, et al. Poxvirus CD8+ T-cell determinants and cross-reactivity in BALB/c mice. Journal of virology. 2006;80(13):6318-23. 39. Draper SJ, Moore AC, Goodman AL, Long CA, Holder AA, Gilbert SC, et al. Effective induction of high-titer antibodies by viral vector vaccines. Nature medicine. 2008;14(8):819-21.
40. Grehan K, Ferrara F, Temperton N. An optimised method for the production of MERS- CoV spike expressing viral pseudotypes. MethodsX. 2015;2:379-84.
41. Song F, Fux R, Provacia LB, Volz A, Eickmann M, Becker S, et al. Middle East respiratory syndrome coronavirus spike protein delivered by modified vaccinia virus Ankara efficiently induces virus-neutralizing antibodies. Journal of virology. 2013;87(21):11950-4.
42. Muthumani K, Falzarano D, Reuschel EL, Tingey C, Flingai S, Villarreal DO, et al. A synthetic consensus anti-spike protein DNA vaccine induces protective immunity against Middle East respiratory syndrome coronavirus in nonhuman primates. Science translational medicine. 2015;7(301):301ral32.
43. Lan J, Yao Y, Deng Y, Chen H, Lu G, Wang W, et al. Recombinant Receptor Binding Domain Protein Induces Partial Protective Immunity in Rhesus Macaques Against Middle East Respiratory Syndrome Coronavirus Challenge. EBioMedicine. 2015;2(10):1438-46.
44. Wang L, Shi W, Joyce MG, Modjarrad K, Zhang Y, Leung K, et al. Evaluation of candidate vaccine approaches for MERS-CoV. Nature communications. 2015;6:7712.
45. Haagmans BL, van den Brand JM, Raj VS, Volz A, Wohlsein P, Smits SL, et al. An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels. Science. 2016;351(6268):77-81.
46. Pellerin C. Army Scientists Begin First MERS Vaccine Clinical Trial. DoD News, Defense Media Activity. 2016.
47. Warimwe GM, Lorenzo G, Lopez-Gil E, Reyes-Sandoval A, Cottingham MG, Spencer AJ, et al. Immunogenicity and efficacy of a chimpanzee adenovirus-vectored Rift Valley fever vaccine in mice. Virology journal. 2013;10:349.
48. Wang W, Wang H, Deng Y, Song T, Lan J, Wu G, et al. Characterization of anti-MERS- CoV antibodies against various recombinant structural antigens of MERS-CoV in an imported case in China. Emerging microbes & infections. 2016;5(ll):ell3.
49. Min CK, Cheon S, Ha NY, Sohn KM, Kim Y, Aigerim A, et al. Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity. Scientific reports. 2016;6:25359.
50. Park WB, Perera RA, Choe PG, Lau EH, Choi SJ, Chun JY, et al. Kinetics of Serologic Responses to MERS Coronavirus Infection in Humans, South Korea. Emerging infectious diseases. 2015;21(12):2186-9.
51. Corman VM, Albarrak AM, Omrani AS, Albarrak MM, Farah M E, Almasri M, et al. Viral Shedding and Antibody Response in 37 Patients With Middle East Respiratory Syndrome
Coronavirus Infection. Clin Infect Dis. 2016;62(4):477-83.
EXAMPLE 2: Protective Efficacy of a Novel Simian Adenovirus Vaccine Against Lethal MERS-CoV Challenge in a Transgenic Human DPP4 Mouse Model: A Simian
Adenovirus MERS-CoV Vaccine
Middle East respiratory syndrome coronavirus (MERS-CoV) is a novel zoonotic virus that causes severe respiratory disease in humans with case fatality rates close to 40%, but for which no vaccines are available. Here, we evaluated the utility of ChAdOxi, a promising replication-deficient simian adenovirus vaccine vector platform with an established safety profile in humans and dromedary camels, for MERS-CoV vaccine development. Using a transgenic lethal BALB/c MERS-CoV mouse model we show that single dose intranasal or intramuscular immunization with ChAdOxi MERS, encoding full-length MERS-CoV Spike glycoprotein, is highly immunogenic and confers protection against lethal viral challenge. Immunogenicity and efficacy were comparable between immunisation routes. Together these data show credible utility of ChAdOxi MERS vaccine in humans and dromedary camels, the animal reservoir of infection.
One ChAd vector, termed ChAdOxi 6-, has undergone testing in dromedary camels, showing excellent safety and immunogenicity when encoding Rift Valley Fever viral glycoproteins Here, using standard adenovirus production methods β we made a vaccine construct, ChAdOxi MERS, encoding the full-length MERS-CoV spike glycoprotein (GenBank accession number KJ650098.1) targeted by protective neutralising antibodies a. To determine vaccine immunogenicity and efficacy we utilised a recently developed transgenic lethal BALB/c mouse model expressing the human dipeptidyl peptidase (hDPP4) gene in the Rosa26 locus, which renders mice susceptible to MERS-CoV infection
hDPP4 mice were vaccinated intranasally or intramuscularly with 108 infectious units of either a control ChAdOxi vaccine encoding enhanced green fluorescent protein (ChAdOxi GFP) or the ChAdOxi MERS vaccine. Sera were obtained before vaccination and 28 days post-vaccination. ChAdOxi MERS vaccination elicited high titre neutralizing antibodies with both immunisation routes being equally immunogenic (Kolomgorov-Smirnov test, p=o.657i) (Fig. 6A). No MERS-CoV neutralizing antibody response was observed among the ChAdOxi GFP vaccinees.
To evaluate vaccine efficacy animals were challenged intranasally with ιο^ TCID50 of the HCoV-EMC/2012 MERS-CoV strain in a total volume of 25 μΐ and observed daily for signs of disease. Euthanasia was indicated at >20% loss of initial body weight. At 3 days post-inoculation (dpi), four animals from each group were sacrificed and lungs collected for analyses. The remaining six animals per group were sacrificed 28 dpi, or when they reached the humane endpoint criteria. ChAdOxi GFP vaccinees developed signs of disease, including loss of body weight, ruffled fur and lethargy (Fig. 6B).
Weight loss begun 3 dpi and at 7-8 dpi all mice in the ChAdOxi GFP groups either succumbed to infection or reached the predefined euthanasia endpoint (Figure 7C). No signs of disease or significant loss of body weight were observed in mice vaccinated with ChAdOxi MERS (Fig. 6B and C).
The presence of MERS-CoV RNA in the lungs was analyzed by qRT-PCR on mice (n=4/group) sacrificed at 3 dpi. High viral loads were found in the lower respiratory tract of the ChAdOxi GFP vaccinated mice (intranasal 105·32 TCID50/gram tissue, 95% confidence interval (CI): io2 28to IOS
Figure imgf000042_0001
tissue, 95% CI: lo^to 104-96). No viral RNA was detected in any of the ChAdOxi MERS vaccinated mice (Figure 6D). Immunohistochemistry staining for MERS-CoV in lung tissue showed abundance of antigen in the ChAdOxi GFP vaccinated mice, but not the ChAdOxi-MERS vaccinees (Figure 7). MERS-CoV staining was observed in the type I and type II pneumocytes (Figure 7 inserts) but not in any of the other respiratory cells such as endothelial cells, bronchiolar epithelium or macrophages.
Together these data provide support for applicability of ChAdOxi MERS vaccine in humans and dromedary camels. This is advantageous given the established safety profile of the ChAdOxi platform in humans s and dromedary camels 2. A deployable human MERS-CoV vaccine will need to be safe and efficacious in at-risk populations, including healthcare workers, camel herders and those with comorbidities as highlighted in the ongoing WHO-led consultation on an ideal target product profile for MERS-CoV vaccines.
ChAdOxi MERS induces T cell responses against MERS Spike protein as well as neutralizing antibodies (example 1) . Identification of immune correlates of protection against MERS-CoV in humans and camels allows cost-effective disease surveillance and vaccine monitoring.
In summary, we have demonstrated the utility of the ChAdOxi platform for MERS-CoV vaccine development in a lethal mouse model. The excellent immunogenicity and efficacy observed here show the value of ChAdOxi MERS in dromedary camels and humans.
Ethics statem ent
Approval of animal experiments was obtained from the Institutional Animal Care and Use Committee of the Rocky Mountain Laboratories. The performance of experiments was done following the guidelines of the Association for Assessment and Accreditation of Laboratory Animal Care, International (AAALAC) by certified staff in an AAALAC- approved facility, following the guidelines and basic principles in the United States Public Health Service Policy on Humane Care and Use of Laboratory Animals and the Guide for the Care and Use of Laboratory Animals. Work with infectious MERS-CoV strains under BSL3 conditions was approved by the Institutional Biosafety Committee (IBC). Inactivation and removal of samples from high containment was performed according to IBC-approved standards. REFERENCES TO EXAMPLE 2
1 Arabi, Y. M. et al. Middle East Respiratory Syndrome. N Engl J Med 376 , 584- 594, doi:io.i056/NEJMsri4o8795 (2017). 2 Oboho, I. K. et al. 2014 MERS-CoV outbreak in Jeddah~a link to health care facilities. N Engl J Med 372 , 846-854, doi:io.i056/NEJMoai4o8636 (2015).
3 Reusken, C. B. et al. Geographic distribution of MERS coronavirus among dromedary camels, Africa. Em erg Infect Dis 20 , 1370-1374,
doi:io.320i/eid20o8.140590 (2014).
4 Modjarrad, K. et al. A roadmap for MERS-CoV research and product development: report from a World Health Organization consultation. Nat Med 22 , 701- 705, doi:io.i038/nm.4i3i (2016).
5 Ewer, K. J. et al. Viral vectors as vaccine platforms: from immunogenicity to impact. Curr Opin Im m unol 41, 47-54, doi:l0.10l6/j.coi.20l6.05.014 (2016).
6 Dicks, M. D. et al. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative
immunogenicity. PLoS One 7, 640385, doi:io.i37i/journal.pone.oo40385 (2012).
7 Warimwe, G. M. et al. Chimpanzee Adenovirus Vaccine Provides Multispecies Protection against Rift Valley Fever. 5c/ Rep 6 , 20617, doi:io.i038/srep2o6i7 (2016).
8 Morris, S. J., Turner, A. V., Green, N. & Warimwe, G. M. Laboratory-Scale Production of Replication-Deficient Adenovirus Vectored Vaccines. Methods Mol Biol 1349 , 121-135, doi:io.ioo7/978-i-4939-30o8-i_8 (2016).
9 de Wit, E., van Doremalen, N., Falzarano, D. & Munster, V. J. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol 14 , 523-534, doi:io.i038/nrmicro.20i6.8i (2016).
10 Agrawal, A. S. et al. Generation of a transgenic mouse model of Middle East respiratory syndrome coronavirus infection and disease. / Virol 89 , 3659-3670, doi:io.ii28/JVI.03427-i4 (2015).
11 van Doremalen, N. et al. High Prevalence of Middle East Respiratory
Coronavirus in Young Dromedary Camels in Jordan. Vector Borne Zoonotic Dis 17, 155-159, doi:io.io89/vbz.20i6.2o62 (2017).
12 Reusken, C. B. et al. Middle East respiratory syndrome coronavirus neutralising serum antibodies in dromedary camels: a comparative serological study. Lancet Infect Dis 13 , 859-866, doi:io.ioi6/Si473-3099(i3)70i64-6 (2013).
13 Ali, M. A. et al. Systematic, active surveillance for Middle East respiratory syndrome coronavirus in camels in Egypt. Em erg Microbes Infect 6 , el,
doi:io.i038/emi.20i6.i30 (2017).
14 de Wit, E. et al. Middle East respiratory syndrome coronavirus (MERS-CoV) causes transient lower respiratory tract infection in rhesus macaques. Proc Natl Acad
Sci U S A 110 , 16598-16603, doi:io.i073/pnas.i3i0744iio (2013). Example 3 - Clinical Trial
1. SYNOPSIS
A phase I clinical trial to determine the safety and immunogenicity of the candidate MERS-CoV vaccine ChAdOxi MERS-S in UK healthy adult volunteers.
Trial Centre Centre for Clinical Vaccinology & Tropical Medicine, University of
Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE
Trial Identifier MERS 001
Clinical phase I
Study Design Open-labelled, non-randomised, dose escalation, phase I clinical trial
Population Healthy adults aged 18 - 50 years
Planned Sam ple 24 volunteers
Size Group Dose of ChAdOxi MERS-S
Group 1 (n=6) 5 x 10? vp
Group 2 (n=9) 2.5 x 1010 vp
Group 3 (n=9) 5 x 1010 vp
Follow -up duration 26 weeks post vaccine administration
Planned Trial Q4 2017 to Q4 2018
Period
Prim ary Objective To assess the safety profile of the candidate vaccine ChAdOxi MERS-
S in healthy adult volunteers dary Objective To assess the immunogenicity of the candidate vaccine ChAdOxi MERS-S in healthy adult volunteers
Investigational ChAdOxi MERS-S, a replication-deficient simian adenoviral vector
Products expressing the spike (S) protein of MERS Coronavirus.
Dose per ChAdOxi MERS-S 5 x io? vp
Ad m in is tr a tio n ChAdOxi MERS-S 2.5 x 1010 vp
ChAdOxi MERS-S 5 x 1010 vp
Form Liquid (all finished products)
Route Intramuscularly (IM) into the deltoid region of the arm
2. ABBREVIATIONS
AE Adverse event
AR Adverse reaction
CBF Clinical Bio-Manufacturing Facility
CCVTM Centre for Clinical Vaccinology and Tropical Medicine
ChAdOxi Chimpanzee Adenovirus Oxi
ChAdOxi MERS-S Recombinant Chimpanzee Adenovirus Oxi with MERS spike antigen
CI Chief Investigator
CRF Case report form or Clinical Research Facility
CTRG Clinical Trials Research Governance
Da Dalton
DSUR Development Safety Update Report
EC Ethics committee
ELISA Enzyme linked immunosorbent assay
ELISPOT Enzyme linked immunospot assay
FBC Full blood count
GCP Good Clinical Practice
GMO Genetically modified organism
GMP Good Manufacturing Practice
HA Haemagglutinin
HBsAg Hepatitis B surface antigen HCG Human Chorionic Gonadotrophin
HCV Hepatitis C virus
HI Haemagglutination inhibition
HIV Human Immunodeficiency virus
HLA Human leukocyte antigen
IB Investigators Brochure
ID Intradermal
IEC Independent Ethics Committee
ICH International Conference on Harmonisation
IM Intramuscular
IMP Investigational Medicinal Product
LSC Local Safety Committee
LSM Local Safety Monitor
MERS Middle East Respiratory Syndrome
MHRA Medicines and Healthcare products Regulatory Agency
MRC Medical Research Council
MVA Modified Vaccinia Virus Ankara
g microgram
nm nanometer
pfu plaque forming units
PIS Participant information sheet
ppm parts per million
QP Qualified Person
SAE Serious Adverse Event
SAR Serious Adverse Reaction
SFU Spot forming units
SOP Standard Operating Procedure
SUSAR Suspected Unexpected Serious Adverse Reaction
TMF Trial Master File
UOXF University of Oxford
VE Vaccine efficacy
Vp Viral particles
WHO World Health Organisation 3. BACKGROUND & RATIONALE
3.1 Im pact of MERS-CoV and the nee d for a vaccine The dipeptidyl peptidase 4 (DPP4) receptor is used by the MERS-CoV virus during infection and is highly conserved between Camels and Humans. The MERS-CoV spike (S) protein is a characteristic structural component of the virion membrane and its Si domain mediates binding to DDP4. The spike protein has been chosen as the target antigen for use in the replication-deficient simian adenovirus developed by the University of Oxford, ChAdOxi vaccine vector. ChAdOxi has shown successful results in the development of Oxford lead vaccines which have gone onto enter phase I trials within the UK (7). In this study we propose the manufacture to GMP of the ChAdOXi MERS-S vaccine and its use in a small open labelled phase I trial in Oxford.
3.1. Progress towards a MERS vaccine
Global efforts to develop a coronavirus vaccine faded in the aftermath of SARS-CoV pandemic but has since gained renewed momentum in the face of the current MERS- CoV outbreak. Most of the developed vaccines were based on the S surface
glycoprotein, the primary target for neutralizing antibodies during any natural coronavirus infection. A number of preclinical and clinical studies showed that the SARS-CoV Si protein subunit, and specifically the RBD at its core, could serve as a dominant target for neutralizing antibodies in mice, non-human primates, and humans. Si, therefore, became the basis for a number of promising SARS-CoV vaccine candidates (8).
The Si protein subunit and RBD have also been the basis for several MERS-CoV vaccine candidates. Both constructs have elicited neutralizing antibodies of high potency across multiple viral strains. Despite their demonstrated immunogenicity in animal models and anticipated safety in humans, RBD or Si-subunit based vaccine candidates are limited in their epitope breadth. Vaccine candidates that elicit a more diverse antibody repertoire as well as a robust cellular immuneresponse may offer the advantage of broader and more durable protection (8).
Live attenuated viruses have historically been among the most immunogenic platforms available, as they have the capacity to present multiple antigens across the viral life cycle in their native conformations. However, manufacturing live-attenuated viruses requires complex containment and biosafety measures. Furthermore, live-attenuated viruses carry the risks of inadequate attenuation causing disseminated disease, particularly in immunocompromised hosts. Given that moderately immunocompromised adults with co-morbidities have suffered the most severe MERS-CoV disease, making a live-attenuated virus vaccine is a less viable option. Replication competent viral vectors could pose a similar threat for disseminated disease in the immuno-suppressed. Replication deficient vectors, however, avoid that risk while maintaining the advantages of native antigen presentation, elicitation of T cell immunity and the ability to express multiple antigens (8).
Several recombinant DNA, protein and viral vectored MERS candidate vaccines have been developed and tested in animal models (mice, non-human primates and camels) with varied efficacy results. Recently, a recombinant MVA encoding the full length Spike protein antigen (S) showed partial efficacy by significantly reducing MERS-CoV viral shedding in a camel challenge study. (9, 10)
The first MERS-CoV vaccine to be used in humans has recently entered a phase I dose ranging safety study in January 2016. The GLS-5300, a DNA plasmid vaccine that expresses the MERS-CoV spike (S) glycoprotein, is being administered to 75 healthy adult volunteers in the USA, by the Walter Reed Army Institute of Research. Safety and immunogenicity data are expected to be reported by the end of 2017.
3.2. MERS spike protein as a vaccine antigen
Coronaviruses (CoVs) are spherical and enveloped viruses with large, unsegmented, single positive RNA genomes. One-third of the genome is responsible for coding the structural proteins: spike (S) glycoprotein, small envelope protein (E), integral membrane protein (M), and genome-associated nucleocapsid protein (N). The proteins E, M, and N are mainly responsible for the assembly of the virions, while the S protein is involved in receptor binding and bears membrane fusion capabilities during CoVs infection. Thus, the S protein has an essential role in virus entry and determines tissue and cell tropism, as well as host range (11).
S is a type I, trimeric, transmembrane protein located at the surface of the viral envelope, giving rise to spikeshaped protrusions from the virion. S is 1353 amino acids in length, heavily glycosylated (with 21 predicted N-linked glycosylation sites), and consists of a large ectodomain and a short cytosolic tail. The S proteins of CoVs can be divided into two functional subunits: the N-terminal Si subunit forms the globular head, and the membrane-embedded C-terminal S2 (11). Si and S2 subunits are respectively, responsible for cellular receptor DPP4 binding via the receptor-binding domain (RBD), and fusion of virus and cell membranes, thereby mediating the entry of MERS-CoV into the target cells. The MERS-CoV RBD consists of a core structure, which is homologous to that of the SARS-CoV S protein RBD, and a receptor-binding motif, which is unique to MERS-CoV, thus determining viral pathogenesis and receptor recognition (12).
The roles of S in receptor binding and membrane fusion make it a perfect target for vaccine and antiviral development. Previous studies on SARS-CoV reveal that vaccines based on the S protein can induce antibodies to block virus binding and fusion or neutralize virus infection (11). ChAdOxi MERS-S expresses a codon-optimised coding sequence for Spike protein from the MERS-CoV isolate Camel/Qatar_2_20i4
(GenBank:KJ650098.i),
3.3.Adenovirus-vectored Vaccines
Adenoviruses are attractive vectors for human vaccination. They possess a stable genome so that inserts of foreign genes are not deleted and they can infect large numbers of cells without any evidence of insertional mutagenesis.
Replication defective adenovirus can be engineered by deletion of genes from the El locus, which is required for viral replication, and these viruses can be propagated easily with good yields in cell lines expressing El from AdHu5 such as human embryonic kidney cells 293 (HEK 293 cells) (13).
Previous mass vaccination campaigns in over 2 million adult US military personnel using orally administered live human adenovirus serotype 4 and 7 have shown good safety and efficacy data (14). Human adenoviruses are under development as vectors for malaria, HIV and hepatitis C vaccines, amongst others. They have been used extensively in human trials with excellent safety profile mainly as vectors for HIV vaccines.
A limiting factor to widespread use of human adenovirus as vaccine vectors has been the level of anti-vector immunity present in humans where adenovirus is a ubiquitous infection. The prevalence of immunity to human adenoviruses prompted the consideration of simian adenoviruses as vectors, as they exhibit hexon structures homologous to human adenoviruses (15). Simian adenoviruses are not known to cause pathological illness in humans and the prevalence of antibodies to chimpanzee origin adenoviruses is less than 5% in humans residing in the US.
3.4 ChAdOxi
ChAdOxi is a novel recombinant chimpanzee adenovirus designed as a vaccine vector, developed by The Jenner Institute at the University of Oxford. This viral vector has been used by researchers at the University of Oxford to produce a number of vaccines expressing a range of different antigens. Three phase I clinical trials have been completed in the UK using ChAdOxi with different inserts (two influenza trials and one TB trial).
