Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
STABILISED SUPEROXIDE DISMUTASE (SOD) HOMOLOG GENE AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2019/220403
Kind Code:
A2
Abstract:
The present invention relates to a stabilised SOD-homolog (SODis) gene of SEQ ID NO:2 and recombinant viruses comprising the gene. The invention also relates to a recombinant lumpy skin disease virus (LSDV) wherein a gene encoding a SOD-homolog polypeptide has been inactivated by deletion from the viral genome. The invention further relates to pharmaceutical compositions comprising the recombinant viruses and to the use of the recombinant viruses in methods of preventing a disease in a subject.

Inventors:
WILLIAMSON ANNA-LISE (ZA)
MUNYANDUKI HENRY (ZA)
DOUGLASS NICOLA JENNIFER (ZA)
Application Number:
PCT/IB2019/054090
Publication Date:
November 21, 2019
Filing Date:
May 17, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CAPE TOWN (ZA)
International Classes:
A61K39/275
Other References:
KARA, P.D.AFONSO, C.L.WALLACE, D.B.KUTISH, G.F.ABOLNIK,C.LU, Z.VREEDE, F.T.TALJAARD, L.C.ZSAK, A.VILJOEN, G.J.: "Comparative sequence analysis of the South African vaccine strain and two virulent field isolates of lumpy skin disease virus", ARCH. VIROL., vol. 148, 2003, pages 1335 - 1356, XP008165302
TULMAN, ERAFONSO, CLZSAK, LKUTISH, GFROCK, DL: "Genome of Lumpy Skin Disease Virus", J VIROL., vol. 75, no. 15, August 2001 (2001-08-01), pages 7122 - 30, XP055280347, DOI: doi:10.1128/JVI.75.15.7122-7130.2001
Attorney, Agent or Firm:
SPOOR & FISHER et al. (ZA)
Download PDF:
Claims:
CLAIMS

1 . A stabilised SOD-homolog (SODis) gene of SEQ ID NO:2.

2. A recombinant virus comprising the SODis gene of claim 1 .

3. The recombinant virus of claim 2, further comprising a marker gene selected from the group consisting of a guanine phosphoribosyltransferase gene, a green fluorescent protein gene, an enhanced green fluorescent protein gene and a red fluorescence protein gene.

4. A pharmaceutical composition, comprising:

i. a recombinant virus of claim 2; and

ii. a pharmaceutically acceptable diluent or excipient.

5. The pharmaceutical composition of claim 4, wherein the pharmaceutical composition is capable of eliciting a protective immune response in a subject against a disease caused by a microorganism.

6. The pharmaceutical composition of claim 5, wherein the microorganism is a virus from the viral genus Caphpoxvihdae.

7. The pharmaceutical composition of claim 5 or 6, wherein the disease is selected from the group consisting of lumpy skin disease, sheep pox, and goat pox.

8. The pharmaceutical composition of any one of claims 5 to 7, wherein the subject is a ruminant.

9. The pharmaceutical composition of claim 8, wherein the ruminant is selected from the group consisting of cattle, sheep, goats, and wild ungulates.

10. The pharmaceutical composition of any one of claims 4 to 9, which is formulated for intramuscular, intradermal, intravenous or subcutaneous administration.

1 1 . A recombinant virus for use in a method of preventing a disease in a subject, wherein the recombinant virus comprises a SODis gene of SEQ ID NO:2 in its viral genome.

12. The recombinant virus for use of claim 1 1 , wherein the disease is caused by a microorganism.

13. The recombinant virus for use of claim 12, wherein the microorganism is a virus from the viral genus Caphpoxvihdae.

14. The recombinant virus for use of any one of claims 1 1 to 13, wherein the disease is selected from lumpy skin disease, sheep pox and goat pox.

15. The recombinant virus for use of any one of claims 1 1 to 14, wherein the subject is a ruminant.

16. The recombinant virus for use of claim 15, wherein the ruminant is selected from the group consisting of cattle, sheep, goats and wild ungulates.

17. A recombinant lumpy skin disease virus (LSDV), wherein a gene encoding a SOD-homolog polypeptide has been inactivated by deletion from the viral genome.

18. The recombinant LSDV of claim 1 7, wherein the SOD-homolog polypeptide is selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:17, SEQ ID NO:18, and SEQ ID NO:19.

19. The recombinant LSDV of claim 1 7 or 18, further comprising a marker gene selected from the group consisting of a guanine phosphoribosyltransferase gene, a green fluorescent protein gene, an enhanced green fluorescent protein gene and a red fluorescence protein gene.

20. A pharmaceutical composition, comprising:

i. a recombinant LSDV wherein a gene encoding a SOD-homolog polypeptide has been inactivated by deletion from the viral genome; and

ii. a pharmaceutically acceptable diluent or excipient.

21 . The pharmaceutical composition of claim 20, wherein the pharmaceutical composition is capable of eliciting a protective immune response in a subject against a disease caused by a microorganism.

22. The pharmaceutical composition of claim 21 , wherein the microorganism is a virus from the viral genus Caphpoxvihdae.

23. The pharmaceutical composition of claim 21 or 22, wherein the disease is selected from the group consisting of lumpy skin disease, sheep pox, and goat pox.

24. The pharmaceutical composition of any one of claims 21 to 23, wherein the subject is a ruminant.

25. The pharmaceutical composition of claim 24, wherein the ruminant is selected from the group consisting of cattle, sheep, goats, and wild