ChAdOxi is produced from a replication-deficient (El and E3 deleted) simian adenovirus and it has been described by Dicks et al (16). The vector was constructed in a bacterial artificial chromosome (BAC) to facilitate genetic manipulation of genomic clones with improved stability and flexibility. Cellular immunogenicity of recombinant Ei E3-deleted ChAdOxi was comparable to that of other species E derived chimpanzee adenovirus vectors including ChAd63, the first simian adenovirus vector to enter clinical trials in humans. The El region is essential for viral replication, hence the ability to delete Ei renders the new vector immediately replication incompetent. The deletion of the non-essential adenovirus E3 region increases the insert capacity of the new vector by approximately 5Mb. It is known that the proteins encoded by the E4 region of adenoviruses interact with El during viral replication, and the imperfect interaction between the gene products of the AdHu5 Ei gene produced by HEK293 cells and simian E4 gene products has been found to result in impaired viral replication in this cell line, and consequently lower virus yields. In ChAdOxi, Ad5 E40rf4 has been inserted to replace the homologous simian virus coding sequence, resulting in improved viral replication during vaccine production. Since no replication of the virus takes place after immunization, this replacement has no effect on immunogenicity of the viral vector. Insertion of recombinant antigens at the El locus is performed using Gateway® site specific recombination technology (Invitrogen).
3.5 Developm ent of ChAdOxi MERS-S
ChAdOxi MERS-S encodes the Spike (S) surface glycoprotein of the coronavirus. A genomic clone of ChAdOxi MERS-S was prepared by Gateway® recombination between an entry plasmid containing the codon-optimised coding sequence for Spike protein from the MERS-CoV isolate Camel/Qatar_2_20i4 (GenBank:KJ650098.i), and the Ei-and E3-deleted ChAdOxi destination vector.
3.6 Preclinical Studies
3.6.1 Efficacy and Im m unogenicity
Mice (balb/c) were immunised with ChAdOxi or MVA vectored vaccines expressing
MERS-CoV Spike protein. Serum samples were taken after 28 days and endpoint titres measured by ELISA. This study showed that a single dose of ChAdOxi results in equivalent immunogenicity to two doses of MVA. Fig. 8 shows Immunogenicity of viral vectored vaccines MERS vaccines in mice.
An efficacy preclinical study has been conducted where mice transgenic for the hDPP4 receptor were immunised with a single dose of ChAdOxi MERS-S by either intranasal or intramuscular injection. The control ChAdOxi vaccine expressed eGFP as the vaccine antigen. Serum neutralising titres were measured 28 days after vaccination, when the mice were then challenged with MERS CoV by intranasal inoculation. The results showed that mice immunised with the MERS vaccine by either route were completely protected against MERS CoV infection. No virus was detected in the lungs of the mice receiving the MERS vaccine and they all survived, whereas all of the sham- vaccinated mice succumbed to infection within 8 days (Vincent Munster,unpublished data).
Fig. 9 A. Virus neutralising titres in mice amongst ChAdOxi MERS-S and controls administered via intranasal or intramuscularly. B. Viral load after MERS-CoV challenge. C Survival amongst ChAdOxi MERS-S and control mice after intranasal MERS-CoV challenge.
3.6.2 Toxicity
A GLP toxicology study performed on BALB/c mice revealed that intramuscular administration of ChAdOxi MERS-S is safe and well tolerated. Clinical observations, inoculation site reactogenicity, clinical chemistry, clinical haematology, gross necropsy, organ weights and histopathology indicated no overt toxicity related to ChAdOxi MERS-S vaccine administration.
3.7 Previous clinical experience
This will be the first-in-human study employing ChAdOxi MERS-S. However, ChAdOxi vectored vaccines expressing different inserts have previously been used in 126 healthy volunteers taking part in clinical trials conducted by the University of Oxford in the UK (table 1).
ChAdOxi encoding the influenza fusion protein NP+Mi has been safely administered to 84 healthy adult volunteers in the UK in two completed clinical trials conducted at The Jenner Institute (FLU004 and FLU005). FLU004 was a phase I, open-label, non- randomised dose escalation study of ChAdOxi NP+Mi. The vaccine was safe, well tolerated and immunogenic, inducing ELISpot responses at all doses. The dose of 2.5 χ io10 vp was chosen for further studies of ChAdOxi NP+Mi (7). FLU005 was a multicentre phase I, randomised study to determine the safety and immunogenicity of vaccination regimens employing the candidate influenza vaccines MVA-NP+Mi and ChAdOxi NP+Mi. Sixty-nine (69) healthy adult volunteers have received ChAdOxi NP+Mi at a concentration of 2.5 x 1010 vp. Administrations of ChAdOxi NP+Mi and MVA-NP+Mi vaccines were found to be safe and well-tolerated, in agreement with our previous studies (7, 17-20). The majority of adverse events were mild to moderate in nature and lasted for 1-2 days. The most common local adverse event was arm pain at the site of injection and the most common systemic adverse event was mild fatigue and headache.
TB034 was an open-label, phase I clinical trial in which 42 healthy adult volunteers received the ChAdOxi viral vector expressing the Mycobacterium tuberculosis antigen 85A (ChAdOxi 85A). No major safety concerns associated with ChAdOxi 85A
administration have been reported.
ChAdOxi 5T4 has been given in the VANCEoi study which is an ongoing first-in-man open label randomized phase I study to determine the safety and immunogenicity of heterologous prime boost ChAd-MVA vaccination against oncofetal antigen 5T4. To date, 34 participants have received the ChAdOxi 5T4 vaccine at a dose of 2.5 x 1010 vp and only mild AEs related to the vaccination have been reported
VAC067 is an ongoing first-in-man study of the ChAdOxi viral vector expressing dual second generation liver-stage malaria antigens LSAi and LSAP2 (ChAdOxi LS2). No significant safety concerns have been reported until this date.
None of the above mentioned clinical trials reported serious adverse events associated with the administration of ChAdOxi, which was shown to have a good safety profile.
Table 1. Clinical experience with ChAdOxi viral vector vaccines.
Num ber of Volunteers
Country Trial Vaccine Age Route Dose
(Received ChAdOxi)
5x1ο8 vp 3
5x10? vp 3
UK FLU004 ChAdOxi NP+Mi 18-50 IM
2.5x1ο10 vp 3
5x1ο10 vp 6
ChAdOxi NP+Mi
18-50 IM 2.5x1ο10 vp 12
MVANP+Mi (week 8)
ChAdOxi NP+Mi
MVA NP+Mi (week 18-50 IM 2.5x1ο10 vp 12
52)
UK FLU005 MVA NP+Ml
ChAdOxi NP+Mi 18-50 IM 2.5x1ο10 vp 12
(week 8)
MVA NP+Ml
ChAdOxi NP+Mi 18-50 IM 2.5x1ο10 vp 9
(week 52) ChAdOxi NP+Mi >50 IM 2.5x1ο10 vp 12
ChAdOxi NP+Mi
>50 IM 2.5x1ο10 vp 12 MVANP+Mi (week 8)
ChAdOxi 85A 18-50 IM 5x1ο9 vp 6
ChAdOxi 85A 18-50 IM 2.5x1ο10 vp 12
UK TB034
ChAdOxi 85A
18-50 IM 2.5x1ο10 vp 12 MVA85A (week 8)
VANCE01 ChAdOxi.5T4 34 (*as of
UK 18 - 75 IM 2.5x1ο10 vp
(ongoing) MVA.5T4 May 2017)
ChAdOxi LS2 18-45 5x1ο9 vp X
UK VAC067 IM
ChAdOxi LS2 18-45 5x1ο10 vp X
3.8 Rationale
MERS-CoV is an emerging zoonotic viral disease considered a global threat and listed as a priority pathogen for urgent Research and Development. The recent MERS-CoV outbreaks in the Middle East (from 2012 and still ongoing) and South Korea (2015) have caused a total of 652 deaths representing a case fatality rate of approximately 36% and imported cases have now been reported in 27 countries.
Chimpanzee adenovirus vaccine vectors have been safely administered to over 1000 people using a wide range of infectious disease targets including malaria (21), HIV (22), tuberculosis, influenza (7), hepatitis C (23), RSV (24) and, most recently, Ebola (25). ChAdOxi viral vectored vaccines have shown to be both safe and immunogenic in previous clinical trials in the UK (FLU004, FLU005 and TB034). Single-dose
immunisation with ChAdOxi MERS-S vaccine has shown to elicit high levels of neutralising antibody in animal model.
Finally, the One Health vaccine development approach used here, by which the same vaccine is co-developed for humans and susceptible animal species, is well suited to many emerging outbreak pathogens, most of which involve zoonotic transmission (26). The approach allows the possibility of cost reductions for the final product by
increasing the scale of manufacture (27). Ultimately the vaccine could be licensed for use in camels in the Middle East and North Africa. If licensed, human vaccines could be deployed for occupationally exposed individuals such as camel workers and health care professionals, with stockpiles available for use in the case of an outbreak.
3.9 Vaccine Developm ent Strategy
The data from this first in human study will then be used to support a further phase I and phase II study with our collaborators based in the Middle East. This study would be ran in parallel with a study in camels with our collaborators at Qassim University, Saudi Arabia. Testing of the MERS vaccine in camels is funded by the Liverpool School of Tropical Medicine Confidence in Concept Scheme.
4. STUDY OVERVIEW
This is a first-in-human, open-label, dose escalation, phase I clinical trial to assess the safety and immunogenicity of the candidate ChAdOxi MERS-S vaccine in healthy volunteers aged 18-50. The vaccine will be administered intramuscularly.
Volunteers will be recruited and vaccinated at the CCVTM, Oxford. There will be 3 study groups and a total of 24 volunteers will be enrolled (table 2). Staggered enrolment will apply for the first three volunteers within each group. Volunteers will be first recruited into Group 1 and subsequently into Groups 2 and 3 following interim clinical safety reviews (see section 8.4.2). Volunteers will be allocated to a study group by selecting eligible volunteers for enrolment in the order in which they were deemed eligible, following screening.
4.1 Rationale for Selected Doses
Doses to be administered in this trial have been selected on the basis of clinical experience with the ChAdOxi adenovirus vector expressing different inserts and similar adenovirus vectored vaccines (eg. ChAd63).
A first-in-man dose escalation study using the ChAdOxi vector encoding an influenza antigen (FLU004), safely administered ChAdOxi NP+Mi at doses ranging from 5 x 108 to 5 x 1010 vp. Subsequent review of the data identified an optimal dose of 2.5 x 1010 vp balancing immunogenicity and reactogenicity. This dose has subsequently been given to over 100 volunteers in numerous larger phase 1 studies at the Jenner Institute (FLU005, TB034 and VANCE01) and ChAdOxi vectored vaccines have thus far demonstrated to be very well tolerated. The vast majority of AEs have been mild- moderate and there have been no SARs until this date.
Another simian adenovirus vector (ChAd63) has been safely administered at doses up to 2 x 1011 vp with an optimal dose of 5 x 1010 vp, balancing immunogenicity and reactogenicity.
As this is a first-in-human assessment of the MERS-S antigenic insert, the first dose of ChAdOxi MERS-S proposed in this study (5 x 109 vp) is therefore at least 10 fold less than what this new insert is expected to be tolerated (5X 1010 vp). Doses will be gradually increased aiming to provide an optimal dose of ChAdOxi MERS-S
considering the tolerability, reactogenicity and immunogenicity profiles.
4.2 Study Groups
Table 2. Study Groups
Figure imgf000056_0001
4.2.1 First Volunteers
Volunteers will be enrolled and doses will be escalated according to the plan outlined below.
The first volunteer in the study will receive 5 xio9 vp of ChAdOxi MERS-S (group 1). This volunteer will be vaccinated ahead of any other volunteers and the profile of adverse events will be examined after 48h. Provided there are no safety concerns as assessed by the Chief Investigator (CI) and the Local Safety Monitor (LSM), another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following vaccination of the first volunteer and at least 1 hour apart from each other. An independent safety review will be conducted by the LSM after vaccination of the first 3 volunteers in group 1. This review will include the results of safety blood tests at day 7 post vaccination and an assessment of the profile of the adverse events reported. The CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 1 and the first volunteer to receive the next incremental dose in group 2. If there are no safety concerns, the remaining volunteers in Group 1 and the first volunteer in group 2 may be vaccinated.
The same procedure will apply for each of the first 3 volunteers enrolled at higher dosage groups and prior to dose escalation (groups 2 and 3).
4.2.2 Duration of study
The total duration of the study will be 26 weeks from the day of enrolment for all volunteers.
4.2.3 Definition of Start and End of Trial The start of the trial is defined as the date of the first vaccination of the first volunteer. The end of the trial is the date of the last visit of the last volunteer.
4.3 Potential Risks for volunteers
The potential risk to participants is considered as low. The potential risks are those associated with phlebotomy and vaccination. In general, recombinant adenoviral vectors are safe. Similar vaccines encoding different antigens have been given to several thousand volunteers (including children) with a good safety profile.
Phlebotomy:
The maximum volume of blood drawn over the study period (approximately 500ml) should not compromise these otherwise healthy volunteers. There may be minor bruising, local tenderness or pre-syncopal symptoms associated with venepuncture, which will not be documented as AEs if they occur. Vaccination:
ChAdOxi MERS-S has not been used in humans before and therefore will be initially administered at the lower dose of 5 x 109 vp before progressing to the higher doses of 2.5 x 1010 and 5 x 1010 in Groups 2 and 3. Potential expected risks from vaccination include local effects such as pain, redness, warmth, swelling, tenderness or itching. Systemic reactions that could potentially occur following immunisation with a recombinant adenovirus vaccine include a flu-like illness with feverishness, fatigue, malaise, arthralgia, myalgia and headache.
As with any vaccine, Guillain-Barre syndrome or immune-mediated reactions that can lead to organ damage may occur, but this should be extremely rare. Serious allergic reactions including anaphylaxis could also occur and for this reason volunteers will be vaccinated in a clinical area where Advanced Life Support trained physicians, equipment and drugs are immediately available for the management of any serious adverse reactions (SAR). 4.4 Known Potential Benefits
Volunteers will not benefit directly from participation in this study. However, it is hoped that the information gained from this study will contribute to the development of a safe and effective MERS vaccine regime. The only benefits for participants would be information about their general health status.
5 OBJECTIVES AND ENDPOINTS The number of volunteers has been chosen to generate adequate safety and
immunogenicity data to meet these objectives, whilst minimising the number of volunteers exposed to a new vaccination regimen. 5.1 Prim ary Objective
To assess the safety and tolerability of ChAdOxi MERS-S in healthy volunteers.
5.1.1 Prim ary Outcom e Measures
The specific endpoints for safety and reactogenicity will be actively and passively collected data on adverse events.
The following parameters will be assessed for all study groups
• Occurrence of solicited local reactogenicity signs and symptoms for 7 days
following the vaccination
• Occurrence of solicited systemic reactogenicity signs and symptoms for 7 days following the vaccination
• Occurrence of unsolicited adverse events for 28 days following the vaccination
• Change from baseline for safety laboratory measures
• Occurrence of serious adverse events during the whole study duration
Volunteers will undergo clinical follow up for adverse events for a further 182 days following completion of the vaccination regimen. SAEs will be collected throughout the study. The duration of follow up reflects the desire to obtain longer term safety data with the first use of ChAdOxi MERS-S in humans.
5.2 Secondary Objective
To assess the cellular and humoral immunogenicity of ChAdOxi MERS-S in healthy adult volunteers.
5.2.1 Secondary Outcom e Measures
Measures of immunogenicity to the ChAdOxi MERS-S vaccine may include:
· ELISA to quantify antibodies to MERS-S protein
• Ex vivo ELISpot responses to MERS-S
Other exploratory immunology may be carried out in collaboration with other specialist laboratories, including laboratories outside of Europe. This would involve transfer of serum/plasma and/or peripheral blood mononuclear cells (PBMC), but samples would be anonymised. Volunteers will be consented for this.
6 INVESTIGATIONAL PRODUCTS The following vaccinations will be given in this study:
ChAdOxi MERS-S 5 x lo^vp
ChAdOxi MERS-S 2.5 x io10vp
ChAdOxi MERS-S 5 x io10vp
6.1. Manufacturing and Presentation
6.1.1 Description of ChAdOxi MERS-S
ChAdOxi MERS-S vaccine consists of the replication-deficient simian adenovirus vector ChAdOxi, containing the structural surface glycoprotein (Spike protein) antigens of the MERS-CoV expressed from the strong CMV IE promoter.
6.1.2. ChAdOxi MERS-S form ulation and packaging
ChAdOxi MERS-S is manufactured in formulation buffer at a nominal concentration of >i.i x 1011 vp/mL. The drug product is filled into 2mL glass vials with a 13 mm grey bromobutyl rubber freeze-dry stopper (CE Marked, supplied by Adelphi Tubes) and a 13 mm aluminium seal. The nitrogen filled vials are supplied sterile. The containers and closures are tested for compliance with defined specifications. The vials are made from Ph Eur Type 1 glass.
6.2 Supply
ChAdOxi MERS-S has been formulated and vialed under Good Manufacturing Practice conditions at the Clinical Bio-Manufacturing Facility (CBF), University of Oxford. At the CBF the vaccine will be certified and labelled for the trial by a Qualified Person (QP) before transfer to the clinical site.
6.3 Storage
The vaccine is stored at nominal -8o°C in a locked freezer, at the clinical site. All movements of the study vaccines will be documented in accordance with existing standard operating procedure (SOP). Vaccine accountability, storage, shipment and handling will be in accordance with relevant SOPs and forms.
6.4 Adm inistration of Investigational Medicinal Products
On vaccination day, ChAdOxi MERS-S will be allowed to thaw to room temperature and will be administered within 1 hour of removal from the freezer. The vaccine will be administered intramuscularly into the deltoid of the non-dominant arm (preferably). All volunteers will be observed in the unit for 1 hour (±10 minutes) after vaccination. During administration of the investigational products, Advanced Life Support drugs and resuscitation equipment will be immediately available for the management of anaphylaxis. Vaccination will be performed and the IMPs handled according to the relevant SOPs.
6.5 Minim ising environm ental contam ination with genetically m odified organism s (GMO)
The study will be performed in accordance with UK Genetically Modified Organisms (Contained Use) Regulations (2014). In order to minimise dissemination of the recombinant vectored vaccine virus into the environment, inoculation sites will be covered with a dressing after immunisation. This should absorb any virus that may leak out through the needle track. The dressing will be removed from the injection site after 30 minutes (+15/- 5 minutes) and will be disposed as GMO waste by autoclaving.
7.3 Inclusion and exclusion criteria
This study will be conducted in healthy adults, with informed consent, who meet the following inclusion and exclusion criteria:
7.3.1 Inclusion Criteria
The volunteer must satisfy all the following criteria to be eligible for the study:
1. Healthy adults aged 18 to 50 years
2. Able and willing (in the Investigator's opinion) to comply with all study
requirements
3. Willing to allow the investigators to discuss the volunteer's medical history with their General Practitioner
4. For females only, willingness to practice continuous effective contraception (see below) during the study and a negative pregnancy test on the day(s) of screening and vaccination
5. Agreement to refrain from blood donation during the course of the study
6. Provide written informed consent
7.3.2 Exclusion Criteria
The volunteer may not enter the study if any of the following apply:
1. Participation in another research study involving receipt of an investigational product in the 30 days preceding enrolment, or planned use during the study period
2. Prior receipt of an investigational vaccine likely to impact on interpretation of the trial data. 3. Administration of immunoglobulins and/ or any blood products within the three months preceding the planned administration of the vaccine candidate
4. Any confirmed or suspected immunosuppressive or immunodeficient state, including HIV infection; asplenia; recurrent, severe infections and chronic (more than 14 days) immunosuppressant medication within the past 6 months
(inhaled and topical steroids are allowed)
5. History of allergic disease or reactions likely to be exacerbated by any
component of the vaccine
6. History of allergic reaction to Amynoglycoside antibiotics
7. Any history of hereditary angioedema, acquired angioedema, or idiopathic angioedema.
8. Any history of anaphylaxis in relation to vaccination
9. Pregnancy, lactation or willingness/intention to become pregnant during the study
10. History of cancer (except basal cell carcinoma of the skin and cervical
carcinoma in situ)
11. History of serious psychiatric condition likely to affect participation in the study
12. Bleeding disorder (eg. factor deficiency, coagulopathy or platelet disorder), or prior history of significant bleeding or bruising following IM injections or venepuncture
13. Any other serious chronic illness requiring hospital specialist supervision
14. Suspected or known current alcohol abuse as defined by an alcohol intake of greater than 42 units every week
15. Suspected or known injecting drug abuse in the 5 years preceding enrolment 16. Seropositive for hepatitis B surface antigen (HBsAg)
17. Seropositive for hepatitis C virus (antibodies to HCV)
18. Any clinically significant abnormal finding on screening biochemistry or
haematology blood tests or urinalysis
19. Any other significant disease, disorder or finding which may significantly
increase the risk to the volunteer because of participation in the study, affect the ability of the volunteer to participate in the study or impair interpretation of the study data
0. Inability of the study team to contact the volunteer's GP to confirm medical history and safety to participate .3.5 Criteria for postponem ent of vaccination The following events constitute contraindications to administration of the vaccine at that point in time; if any one of these events occurs at the time scheduled for vaccination, the subject may be vaccinated at a later date, or withdrawn at the discretion of the Investigator.
· Acute disease at the time of vaccination. Acute disease is defined as the presence of a moderate or severe illness with or without fever. All vaccines can be administered to persons with a minor illness such as diarrhoea, mild upper respiratory infection with or without low-grade febrile illness, i.e. temperature of≤37.5°C/99.5°F.
· Temperature of >37.5°C (99.5°F) at the time of vaccination.
7.4 Com pliance with Dosing Regim e
All doses in this vaccine study will be administered by the Investigator and recorded in the CRF. The study medication will be at no time in the possession of the volunteer and compliance will not, therefore, be an issue.
7.5 Pregnancy
Should a volunteer become pregnant during the trial, she will be followed up as other volunteers and in addition will be followed until pregnancy outcome. We will not routinely perform venepuncture in a pregnant volunteer.
8 TREATMENT OF TRIAL VOLUNTEERS
This section describes the clinical procedures for evaluating study participants and follow-up after administration of study vaccine.
8.1 Study procedures
All volunteers will have the same schedule of clinic attendances and procedures as indicated in the schedules of attendance (Table 4). All subjects will receive the
ChAdOxi MERS-S vaccine, and undergo follow-up for a total of 26 weeks. The total volume of blood donated during the study will be mL. Additional visits or procedures may be performed at the discretion of the investigators, e.g., further medical history and physical examination, urine microscopy in the event of positive urinalysis or additional blood tests if clinically relevant. 8.2 Observations Pulse, blood pressure and temperature will be measured at the time-points indicated in the schedule of procedures and may also be measured as part of a physical examination if indicated at other time-points.
8.3 Blood Tests and Urinalysis
Blood will be drawn for the following laboratory tests and processed:
1. At Oxford University Hospitals' NHS Trust, or Hammersmith Hospital using NHS standard procedures:
• Haem atology; Full Blood Count
• Biochem istry; Sodium, Potassium, Urea, Creatinine, Albumin, Liver Function Tests (ALT, ALP, Bilirubin)
• Diagnostic serology; HBsAg, HCV antibodies, ΗΓν antibodies (specific
consent will be gained prior to testing blood for these blood-borne viruses)
• Im m unology; Human Leukocyte Antigen (HLA) typing
Additional safety blood tests may be performed if clinically relevant at the discretion of the medically qualified investigators. These generally include, but are not limited to, AST, GGT and a coagulation screen.
2. At University of Oxford research laboratories:
• Exploratory Im m unology; Immunogenicity will be assessed by a variety of immunological assays. This may include antibodies to MERS Spike protein, ex vivo ELISpot assays for interferon gamma and flow cytometry assays, functional antibody assays and B cell analyses. Other exploratory immunological assays including cytokine analysis and other antibody assays, DNA analysis of genetic polymorphisms potentially relevant to vaccine immunogenicity and gene expression studies amongst others may be performed at the discretion of the Investigators.
Urinalysis ; Urine will be tested for protein, blood and glucose at screening. For female volunteers only, urine will be tested for beta-human chorionic gonadotrophin (β-HCG) at screening and immediately prior to each vaccination.
Collaboration with other specialist laboratories in the UK, Europe and outside of Europe for further exploratory immunological tests may occur. This would involve the transfer of serum or plasma and/ or PBMC to these laboratories, but these would remain anonymised. Informed consent for this will be gained from volunteers.
Immunological assays will be conducted according to local SOPs.
6i Subjects will be informed that there may be leftover samples of their blood (after all testing for this study is completed), and that such samples may be stored indefinitely in the Oxford Vaccine Center Biobank for possible future research (exploratory immunology), including human DNA and RNA analyses to search for correlates of vaccine immunogenicity and efficacy. Subjects will be able to decide if they will permit such future use of any leftover samples. With the volunteers' informed consent, any leftover cells, urine and serum/plasma will be frozen indefinitely for future ethically approved research studies of MERS specific or vaccine-related responses. If a subject elects not to permit this, all of that subject's leftover samples will be discarded after the required period of storage to meet Good Clinical Practice (GCP) and regulatory requirements.
8.4 Study visits 8.4.2 Day 0 : Enrolm ent and Vaccination Visit
Volunteers will not be considered enrolled in the study until they have received a vaccine. Before vaccination, the eligibility of the volunteer will be reviewed. Pulse, blood pressure and temperature will be observed and if necessary, a medical history and physical examination may be undertaken to determine need to postpone vaccination depending on criteria listed in section 7.3.5. Vaccinations will be administered as described below.
8.4.2.1 Vaccinations
Before each vaccination, the on-going eligibility of the volunteer will be reviewed. All vaccines will be administered intramuscularly according to SOP VC002 Vaccination as described above in section 6.4. The injection site will be covered with a sterile dressing and the volunteer will stay in the CCVTM for observation, in case of immediate adverse events. Observations will be taken 30 minutes after vaccination (+/- 5 minutes) and the sterile dressing removed and injection site inspected. Observations will also be taken at 60 minutes (+/- 10 minutes) before the volunteer leaves. An oral thermometer, tape measure and diary card (paper or electronic) will be given to each volunteer, with instructions on use, along with the emergency 24 hour telephone number to contact the on-call study physician if needed.
Diary cards will collect information on the timing and severity of the following solicited AEs:
Table 3. Solicited AEs as collected on post vaccination diary cards Local solicited AEs System ic solicited AEs
Pain Fever
Redness Feverishness
Warmth Joint pains
Itch Muscle pains
Fatigue
Headache
Nausea
Malaise
Volunteers will be instructed on how to self-assess the severity of these AEs. There will also be space on the diary card to self-document unsolicited AEs, and whether medication was taken to relieve the symptoms.
8.4.2.2 Sequence of Enrolm ent and Vaccination of Volunteers
For safety reasons, the first volunteer in Group ι will be vaccinated ahead of any other volunteers and the profile of adverse events will be reviewed after 48 hours post vaccination. Provided there are no safety concerns, as assessed by the CI and the LSM, another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following the first volunteer being vaccinated and at least 1 hour apart from each other. An independent safety review will be conducted by the LSM after vaccination of the first three volunteers. This review will include an assessment of the profile of adverse events and the results of safety blood tests at day 7 post vaccination. The CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 1 and the first volunteer to receive the next incremental dose in group 2. If there are no safety concerns, the remaining volunteers in Group 1 and the first volunteer in group 2 may be vaccinated.
Enrolment of the first volunteer in Group 2 will only proceed if the CI and LSM assess the data from the first three vaccinees in Group 1 as indicating that it is safe to do so. The first subject in Group 2 will be vaccinated alone, and a 48 hour gap allowed before vaccinating further subjects in this group. Provided there are no safety concerns, as assessed by the CI and the LSM, another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following the first volunteer being vaccinated and at least 1 hour apart from each other. An independent safety review will be conducted by the LSM after vaccination of the first three volunteers. This review will include an assessment of the profile of adverse events and the results of safety blood tests at day 7 post vaccination. The CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 2 and the first volunteer to receive the next incremental dose in group 3. If there are no safety concerns, the remaining volunteers in Group 2 and the first volunteer in group 3 may be vaccinated.
Enrolment of the first volunteer in Group 3 will only proceed if the CI and LSM assess the data from the first three vaccinees in Group 2 as indicating that it is safe to do so. The first subject in Group 3 will be vaccinated alone, and a 48 hour gap allowed before vaccinating further subjects in this group. Provided there are no safety concerns, as assessed by the CI and the LSM, another 2 volunteers will be vaccinated at the same dose after at least 48 hours has elapsed following the first volunteer being vaccinated and at least 1 hour apart from each other. An independent safety review will be conducted by the LSM after vaccination of the first three volunteers. This review will include an assessment of the profile of adverse events and the results of safety blood tests at day 7 post vaccination. The CI and the LSM will be asked to provide the decision on whether to proceed with vaccinations of the remaining participants in group 3. If there are no safety concerns, the remaining volunteers in Group 3 may be vaccinated. 8.4.3 Subsequent visits : days 2, 7, 14, 28 , 56 and 182.
Follow-up visits will take place 48 hours (±24h), 7 days (± 2 days), 14 days (± 3 days), 28 days (± 3 days), 56 days (± 7 days) and 182 (± 14 days) after vaccination. Volunteers will be assessed for local and systemic adverse events, interim history, physical examination, review of diary cards (paper or electronic) and blood tests at these time points as detailed in the schedule of attendances. Blood will also be taken for exploratory immunology purposes.
If volunteers experience adverse events (laboratory or clinical), which the investigator (physician), CI and/or LSM determine necessary for further close observation, the volunteer may be admitted to an NHS hospital for observation and further medical management under the care of the Consultant on call.
Table 4. Schedule of attendances Attendance Number Is 2 3 4 5 6 7 8
Timeline** <
0 2 7 14 28 56 182 (days) 90
Time window (days) ±1 ±2 ±3 ±3 ±7 ±14
Informed Consent X
Review contraindications, inclusion
X X
and exclusion criteria
Vaccination X
Vital signsA X X X X X X X X
Ascertainment of adverse events X X X X X X X
Diary cards provided X
Diary cards collected X
(X
Medical History, Physical Examination X (X) (X) (X) (X) (X) (X)
)
Biochemistry®, Haematology (mL) 5 5 5 5 5
Exploratory immunology£ (mL) 50 50 50 50 50
Urinalysis X
Urinary HCG (women only) X X
HLA typing (mL) 4
HBsAg, HCV Ab, HIV serology (mL) 5
Blood volume per visit 10 59 5 50 55 50 50
12
Cumulative blood volume% 10 69 74 79 184 234 284
9
S = screening visit; (X) = if considered necessary A = Vital signs includes pulse, blood pressure and temperature; $ = Biochemistry will include Sodium, Potassium, Urea, Creatinine, Albumin and Liver function tests. £ = Exploratory immunology includes antibodies to MERS S, ex vivo interferon-gamma ELISpot responses to MERS S ** Timeline is approximate only. Exact timings of visits relate to the day on enrolment, e, each visit must occur at indicated number of days after enrolment ± time window. % Cumulative blood volume for Oxford volunteers if blood taken as per schedule, and excluding any repeat safety blood test that may be necessary. 9 ASSESSMENT OF SAFETY
Safety will be assessed by the frequency, incidence and nature of adverse events and serious adverse events arising during the study. 9.2 Foreseeable Adverse Reactions :
The foreseeable ARs following vaccination with ChAdOxi MERS-S include injection site pain, erythema, warmth, swelling, pruritus, myalgia, arthralgia, headache, fatigue, fever, feverishness, malaise and nausea. 9.3 Expected Serious Adverse Events
No serious adverse events are expected in this study.
9.6 Assessm ent of severity
The severity of clinical and laboratory adverse events will be assessed according to the scales in Tables 6-8, also described in the SOP VC027.
Table 6. Severity grading criteria for local adverse events.
Figure imgf000068_0001
*erythema or swelling≤3mm is an expected consequence of skin puncture and will therefore not be considered an adverse event.
Table 7. Severity grading criteria for physical observations
Figure imgf000068_0002
Systolic hypertension (mmHg) 141 - 159 160 - 179 ≥i8o
Systolic hypotension 85 - 89 80 - 84 <8o
(mmHg)***
Diastolic hypertension (mmHg) 91 - 99 100 - 109 ≥110
Taken after≥io minutes at rest
**Use clinical judgement when characterising bradycardia among some healthy subject populations, for example, conditioned athletes.
***Only if symptomatic (e.g. dizzy/ light-headed)
Table 8. Severity grading criteria for local and systemic AEs.
Figure imgf000069_0001
10. STATISTICS
This is a descriptive safety study, where volunteers will be vaccinated with a single dose of ChAdOxi MERS-S. Twenty four (24) volunteers will be vaccinated in total. This sample size should allow an estimation to be made of the frequency and magnitude of outcome measures, rather than aiming to obtain statistical significance for differences between groups. Safety data will be presented according to frequency, severity and duration of adverse events.
Non-parametric tests will be used to determine differences in the primary
immunogenicity outcome (ELISpot) data. A Wilcoxon signed rank test will permit testing of differences in responses between time points within a group and a Mann- Whitney U test will permit testing of differences in responses between different groups.
Sam ple Size Selection
This is a descriptive phase I first in human trial that will balance the safety of volunteers with the aims to assess the vaccine's safety profile and immunogenicity after selected doses of the vaccines. The primary dose comparison will be between Groups 1, 2 and 3, which will have 6-9 subjects each. MERS S-specific immunogenicity will be the key immunological readout assessed by a variety of immunological assays.
12. ETHICS 12.1 Declaration of Helsinki
The Investigators will ensure that this study is conducted according to the principles of the current revision of the Declaration of Helsinki. 12.2 Guidelines for Good Clinical Practice
The Investigators will ensure that this study is conducted in full conformity with the Good Clinical Practice (GCP).
12.3 Approvals
The protocol, informed consent form, participant information sheet and any proposed advertising material will be submitted to an appropriate Research Ethics Committee (PvEC), HPvA (where required), regulatory authorities (MHRA in the UK), and host institution(s) for written approval.
The Investigator will submit and, where necessary, obtain approval from the above parties for all substantial amendments to the original approved documents
12.5 Volunteer Confidentiality
All data will be anonymised: volunteer data will be identified by a unique study number in the CRF and database.
Exam ple 4 - Im m unohistopathology
We refer to Figure 10. Intramuscular (IM)
A-B: Hematoxylin & Eosin
A: Vaccinated with unrelated Adenovirus; Lung: Normal - 100X
B: Chimp Adenovirus/MERS-CoV vaccine; Lung: Normal - 100X Intranasal (IN)
C-D: Hematoxylin & Eosin
C: Vaccinated with unrelated Adenovirus; Lung: Multifocal, mild, interstitial infiltrates with lymphocytic perivascular cuffing- 100X.
D: Chimp Adenovirus/MERS-CoV vaccine; Lung: rare interstitial infiltrates with lymphocytic perivascular culling - 100X Thus it is demonstrated that there is clearly no evidence of enhanced pathology or hypersensitivity in the MERS vaccinated groups.
Exam ple 5 - Safety Report
This example presents safety and immunogenicity data for the MERS-CoV vaccine ChAdOxi MERS in both human volunteers (MERSooi phase I clinical trial) and camels, the principal animal reservoir for MERS coronavirus (MERS-CoV).
Safety report for MERS00 1 phase I clinical trial
MERSooi is a phase I clinical trial to determine the safety and immunogenicity of the candidate MERS-CoV vaccine ChAdOxi MERS in UK healthy adult volunteers. The study groups are shown in the table below.
Table : Study groups for low (5 x 109 viral particles (vp)), m edium (2.5 x 10 vp) and high (5 x 1010 vp) dose vaccinations.
Group ChAdOxi MERS dose
1 (n=6) 5 x 109 vp
2 (n=9) 2.5 x 101G vp
3 (n=9¾ 5 x 101C> vp
Safety reviews are required prior to each dose escalation of ChAdOxi MERS and prior to enrolment of the remaining volunteers in the group. The first review takes place after 3 volunteers in Group l have all received their single dose (5 x 109 vp) of ChAdOxi MERS and completed 7 days of follow-up post vaccine administration. The second review takes place after 3 volunteers in Group 2 have all received their single dose (2.5 x 1010 vp) of ChAdOxi MERS and completed 7 days of follow-up post vaccine administration. At the time of writing, 3 volunteers have been enrolled in group 1, and 3 volunteers have been enrolled in group 2. All enrolled volunteers to date have reached their day 7 follow-up post vaccination. The table below shows the current progress of enrolment (completed follow-up visits in bold text). Table : Current progress of enrolm ent (com pleted follow -up visits in bold text) .
Participant DO D2 D7
MERS-00101006 14/03/20 18 16/03/20 18 21/03/20 18 Group 1 MERS-00101004 20/03/2018 22/03/2018 27/03/2018
MERS-ooioioii 12/04/2018 14/04/2018 19/04/2018
MERS-00101012 25/04/2018 27/04/2018 02/05/2018
Group 2 MERS-ooioioio 02/05/2018 04/05/2018 09/05/2018
MERS-00101013 03/05/2018 05/05/2018 10/05/2018
Participant D14 D28 D56 D182
MERS-00101006 28/03/2018 11/04/2018 09/05/2018 12/09/2018
Group 1 MERS-00101004 03/04/2018 17/04/2018 15/05/2018 18/09/2018
MERS-ooioioii 26/04/2018 10/05/2018 07/06/2018 11/10/2018
MERS-00101012 09/05/2018 23/05/2018 20/06/2018 24/10/2018
Group 2 MERS-ooioioio 16/05/2018 30/05/2018 27/06/2018 31/10/2018
MERS-00101013 17/05/2018 31/05/2018 28/06/2018 01/11/2018
Adverse events (5 x 109 p, low dose)
Solicited adverse events (AEs) after single dose of ChAdOxl MERS (5 x 109 vp):
The 7-day data from the E-diaries of the first 3 participants in Group 1, following their single vaccination, is shownin the table below (mild grade 1 AEs in yellow, moderate grade 2 AEs in orange). There have been no severe AEs and no SAEs following vaccine administration.
Volunteers MER-00101006 and MER-00101011 reported a short-lived mild pruritus at injection site at their 30 minutes observation in clinic. A Grade 1 mild redness at injection site (4mm) was observed at the 60 minutes review following vaccine
administration in volunteer MER-00101011. All volunteers reported at least one systemic AE after vaccination. Systemic AEs were mild or moderate in nature and resolved within 24h.
Table: Solicited AEs self-reported (e-diaries) during 7 days after vaccine administration (5 x 10? vp ChAdOxl MERS)
Figure imgf000072_0001
MER- 00101006 0 2 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 2 2 0 0 0 0 0
Pain DO Dl D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 0 0
Itch DO Dl D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 0 0
Warm th DO Dl D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 0 0
Tem perature DO D l D2 D3 D4 D5 D6
MER- 00101006 36.2 36.3 36.I 36 36.I 35-8 36
MER- 00101004 36.5 36.3 36.4 36.4 36.4 36.7 36.8 MER- 00101011 35-3 35-7 35-8 36.8 35-8 36.3 36.9
Arthralgia DO Dl D2 D3 D4 D5 D6
MER- 00101006 1 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 0 0
Myalgia DO Dl D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 1 0
Feverishness DO Dl D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 1
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 0 0
Headache DO D l D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 2 0 0 0 0 0 0
MER- 00101011 1 0 0 0 1 0 0
Fatigue DO Dl D2 D3 D4 D5 D6 MER- 00101006 0 1 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 1 0 0 0 0 0
Nausea DO Dl D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 0 0 0 0 0
Malaise DO D l D2 D3 D4 D5 D6
MER- 00101006 0 0 0 0 0 0 0
MER- 00101004 0 0 0 0 0 0 0
MER- 00101011 0 0 1 0 0 0 0
Uns o licited AEs after single dos e of ChAdOxl MERS (5 x 109 vp )
Volunteer MER-00101006 reported mild (grade 1) 'Blocked nose', 'Running nose' and 'Cold symptoms' from Dl until Dio post vaccination. They also reported a mild and self-limited 'Loose bowel movement' episode on D5. Volunteer MER-00101011 reported mild and short-lived 'Back pain' on Dl. They also reported mild 'Swollen glands / soreness when swallowing' symptoms from D2 until D6.
Serio us Adv ers e Ev ents
No SAEs have been reported until this date.
Clin ica l Ob serv a tio ns
Observations are taken at each clinic visit from day o - 28. All three volunteers have been reviewed at the day 2 and day 7. Observations at all these time-points have been unremarkable. Lab ora tory AEs
There are no Laboratory AEs reported to date. Adverse events (2.5 x 10 10 p, m edium dose)
So licited AEs after s ingle dos e of ChAdOxl MERS (2.5 x 1010 vp )
The 7-day data from the E-diaries of the first 3 participants in Group 2, following their single vaccination, is shown below in table (mild grade 1 AEs in yellow, moderate grade 2 AEs in orange). There have been no severe solicited AEs and no SAEs following vaccine administration.
Volunteers MER-00101016 reported mild and short-lived warmth at injection site at their 30 min observation in clinic which completely resolved by their 60 min review. All volunteers reported at least one systemic AE after vaccination. Systemic AEs were mild or moderate in nature and resolved within 5 days.
Table : Solicited AEs self-reported (e-diaries) during 7 days after vaccine adm inistration (2.5 x 1010 vp ChAdOxl MERS) .
Redness DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 0 0 0 0 0 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 0 0 0 0 0 0 0
Pain DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 1 0 0 0 0 0
MER- 00101010 1 1 0 0 0 0 0
MER- 00101013 0 1 0 0 0 0 0
Itch DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 0 0 0 0 0 0 MER- OOIOIOIO 0 0 0 0 0 0 0
MER- 00101013 0 0 0 0 0 0 0
Warm th DO D l D2 D3 D4 D5 D6
MER-
00101012 0 0 0 0 0 0 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 0 0 0 0 0 0 0
Tem perature DO D l D2 D3 D4 D5 D6
MER-
00101012 36.3 36.7 35-9 36.3 36.9 36.9 36.9
MER- 00101010 35-9 36.6 35-9 35-3 36.2 35-8 36.1
MER- 00101013 36.8 36.5 36.8 36.9 36.3 37 36.3
Arthralgia DO D l D2 D3 D4 D5 D6
MER-
00101012 0 1 1 1 1 1 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 0 0 0 0 0 0 0
Myalgia DO D l D2 D3 D4 D5 D6
MER-
00101012 0 0 0 0 0 0 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 0 0 0 0 0 0 0 Feverishness DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 1 0 0 0 0 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 0 0 0 0 0 0 0
Headache DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 1 0 0 0 0 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 0 1 0 0 0 0 0
Fatigue DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 1 1 0 0 0 0
MER- 00101010 0 0 0 0 0 0 0
MER- 00101013 1 1 0 0 0 0 0
Nausea DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 0 0 0 0 0 0
MER- 00101010 0 0 0 0 1 1 2
MER- 00101013 0 0 0 0 0 0 0
Malaise DO Dl D2 D3 D4 D5 D6
MER-
00101012 0 0 0 0 0 0 0 MER- OOIOIOIO 0 0 0 0 1 1 2
MER- 00101013 0 0 0 0 0 0 0
Uns o licited AEs after single dos e of ChAdOxl MERS (2.5 x 1010 vp)
Volunteer MER-00101012 reported mild (grade 1) Photophobia and Earache on Di which resolved within 24.I1. Volunteer MER-00101010 reported mild Lack of appetite on Di and moderate 'Diarrhoea' on D2. They remained asymptomatic on D3, but then reported gastroenteritis symptoms starting with mild Nausea, Malaise & Vomiting on D4 which got progressively worse until D7 when they experienced severe (grade 3) Nausea, Abdominal Pain, Vomiting and Diarrhoea. Volunteer called their GP who prescribed anti-emetics and advised increased fluids intake. Contacted over the phone on D8 (date of this report) and volunteer reported significant improvement on frequency of Diarrhoeal episodes and Nausea, Vomiting and Abdominal Pain have now ceased. Volunteer MER-00101013 reported mild Light-headedness on D2, which resolved within 24h. Serio us Adv ers e Ev ents
No SAEs have been reported until this date.
Clin ica l Ob serv a tio ns
Observations are taken at each clinic visit throughout the trial. All three volunteers have been reviewed at days 2 and 7 following vaccination. Volunteer MERS-00101010 had a grade 1 (mild) Bradycardia with a heart rate of 53 bpm at 60 min post vaccine administration and 54 bpm at D7 (baseline heart rate = 67 bpm). Volunteer was well and asymptomatic. There are no further significant changes in their clinical observations.
Lab ora tory AEs
Volunteer MERS-00101012 had a grade 2 (moderate) microcytic anaemia (Hb = 101 g/L, MCV = 75.2 fl on D2; Hb = 104 g/L, MCV 74.3 fl on D7). This represents a nonsignificant change from their baseline microcytic anaemia likely due to iron deficiency (Hb = 105 g/L, MCV = 75.7 on Do).
Volunteer MERS-00101013 had a grade 2 (moderate) normocytic anaemia (Hb = 104 g/L, MCV = 85 fl on D7). This represents a non-significant change from their baseline normocytic anaemia (Hb = 106 g/L, MCV = 86 on Do). This volunteer also had a grade l (mild) Neutropenia (N = 1.4 X109/L). All safety blood tests will be repeated at D28, as per protocol.
Adverse events (5 x 10 10 p, high dose)
The first Group 3 volunteer is scheduled to be vaccinated with the highest dose of ChAdOxi MERS (5 x 1010 vp) on the 14th May 2018, pending the review of the safety report data.
Exam ple 6 : MERS00 1 Interim analysis of im m unolo gy data.
Methods
ELISPOT
The Enzyme-Linked ImmunoSpot (ELISPOT) assay is a widely used method for monitoring cellular immune responses. Herein it is used to measure interferon gamma (IFNy produced by T cells (activated in volunteers in response to vaccination) responding to MERS CoV peptides in culture. Ex vivo (18-h stimulation) ELISPOT assays were performed using Multiscreen IP ELISPOT plates (Millipore), human IFN SA-ALP antibody kits (Mabtech) and BCIP NBT-plus chromogenic substrate (Moss Inc). Cells were cultured in RPMI (Sigma) containing 10% heat-inactivated, sterile- filtered fetal calf serum, previously screened for low reactivity (Labtech International). Antigens were tested in duplicate with 250,000 PBMC added to each well of the ex vivo ELISPOT plate. Peptides were 20 amino acids in length, overlapping by 10 amino acids (Neopeptide), assayed in 13 pools of peptides at 10 mg/ml (tables below).
Tables: MERS-CoV Si peptide pools for ELISPOT assay. tPA indicates peptides covering the leader
sequence/ secretory tissue plasminogen activator amino acid sequence which is fused to the N-terminus of the MERS-CoV spike protein antigen. Responses to tPA are not included in the ELISPOT response SFC counts.
Peptide pool - tPA
MDAMKRGLCCVLLLC RGLCCVLLLCGAVFV VLLLCGAVFVSASQE GAVFVSASQEIHARF SASQEIHARFRRIHS Peptide pools
2 3 4 5 6
IHARFPvRIHSVFLLM LFVANYSQ D VKQ FAN ATYHTPATDCSDGNY WAAFYVYKLQPLTFL LSLFSVNDFTCSQIS ANQYSPCVSrVPSTV
RRIH SVFLLMFLLTP YSQDVKQFANGFWR PATDCSDGNYNRNAS VYKLQPLTFLLDFSV VNDFTCSQISPAAIA PCVSrVPSTVWEDGD
VFLLMFLLTPTESYV KQFANGFWRIGAAA SDGNYNRNASLNSFK PLTFLLDFSVDGYIR CSQISPAAIASNCYS VPSTVWEDGDYYRK
FLLTPTESYVDVGPD GFWRIGAAANSTGT NRNASLNSFKEYFNL LDFSVDGYIRRAIDC PAAIASNCYSSLILD WEDGDYYRKQLSPL
TESYVDVGPDSVKSA IGAAANSTGTVIISP LNSFKEYFNLRNCTF DGYIRRAIDCGFNDL SNCYSSLILDYFSYP YYRKQLSPLEGGGW
DVGPDSVKSACIEVD NSTGTVIISPSTSAT EYFNLRNCTFMYTYN RAIDCGFNDLSQLHC SLILDYFSYPLSMKS LSPLEGGGWLVASG
SVKS ACIEVD IQ QTF VIISPSTSATIRKrf RNCTFMYTYNITEDE GFNDLSQLHCSYESF YFSYPLSMKSDLSVS GGGWLVASGSTVAM
CIEVDIQQTFFDKTW STSATIRKIYPAFML MYTYNITEDEILEWF SQLHCSYESFDVESG LSMKSDLSVSSAGPI VASGSTVAMTEQLQ
IQQTFFDKTWPRPID IRKrYPAFMLGSSVG ITEDEILEWFGITQT SYESFDVESGVYSVS DLSVSSAGPISQFNY TVAMTEQLQMGFGI
FDKTWPRPIDVSKAD PAFMLGSSVGNFSDG ILEWFGITQTAQGVH DVESGVYSVSSFEAK SAGPISQFNYKQSFS EQLQMGFGITVQYG
PRPIDVSKADGIIYP GSSVGNFSDGKMGRF GITQTAQGVHLFSSR VYSVSSFEAKPSGSV SQFNYKQSFSNPTCL GFGITVQYGTDTNSV
VSKADGIIYPQGRTY NFSDGKMGRFFNHTL AQGVHLFSSRYVDLY SFEAKPSGSWEQAE KQSFSNPTCLILATV VQYGTDTNSVCPKL
GirfPQGRTYSNITI KMGRFFNHTLVLLPD LFSSRYVDLYGGNMF PSGSWEQAEGVECD NPTCLILATVPHNLT DTNSVCPKLEFAND
QGRTYSNITITYQGL FNHTLVLLPDGCGTL YVDLYGGNMFQFATL VEQAEGVECDFSPLL ILATVPHNLTTITKP CPKLEFANDTKIASQ
SNITITYQGLFPYQG VLLPDGCGTLLRAFY GGNMFQFATLPVYDT GVECDFSPLLSGTPP PHNLTTITKPLKYSY FANDTKIASQLGNC
TYQGLFPYQGDHGDM GCGTLLRAFYCILEP QFATLPVYDTIKYYS FSPLLSGTPPQVYNF TITKPLKYSYINKCS KIASQLGNCVEYSLY
FPYQGDHGDMYVYSA LRAFYCILEPRSGNH PVYDTIKYYSIIPHS SGTPPQVYNFKRLVF LKYSYINKCSRLLSD LGNCVEYSLYGVSG
DHGDMYVYSAGHATG CILEPRSGNH CPAGN IKYYSIIPHSIRSIQ QVYNFKRLVFTNCNY INKCSRLLSDDRTEV EYSLYGVSGRGVFQ
YVYSAGHATGTTPQK RSGNHCPAGNSHTSF IIPHSIRSIQSDRKA KRLVFTNCNYNLTKL RLLSDDRTEVPQLVN GVSGRGVFQNCTAV
GHATGTTPQKLFVAN CPAGNSHTSFATYHT IRSIQSDRKAWAAFY TNCNYNLTKLLSLFS DRTEVPQLVNANQYS GVFQNCTAVGVRQQ
TTPQ K LFVAN YSQ D V SHTSFATYHTPATDC SDRKAWAAFYVYKLQ NLTKLLSLFSVNDFT PQLVNANQYSPCVSI CTAVGVRQQRFVYD
Peptide pools continued
7 8 9 10 11
VRQQRFVYDAYQNLV PLGQSLCALPDTPST LHGANLRQDDSVRNL D VN M E AAYTSS LLCS GAISASIGDIIQRLD
FVYDAYQNLVGYYSD LCALPDTPSTLTPRS LRQDDSVRNLFASVK AAYTSSLLGSIAGVG SIGDIIQRLDVLEQD
YQNLVGYYSDDGNYY DTPSTLTPRSVRSVP SVR LFASVKSSQSS SLLGSIAGVGWTAGL IQRLDVLEQDAQIDR
GYYSDDGNYYCLRAC LTPRSVRSVPGEMRL FASVKSSQSSPIIPG IAGVGWTAGLSSFAA VLEQDAQIDRLINGR
DGNYYCLRACVSVPV VRSVPGEMRLASIAF SSQSSPIIPGFGGDF WTAGLSSFAAIPFAQ AQIDRLINGRLTTLN
CLRACVSVPVSVT D GEMRLASIAFNHPIQ PIIPGFGGDFNLTLL SSFAAIPFAQSIFYR LINGRLTTLNAFVAQ
VSVPVSVIYDKET T ASIAFNHPIQVDQLN FGGDFNLTLLEPVSI IPFAQSIFYRLNGVG LTTLNAFVAQQLVRS
SWDKETKTHATLF NHPIQVDQLNSSYFK NLTLLEPVSISTGSR SIFYRLNGVGITQQV AFVAQQLVRSESAAL
KETKTHATLFGSVAC VDQLNSSYFKLSIPT EPVSISTGSRSARSA LNGVGITQ QVLSENQ QLVRSESAALSAQLA
HATLFGSVACEHISS SSYFKLSIPTNFSFG STGSRSARSAIEDLL ITQQVLSENQKLIAN ESAALSAQLAKD KVN
GSVACEHISSTMSQY LSIPTNFSFGVTQEY SARSAIEDLLFDKVT LSENQKLIANKFNQA SAQLAKDKVNECVKA
EHISSTMSQYSRSTR NFSFGVTQEYIQTTI IEDLLFDKVTIADPG KLIANKFNQALGAMQ KDKVNECVKAQSKRS
TMSQYSRSTRSMLKR VTQEYIQTTIQKVTV FDKVTIADPGYMQGY KFNQALGAMQTGFTT ECVKAQSKRSGFCGQ
SRSTRSMLKRRD STY IQTTIQKVTVDCKQY IADPGYMQGYDDCMQ LGAMQTGFTTTNEAF QSKRSGFCGQGTHIV
SMLKRRDSTYGPLQT QKVTVDCKQYVCNGF YMQGYDDCMQQGPAS TGFTTTNEAFRKVQD GFCGQGTHrVSFWN
RDSTYGPLQTPVGCV DCKQYVCNGFQKCEQ DDCMQQGPASARDLI TNEAFRKVQDAVNNN GTH IVSFVVNAPNGL
GPLQTPVGCVLGLVN VCNGFQKCEQLLREY QGPASARDLICAQYV RKVQDAVNNNAQALS SFWNAPNGLYFMHV
PVGCVLGLVNSSLFV QKCEQLLREYGQFCS ARDLICAQYVAGYKV AVNNNAQALSKLASE APNGLYFMHVGYYPS
LGLVNSSLFVEDCKL LLPvEYGQFCSKINQA CAQYVAGYKVLPPLM AQALSKLASELSNTF YFMH VGYYPSNH IEV SSLFVEDCKLPLGQS GQFCSKINQALHGAN AGYKVLPPLMDVNME KLASELSNTFGAISA GYYPSNH IEWSAYG EDCKLPLGQSLCALP KINQALHGANLRQDD LPPLMDVNMEAAYTS LSNTFGAISASIGDI NHIEWSAYGLCDAA
Peptide pools continued
12 13
VSAYGLCDAANPTNC DFQDELDEFFKNVST INTTLLDLTYEMLSL LALCVFFILCCTGCG
LCDAANPTNCIAPVN LDEFFKNVSTSIPNF LDLTYEMLSLQQWK FFILCCTGCGTNCMG
NPTNCIAPVNGYFIK KNVSTSIPNFGSLTQ EMLSLQQWKALNES CTGCGTNCMGKLKCN
IAPVNGYFIKTNNTR SIPNFGSLTQINTTL QQWKALNESYIDLK TNCMGKLKCNRCCDR
GYFIKTNNTRIVDEW GSLTQINTTLLDLTY ALNESYIDLKELGNY KLKCNRCCDRYEEYD
TNNTRIVDEWSYTGS TYQNISTNLPPPLLG YIDLKELGNYTYYNK RCCDRYEEYDLEPHK
rVDEWSYTGSSFYAP STNLPPPLLGNSTGI ELGNYTYYNKWPWYI YEEYDLEPHKVHVH
SYTGSSFYAPEPITS PPLLGNSTGIDFQDE TYYNKWPWYIWLGFI
SFYAPEPITSLNTKY NSTGIDFQDELDEFF WPWYIWLGFIAGLVA
EPITSLNTKYVAPQV VAPQVTYQNISTNLP WLGFIAGLVALALCV
LNTKYVAPQVTYQNI AGLVALALCVFFILC
Responses were averaged across triplicates, responses in unstimulated (negative control) wells were subtracted and then responses in individual pools were summed. Staphylococcal enzyme B (0.04 mg/ml) and phytohaemmagglutinin-L (20 ug/ml) were used as a positive control. Plates were counted using an AID automated ELISPOT counter (AID Diagnostika GmbH, algorithm C), using identical settings for all plates and counts were adjusted only to remove artefacts. Responses to the negative control were always <8o spot forming cells (SFC) per million peripheral blood mononuclear cells (PBMC) and >8oo SFC for the positive control. Results are shown in figure 11. Results
T cell resp o ns es to ChAdOxl MERS (5 x 109 vp, lo w dose)
ELISPOT responses as spot forming cells (IFN/ producing activated T cells) per million PBMCs were determined for each volunteer pre vaccination, and at 14 and 28 days post vaccination. Results are displayed in figure 11.
Figure 11 shows spot forming cells (T cells activated in response to MERS CoV peptides) per million peripheral blood mononuclear cells (the rest of the cells in a given volume of blood). Group 1 (n=3) volunteers were vaccinated with a single dose of ChAdOxl MERS (5 x io? vp, low dose). Data points indicate the summed response for each volunteer to all pools of peptides in the MERS Si protein component of the
ChAdOxl MERS vaccine. All volunteers showed a significant increase in response at day 14 after vaccination (p<o.05, Kruskall-Wallis test).
Responses to MERS Si protein peptide pools prior to vaccination were low, with a median response of 85 SFC per million PBMC, increasing to a median of 1411 SFC at day 14 in the low dose group. Individual responses are tabulated below.
Individual ELISPOT responses (m ean per patient) to MERS CoV peptide pools :
SFC per m illion PBMC
Dose D56
Patient Group
(v. p.) DO D14 D28
004 84 1411 984 X
006 1 5X109 52 3304 2895 X
Oil 291 1359 X
012 2 2.5x1ο10 85 X X 010 X X
013 X X
X indicates data due in the next two weeks.
Exam ple 7: Cam el study Methods
ChAdOxl MERS
Average from 3 vials batch 01N 17-01: 1.74 x 1011 vp/mL and 1.61 x 109 ifu/mL.
Co m m ercia l MERS-Co V SI sp ecific ELISA fro m Euro iin in 11 n
Antibody responses in vaccinated camels were evaluated using a commercial camel MERS-CoV Si specific ELISA kit from Euroimmun (1). The results of this kit are presented as ratios of optical density (OD) of each serum sample over the OD of a provided positive calibrator, the positive cut-off is 1.1 following manufacturers recommendation.
In ho use MERS-Co V SI sp ecific ELISA
Enzyme-linked immunosorbent assay (ELISA) is an assay technique designed for detecting and quantifying substances such as peptides, proteins, antibodies and hormones. Herein it is used to measure serum antibody responses to the MERS CoV Si protein. ELISA plates were coated with 2 μg/ml capturing antigen (Si recombinant protein from ATGen Co. Ltd., CA, USA, provided as part of a collaboration with the International Vaccine Institute of Seoul National University, Seoul, Korea), and standard endpoint ELISA protocol was followed, as previously described (2). Sera were prepared in a 10-fold serial dilution in PBS/T and then 50 μΐ were plated in duplicate wells. Anti-camel IgG conjugated to alkaline phosphatase (Sigma) and PNPP tablet (20 mg p-nitrophenylphosphate, SIGMA) substrate were used in the assay.
Virus n e utra lisa tio n assay
Induction of virus-neutralising antibodies was confirmed according to previously published protocols (3, 4). Serum samples were tested for their capacity to neutralize MERS-CoV in tissue culture infections by using 100 50% tissue culture infective doses (TCID50) in in Huh-7 cells. Virus neutralisation titre is calculated as the reciprocal of the serum dilution that neutralizes the virus infection. Exp erim enta l a n im a Is
Camels were purchased from rural markets in the north of Saudi Arabia and maintained at a camel research farm, managed by the Saudi Ministry of Water, Environment, and Agriculture (MEWA). Animal maintenance and procedures were in accordance with King Abdullah International Medical Research Centre (KAIMRC, Riyadh, Kingdom of Saudi Arabia) Institutional Animal Care and Use Committee (IACUC) recommendations, under the regulation of MEWA. Project designation is KAIMRC research project RC16-093.
Pilot study: Nine na'ive calves, with Euroimmun ELISA ratios below the positive cut-off value of 1.1 were divided into 3 groups (n=3 per group). In the control group, two calves received phosphate buffered saline (PBS) and one calf received lxio? Infectious Unit (IU) of ChAdOxi-eGFP, intramuscularly as a vector control. The second group comprised 3 young calves of 8-10 months old that received lxio9 IU of ChAdOxi MERS vaccine via the intramuscular route. The third group comprised 3 older calves of 18-24 months old that received same vaccine regimen as the second group.
Main study: Ten na'ive calves were divided into 2 groups (n=5 per group). Group 1 na'ive (G01-N) received control injections of PBS (n=3) or lxio9 ChAdOxi-eGFP (n=2) and group 2 na'ive (G02-N) received vaccine injections of lxio9 ChAdOxi MERS (n=5). All vaccination was performed via the intramuscular route. Serum samples were collected pre-immunisation (day o) and at 7, 14, 21, and 28 days post immunisation (d.p.i.). Two calves were boosted with a second dose at 45 d.p.i. and serum samples were collected post-boost (p.b.) at 49, 56, 63, and 70 d.p.i. In addition, 10 seropositive camels were divided into 2 groups (n=5 per group); where Group 1 seropositive (G01-S) group received control injections of PBS (n=3) or lxio9 ChAdOxi-eGFP (n=2) and Group 2 seropositive (G02-S) received lxio9 ChAdOxi MERS (n=5). Serum samples were collected pre-immunisation (day o) and at 7, 14, 21, 28, and 56 d.p.i.
Below is age information for the camels in the main study. The age of na'ive camels are very young. The seropositive camels vary from 1 to 3 years. See the table below - the top set is G01, lower set G02.
Na'r've 6-12 m
Na'r've 6-12 m
Na'r've 6-12 m
Na'r've 6-12 m
Na'r've 6-12 m
Seropositive 6-12 m Seropositive i8-24m
Seropositive 8 months
Seropositive 18-241TL
Seropositive 6-12 m
Na'ive 6-12 m
Na'ive 6-12 m
Na'ive 6-12 m
Na'ive 6-12 m
Na'ive 6-12 m
Seropositive 6-12 m
Seropositive 2-5 - 3 years
Seropositive 2-5 - 3 years
Seropositive 2-5 - 3 years
Seropositive 2-5 - 3 years
Va ccinatio n
Camels received lxio^ Infectious Units (IU) of ChAdOxi-MERS or ChAdOxi-eGFP (as a vector control) per vaccination via the intramuscular route.
Sta tistica l a na ly s is
GraphPad Prism (GraphPad software) was used for statistical analysis and to plot data. Ethics statem ent
All animal procedures were performed in accordance with the terms of the UK Animals (Scientific Procedures) Act (ASPA) and were approved by the University of Oxford Animal Care and Ethical Review Committee. Animal maintenance and procedures were in accordance with King Abdullah International Medical Research Centre (KAIMRC, Riyadh, Kingdom of Saudi Arabia) Institutional Animal Care and Use Committee (IACUC) recommendations, under the regulation of the Saudi Ministry of Water, Environment, and Agriculture (MEWA).
Results
Co m m ercia l ELISA res ults - Pilot study
Antibody responses following vaccination with ChAdOxi-MERS were evaluated using the commercial ELISA kit from Euroimmun. Data are displayed in figure 12: Groups of calves (n=3) in vaccinated groups received lxio9 IU of ChAdOxi-MERS. The control group received either phosphate buffered saline (PBS, n=2) and or lxio? Infectious Unit (IU) of ChAdOxi— eGFP (vector control, n=i). Young calves were 8-10 months old. Older calves were 18-24 months old. Data points show the ratio of the optical density (OD) of test samples over the
manufacturer's positive calibrator OD. All vaccination was performed via the intramuscular route. Data points represent individual camels with the mean shown as a horizontal line.
Antibody responses indicated by ELISA ratios greater than 1.1 were seen from day 7 in the older calves (18-24 months) following a single dose of ChAdOxi-MERS. Younger calves did not respond to the single dose of vaccine to the same extent, with only one animal producing a positive result from day 56. There is a statistically significant difference between the ELISA ratios of the younger and older calves (2 way ANOVA, p
<O.OOOl).
In ho us e ELISA res ults - Pilot study
An in house ELISA was developed to allow calculation of endpoint titres.
Samples from figure 12 were analysed using the in house ELISA, this analysis is shown in figure 13: samples and groups are as figure 12, except data are not available for days 7-21 therefore are not included. ELISA plates were coated with Si recombinant protein (from ATGen Co. Ltd., CA, USA, provided as part of a collaboration with the
International Vaccine Institute of Seoul National University, Seoul, Korea), and standard endpoint ELISA protocol was followed, as previously described (1). Control animals were all negative. Data points represent individual camels with the mean shown as a horizontal line. There is a statistically significant difference between the end point titres of the younger and older calves (2 way ANOVA, p <o.05).
Virus Neutra lisatio n Assay - Pilot study
A virus neutralisation assay was used to assess the ability of vaccine induced antibodies to neutralise MERS CoV infection in tissue culture. Results are displayed in figure 14: virus neutralisation titre (VNT) was calculated for camel serum samples collected at o and 28 d.p.i. Serum samples were tested for their ability to neutralise MERS CoV infection in tissue culture by using 100 50% tissue culture infective doses (TCID50) in in Huh-7 cells. Virus neutralisation titre is calculated as the reciprocal of the serum dilution that neutralizes the virus infection. Data points represent individual camels with the mean shown as a horizontal line.
Sera from older calves neutralised MERS CoV infection at much higher dilutions (mean 1 in 655 dilution) than sera from younger calves. Only serum from one of the younger calves was able to neutralise MERS CoV infection at a dilution of 1 in 16. This indicates the presence of effective neutralising antibodies in the older calves compared to the younger cohort. There is a statistically significant difference between the neutralisation titres of the younger and older calves (2 way ANOVA, p <o.05).
Co m m ercia l ELISA res u lts - Ma in study
Antibody responses following vaccination with ChAdOxi-MERS were evaluated using the commercial ELISA kit from Euroimmun. Data are displayed in figure 15 and figure 16.
Referring to Figure 15, 10 na'ive (negative for MERS antibodies) calves were divided into 2 groups (n=5 per group); where G01-N group received control injections of PBS (n=3) or lxio9 ChAdOxi-eGFP as a vector control (n=2) and G02-N received vaccine injections of lxio9 ChAdOxi MERS (n=5) intramuscularly. Serum samples were collected pre-immunisation (o d.p.i.) and at 7, 14, 21, and 28 days post immunisation (d.p.i.). Two calves were boosted with a second dose at 45 d.p.i. and serum samples were collected weekly post-boost (pb) at 40, 56, 63, and 70 d.p.i. Samples were analysed using the commercial kit from Euroimmun. Data points represent individual camels with the mean shown as a horizontal line.
Responses above the manufacturers recommended cut-off were measurable only in animals that received the boost vaccination at day 45. There is a statistically significant difference between the ELISA ratios of the younger and older calves post boost (2 way ANOVA, p <o.oooi).
Referring to figure 16, 10 camels that tested positive for the presence of anti MERS antibodies (seropositive) were divided into 2 groups (n=5); where G01-S group received control injections of PBS (n=3) or lxio? ChAdOxi-eGFP (n=2) and G02-S received vaccine injections of lxio9 ChAdOxi MERS (n=5) intramuscularly. Serum samples were collected pre-immunisation (od.p.i.) and post-immunisation at 7, 14, 21, 28, and 56 d.p.i. Samples were analysed using the commercial kit from Euroimmun that measures responses against the Si protein. Data points represent individual camels with the mean shown as a horizontal line.
Responses above the manufacturers recommended cut-off were measurable in most of the control animals and in all of the vaccinated camels with the exception of one animal at day 7. Responses in the vaccinated animals were generally higher than in the control animals, indicating that vaccination with ChAdOxi MERS can boost responses to the Si protein in animals previously exposed to MERS CoV. There is a statistically significant difference between the ELISA ratios of the control and vaccinated calves (2 way ANOVA, p <o.oooi). In ho use ELISA res ults - Main study
Antibody responses following vaccination with ChAdOxi-MERS were evaluated using an in house ELISA. Data are displayed in figure 17 and figure 18.
Referring to figure 17, 10 na'ive (negative for MERS antibodies) calves were divided into 2 groups (n=5 per group); where G01-N group received control injections of PBS (n=3) or lxio9 ChAdOxi-eGFP as a vector control (n=2) and G02-N received vaccine injections of lxio9 ChAdOxi MERS (n=5) intramuscularly. Serum samples were collected pre-immunisation (o d.p.i.) and at 7, 14, 21, and 28 days post immunisation (d.p.i.). Two calves were boosted with a second dose at 45 d.p.i. and serum samples were collected post-boost (pb) at 49, 56, 63, and 70 d.p.i. Samples were analysed using an in house end point titre ELISA that measures responses against the Si protein. These ELISA data are for the same serum samples as in figure 15. Data points represent individual camels with the mean shown as a horizontal line. Responses were measurable only in animals that received the boost vaccination at day 45·
Referring to figure 18, 10 camels that tested positive for the presence of anti MERS antibodies (seropositive) were divided into 2 groups (n=5); where G01-S group received control injections of PBS (n=3) or lxio9 ChAdOxi-eGFP (n=2) and G02-S received vaccine injections of lxio9 ChAdOxi MERS (n=5) intramuscularly. Serum samples were collected pre-immunisation (o d.p.i.) and post-immunisation at 7, 14, 21, 28, and 56 d.p.i. Samples were analysed using an in house end point titre ELISA that measures responses against the Si protein. These ELISA data are for the same serum samples as in figure 16. Data points represent individual camels with the mean shown as a horizontal line.
Responses were measurable in all of the control animals and in all of the vaccinated camels at all time-points. Responses in the vaccinated animals were generally higher than in the control animals, indicating that vaccination with ChAdOxi MERS can boost responses to the Si protein in animals previously exposed to MERS CoV. These differences were not statistically significant.
Sum m ary
The MERS001 phase I clinical trial has demonstrated safety at (5 x io? and 2.5 x 1010 vp (low and medium doses) and immunogenicity at 5 x 109 vp. Adverse events following vaccine administration have been mild or moderate in nature and self-resolving at both doses. T cell ELISPOT responses have been relatively low but significant at the low dose.
In camels, antibody responses were detectable by both commercial and in house ELISAs in older calves following a single vaccination and in younger animals that received a boost at 45 dpi. In seropositive animals ChAdOxi MERS boosted preexisting antibody responses. Antibodies generated by ChAdOxi MERS were able to neutralise MERS CoV in tissue culture.
Thus it can be appreciated that there are no adverse effects, and only very mild side effects.
References to exam ples 5 to 7
1 Muller MA, Corman VM, Jores J, Meyer B, Younan M, Liljander A, et al. MERS coronavirus neutralizing antibodies in camels, Eastern Africa, 1983- 1997. Emerg Infect Dis. 20i4;2o(i2):2093-5.
2 Draper SJ, Moore AC, Goodman AL, Long CA, Holder AA, Gilbert SC, et al.
Effective induction of high-titer antibodies by viral vector vaccines. Nature medicine. 2θθ8;ΐ4(8):8ΐ9-2ΐ.
3 Volz A, Kupke A, Song F, Jany S, Fux R, Shams-Eldin H, et al. Protective
efficacy of recombinant Modified Vaccinia virus Ankara (MVA) delivering
Middle East Respiratory Syndrome coronavirus spike glycoprotein. Journal of virology. 2015. 4 Song F, Fux R, Provacia LB, Volz A, Eickmann M, Becker S, et al. Middle East respiratory syndrome coronavirus spike protein delivered by modified vaccinia virus Ankara efficiently induces virus-neutralizing antibodies. Journal of virology. 20i3;87(2i):ii950-4.
Although illustrative embodiments of the invention have been disclosed in detail herein, with reference to the accompanying drawings, it is understood that the invention is not limited to those precise embodiments and that various changes and modifications can be effected therein by one skilled in the art without departing from the scope of the invention as defined by the appended claims and their equivalents.
Table of sequences
Figure imgf000093_0001
SEQUENCE LISTING SEQ ID NO: 2
ChAd0x2: Viral vector based on Chimpanzee adenovirus C68 ccatcttcaa taatatacct caaacttttt gtgcgcgtta atatgcaaat gaggcgtttg 60 aatttgggga ggaagggcgg tgattggtcg agggatgagc gaccgttagg ggcggggcga 120 gtgacgtttt gatgacgtgg ttgcgaggag gagccagttt gcaagttctc gtgggaaaag 180 tgacgtcaaa cgaggtgtgg tttgaacacg gaaatactca attttcccgc gctctctgac 240 aggaaatgag gtgtttctgg gcggatgcaa gtgaaaacgg gccattttcg cgcgaaaact 300 gaatgaggaa gtgaaaatct gagtaatttc gcgtttatgg cagggaggag tatttgccga 360 gggccgagta gactttgacc gattacgtgg gggtttcgat taccgtgttt ttcacctaaa 420 tttccgcgta cggtgtcaaa gtccggtgtt tttacgcgat cgctagcgac atcgatcaca 480 agtttgtaca aaaaagctga acgagaaacg taaaatgata taaatatcaa tatattaaat 540 tagattttgc ataaaaaaca gactacataa tactgtaaaa cacaacatat ccagtcacta 600 tggcggccgc cgatttattc aacaaagcca cgttgtgtct caaaatctct gatgttacat 660 tgcacaagat aaaaatatat catcatgaac aataaaactg tctgcttaca taaacagtaa 720 tacaaggggt gttatgagcc atattcaacg ggaaacgtct tgctcgaggc cgcgattaaa 780 ttccaacatg gatgctgatt tatatgggta taaatgggct cgtgataatg tcgggcaatc 840 aggtgcgaca atctatcgat tgtatgggaa gcccgatgcg ccagagttgt ttctgaaaca 900 tggcaaaggt agcgttgcca atgatgttac agatgagatg gtcagactaa actggctgac 960 ggaatttatg cctcttccga ccatcaagca ttttatccgt actcctgatg atgcatggtt 1020 actcaccact gcgatccccg ggaaaacagc attccaggta ttagaagaat atcctgattc 1080 aggtgaaaat attgttgatg cgctggcagt gttcctgcgc cggttgcatt cgattcctgt 1140 ttgtaattgt ccttttaaca gcgatcgcgt atttcgtctc gctcaggcgc aatcacgaat 1200 gaataacggt ttggttgatg cgagtgattt tgatgacgag cgtaatggct ggcctgttga 1260 acaagtctgg aaagaaatgc ataagctttt gccattctca ccggattcag tcgtcactca 1320 tggtgatttc tcacttgata accttatttt tgacgagggg aaattaatag gttgtattga 1380 tgttggacga gtcggaatcg cagaccgata ccaggatctt gccatcctat ggaactgcct 1440 cggtgagttt tctccttcat tacagaaacg gctttttcaa aaatatggta ttgataatcc 1500 tgatatgaat aaattgcagt ttcatttgat gctcgatgag tttttctaat cagaattggt 1560 taattggttg taacactggc acgcgtggat ccggcttact aaaagccaga taacagtatg 1620 cgtatttgcg cgctgatttt tgcggtataa gaatatatac tgatatgtat acccgaagta 1680 tgtcaaaaag aggtatgcta tgaagcagcg tattacagtg acagttgaca gcgacagcta 1740 tcagttgctc aaggcatata tgatgtcaat atctccggtc tggtaagcac aaccatgcag 1800 aatgaagccc gtcgtctgcg tgccgaacgc tggaaagcgg aaaatcagga agggatggct i860 gaggtcgccc ggtttattga aatgaacggc tcttttgctg acgagaacag gggctggtga 1920 aatgcagttt aaggtttaca cctataaaag agagagccgt tatcgtctgt ttgtggatgt 1980 acagagtgat attattgaca cgcccgggcg acggatggtg atccccctgg ccagtgcacg 2040 tctgctgtca gataaagtct cccgtgaact ttacccggtg gtgcatatcg gggatgaaag 2100 ctggcgcatg atgaccaccg atatggccag tgtgccggtc tccgttatcg gggaagaagt 2160 ggctgatctc agccaccgcg aaaatgacat caaaaacgcc attaacctga tgttctgggg 2220 aatataaatg tcaggctccc ttatacacag ccagtctgca ggtcgaccat agtgactgga 2280 tatgttgtgt tttacagtat tatgtagtct gttttttatg caaaatctaa tttaatatat 2340 tgatatttat atcattttac gtttctcgtt cagctttctt gtacaaagtg gtgatcgatt 2400 cgacagatcg cgatcgcaag tgagtagtgt tctggggcgg gggaggacct gcatgagggc 2460 cagaataact gaaatctgtg cttttctgtg tgttgcagca gcatgagcgg aagcggctcc 2520 tttgagggag gggtattcag cccttatctg acggggcgtc tcccctcctg ggcgggagtg 2580 cgtcagaatg tgatgggatc cacggtggac ggccggcccg tgcagcccgc gaactcttca 2640 accctgacct atgcaaccct gagctcttcg tcgttggacg cagctgccgc cgcagctgct 2700 gcatctgccg ccagcgccgt gcgcggaatg gccatgggcg ccggctacta cggcactctg 2760 gtggccaact cgagttccac caataatccc gccagcctga acgaggagaa gctgttgctg 2820 ctgatggccc agctcgaggc cttgacccag cgcctgggcg agctgaccca gcaggtggct 2880 cagctgcagg agcagacgcg ggccgcggtt gccacggtga aatccaaata aaaaatgaat 2940 caataaataa acggagacgg ttgttgattt taacacagag tctgaatctt tatttgattt 3000 ttcgcgcgcg gtaggccctg gaccaccggt ctcgatcatt gagcacccgg tggatctttt 3060 ccaggacccg gtagaggtgg gcttggatgt tgaggtacat gggcatgagc ccgtcccggg 3120 ggtggaggta gctccattgc agggcctcgt gctcgggggt ggtgttgtaa atcacccagt 3180 catagcaggg gcgcagggca tggtgttgca caatatcttt gaggaggaga ctgatggcca 3240 cgggcagccc tttggtgtag gtgtttacaa atctgttgag ctgggaggga tgcatgcggg 3300 gggagatgag gtgcatcttg gcctggatct tgagattggc gatgttaccg cccagatccc 3360 gcctggggtt catgttgtgc aggaccacca gcacggtgta tccggtgcac ttggggaatt 3420 tatcatgcaa cttggaaggg aaggcgtgaa agaatttggc gacgcctttg tgcccgccca 3480 ggttttccat gcactcatcc atgatgatgg cgatgggccc gtgggcggcg gcctgggcaa 3540 agacgtttcg ggggtcggac acatcatagt tgtggtcctg ggtgaggtca tcataggcca 3600 ttttaatgaa tttggggcgg agggtgccgg actgggggac aaaggtaccc tcgatcccgg 3660 gggcgtagtt cccctcacag atctgcatct cccaggcttt gagctcggag ggggggatca 3720 tgtccacctg cggggcgata aagaacacgg tttccggggc gggggagatg agctgggccg 3780 aaagcaagtt ccggagcagc tgggacttgc cgcagccggt ggggccgtag atgaccccga 3840 tgaccggctg caggtggtag ttgagggaga gacagctgcc gtcctcccgg aggagggggg 3900 ccacctcgtt catcatctcg cgcacgtgca tgttctcgcg caccagttcc gccaggaggc 3960 gctctccccc cagggatagg agctcctgga gcgaggcgaa gtttttcagc ggcttgagtc 4020 cgtcggccat gggcattttg gagagggttt gttgcaagag ttccaggcgg tcccagagct 4080 cggtgatgtg ctctacggca tctcgatcca gcagacctcc tcgtttcgcg ggttgggacg 4140 gctgcgggag tagggcacca gacgatgggc gtccagcgca gccagggtcc ggtccttcca 4200 gggtcgcagc gtccgcgtca gggtggtctc cgtcacggtg aaggggtgcg cgccgggctg 4260 ggcgcttgcg agggtgcgct tcaggctcat ccggctggtc gaaaaccgct cccgatcggc 4320 gccctgcgcg tcggccaggt agcaattgac catgagttcg tagttgagcg cctcggccgc 4380 gtggcctttg gcgcggagct tacctttgga agtctgcccg caggcgggac agaggaggga 4440 cttgagggcg tagagcttgg gggcgaggaa gacggactcg ggggcgtagg cgtccgcgcc 4500 gcagtgggcg cagacggtct cgcactccac gagccaggtg aggtcgggct ggtcggggtc 4560 aaaaaccagt ttcccgccgt tctttttgat gcgtttctta cctttggtct ccatgagctc 4620 gtgtccccgc tgggtgacaa agaggctgtc cgtgtccccg tagaccgact ttatgggccg 4680 gtcctcgagc ggtgtgccgc ggtcctcctc gtagaggaac cccgcccact ccgagacgaa 4740 agcccgggtc caggccagca cgaaggaggc cacgtgggac gggtagcggt cgttgtccac 4800 cagcgggtcc accttttcca gggtatgcaa acacatgtcc ccctcgtcca catccaggaa 4860 ggtgattggc ttgtaagtgt aggccacgtg accgggggtc ccggccgggg gggtataaaa 4920 gggtgcgggt ccctgctcgt cctcactgtc ttccggatcg ctgtccagga gcgccagctg 4980 ttggggtagg tattccctct cgaaggcggg catgacctcg gcactcaggt tgtcagtttc 5040 tagaaacgag gaggatttga tattgacggt gccggcggag atgcctttca agagcccctc 5100 gtccatctgg tcagaaaaga cgatcttttt gttgtcgagc ttggtggcga aggagccgta 5160 gagggcgttg gagaggagct tggcgatgga gcgcatggtc tggttttttt ccttgtcggc 5220 gcgctccttg gcggcgatgt tgagctgcac gtactcgcgc gccacgcact tccattcggg 5280 gaagacggtg gtcagctcgt cgggcacgat tctgacctgc cagccccgat tatgcagggt 5340 gatgaggtcc acactggtgg ccacctcgcc gcgcaggggc tcattagtcc agcagaggcg 5400 tccgcccttg cgcgagcaga aggggggcag ggggtccagc atgacctcgt cgggggggtc 5460 ggcatcgatg gtgaagatgc cgggcaggag gtcggggtca aagtagctga tggaagtggc 5520 cagatcgtcc agggcagctt gccattcgcg cacggccagc gcgcgctcgt agggactgag 5580 gggcgtgccc cagggcatgg gatgggtaag cgcggaggcg tacatgccgc agatgtcgta 5640 gacgtagagg ggctcctcga ggatgccgat gtaggtgggg tagcagcgcc ccccgcggat 5700 gctggcgcgc acgtagtcat acagctcgtg cgagggggcg aggagccccg ggcccaggtt 5760 ggtgcgactg ggcttttcgg cgcggtagac gatctggcgg aaaatggcat gcgagttgga 5820 ggagatggtg ggcctttgga agatgttgaa gtgggcgtgg ggcagtccga ccgagtcgcg 5880 gatgaagtgg gcgtaggagt cttgcagctt ggcgacgagc tcggcggtga ctaggacgtc 5940 cagagcgcag tagtcgaggg tctcctggat gatgtcatac ttgagctgtc ccttttgttt 6000 ccacagctcg cggttgagaa ggaactcttc gcggtccttc cagtactctt cgagggggaa 6060 cccgtcctga tctgcacggt aagagcctag catgtagaac tggttgacgg ccttgtaggc 6120 gcagcagccc ttctccacgg ggagggcgta ggcctgggcg gccttgcgca gggaggtgtg 6180 cgtgagggcg aaagtgtccc tgaccatgac cttgaggaac tggtgcttga agtcgatatc 6240 gtcgcagccc ccctgctccc agagctggaa gtccgtgcgc ttcttgtagg cggggttggg 6300 caaagcgaaa gtaacatcgt tgaagaggat cttgcccgcg cggggcataa agttgcgagt 6360 gatgcggaaa ggttggggca cctcggcccg gttgttgatg acctgggcgg cgagcacgat 6420 ctcgtcgaag ccgttgatgt tgtggcccac gatgtagagt tccacgaatc gcggacggcc 6480 cttgacgtgg ggcagtttct tgagctcctc gtaggtgagc tcgtcggggt cgctgagccc 6540 gtgctgctcg agcgcccagt cggcgagatg ggggttggcg cggaggaagg aagtccagag 6600 atccacggcc agggcggttt gcagacggtc ccggtactga cggaactgct gcccgacggc 6660 cattttttcg ggggtgacgc agtagaaggt gcgggggtcc ccgtgccagc gatcccattt 6720 gagctggagg gcgagatcga gggcgagctc gacgagccgg tcgtccccgg agagtttcat 6780 gaccagcatg aaggggacga gctgcttgcc gaaggacccc atccaggtgt aggtttccac 6840 atcgtaggtg aggaagagcc tttcggtgcg aggatgcgag ccgatgggga agaactggat 6900 ctcctgccac caattggagg aatggctgtt gatgtgatgg aagtagaaat gccgacggcg 6960 cgccgaacac tcgtgcttgt gtttatacaa gcggccacag tgctcgcaac gctgcacggg 7020 atgcacgtgc tgcacgagct gtacctgagt tcctttgacg aggaatttca gtgggaagtg 7080 gagtcgtggc gcctgcatct cgtgctgtac tacgtcgtgg tggtcggcct ggccctcttc 7140 tgcctcgatg gtggtcatgc tgacgagccc gcgcgggagg caggtccaga cctcggcgcg 7200 agcgggtcgg agagcgagga cgagggcgcg caggccggag ctgtccaggg tcctgagacg 7260 ctgcggagtc aggtcagtgg gcagcggcgg cgcgcggttg acttgcagga gtttttccag 7320 ggcgcgcggg aggtccagat ggtacttgat ctccaccgcg ccattggtgg cgacgtcgat 7380 ggcttgcagg gtcccgtgcc cctggggtgt gaccaccgtc ccccgtttct tcttgggcgg 7440 ctggggcgac gggggcggtg cctcttccat ggttagaagc ggcggcgagg acgcgcgccg 7500 ggcggcaggg gcggctcggg gcccggaggc aggggcggca ggggcacgtc ggcgccgcgc 7560 gcgggtaggt tctggtactg cgcccggaga agactggcgt gagcgacgac gcgacggttg 7620 acgtcctgga tctgacgcct ctgggtgaag gccacgggac ccgtgagttt gaacctgaaa 7680 gagagttcga cagaatcaat ctcggtatcg ttgacggcgg cctgccgcag gatctcttgc 7740 acgtcgcccg agttgtcctg gtaggcgatc tcggtcatga actgctcgat ctcctcctct 7800 tgaaggtctc cgcggccggc gcgctccacg gtggccgcga ggtcgttgga gatgcggccc 7860 atgagctgcg agaaggcgtt catgcccgcc tcgttccaga cgcggctgta gaccacgacg 7920 ccctcgggat cgccggcgcg catgaccacc tgggcgaggt tgagctccac gtggcgcgtg 7980 aagaccgcgt agttgcagag gcgctggtag aggtagttga gcgtggtggc gatgtgctcg 8040 gtgacgaaga aatacatgat ccagcggcgg agcggcatct cgctgacgtc gcccagcgcc 8100 tccaaacgtt ccatggcctc gtaaaagtcc acggcgaagt tgaaaaactg ggagttgcgc 8160 gccgagacgg tcaactcctc ctccagaaga cggatgagct cggcgatggt ggcgcgcacc 8220 tcgcgctcga aggcccccgg gagttcctcc acttcctctt cttcctcctc cactaacatc 8280 tcttctactt cctcctcagg cggcagtggt ggcgggggag ggggcctgcg tcgccggcgg 8340 cgcacgggca gacggtcgat gaagcgctcg atggtctcgc cgcgccggcg tcgcatggtc 8400 tcggtgacgg cgcgcccgtc ctcgcggggc cgcagcgtga agacgccgcc gcgcatctcc 8460 aggtggccgg gggggtcccc gttgggcagg gagagggcgc tgacgatgca tcttatcaat 8520 tgccccgtag ggactccgcg caaggacctg agcgtctcga gatccacggg atctgaaaac 8580 cgctgaacga aggcttcgag ccagtcgcag tcgcaaggta ggctgagcac ggtttcttct 8640 ggcgggtcat gttggttggg agcggggcgg gcgatgctgc tggtgatgaa gttgaaatag 8700 gcggttctga gacggcggat ggtggcgagg agcaccaggt ctttgggccc ggcttgctgg 8760 atgcgcagac ggtcggccat gccccaggcg tggtcctgac acctggccag gtccttgtag 8820 tagtcctgca tgagccgctc cacgggcacc tcctcctcgc ccgcgcggcc gtgcatgcgc 8880 gtgagcccga agccgcgctg gggctggacg agcgccaggt cggcgacgac gcgctcggcg 8940 aggatggctt gctggatctg ggtgagggtg gtctggaagt catcaaagtc gacgaagcgg 9000 tggtaggctc cggtgttgat ggtgtaggag cagttggcca tgacggacca gttgacggtc 9060 tggtggcccg gacgcacgag ctcgtggtac ttgaggcgcg agtaggcgcg cgtgtcgaag 9120 atgtagtcgt tgcaggtgcg caccaggtac tggtagccga tgaggaagtg cggcggcggc 9180 tggcggtaga gcggccatcg ctcggtggcg ggggcgccgg gcgcgaggtc ctcgagcatg 9240 gtgcggtggt agccgtagat gtacctggac atccaggtga tgccggcggc ggtggtggag 9300 gcgcgcggga actcgcggac gcggttccag atgttgcgca gcggcaggaa gtagttcatg 9360 gtgggcacgg tctggcccgt gaggcgcgcg cagtcgtgga tgctctatac gggcaaaaac 9420 gaaagcggtc agcggctcga ctccgtggcc tggaggctaa gcgaacgggt tgggctgcgc 9480 gtgtaccccg gttcgaatct cgaatcaggc tggagccgca gctaacgtgg tattggcact 9540 cccgtctcga cccaagcctg caccaaccct ccaggatacg gaggcgggtc gttttgcaac 9600 ttttttttgg aggccggatg agactagtaa gcgcggaaag cggccgaccg cgatggctcg 9660 ctgccgtagt ctggagaaga atcgccaggg ttgcgttgcg gtgtgccccg gttcgaggcc 9720 ggccggattc cgcggctaac gagggcgtgg ctgccccgtc gtttccaaga ccccatagcc 9780 agccgacttc tccagttacg gagcgagccc ctcttttgtt ttgtttgttt ttgccagatg 9840 catcccgtac tgcggcagat gcgcccccac caccctccac cgcaacaaca gccccctcca 9900 cagccggcgc ttctgccccc gccccagcag caacttccag ccacgaccgc cgcggccgcc 9960 gtgagcgggg ctggacagag ttatgatcac cagctggcct tggaagaggg cgaggggctg 10020 gcgcgcctgg gggcgtcgtc gccggagcgg cacccgcgcg tgcagatgaa aagggacgct 10080 cgcgaggcct acgtgcccaa gcagaacctg ttcagagaca ggagcggcga ggagcccgag 10140 gagatgcgcg cggcccggtt ccacgcgggg cgggagctgc ggcgcggcct ggaccgaaag 10200 agggtgctga gggacgagga tttcgaggcg gacgagctga cggggatcag ccccgcgcgc 10260 gcgcacgtgg ccgcggccaa cctggtcacg gcgtacgagc agaccgtgaa ggaggagagc 10320 aacttccaaa aatccttcaa caaccacgtg cgcaccctga tcgcgcgcga ggaggtgacc 10380 ctgggcctga tgcacctgtg ggacctgctg gaggccatcg tgcagaaccc caccagcaag 10440 ccgctgacgg cgcagctgtt cctggtggtg cagcatagtc gggacaacga agcgttcagg 10500 gaggcgctgc tgaatatcac cgagcccgag ggccgctggc tcctggacct ggtgaacatt 10560 ctgcagagca tcgtggtgca ggagcgcggg ctgccgctgt ccgagaagct ggcggccatc 10620 aacttctcgg tgctgagttt gggcaagtac tacgctagga agatctacaa gaccccgtac 10680 gtgcccatag acaaggaggt gaagatcgac gggttttaca tgcgcatgac cctgaaagtg 10740 ctgaccctga gcgacgatct gggggtgtac cgcaacgaca ggatgcaccg tgcggtgagc 10800 gccagcaggc ggcgcgagct gagcgaccag gagctgatgc atagtctgca gcgggccctg 10860 accggggccg ggaccgaggg ggagagctac tttgacatgg gcgcggacct gcactggcag 10920 cccagccgcc gggccttgga ggcggcggca ggaccctacg tagaagaggt ggacgatgag 10980 gtggacgagg agggcgagta cctggaagac tgatggcgcg accgtatttt tgctagatgc 11040 aacaacaaca gccacctcct gatcccgcga tgcgggcggc gctgcagagc cagccgtccg 11100 gcattaactc ctcggacgat tggacccagg ccatgcaacg catcatggcg ctgacgaccc 11160 gcaaccccga agcctttaga cagcagcccc aggccaaccg gctctcggcc atcctggagg 11220 ccgtggtgcc ctcgcgctcc aaccccacgc acgagaaggt cctggccatc gtgaacgcgc 11280 tggtggagaa caaggccatc cgcggcgacg aggccggcct ggtgtacaac gcgctgctgg 11340 agcgcgtggc ccgctacaac agcaccaacg tgcagaccaa cctggaccgc atggtgaccg 11400 acgtgcgcga ggccgtggcc cagcgcgagc ggttccaccg cgagtccaac ctgggatcca 11460 tggtggcgct gaacgccttc ctcagcaccc agcccgccaa cgtgccccgg ggccaggagg 11520 actacaccaa cttcatcagc gccctgcgcc tgatggtgac cgaggtgccc cagagcgagg 11580 tgtaccagtc cgggccggac tacttcttcc agaccagtcg ccagggcttg cagaccgtga 11640 acctgagcca ggctttcaag aacttgcagg gcctgtgggg cgtgcaggcc ccggtcgggg 11700 accgcgcgac ggtgtcgagc ctgctgacgc cgaactcgcg cctgctgctg ctgctggtgg 11760 cccccttcac ggacagcggc agcatcaacc gcaactcgta cctgggctac ctgattaacc 11820 tgtaccgcga ggccatcggc caggcgcacg tggacgagca gacctaccag gagatcaccc 11880 acgtgagccg cgccctgggc caggacgacc cgggcaacct ggaagccacc ctgaactttt 11940 tgctgaccaa ccggtcgcag aagatcccgc cccagtacgc gctcagcacc gaggaggagc 12000 gcatcctgcg ttacgtgcag cagagcgtgg gcctgttcct gatgcaggag ggggccaccc 12060 ccagcgccgc gctcgacatg accgcgcgca acatggagcc cagcatgtac gccagcaacc 12120 gcccgttcat caataaactg atggactact tgcatcgggc ggccgccatg aactctgact 12180 atttcaccaa cgccatcctg aatccccact ggctcccgcc gccggggttc tacacgggcg 12240 agtacgacat gcccgacccc aatgacgggt tcctgtggga cgatgtggac agcagcgtgt 12300 tctccccccg accgggtgct aacgagcgcc ccttgtggaa gaaggaaggc agcgaccgac 12360 gcccgtcctc ggcgctgtcc ggccgcgagg gtgctgccgc ggcggtgccc gaggccgcca 12420 gtcctttccc gagcttgccc ttctcgctga acagtatccg cagcagcgag ctgggcagga 12480 tcacgcgccc gcgcttgctg ggcgaagagg agtacttgaa tgactcgctg ttgagacccg 12540 agcgggagaa gaacttcccc aataacggga tagaaagcct ggtggacaag atgagccgct 12600 ggaagacgta tgcgcaggag cacagggacg atccccgggc gtcgcagggg gccacgagcc 12660 ggggcagcgc cgcccgtaaa cgccggtggc acgacaggca gcggggacag atgtgggacg 12720 atgaggactc cgccgacgac agcagcgtgt tggacttggg tgggagtggt aacccgttcg 12780 ctcacctgcg cccccgtatc gggcgcatga tgtaagagaa accgaaaata aatgatactc 12840 accaaggcca tggcgaccag cgtgcgttcg tttcttctct gttgttgttg tatctagtat 12900 gatgaggcgt gcgtacccgg agggtcctcc tccctcgtac gagagcgtga tgcagcaggc 12960 gatggcggcg gcggcgatgc agcccccgct ggaggctcct tacgtgcccc cgcggtacct 13020 ggcgcctacg gaggggcgga acagcattcg ttactcggag ctggcaccct tgtacgatac 13080 cacccggttg tacctggtgg acaacaagtc ggcggacatc gcctcgctga actaccagaa 13140 cgaccacagc aacttcctga ccaccgtggt gcagaacaat gacttcaccc ccacggaggc 13200 cagcacccag accatcaact ttgacgagcg ctcgcggtgg ggcggccagc tgaaaaccat 13260 catgcacacc aacatgccca acgtgaacga gttcatgtac agcaacaagt tcaaggcgcg 13320 ggtgatggtc tcccgcaaga cccccaatgg ggtgacagtg acagaggatt atgatggtag 13380 tcaggatgag ctgaagtatg aatgggtgga atttgagctg cccgaaggca acttctcggt 13440 gaccatgacc atcgacctga tgaacaacgc catcatcgac aattacttgg cggtggggcg 13500 gcagaacggg gtgctggaga gcgacatcgg cgtgaagttc gacactagga acttcaggct 13560 gggctgggac cccgtgaccg agctggtcat gcccggggtg tacaccaacg aggctttcca 13620 tcccgatatt gtcttgctgc ccggctgcgg ggtggacttc accgagagcc gcctcagcaa 13680 cctgctgggc attcgcaaga ggcagccctt ccaggaaggc ttccagatca tgtacgagga 13740 tctggagggg ggcaacatcc ccgcgctcct ggatgtcgac gcctatgaga aaagcaagga 13800 ggatgcagca gctgaagcaa ctgcagccgt agctaccgcc tctaccgagg tcaggggcga 13860 taattttgca agcgccgcag cagtggcagc ggccgaggcg gctgaaaccg aaagtaagat 13920 agtcattcag ccggtggaga aggatagcaa gaacaggagc tacaacgtac taccggacaa 13980 gataaacacc gcctaccgca gctggtacct agcctacaac tatggcgacc ccgagaaggg 14040 cgtgcgctcc tggacgctgc tcaccacctc ggacgtcacc tgcggcgtgg agcaagtcta 14100 ctggtcgctg cccgacatga tgcaagaccc ggtcaccttc cgctccacgc gtcaagttag 14160 caactacccg gtggtgggcg ccgagctcct gcccgtctac tccaagagct tcttcaacga 14220 gcaggccgtc tactcgcagc agctgcgcgc cttcacctcg cttacgcacg tcttcaaccg 14280 cttccccgag aaccagatcc tcgtccgccc gcccgcgccc accattacca ccgtcagtga 14340 aaacgttcct gctctcacag atcacgggac cctgccgctg cgcagcagta tccggggagt 14400 ccagcgcgtg accgttactg acgccagacg ccgcacctgc ccctacgtct acaaggccct 14460 gggcatagtc gcgccgcgcg tcctctcgag ccgcaccttc taaatgtcca ttctcatctc 14520 gcccagtaat aacaccggtt ggggcctgcg cgcgcccagc aagatgtacg gaggcgctcg 14580 ccaacgctcc acgcaacacc ccgtgcgcgt gcgcgggcac ttccgcgctc cctggggcgc 14640 cctcaagggc cgcgtgcggt cgcgcaccac cgtcgacgac gtgatcgacc aggtggtggc 14700 cgacgcgcgc aactacaccc ccgccgccgc gcccgtctcc accgtggacg ccgtcatcga 14760 cagcgtggtg gcggacgcgc gccggtacgc ccgcgccaag agccggcggc ggcgcatcgc 14820 ccggcggcac cggagcaccc ccgccatgcg cgcggcgcga gccttgctgc gcagggccag 14880 gcgcacggga cgcagggcca tgctcagggc ggccagacgc gcggcttcag gcgccagcgc 14940 cggcaggacc cggagacgcg cggccacggc ggcggcagcg gccatcgcca gcatgtcccg 15000 cccgcggcga gggaacgtgt actgggtgcg cgacgccgcc accggtgtgc gcgtgcccgt 15060 gcgcacccgc ccccctcgca cttgaagatg ttcacttcgc gatgttgatg tgtcccagcg 15120 gcgaggagga tgtccaagcg caaattcaag gaagagatgc tccaggtcat cgcgcctgag 15180 atctacggcc ctgcggtggt gaaggaggaa agaaagcccc gcaaaatcaa gcgggtcaaa 15240 aaggacaaaa aggaagaaga aagtgatgtg gacggattgg tggagtttgt gcgcgagttc 15300 gccccccggc ggcgcgtgca gtggcgcggg cggaaggtgc aaccggtgct gagacccggc 15360 accaccgtgg tcttcacgcc cggcgagcgc tccggcaccg cttccaagcg ctcctacgac 15420 gaggtgtacg gggatgatga tattctggag caggcggccg agcgcctggg cgagtttgct 15480 tacggcaagc gcagccgttc cgcaccgaag gaagaggcgg tgtccatccc gctggaccac 15540 ggcaacccca cgccgagcct caagcccgtg accttgcagc aggtgctgcc gaccgcggcg 15600 ccgcgccggg ggttcaagcg cgagggcgag gatctgtacc ccaccatgca gctgatggtg 15660 cccaagcgcc agaagctgga agacgtgctg gagaccatga aggtggaccc ggacgtgcag 15720 cccgaggtca aggtgcggcc catcaagcag gtggccccgg gcctgggcgt gcagaccgtg 15780 gacatcaaga ttcccacgga gcccatggaa acgcagaccg agcccatgat caagcccagc 15840 accagcacca tggaggtgca gacggatccc tggatgccat cggctcctag tcgaagaccc 15900 cggcgcaagt acggcgcggc cagcctgctg atgcccaact acgcgctgca tccttccatc 15960 atccccacgc cgggctaccg cggcacgcgc ttctaccgcg gtcataccag cagccgccgc 16020 cgcaagacca ccactcgccg ccgccgtcgc cgcaccgccg ctgcaaccac ccctgccgcc 16080 ctggtgcgga gagtgtaccg ccgcggccgc gcacctctga ccctgccgcg cgcgcgctac 16140 cacccgagca tcgccattta aactttcgcc agctttgcag atcaatggcc ctcacatgcc 16200 gccttcgcgt tcccattacg ggctaccgag gaagaaaacc gcgccgtaga aggctggcgg 16260 ggaacgggat gcgtcgccac caccaccggc ggcggcgcgc catcagcaag cggttggggg 16320 gaggcttcct gcccgcgctg atccccatca tcgccgcggc gatcggggcg atccccggca 16380 ttgcttccgt ggcggtgcag gcctctcagc gccactgaga cacacttgga aacatcttgt 16440 aataaaccca tggactctga cgctcctggt cctgtgatgt gttttcgtag acagatggaa 16500 gacatcaatt tttcgtccct ggctccgcga cacggcacgc ggccgttcat gggcacctgg 16560 agcgacatcg gcaccagcca actgaacggg ggcgccttca attggagcag tctctggagc 16620 gggcttaaga atttcgggtc cacgcttaaa acctatggca gcaaggcgtg gaacagcacc 16680 acagggcagg cgctgaggga taagctgaaa gagcagaact tccagcagaa ggtggtcgat 16740 gggctcgcct cgggcatcaa cggggtggtg gacctggcca accaggccgt gcagcggcag 16800 atcaacagcc gcctggaccc ggtgccgccc gccggctccg tggagatgcc gcaggtggag 16860 gaggagctgc ctcccctgga caagcggggc gagaagcgac cccgccccga tgcggaggag 16920 acgctgctga cgcacacgga cgagccgccc ccgtacgagg aggcggtgaa actgggtctg 16980 cccaccacgc ggcccatcgc gcccctggcc accggggtgc tgaaacccga aaagcccgcg 17040 accctggact tgcctcctcc ccagccttcc cgcccctcta cagtggctaa gcccctgccg 17100 ccggtggccg tggcccgcgc gcgacccggg ggcaccgccc gccctcatgc gaactggcag 17160 agcactctga acagcatcgt gggtctggga gtgcagagtg tgaagcgccg ccgctgctat 17220 taaacctacc gtagcgctta acttgcttgt ctgtgtgtgt atgtattatg tcgccgccgc 17280 cgctgtccac cagaaggagg agtgaagagg cgcgtcgccg agttgcaaga tggccacccc 17340 atcgatgctg ccccagtggg cgtacatgca catcgccgga caggacgctt cggagtacct 17400 gagtccgggt ctggtgcagt ttgcccgcgc cacagacacc tacttcagtc tggggaacaa 17460 gtttaggaac cccacggtgg cgcccacgca cgatgtgacc accgaccgca gccagcggct 17520 gacgctgcgc ttcgtgcccg tggaccgcga ggacaacacc tactcgtaca aagtgcgcta 17580 cacgctggcc gtgggcgaca accgcgtgct ggacatggcc agcacctact ttgacatccg 17640 cggcgtgctg gatcggggcc ctagcttcaa accctactcc ggcaccgcct acaacagtct 17700 ggcccccaag ggagcaccca acacttgtca gtggacatat aaagccgatg gtgaaactgc 17760 cacagaaaaa acctatacat atggaaatgc acccgtgcag ggcattaaca tcacaaaaga 17820 tggtattcaa cttggaactg acaccgatga tcagccaatc tacgcagata aaacctatca 17880 gcctgaacct caagtgggtg atgctgaatg gcatgacatc actggtactg atgaaaagta 17940 tggaggcaga gctcttaagc ctgataccaa aatgaagcct tgttatggtt cttttgccaa 18000 gcctactaat aaagaaggag gtcaggcaaa tgtgaaaaca ggaacaggca ctactaaaga 18060 atatgacata gacatggctt tctttgacaa cagaagtgcg gctgctgctg gcctagctcc 18120 agaaattgtt ttgtatactg aaaatgtgga tttggaaact ccagataccc atattgtata 18180 caaagcaggc acagatgaca gcagctcttc tattaatttg ggtcagcaag ccatgcccaa 18240 cagacctaac tacattggtt tcagagacaa ctttatcggg ctcatgtact acaacagcac 18300 tggcaatatg ggggtgctgg ccggtcaggc ttctcagctg aatgctgtgg ttgacttgca 18360 agacagaaac accgagctgt cctaccagct cttgcttgac tctctgggtg acagaacccg 18420 gtatttcagt atgtggaatc aggcggtgga cagctatgat cctgatgtgc gcattattga 18480 aaatcatggt gtggaggatg aacttcccaa ctattgtttc cctctggatg ctgttggcag 18540 aacagatact tatcagggaa ttaaggctaa tggaactgat caaaccacat ggaccaaaga 18600 tgacagtgtc aatgatgcta atgagatagg caagggtaat ccattcgcca tggaaatcaa 18660 catccaagcc aacctgtgga ggaacttcct ctacgccaac gtggccctgt acctgcccga 18720 ctcttacaag tacacgccgg ccaatgttac cctgcccacc aacaccaaca cctacgatta 18780 catgaacggc cgggtggtgg cgccctcgct ggtggactcc tacatcaaca tcggggcgcg 18840 ctggtcgctg gatcccatgg acaacgtgaa ccccttcaac caccaccgca atgcggggct 18900 gcgctaccgc tccatgctcc tgggcaacgg gcgctacgtg cccttccaca tccaggtgcc 18960 ccagaaattt ttcgccatca agagcctcct gctcctgccc gggtcctaca cctacgagtg 19020 gaacttccgc aaggacgtca acatgatcct gcagagctcc ctcggcaacg acctgcgcac 19080 ggacggggcc tccatctcct tcaccagcat caacctctac gccaccttct tccccatggc 19140 gcacaacacg gcctccacgc tcgaggccat gctgcgcaac gacaccaacg accagtcctt 19200 caacgactac ctctcggcgg ccaacatgct ctaccccatc ccggccaacg ccaccaacgt 19260 gcccatctcc atcccctcgc gcaactgggc cgccttccgc ggctggtcct tcacgcgtct 19320 caagaccaag gagacgccct cgctgggctc cgggttcgac ccctacttcg tctactcggg 19380 ctccatcccc tacctcgacg gcaccttcta cctcaaccac accttcaaga aggtctccat 19440 caccttcgac tcctccgtca gctggcccgg caacgaccgg ctcctgacgc ccaacgagtt 19500 cgaaatcaag cgcaccgtcg acggcgaggg ctacaacgtg gcccagtgca acatgaccaa 19560 ggactggttc ctggtccaga tgctggccca ctacaacatc ggctaccagg gcttctacgt 19620 gcccgagggc tacaaggacc gcatgtactc cttcttccgc aacttccagc ccatgagccg 19680 ccaggtggtg gacgaggtca actacaagga ctaccaggcc gtcaccctgg cctaccagca 19740 caacaactcg ggcttcgtcg gctacctcgc gcccaccatg cgccagggcc agccctaccc 19800 cgccaactac ccctacccgc tcatcggcaa gagcgccgtc accagcgtca cccagaaaaa 19860 gttcctctgc gacagggtca tgtggcgcat ccccttctcc agcaacttca tgtccatggg 19920 cgcgctcacc gacctcggcc agaacatgct ctatgccaac tccgcccacg cgctagacat 19980 gaatttcgaa gtcgacccca tggatgagtc cacccttctc tatgttgtct tcgaagtctt 20040 cgacgtcgtc cgagtgcacc agccccaccg cggcgtcatc gaggccgtct acctgcgcac 20100 ccccttctcg gccggtaacg ccaccaccta agctcttgct tcttgcaagc catggccgcg 20160 ggctccggcg agcaggagct cagggccatc atccgcgacc tgggctgcgg gccctacttc 20220 ctgggcacct tcgataagcg cttcccggga ttcatggccc cgcacaagct ggcctgcgcc 20280 atcgtcaaca cggccggccg cgagaccggg ggcgagcact ggctggcctt cgcctggaac 20340 ccgcgctcga acacctgcta cctcttcgac cccttcgggt tctcggacga gcgcctcaag 20400 cagatctacc agttcgagta cgagggcctg ctgcgccgca gcgccctggc caccgaggac 20460 cgctgcgtca ccctggaaaa gtccacccag accgtgcagg gtccgcgctc ggccgcctgc 20520 gggctcttct gctgcatgtt cctgcacgcc ttcgtgcact ggcccgaccg ccccatggac 20580 aagaacccca ccatgaactt gctgacgggg gtgcccaacg gcatgctcca gtcgccccag 20640 gtggaaccca ccctgcgccg caaccaggag gcgctctacc gcttcctcaa ctcccactcc 20700 gcctactttc gctcccaccg cgcgcgcatc gagaaggcca ccgccttcga ccgcatgaat 20760 caagacatgt aaaccgtgtg tgtatgttaa atgtctttaa taaacagcac tttcatgtta 20820 cacatgcatc tgagatgatt tatttagaaa tcgaaagggt tctgccgggt ctcggcatgg 20880 cccgcgggca gggacacgtt gcggaactgg tacttggcca gccacttgaa ctcggggatc 20940 agcagtttgg gcagcggggt gtcggggaag gagtcggtcc acagcttccg cgtcagttgc 21000 agggcgccca gcaggtcggg cgcggagatc ttgaaatcgc agttgggacc cgcgttctgc 21060 gcgcgggagt tgcggtacac ggggttgcag cactggaaca ccatcagggc cgggtgcttc 21120 acgctcgcca gcaccgtcgc gtcggtgatg ctctccacgt cgaggtcctc ggcgttggcc 21180 atcccgaagg gggtcatctt gcaggtctgc cttcccatgg tgggcacgca cccgggcttg 21240 tggttgcaat cgcagtgcag ggggatcagc atcatctggg cctggtcggc gttcatcccc 21300 gggtacatgg ccttcatgaa agcctccaat tgcctgaacg cctgctgggc cttggctccc 21360 tcggtgaaga agaccccgca ggacttgcta gagaactggt tggtggcgca cccggcgtcg 21420 tgcacgcagc agcgcgcgtc gttgttggcc agctgcacca cgctgcgccc ccagcggttc 21480 tgggtgatct tggcccggtc ggggttctcc ttcagcgcgc gctgcccgtt ctcgctcgcc 21540 acatccatct cgatcatgtg ctccttctgg atcatggtgg tcccgtgcag gcaccgcagc 21600 ttgccctcgg cctcggtgca cccgtgcagc cacagcgcgc acccggtgca ctcccagttc 21660 ttgtgggcga tctgggaatg cgcgtgcacg aagccctgca ggaagcggcc catcatggtg 21720 gtcagggtct tgttgctagt gaaggtcagc ggaatgccgc ggtgctcctc gttgatgtac 21780 aggtggcaga tgcggcggta cacctcgccc tgctcgggca tcagctggaa gttggctttc 21840 aggtcggtct ccacgcggta gcggtccatc agcatagtca tgatttccat acccttctcc 21900 caggccgaga cgatgggcag gctcataggg ttcttcacca tcatcttagc gctagcagcc 21960 gcggccaggg ggtcgctctc gtccagggtc tcaaagctcc gcttgccgtc cttctcggtg 22020 atccgcaccg gggggtagct gaagcccacg gccgccagct cctcctcggc ctgtctttcg 22080 tcctcgctgt cctggctgac gtcctgcagg accacatgct tggtcttgcg gggtttcttc 22140 ttgggcggca gcggcggcgg agatgttgga gatggcgagg gggagcgcga gttctcgctc 22200 accactacta tctcttcctc ttcttggtcc gaggccacgc ggcggtaggt atgtctcttc 22260 gggggcagag gcggaggcga cgggctctcg ccgccgcgac ttggcggatg gctggcagag 22320 ccccttccgc gttcgggggt gcgctcccgg cggcgctctg actgacttcc tccgcggccg 22380 gccattgtgt tctcctaggg aggaacaaca agcatggaga ctcagccatc gccaacctcg 22440 ccatctgccc ccaccgccga cgagaagcag cagcagcaga atgaaagctt aaccgccccg 22500 ccgcccagcc ccgccacctc cgacgcggcc gtcccagaca tgcaagagat ggaggaatcc 22560 atcgagattg acctgggcta tgtgacgccc gcggagcacg aggaggagct ggcagtgcgc 22620 ttttcacaag aagagataca ccaagaacag ccagagcagg aagcagagaa tgagcagagt 22680 caggctgggc tcgagcatga cggcgactac ctccacctga gcggggggga ggacgcgctc 22740 atcaagcatc tggcccggca ggccaccatc gtcaaggatg cgctgctcga ccgcaccgag 22800 gtgcccctca gcgtggagga gctcagccgc gcctacgagt tgaacctctt ctcgccgcgc 22860 gtgcccccca agcgccagcc caatggcacc tgcgagccca acccgcgcct caacttctac 22920 ccggtcttcg cggtgcccga ggccctggcc acctaccaca tctttttcaa gaaccaaaag 22980 atccccgtct cctgccgcgc caaccgcacc cgcgccgacg cccttttcaa cctgggtccc 23040 ggcgcccgcc tacctgatat cgcctccttg gaagaggttc ccaagatctt cgagggtctg 23100 ggcagcgacg agactcgggc cgcgaacgct ctgcaaggag aaggaggaga gcatgagcac 23160 cacagcgccc tggtcgagtt ggaaggcgac aacgcgcggc tggcggtgct caaacgcacg 23220 gtcgagctga cccatttcgc ctacccggct ctgaacctgc cccccaaagt catgagcgcg 23280 gtcatggacc aggtgctcat caagcgcgcg tcgcccatct ccgaggacga gggcatgcaa 23340 gactccgagg agggcaagcc cgtggtcagc gacgagcagc tggcccggtg gctgggtcct 23400 aatgctagtc cccagagttt ggaagagcgg cgcaaactca tgatggccgt ggtcctggtg 23460 accgtggagc tggagtgcct gcgccgcttc ttcgccgacg cggagaccct gcgcaaggtc 23520 gaggagaacc tgcactacct cttcaggcac gggttcgtgc gccaggcctg caagatctcc 23580 aacgtggagc tgaccaacct ggtctcctac atgggcatct tgcacgagaa ccgcctgggg 23640 cagaacgtgc tgcacaccac cctgcgcggg gaggcccggc gcgactacat ccgcgactgc 23700 gtctacctct acctctgcca cacctggcag acgggcatgg gcgtgtggca gcagtgtctg 23760 gaggagcaga acctgaaaga gctctgcaag ctcctgcaga agaacctcaa gggtctgtgg 23820 accgggttcg acgagcgcac caccgcctcg gacctggccg acctcatttt ccccgagcgc 23880 ctcaggctga cgctgcgcaa cggcctgccc gactttatga gccaaagcat gttgcaaaac 23940 tttcgctctt tcatcctcga acgctccgga atcctgcccg ccacctgctc cgcgctgccc 24000 tcggacttcg tgccgctgac cttccgcgag tgccccccgc cgctgtggag ccactgctac 24060 ctgctgcgcc tggccaacta cctggcctac cactcggacg tgatcgagga cgtcagcggc 24120 gagggcctgc tcgagtgcca ctgccgctgc aacctctgca cgccgcaccg ctccctggcc 24180 tgcaaccccc agctgctgag cgagacccag atcatcggca ccttcgagtt gcaagggccc 24240 agcgaaggcg agggttcagc cgccaagggg ggtctgaaac tcaccccggg gctgtggacc 24300 tcggcctact tgcgcaagtt cgtgcccgag gactaccatc ccttcgagat caggttctac 24360 gaggaccaat cccatccgcc caaggccgag ctgtcggcct gcgtcatcac ccagggggcg 24420 atcctggccc aattgcaagc catccagaaa tcccgccaag aattcttgct gaaaaagggc 24480 cgcggggtct acctcgaccc ccagaccggt gaggagctca accccggctt cccccaggat 24540 gccccgagga aacaagaagc tgaaagtgga gctgccgccc gtggaggatt tggaggaaga 24600 ctgggagaac agcagtcagg cagaggagga ggagatggag gaagactggg acagcactca 24660 ggcagaggag gacagcctgc aagacagtct ggaggaagac gaggaggagg cagaggagga 24720 ggtggaagaa gcagccgccg ccagaccgtc gtcctcggcg ggggagaaag caagcagcac 24780 ggataccatc tccgctccgg gtcggggtcc cgctcgacca cacagtagat gggacgagac 24840 cggacgattc ccgaacccca ccacccagac cggtaagaag gagcggcagg gatacaagtc 24900 ctggcggggg cacaaaaacg ccatcgtctc ctgcttgcag gcctgcgggg gcaacatctc 24960 cttcacccgg cgctacctgc tcttccaccg cggggtgaac tttccccgca acatcttgca 25020 ttactaccgt cacctccaca gcccctacta cttccaagaa gaggcagcag cagcagaaaa 25080 agaccagcag aaaaccagca gctagaaaat ccacagcggc ggcagcaggt ggactgagga 25140 tcgcggcgaa cgagccggcg caaacccggg agctgaggaa ccggatcttt cccaccctct 25200 atgccatctt ccagcagagt cgggggcagg agcaggaact gaaagtcaag aaccgttctc 25260 tgcgctcgct cacccgcagt tgtctgtatc acaagagcga agaccaactt cagcgcactc 25320 tcgaggacgc cgaggctctc ttcaacaagt actgcgcgct cactcttaaa gagtagcccg 25380 cgcccgccca gtcgcagaaa aaggcgggaa ttacgtcacc tgtgcccttc gccctagccg 25440 cctccaccca tcatcatgag caaagagatt cccacgcctt acatgtggag ctaccagccc 25500 cagatgggcc tggccgccgg tgccgcccag gactactcca cccgcatgaa ttggctcagc 25560 gccgggcccg cgatgatctc acgggtgaat gacatccgcg cccaccgaaa ccagatactc 25620 ctagaacagt cagcgctcac cgccacgccc cgcaatcacc tcaatccgcg taattggccc 25680 gccgccctgg tgtaccagga aattccccag cccacgaccg tactacttcc gcgagacgcc 25740 caggccgaag tccagctgac taactcaggt gtccagctgg cgggcggcgc caccctgtgt 25800 cgtcaccgcc ccgctcaggg tataaagcgg ctggtgatcc ggggcagagg cacacagctc 25860 aacgacgagg tggtgagctc ttcgctgggt ctgcgacctg acggagtctt ccaactcgcc 25920 ggatcgggga gatcttcctt cacgcctcgt caggccgtcc tgactttgga gagttcgtcc 25980 tcgcagcccc gctcgggtgg catcggcact ctccagttcg tggaggagtt cactccctcg 26040 gtctacttca accccttctc cggctccccc ggccactacc cggacgagtt catcccgaac 26100 ttcgacgcca tcagcgagtc ggtggacggc tacgattgag tttaaactca cccccttatc 26160 cagtgaaata aagatcatat tgatgatgat tttacagaaa taaaaaataa tcatttgatt 26220 tgaaataaag atacaatcat attgatgatt tgagtttaac aaaaaaataa agaatcactt 26280 acttgaaatc tgataccagg tctctgtcca tgttttctgc caacaccact tcactcccct 26340 cttcccagct ctggtactgc aggccccggc gggctgcaaa cttcctccac acgctgaagg 26400 ggatgtcaaa ttcctcctgt ccctcaatct tcattttatc ttctatcaga tgtccaaaaa 26460 gcgcgtccgg gtggatgatg acttcgaccc cgtctacccc tacgatgcag acaacgcacc 26520 gaccgtgccc ttcatcaacc cccccttcgt ctcttcagat ggattccaag agaagcccct 26580 gggggtgttg tccctgcgac tggccgaccc cgtcaccacc aagaacgggg aaatcaccct 26640 caagctggga gagggggtgg acctcgattc ctcgggaaaa ctcatctcca acacggccac 26700 caaggccgcc gcccctctca gtttttccaa caacaccatt tcccttaaca tggatcaccc 26760 cttttacact aaagatggaa aattatcctt acaagtttct ccaccattaa atatactgag 26820 aacaagcatt ctaaacacac tagctttagg ttttggatca ggtttaggac tccgtggctc 26880 tgccttggca gtacagttag tctctccact tacatttgat actgatggaa acataaagct 26940 taccttagac agaggtttgc atgttacaac aggagatgca attgaaagca acataagctg 27000 ggctaaaggt ttaaaatttg aagatggagc catagcaacc aacattggaa atgggttaga 27060 gtttggaagc agtagtacag aaacaggtgt tgatgatgct tacccaatcc aagttaaact 27120 tggatctggc cttagctttg acagtacagg agccataatg gctggtaaca aagaagacga 27180 taaactcact ttgtggacaa cacctgatcc atcaccaaac tgtcaaatac tcgcagaaaa 27240 tgatgcaaaa ctaacacttt gcttgactaa atgtggtagt caaatactgg ccactgtgtc 27300 agtcttagtt gtaggaagtg gaaacctaaa ccccattact ggcaccgtaa gcagtgctca 27360 ggtgtttcta cgttttgatg caaacggtgt tcttttaaca gaacattcta cactaaaaaa 27420 atactggggg tataggcagg gagatagcat agatggcact ccatatacca atgctgtagg 27480 attcatgccc aatttaaaag cttatccaaa gtcacaaagt tctactacta aaaataatat 27540 agtagggcaa gtatacatga atggagatgt ttcaaaacct atgcttctca ctataaccct 27600 caatggtact gatgacagca acagtacata ttcaatgtca ttttcataca cctggactaa 27660 tggaagctat gttggagcaa catttggggc taactcttat accttctcat acatcgccca 27720 agaatgaaca ctgtatccca ccctgcatgc caacccttcc caccccactc tgtggaacaa 27780 actctgaaac acaaaataaa ataaagttca agtgttttat tgattcaaca gtttcacaga 27840 accctagtat tcaacctgcc acctccctcc caacacacag agtacacagt cctttctccc 27900 cggctggcct taaaaagcat catatcatgg gtaacagaca tattcttagg tgttatattc 27960 cacacggttt cctgtcgagc caaacgctca tcagtgatat taataaactc cccgggcagc 28020 tcacttaagt tcatgtcgct gtccagctgc tgagccacag gctgctgtcc aacttgcggt 28080 tgcttaacgg gcggcgaagg agaagtccac gcctacatgg gggtagagtc ataatcgtgc 28140 atcaggatag ggcggtggtg ctgcagcagc gcgcgaataa actgctgccg ccgccgctcc 28200 gtcctgcagg aatacaacat ggcagtggtc tcctcagcga tgattcgcac cgcccgcagc 28260 ataaggcgcc ttgtcctccg ggcacagcag cgcaccctga tctcacttaa atcagcacag 28320 taactgcagc acagcaccac aatattgttc aaaatcccac agtgcaaggc gctgtatcca 28380 aagctcatgg cggggaccac agaacccacg tggccatcat accacaagcg caggtagatt 28440 aagtggcgac ccctcataaa cacgctggac ataaacatta cctcttttgg catgttgtaa 28500 ttcaccacct cccggtacca tataaacctc tgattaaaca tggcgccatc caccaccatc 28560 ctaaaccagc tggccaaaac ctgcccgccg gctatacact gcagggaacc gggactggaa 28620 caatgacagt ggagagccca ggactcgtaa ccatggatca tcatgctcgt catgatatca 28680 atgttggcac aacacaggca cacgtgcata cacttcctca ggattacaag ctcctcccgc 28740 gttagaacca tatcccaggg aacaacccat tcctgaatca gcgtaaatcc cacactgcag 28800 ggaagacctc gcacgtaact cacgttgtgc attgtcaaag tgttacattc gggcagcagc 28860 ggatgatcct ccagtatggt agcgcgggtt tctgtctcaa aaggaggtag acgatcccta 28920 ctgtacggag tgcgccgaga caaccgagat cgtgttggtc gtagtgtcat gccaaatgga 28980 acgccggacg tagtcatatt tcctgaagca aaaccaggtg cgggcgtgac aaacagatct 29040 gcgtctccgg tctcgccgct tagatcgctc tgtgtagtag ttgtagtata tccactctct 29100 caaagcatcc aggcgccccc tggcttcggg ttctatgtaa actccttcat gcgccgctgc 29160 cctgataaca tccaccaccg cagaataagc cacacccagc caacctacac attcgttctg 29220 cgagtcacac acgggaggag cgggaagagc tggaagaacc atgattaact ttattccaaa 29280 cggtctcgga gcacttcaaa atgcaggtcc cggaggtggc acctctcgcc cccactgtgt 29340 tggtggaaaa taacagccag gtcaaaggtg acacggttct cgagatgttc cacggtggct 29400 tccagcaaag cctccacgcg cacatccaga aacaagagga cagcgaaagc gggagcgttt 29460 tctaattcct caatcatcat attacactcc tgcaccatcc ccagataatt ttcatttttc 29520 cagccttgaa tgattcgtat tagttcctga ggtaaatcca agccagccat gataaaaagc 29580 tcgcgcagag cgccctccac cggcattctt aagcacaccc tcataattcc aagagattct 29640 gctcctggtt cacctgcagc agattaacaa tgggaatatc aaaatctctg ccgcgatccc 29700 taagctcctc cctcaacaat aactgtatgt aatctttcat atcatctccg aaatttttag 29760 ccatagggcc gccaggaata agagcagggc aagccacatt acagataaag cgaagtcctc 29820 cccagtgwgc attgccaaat gtaagattga aataagcatg ctggctagac cctgtgatat 29880 cttccagata actggacaga aaatcaggca agcaattttt aagaaaatca acaaaagaaa 29940 agtcgtccag gtgcaggttt agagcctcag gaacaacgat ggaataagtg caaggagtgc 30000 gttccagcat ggttagtgtt tttttggtga tctgtagaac aaaaaataaa catgcaatat 30060 taaaccatgc tagcctggcg aacaggtggg taaatcactc tttccagcac caggcaggct 30120 acggggtctc cggcgcgacc ctcgtagaag ctgtcgccat gattgaaaag catcaccgag 30180 agaccttccc ggtggccggc atggatgatt cgagaagaag catacactcc gggaacattg 30240 gcatccgtga gtgaaaaaaa gcgacctata aagcctcggg gcactacaat gctcaatctc 30300 aattccagca aagccacccc atgcggatgg agcacaaaat tggcaggtgc gtaaaaaatg 30360 taattactcc cctcctgcac aggcagcaaa gcccccgctc cctccagaaa cacatacaaa 30420 gcctcagcgt ccatagctta ccgagcacgg caggcgcaag agtcagagaa aaggctgagc 30480 tctaacctga ctgcccgctc ctgtgctcaa tatatagccc taacctacac tgacgtaaag 30540 gccaaagtct aaaaataccc gccaaataat cacacacgcc cagcacacgc ccagaaaccg 30600 gtgacacact caaaaaaata cgcgcacttc ctcaaacgcc caaaactgcc gtcatttccg 30660 ggttcccacg ctacgtcatc aaaacacgac tttcaaattc cgtcgaccgt taaaaacgtc 30720 acccgccccg cccctaacgg tcgcccgtct ctcagccaat cagcgccccg catccccaaa 30780 ttcaaacacc tcatttgcat attaacgcgc acaaaaagtt tgaggtatat tattgatgat 30840 gg 30842

Claims

1. A composition comprising a viral vector, the viral vector comprising nucleic acid having a polynucleotide sequence encoding the spike protein from the middle eastern respiratory syndrome coronavirus (MERS-CoV), characterised in that said viral vector is an adenovirus based vector.
2. A composition according to claim ι wherein said adenovirus based vector is ChAdOx l.
3. A composition according to claim 1 or claim 2 wherein said spike protein comprises the receptor binding domains (RBDs).
4. A composition according to any of claims 1 to 3 wherein said spike protein is full length spike protein.
5. A composition according to any preceding claim wherein said spike protein is present as a fusion with the tissue plasminogen activator (tPA) sequence in the order N-terminus - tPA - spike protein - C-terminus.
6. A composition according to claim 5 wherein said tPA has the amino acid sequence SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, or SEQ ID NO: 8.
7. A composition according to any preceding claim wherein said spike protein has the amino acid sequence SEQ ID NO: 1.
8. A composition according to any preceding claim wherein said polynucleotide sequence comprises the sequence of SEQ ID NO: 3 or SEQ ID NO: 4.
9. A composition according to any of claims 2 to 8 wherein said viral vector sequence is as in ECACC accession number 12052403.
10. A composition according to any preceding claim wherein administration of a single dose of said composition to a mammalian subject induces protective immunity in said subject.
11. A composition according to any preceding claim for induction of an immune response against MERS-CoV.
12. A composition according to any preceding claim for induction of an immune response against MERS-CoV, wherein a single dose of said composition is
administered.
13. A composition according to any preceding claim for induction of an immune response against MERS-CoV, wherein said composition is administered once.
14. A composition according to claim 13, wherein said composition is administered once per 6 months.
15. A composition according to claim 13, wherein said composition is administered once per 12 months.
16. A composition according to any preceding claim for preventing MERS-CoV infection.
17. A composition according to any preceding claim for preventing MERS-CoV infection, wherein a single dose of said composition is administered.
18. A composition according to any preceding claim for preventing MERS-CoV infection, wherein said composition is administered once.
19. A composition according to claim 18, wherein said composition is administered once per 6 months.
20. A composition according to claim 18, wherein said composition is administered once per 12 months.
21. Use of a composition according to any preceding claim in medicine.
22. Use of a composition according to any preceding claim in the preparation of a medicament for prevention of MERS-CoV infection.
23. A method of inducing an immune response against middle eastern respiratory syndrome coronaviras (MERS-CoV) in a mammalian subject, the method comprising administering a composition according to any preceding claim to said subject.
24. A method according to claim 23 wherein a single dose of said composition is administered to said subject.
25. A method according to claim 23 or 24 wherein said composition is administered once.
26. A method according to claim 25 wherein said composition is administered once per 6 months.
27. A method according to claim 25 wherein said composition is administered once per 12 months.
28. A method according to any of claims 23 to 27 wherein said composition is administered by a route of administration selected from a group consisting of subcutaneous, intranasal, aerosol, nebuliser, intradermal and intramuscular.
29. A method according to claim 28 wherein said administration is intramuscular.
PCT/GB2018/051399 2017-05-26 2018-05-23 Compositions and methods for inducing an immune response WO2018215766A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1708444.3 2017-05-26
GBGB1708444.3A GB201708444D0 (en) 2017-05-26 2017-05-26 Compositions and methods for inducing an immune response

Publications (1)

Publication Number Publication Date
WO2018215766A1 true WO2018215766A1 (en) 2018-11-29

Family

ID=59271005

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2018/051399 WO2018215766A1 (en) 2017-05-26 2018-05-23 Compositions and methods for inducing an immune response

Country Status (2)

Country Link
GB (1) GB201708444D0 (en)
WO (1) WO2018215766A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20210108331A (en) * 2020-02-25 2021-09-02 (주)지뉴인텍 Recombinant adenovirus vaccine for corona virus disease 19 and combination therapy using the same
WO2021181100A1 (en) * 2020-03-13 2021-09-16 Oxford University Innovation Limited Compositions and methods for inducing an immune response
WO2021195694A1 (en) * 2020-03-31 2021-10-07 Sementis Limited Attenuated poxvirus vector based vaccine for protection against covid-19
WO2021207592A1 (en) * 2020-04-09 2021-10-14 4Mvac Llc Use of viral vectors for coronavirus vaccine production
WO2021211279A1 (en) * 2020-04-17 2021-10-21 Regents Of The University Of Minnesota SARS-CoV-2 SPIKE RECEPTOR BINDING DOMAIN AND COMPOSITIONS AND METHODS THEREOF
WO2021228842A1 (en) 2020-05-11 2021-11-18 Janssen Pharmaceuticals, Inc. Stabilized coronavirus spike protein fusion proteins
WO2022002894A1 (en) 2020-06-29 2022-01-06 Janssen Vaccines & Prevention B.V. Vaccine combination against respiratory syncytial virus infection
WO2022008438A1 (en) 2020-07-06 2022-01-13 Janssen Pharmaceuticals, Inc. Stabilized corona virus spike protein fusion proteins
WO2022123030A1 (en) * 2020-12-10 2022-06-16 The Chancellor, Masters And Scholars Of The University Of Oxford Method for purifying virus
WO2022123033A1 (en) * 2020-12-10 2022-06-16 The Chancellor, Masters And Scholars Of The University Of Oxford Method for producing virus
US11384122B2 (en) 2020-01-31 2022-07-12 Janssen Pharmaceuticals, Inc. Compositions and methods for preventing and treating coronavirus infection—SARS-CoV-2 vaccines
WO2022175477A1 (en) 2021-02-19 2022-08-25 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion rsv fb antigens
WO2022175479A1 (en) 2021-02-19 2022-08-25 Janssen Vaccines & Prevention B.V. Vaccine combinations against respiratory syncytial virus strain a and b infections
WO2022180007A1 (en) 2021-02-23 2022-09-01 Janssen Vaccines & Prevention B.V. Trimer stabilizing hiv envelope protein mutation
WO2022207839A2 (en) 2021-04-01 2022-10-06 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion piv3 f proteins
WO2022234283A1 (en) * 2021-05-07 2022-11-10 Astrazeneca Uk Limited Compositions and methods for inducing an immune response
WO2023020939A1 (en) 2021-08-17 2023-02-23 Janssen Pharmaceuticals, Inc. Sars-cov-2 vaccines
WO2023026182A1 (en) 2021-08-24 2023-03-02 Janssen Pharmaceuticals, Inc. Sars-cov-2 vaccines
WO2023047348A1 (en) 2021-09-24 2023-03-30 Janssen Pharmaceuticals, Inc. Stabilized corona virus spike protein fusion proteins
WO2023047349A1 (en) 2021-09-24 2023-03-30 Janssen Pharmaceuticals, Inc. Stabilized coronavirus spike protein fusion proteins
WO2023111725A1 (en) 2021-12-14 2023-06-22 Janssen Pharmaceuticals, Inc. Sars-cov-2 vaccines
WO2023110618A1 (en) 2021-12-16 2023-06-22 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion hmpv fusion proteins
WO2023156505A1 (en) 2022-02-17 2023-08-24 Janssen Vaccines & Prevention B.V. Trimer stabilizing hiv envelope protein mutations r304v, n302m and t320l
EP4103210A4 (en) * 2020-02-14 2024-03-20 Altimmune Inc Coronavirus immunogenic compositions and uses thereof
WO2024061757A1 (en) 2022-09-23 2024-03-28 Janssen Vaccines & Prevention B.V. Pre-fusion human piv1 f proteins
WO2024061759A1 (en) 2022-09-23 2024-03-28 Janssen Vaccines & Prevention B.V. Stabilized coronavirus s proteins

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012172277A1 (en) 2011-05-25 2012-12-20 Isis Innovation Limited Simian adenovirus and hybrid adenoviral vectors
US9273327B2 (en) 2010-04-16 2016-03-01 Isis Innovation Limited Poxvirus expression system
CN105543248A (en) * 2015-03-13 2016-05-04 中国疾病预防控制中心病毒病预防控制所 MERS-CoV spike protein coding gene optimization-based recombinant 5-type adenovirus vector vaccine
WO2017176596A1 (en) * 2016-04-04 2017-10-12 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Multivalent vaccines for rabies virus and coronaviruses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9273327B2 (en) 2010-04-16 2016-03-01 Isis Innovation Limited Poxvirus expression system
WO2012172277A1 (en) 2011-05-25 2012-12-20 Isis Innovation Limited Simian adenovirus and hybrid adenoviral vectors
CN105543248A (en) * 2015-03-13 2016-05-04 中国疾病预防控制中心病毒病预防控制所 MERS-CoV spike protein coding gene optimization-based recombinant 5-type adenovirus vector vaccine
WO2017176596A1 (en) * 2016-04-04 2017-10-12 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Multivalent vaccines for rabies virus and coronaviruses

Non-Patent Citations (89)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. AHX71946.1
"GenBank", Database accession no. KJ650098.1
ADNEY DR; BROWN VR; PORTER SM; BIELEFELDT-OHMANN H; HARTWIG AE; BOWEN RA: "Inoculation of Goats, Sheep, and Horses with MERS-CoV Does Not Result in Productive Viral Shedding", VIRUSES, vol. 8, no. 8, 2016
AGRAWAL, A. S. ET AL.: "Generation of a transgenic mouse model of Middle East respiratory syndrome coronavirus infection and disease", J VIROL, vol. 89, 2015, pages 3659 - 3670, XP055212038, DOI: doi:10.1128/JVI.03427-14
AI HAMMADI ZM; CHU DK; ELTAHIR YM; AI HOSANI F; AI MULLA M; TARNINI W ET AL.: "Asymptomatic MERS-CoV Infection in Humans Possibly Linked to Infected Dromedaries Imported from Oman to United Arab Emirates", EMERGING INFECTIOUS DISEASES, vol. 21, no. 12, May 2015 (2015-05-01), pages 2197 - 200
ALAGAILI AN; BRIESE T; MISHRA N; KAPOOR V; SAMEROFF SC; BURBELO PD ET AL.: "Middle East respiratory syndrome coronavirus infection in dromedary camels in Saudi Arabia", MBIO, vol. 5, no. 2, 2014, pages e00884 - 14
ALHARBI NK: "Vaccines against Middle East respiratory syndrome coronavirus for humans and camels", REVIEWS IN MEDICAL VIROLOGY, 2016
ALHARBI NK; SPENCER AJ; SALMAN AM; TULLY CM; CHINNAKANNAN SK; LAMBE T ET AL.: "Enhancing cellular immunogenicity of MVA-vectored vaccines by utilizing the F11L endogenous promoter", VACCINE, vol. 34, no. 1, 2016, pages 49 - 55, XP029341745, DOI: doi:10.1016/j.vaccine.2015.11.028
ALI MA; SHEHATA MM; GOMAA MR; KANDEIL A; EL-SHESHENY R; KAYED AS ET AL.: "Systematic, active surveillance for Middle East respiratory syndrome coronavirus in camels in Egypt", EMERGING MICROBES & INFECTIONS, vol. 6, no. l, 2017, pages e1
ALI, M. A. ET AL.: "Systematic, active surveillance for Middle East respiratory syndrome coronavirus in camels in Egypt", EMERG MICROBES INFECT, vol. 6, 2017, pages e1
ALRADDADI BM; WATSON JT; ALMARASHI A; ABEDI GR; TURKISTANI A; SADRAN M ET AL.: "Risk Factors for Primary Middle East Respiratory Syndrome Coronavirus Illness in Humans, Saudi Arabia, 2014", EMERGING INFECTIOUS DISEASES, vol. 22, no. 1, 2016, pages 49 - 55
ANNA H. MALCZYK ET AL: "ABSTRACT", JOURNAL OF VIROLOGY., vol. 89, no. 22, 9 September 2015 (2015-09-09), US, pages 11654 - 11667, XP055257597, ISSN: 0022-538X, DOI: 10.1128/JVI.01815-15 *
ANTROBUS RD; COUGHLAN L; BERTHOUD TK; DICKS MD; HILL AV; LAMBE T ET AL.: "Clinical assessment of a novel recombinant simian adenovirus ChAdOx1 as a vectored vaccine expressing conserved Influenza A antigens", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 22, no. 3, 2014, pages 668 - 74, XP055401001, DOI: doi:10.1038/mt.2013.284
ARABI, Y. M. ET AL.: "Middle East Respiratory Syndrome", N ENGL J MED, vol. 376, 2017, pages 584 - 594
ASISA VOLZ ET AL: "Protective Efficacy of Recombinant Modified Vaccinia Virus Ankara Delivering Middle East Respiratory Syndrome Coronavirus Spike Glycoprotein", JOURNAL OF VIROLOGY., vol. 89, no. 16, 27 May 2015 (2015-05-27), US, pages 8651 - 8656, XP055257600, ISSN: 0022-538X, DOI: 10.1128/JVI.00614-15 *
B. L. HAAGMANS ET AL: "An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels", SCIENCE, vol. 351, no. 6268, 1 January 2016 (2016-01-01), US, pages 77 - 81, XP055265765, ISSN: 0036-8075, DOI: 10.1126/science.aad1283 *
CORMAN VM; ALBARRAK AM; OMRANI AS; ALBARRAK MM; FARAH ME; ALMASRI M ET AL.: "Viral Shedding and Antibody Response in 37 Patients With Middle East Respiratory Syndrome Coronavirus Infection", CLIN INFECT DIS., vol. 62, no. 4, 2016, pages 477 - 83
COTTINGHAM MG; ANDERSEN RF; SPENCER AJ; SAURYA S; FURZE J; HILL AV ET AL.: "Recombination-mediated genetic engineering of a bacterial artificial chromosome clone of modified vaccinia virus Ankara (MVA", PLOS ONE, vol. 3, no. 2, 2008, pages e1638, XP002642804, DOI: doi:10.1371/journal.pone.0001638
COTTINGHAM MG; CARROLL F; MORRIS SJ; TURNER AV; VAUGHAN AM; KAPULU MC ET AL.: "Preventing spontaneous genetic rearrangements in the transgene cassettes of adenovirus vectors", BIOTECHNOL BIOENG., vol. 109, no. 3, 2012, pages 719 - 28, XP055401017, DOI: doi:10.1002/bit.24342
DE WIT, E. ET AL.: "Middle East respiratory syndrome coronavirus (MERS-CoV) causes transient lower respiratory tract infection in rhesus macaques", PROC NATL ACAD SCI USA, vol. 110, no. 16, 2013, pages 8 - 16603
DE WIT, E.; VAN DOREMALEN, N.; FALZARANO, D.; MUNSTER, V. J.: "SARS and MERS: recent insights into emerging coronaviruses", NAT REV MICROBIOL, vol. 14, 2016, pages 523 - 534, XP055369190, DOI: doi:10.1038/nrmicro.2016.81
DICKS MD; SPENCER AJ; EDWARDS NJ; WADELL G; BOJANG K; GILBERT SC ET AL.: "A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity", PLOS ONE, vol. 7, no. 7, 2012, pages e40385, XP055037313, DOI: doi:10.1371/journal.pone.0040385
DICKS MDJ; SPENCER AJ; EDWARDS NJ; WADELL G; BOJANG K ET AL.: "A Novel Chimpanzee Adenovirus Vector with Low Human Seroprevalence: Improved Systems for Vector Derivation and Comparative Immunogenicity", PLOS ONE, vol. 7, no. 7, 2012, pages e40385, XP055037313, DOI: doi:10.1371/journal.pone.0040385
DICKS, M. D. ET AL.: "A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity", PLOS ONE, vol. 7, 2012, pages e40385, XP055037313, DOI: doi:10.1371/journal.pone.0040385
DRAPER SJ; HEENEY JL: "Viruses as vaccine vectors for infectious diseases and cancer", NAT REV MICROBIOL., vol. 8, no. 1, 2010, pages 62 - 73, XP055484517, DOI: doi:10.1038/nrmicro2240
DRAPER SJ; MOORE AC; GOODMAN AL; LONG CA; HOLDER AA; GILBERT SC ET AL.: "Effective induction of high-titer antibodies by viral vector vaccines", NATURE MEDICINE, vol. 14, no. 8, 2008, pages 819 - 21, XP002497769, DOI: doi:10.1038/nm.1850
EWER, K. J. ET AL.: "Viral vectors as vaccine platforms: from immunogenicity to impact", CURR OPIN IMMUNOL, vol. 41, 2016, pages 47 - 54, XP029688144, DOI: doi:10.1016/j.coi.2016.05.014
FUNK AL; GOUTARD FL; MIGUEL E; BOURGAREL M; CHEVALIER V; FAYE B ET AL.: "MERS-CoV at the Animal-Human Interface: Inputs on Exposure Pathways from an Expert-Opinion Elicitation", FRONTIERS IN VETERINARY SCIENCE, vol. 3, 2016, pages 88
GEORGE M. WARIMWE ET AL: "Chimpanzee Adenovirus Vaccine Provides Multispecies Protection against Rift Valley Fever", SCIENTIFIC REPORTS, vol. 6, no. 1, 5 February 2016 (2016-02-05), XP055499794, DOI: 10.1038/srep20617 *
GILBERT SC: "Clinical development of Modified Vaccinia virus Ankara vaccines", VACCINE, vol. 31, no. 39, 2013, pages 4241 - 6, XP028697925, DOI: doi:10.1016/j.vaccine.2013.03.020
GOMEZ CE; PERDIGUERO B; GARCIA-ARRIAZA J; ESTEBAN M: "Clinical applications of attenuated MVA poxvirus strain", EXPERT REVIEW OF VACCINES, vol. 12, no. 12, 2013, pages 1395 - 416
GOSSNER C; DANIELSON N; GERVELMEYER A; BERTHE F; FAYE B; KAASIK AASLAV K ET AL.: "Human-Dromedary Camel Interactions and the Risk of Acquiring Zoonotic Middle East Respiratory Syndrome Coronavirus Infection", ZOONOSES AND PUBLIC HEALTH., vol. 63, no. 1, 2016, pages 1 - 9
GREHAN K; FERRARA F; TEMPERTON N: "An optimised method for the production of MERS-CoV spike expressing viral pseudotypes", METHODSX., vol. 2, 2015, pages 379 - 84
GUTIERREZ C; TEJEDOR-JUNCO MT; GONZALEZ M; LATTWEIN E; RENNEKER S: "Presence of antibodies but no evidence for circulation of MERS-CoV in dromedaries on the Canary Islands, 2015", EURO SURVEILLANCE : BULLETIN EUROPEEN SUR LES MALADIES TRANSMISSIBLES = EUROPEAN COMMUNICABLE DISEASE BULLETIN, vol. 20, no. 37, 2015
HAAGMANS BL; VAN DEN BRAND JM; RAJ VS; VOLZ A; WOHLSEIN P; SMITS SL ET AL.: "An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels", SCIENCE, vol. 351, no. 6268, 2016, pages 77 - 81, XP055265765, DOI: doi:10.1126/science.aad1283
HAAGMANS ET AL., SCIENCE, vol. 351, 2016, pages 77
HEMIDA MG; PERERA RA; AI JASSIM RA; KAYALI G; SIU LY; WANG P ET AL.: "Seroepidemiology of Middle East respiratory syndrome (MERS) coronavirus in Saudi Arabia (1993) and Australia (2014) and characterisation of assay specificity", EURO SURVEILLANCE : BULLETIN EUROPEEN SUR LES MALADIES TRANSMISSIBLES = EUROPEAN COMMUNICABLE DISEASE BULLETIN, vol. 19, no. 23, 2014
HEMIDA MG; PERERA RA; WANG P; ALHAMMADI MA; SIU LY; LI M ET AL.: "Middle East Respiratory Syndrome (MERS) coronavirus seroprevalence in domestic livestock in Saudi Arabia, 2010 to 2013", EURO SURVEILLANCE : BULLETIN EUROPEEN SUR LES MALADIES TRANSMISSIBLES = EUROPEAN COMMUNICABLE DISEASE BULLETIN, vol. 18, no. 50, 2013, pages 20659
HONDA-OKUBO ET AL., JOURNAL OF VIROLOGY, vol. 89, 2015, pages 2995
HOTEZ ET AL., MICROBES AND INFECTION, vol. 2016, 2014, pages 529 - 531
IWATA-YOSHIKAWA ET AL., JOURNAL OF VIROLOGY, vol. 88, 2014, pages 8597
JAUME ET AL., HONG KONG MED J, vol. 18, 2012, pages 31
KIM EUN ET AL: "Immunogenicity of an adenoviral-based Middle East Respiratory Syndrome coronavirus vaccine in BALB/c mice", VACCINE, ELSEVIER, AMSTERDAM, NL, vol. 32, no. 45, 3 September 2014 (2014-09-03), pages 5975 - 5982, XP029069689, ISSN: 0264-410X, DOI: 10.1016/J.VACCINE.2014.08.058 *
LAN J; YAO Y; DENG Y; CHEN H; LU G; WANG W ET AL.: "Recombinant Receptor Binding Domain Protein Induces Partial Protective Immunity in Rhesus Macaques Against Middle East Respiratory Syndrome Coronavirus Challenge", EBIOMEDICINE, vol. 2, no. 10, 2015, pages 1438 - 46
LESSLER J; SALJE H; VAN KERKHOVE MD; FERGUSON NM; CAUCHEMEZ S; RODRIQUEZ-BARRAQUER I ET AL.: "Estimating the Severity and Subclinical Burden of Middle East Respiratory Syndrome Coronavirus Infection in the Kingdom of Saudi Arabia", AMERICAN JOURNAL OF EPIDEMIOLOGY, vol. 183, no. 7, 2016, pages 657 - 63
LUO M; TAO P; LI J; ZHOU S; GUO D; PAN Z: "Immunization with plasmid DNA encoding influenza A virus nucleoprotein fused to a tissue plasminogen activator signal sequence elicits strong immune responses and protection against H5N1 challenge in mice", JOURNAL OF VIROLOGICAL METHODS, vol. 154, no. 1-2, 2008, pages 121 - 7, XP025680309, DOI: doi:10.1016/j.jviromet.2008.08.011
MALCZYK ET AL., JOURNAL OF VIROLOGY, vol. 89, 2015, pages 11654
MEMISH ZA; ZUMLA AL; ASSIRI A: "Middle East respiratory syndrome coronavirus infections in health care workers", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 369, no. 9, 2013, pages 884 - 6
MIN CK; CHEON S; HA NY; SOHN KM; KIM Y; AIGERIM A ET AL.: "Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity", SCIENTIFIC REPORTS, vol. 6, 2016, pages 25359
MODJARRAD, K. ET AL.: "A roadmap for MERS-CoV research and product development: report from a World Health Organization consultation", NAT MED, vol. 22, 2016, pages 701 - 705
MORRIS, S. J.; TURNER, A. V.; GREEN, N.; WARIMWE, G. M.: "Laboratory-Scale Production of Replication-Deficient Adenovirus Vectored Vaccines", METHODS MOL BIOL, vol. 1349, 2016, pages 121 - 135
MULLER MA; CORMAN VM; JORES J; MEYER B; YOUNAN M; LILJANDER A ET AL.: "MERS coronavirus neutralizing antibodies in camels, Eastern Africa, 1983-1997", EMERG INFECT DIS., vol. 20, no. i2, 2014, pages 2093 - 5
MULLER MA; CORMAN VM; JORES J; MEYER B; YOUNAN M; LILJANDER A ET AL.: "MERS coronavirus neutralizing antibodies in camels, Eastern Africa, 1983-1997", EMERGING INFECTIOUS DISEASES, vol. 20, no. 12, 2014, pages 2093 - 5
MUTHUMANI K; FALZARANO D; REUSCHEL EL; TINGEY C; FLINGAI S; VILLARREAL DO ET AL.: "A synthetic consensus anti-spike protein DNA vaccine induces protective immunity against Middle East respiratory syndrome coronavirus in nonhuman primates", SCIENCE TRANSLATIONAL MEDICINE, vol. 7, no. 301, 2015, pages 301ral32, XP055271204, DOI: doi:10.1126/scitranslmed.aac7462
NISREEN MA OKBA ET AL: "Middle East respiratory syndrome coronavirus vaccines: current status and novel approaches", CURRENT OPINION IN VIROLOGY, vol. 23, 13 April 2017 (2017-04-13), United Kingdom, pages 49 - 58, XP055499799, ISSN: 1879-6257, DOI: 10.1016/j.coviro.2017.03.007 *
NUCLEIC ACIDS RES., vol. 43, 2015, pages D204 - D212
NUCLEIC ACIDS RESEARCH, vol. 4i, no. Di, January 2013 (2013-01-01), pages D36 - 42
OBOHO IK; TOMCZYK SM; AL-ASMARI AM; BANJAR AA; AL-MUGTI H; ALORAINI MS ET AL.: "MERS-CoV outbreak in Jeddah--a link to health care facilities", N ENGL J MED., vol. 372, no. 9, 2014, pages 846 - 54
OBOHO, I. K. ET AL.: "MERS-CoV outbreak in Jeddah--a link to health care facilities", N ENGL J MED, vol. 372, 2014, pages 846 - 854
PARK WB; PERERA RA; CHOE PG; LAU EH; CHOI SJ; CHUN JY ET AL.: "Kinetics of Serologic Responses to MERS Coronavirus Infection in Humans, South Korea", EMERGING INFECTIOUS DISEASES, vol. 21, no. 12, 2015, pages 2186 - 9
PELLERIN C: "Army Scientists Begin First MERS Vaccine Clinical Trial", DOD NEWS, DEFENSE MEDIA ACTIVITY, 2016
PERERA RA; WANG P; GOMAA MR; EL-SHESHENY R; KANDEIL A; BAGATO O ET AL.: "Seroepidemiology for MERS coronavirus using microneutralisation and pseudoparticle virus neutralisation assays reveal a high prevalence of antibody in dromedary camels in Egypt, June 2013", EURO SURVEILLANCE : BULLETIN EUROPEEN SUR LES MALADIES TRANSMISSIBLES = EUROPEAN COMMUNICABLE DISEASE BULLETIN, vol. 18, no. 36, 2013, pages 20574, XP055354126, DOI: doi:10.2807/1560-7917.ES2013.18.36.20574
REUSKEN CB; ABABNEH M; RAJ VS; MEYER B; ELJARAH A; ABUTARBUSH S ET AL.: "Middle East Respiratory Syndrome coronavirus (MERS-CoV) serology in major livestock species in an affected region in Jordan, June to September 2013", EURO SURVEILLANCE : BULLETIN EUROPEEN SUR LES MALADIES TRANSMISSIBLES = EUROPEAN COMMUNICABLE DISEASE BULLETIN, vol. 18, no. 50, 2013, pages 20662
REUSKEN CB; HAAGMANS BL; MULLER MA; GUTIERREZ C; GODEKE GJ; MEYER B ET AL.: "Middle East respiratory syndrome coronavirus neutralising serum antibodies in dromedary camels: a comparative serological study", THE LANCET INFECTIOUS DISEASES, vol. 13, no. 10, 2013, pages 859 - 66
REUSKEN, C. B. ET AL.: "Geographic distribution of MERS coronavirus among dromedary camels, Africa", EMERG INFECT DIS, vol. 20, 2014, pages 1370 - 1374
REUSKEN, C. B. ET AL.: "Middle East respiratory syndrome coronavirus neutralising serum antibodies in dromedary camels: a comparative serological study", LANCET INFECT DIS, vol. 13, 2013, pages 859 - 866
RICHARD D ANTROBUS ET AL: "Clinical Assessment of a Novel Recombinant Simian Adenovirus ChAdOx1 as a Vectored Vaccine Expressing Conserved Influenza A Antigens", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 22, no. 3, 1 March 2014 (2014-03-01), US, pages 668 - 674, XP055401001, ISSN: 1525-0016, DOI: 10.1038/mt.2013.284 *
SONG F; FUX R; PROVACIA LB; VOLZ A; EICKMANN M; BECKER S ET AL.: "Middle East respiratory syndrome coronavirus spike protein delivered by modified vaccinia virus Ankara efficiently induces virus-neutralizing antibodies", JOURNAL OF VIROLOGY, vol. 87, no. 21, 2013, pages 11950 - 4, XP055157123, DOI: doi:10.1128/JVI.01672-13
SRIDHAR S; REYES-SANDOVAL A; DRAPER SJ; MOORE AC; GILBERT SC; GAO GP ET AL.: "Single-dose protection against Plasmodium berghei by a simian adenovirus vector using a human cytomegalovirus promoter containing intron A", J VIROL., vol. 82, no. 8, 2008, pages 3822 - 33, XP009103960, DOI: doi:10.1128/JVI.02568-07
TAO D; BARBA-SPAETH G; RAI U; NUSSENZWEIG V; RICE CM; NUSSENZWEIG RS: "Yellow fever 17D as a vaccine vector for microbial CTL epitopes: protection in a rodent malaria model", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 201, no. 2, 2005, pages 201 - 9
TSCHARKE DC; WOO WP; SAKALA IG; SIDNEY J; SETTE A; MOSS DJ ET AL.: "Poxvirus CD8+ T-cell determinants and cross-reactivity in BALB/c mice", JOURNAL OF VIROLOGY, vol. 80, no. 13, 2006, pages 6318 - 23
TSENG ET AL., PLOS ONE, vol. 7, 2012, pages e35421
VAN DOREMALEN N; HIJAZEEN ZS; HOLLOWAY P; AL OMARI B; MCDOWELL C; ADNEY D ET AL.: "High Prevalence of Middle East Respiratory Coronavirus in Young Dromedary Camels in Jordan", VECTOR BORNE AND ZOONOTIC DISEASES, vol. 17, no. 2, 2017, pages 155 - 9
VAN DOREMALEN, N. ET AL.: "High Prevalence of Middle East Respiratory Coronavirus in Young Dromedary Camels in Jordan", VECTOR BORNE ZOONOTIC DIS, vol. 17, 2017, pages 155 - 159
VERGARA-ALERT J; VAN DEN BRAND JM; WIDAGDO W; MUNOZ MT; RAJ S; SCHIPPER D ET AL.: "Livestock Susceptibility to Infection with Middle East Respiratory Syndrome Coronavirus", EMERGING INFECTIOUS DISEASES, vol. 23, no. 2, 2017, pages 232 - 40
VOLZ A; KUPKE A; SONG F; JANY S; FUX R; SHAMS-ELDIN H ET AL.: "Protective efficacy of recombinant Modified Vaccinia virus Ankara (MVA) delivering Middle East Respiratory Syndrome coronavirus spike glycoprotein", JOURNAL OF VIROLOGY, 2015
VOLZ ET AL., JOURNAL OF VIROLOGY, vol. 89, 2015, pages 8651
WALLACE A; WEST K; ROTHMAN AL; ENNIS FA; LU S; WANG S: "Post-translational intracellular trafficking determines the type of immune response elicited by DNA vaccines expressing Gag antigen of Human Immunodeficiency Virus Type 1 (HIV-1", HUM VACCIN IMMUNOTHER, vol. 9, no. 10, 2013, pages 2095 - 102
WANG L; SHI W; JOYCE MG; MODJARRAD K; ZHANG Y; LEUNG K ET AL.: "Evaluation of candidate vaccine approaches for MERS-CoV", NATURE COMMUNICATIONS, vol. 6, 2015, pages 7712, XP055271251, DOI: doi:10.1038/ncomms8712
WANG W; WANG H; DENG Y; SONG T; LAN J; WU G ET AL.: "Characterization of anti-MERS-CoV antibodies against various recombinant structural antigens of MERS-CoV in an imported case in China", EMERGING MICROBES & INFECTIONS, vol. 5, no. 11, 2016, pages e113
WARIMWE GM; GESHARISHA J; CARR BV; OTIENO S; OTINGAH K; WRIGHT D ET AL.: "Chimpanzee Adenovirus Vaccine Provides Multispecies Protection against Rift Valley Fever", SCI REP., vol. 6, 2016, pages 20617
WARIMWE GM; LORENZO G; LOPEZ-GIL E; REYES-SANDOVAL A; COTTINGHAM MG; SPENCER AJ ET AL.: "Immunogenicity and efficacy of a chimpanzee adenovirus-vectored Rift Valley fever vaccine in mice", VIROLOGY JOURNAL, vol. 10, 2013, pages 349, XP021171452, DOI: doi:10.1186/1743-422X-10-349
WARIMWE, G. M. ET AL.: "Chimpanzee Adenovirus Vaccine Provides Multispecies Protection against Rift Valley Fever", SCI REP, vol. 6, 2016, pages 20617
WEINGARTL ET AL., JOURNAL OF VIROLOGY, vol. 8, 2004, pages 12672
WHO MERS-COV RESEARCH G: "State of Knowledge and Data Gaps of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) in Humans", PLOS CURRENTS, vol. 5, 2013
WIRBLICH CHRISTOPH ET AL: "One-Health: a Safe, Efficient, Dual-Use Vaccine for Humans and Animals against Middle East Respiratory Syndrome Coronavirus and Rabies Virus", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 91, no. 2, 1 January 2017 (2017-01-01), pages e02040 - 16, XP009194667, ISSN: 0022-538X, DOI: 10.1128/JVI.02040-16 *
WORLD HEALTH ORGANISATION. MIDDLE EAST RESPIRATORY SYNDROME CORONAVIRUS (MERS-COV, 2016, Retrieved from the Internet <URL:http://www.who.int/emergencies/mers-cov/en>
XIAOJUAN GUO ET AL: "Systemic and mucosal immunity in mice elicited by a single immunization with human adenovirus type 5 or 41 vector-based vaccines carrying the spike protein of Middle East respiratory syndrome coronavirus", IMMUNOLOGY, vol. 145, no. 4, 21 April 2015 (2015-04-21), GB, pages 476 - 484, XP055499409, ISSN: 0019-2805, DOI: 10.1111/imm.12462 *
ZAKI AM; VAN BOHEEMEN S; BESTEBROER TM; OSTERHAUS AD; FOUCHIER RA: "Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 367, no. 19, 2012, pages 1814 - 20, XP055274765, DOI: doi:10.1056/NEJMoa1211721

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11498944B2 (en) 2020-01-31 2022-11-15 Janssen Pharmaceuticals, Inc. Compositions and methods for preventing and treating coronavirus infection—SARS-CoV-2 vaccines
US11384122B2 (en) 2020-01-31 2022-07-12 Janssen Pharmaceuticals, Inc. Compositions and methods for preventing and treating coronavirus infection—SARS-CoV-2 vaccines
EP4103210A4 (en) * 2020-02-14 2024-03-20 Altimmune Inc Coronavirus immunogenic compositions and uses thereof
KR20210108331A (en) * 2020-02-25 2021-09-02 (주)지뉴인텍 Recombinant adenovirus vaccine for corona virus disease 19 and combination therapy using the same
KR102601258B1 (en) 2020-02-25 2023-11-13 (주)지뉴인텍 Recombinant adenovirus vaccine for corona virus disease 19 and combination therapy using the same
US20230285532A1 (en) * 2020-03-13 2023-09-14 Oxford University Innovation Limited Compositions and Methods for Inducing an Immune Response
WO2021181100A1 (en) * 2020-03-13 2021-09-16 Oxford University Innovation Limited Compositions and methods for inducing an immune response
WO2021195694A1 (en) * 2020-03-31 2021-10-07 Sementis Limited Attenuated poxvirus vector based vaccine for protection against covid-19
WO2021207592A1 (en) * 2020-04-09 2021-10-14 4Mvac Llc Use of viral vectors for coronavirus vaccine production
WO2021211279A1 (en) * 2020-04-17 2021-10-21 Regents Of The University Of Minnesota SARS-CoV-2 SPIKE RECEPTOR BINDING DOMAIN AND COMPOSITIONS AND METHODS THEREOF
WO2021228842A1 (en) 2020-05-11 2021-11-18 Janssen Pharmaceuticals, Inc. Stabilized coronavirus spike protein fusion proteins
WO2022002894A1 (en) 2020-06-29 2022-01-06 Janssen Vaccines & Prevention B.V. Vaccine combination against respiratory syncytial virus infection
WO2022008438A1 (en) 2020-07-06 2022-01-13 Janssen Pharmaceuticals, Inc. Stabilized corona virus spike protein fusion proteins
WO2022123030A1 (en) * 2020-12-10 2022-06-16 The Chancellor, Masters And Scholars Of The University Of Oxford Method for purifying virus
WO2022123033A1 (en) * 2020-12-10 2022-06-16 The Chancellor, Masters And Scholars Of The University Of Oxford Method for producing virus
WO2022175479A1 (en) 2021-02-19 2022-08-25 Janssen Vaccines & Prevention B.V. Vaccine combinations against respiratory syncytial virus strain a and b infections
WO2022175477A1 (en) 2021-02-19 2022-08-25 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion rsv fb antigens
WO2022180007A1 (en) 2021-02-23 2022-09-01 Janssen Vaccines & Prevention B.V. Trimer stabilizing hiv envelope protein mutation
WO2022207839A2 (en) 2021-04-01 2022-10-06 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion piv3 f proteins
WO2022234283A1 (en) * 2021-05-07 2022-11-10 Astrazeneca Uk Limited Compositions and methods for inducing an immune response
WO2023020939A1 (en) 2021-08-17 2023-02-23 Janssen Pharmaceuticals, Inc. Sars-cov-2 vaccines
WO2023026182A1 (en) 2021-08-24 2023-03-02 Janssen Pharmaceuticals, Inc. Sars-cov-2 vaccines
WO2023047348A1 (en) 2021-09-24 2023-03-30 Janssen Pharmaceuticals, Inc. Stabilized corona virus spike protein fusion proteins
WO2023047349A1 (en) 2021-09-24 2023-03-30 Janssen Pharmaceuticals, Inc. Stabilized coronavirus spike protein fusion proteins
WO2023111725A1 (en) 2021-12-14 2023-06-22 Janssen Pharmaceuticals, Inc. Sars-cov-2 vaccines
WO2023110618A1 (en) 2021-12-16 2023-06-22 Janssen Vaccines & Prevention B.V. Stabilized pre-fusion hmpv fusion proteins
WO2023156505A1 (en) 2022-02-17 2023-08-24 Janssen Vaccines & Prevention B.V. Trimer stabilizing hiv envelope protein mutations r304v, n302m and t320l
WO2024061757A1 (en) 2022-09-23 2024-03-28 Janssen Vaccines & Prevention B.V. Pre-fusion human piv1 f proteins
WO2024061759A1 (en) 2022-09-23 2024-03-28 Janssen Vaccines & Prevention B.V. Stabilized coronavirus s proteins

Also Published As

Publication number Publication date
GB201708444D0 (en) 2017-07-12

Similar Documents

Publication Publication Date Title
WO2018215766A1 (en) Compositions and methods for inducing an immune response
Alharbi et al. ChAdOx1 and MVA based vaccine candidates against MERS-CoV elicit neutralising antibodies and cellular immune responses in mice
US11459583B2 (en) Adenovirus vectors and uses thereof
US11236361B2 (en) Adenovirus and uses thereof
Guo et al. Immunization with a novel human type 5 adenovirus-vectored vaccine expressing the premembrane and envelope proteins of Zika virus provides consistent and sterilizing protection in multiple immunocompetent and immunocompromised animal models
WO2021076010A1 (en) Pharmaceutical agent for inducing specific immunity against sars-cov-2
US10286060B2 (en) Compositions and methods
Grubman et al. Adenovirus serotype 5-vectored foot-and-mouth disease subunit vaccines: the first decade
US20220016234A1 (en) Anti covid-19 therapies using nucleocapsid and spike proteins
KR20220152248A (en) Compositions and methods for inducing an immune response
US20220001007A1 (en) Compositions and methods
CN107530383A (en) Method and composition for research of Ebola vaccine inoculation
Kantarcioglu et al. An update on the status of vaccine development for SARS-CoV-2 including variants. Practical considerations for COVID-19 special populations
Holman et al. A complex adenovirus-vectored vaccine against Rift Valley fever virus protects mice against lethal infection in the presence of preexisting vector immunity
Verardi et al. Long-term sterilizing immunity to rinderpest in cattle vaccinated with a recombinant vaccinia virus expressing high levels of the fusion and hemagglutinin glycoproteins
Hao et al. An adenovirus-vectored RVF vaccine confers complete protection against lethal RVFV challenge in A129 mice
WO2023111725A1 (en) Sars-cov-2 vaccines
Bian et al. Single-dose of a replication-competent adenovirus-vectored vaccine provides sterilizing protection against Rift Valley fever virus challenge
EP4205761A1 (en) Novel coronavirus recombinant spike protein, polynucleotide encoding same, vector comprising polynucleotide, and vaccine for preventing or treating coronavirus infection, comprising vector
Wu et al. Development and evaluation of Newcastle disease-avian influenza bivalent vector vaccines in commercial chickens
JP7360544B2 (en) Pharmaceutical products for inducing specific immunity against SARS-COV-2
US11857620B2 (en) Method of inducing immunity against SARS-CoV-2 using spike (s) and nucleocapsid (N)-ETSD immunogens delivered by a replication-defective adenovirus
CN114891830B (en) Recombinant expression vector based on varicella-zoster virus, recombinant virus and application thereof
Alamri et al. Enhanced immunogenicity of a synthetic DNA vaccine expressing consensus SARS-CoV-2 Spike protein using needle-free immunization
US20220265816A1 (en) Use of the agent for induction of specific immunity against severe acute respiratory syndrome virus sars-cov-2 for revaccination of population (variants)

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: 18728706

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18728706

Country of ref document: EP

Kind code of ref document: A1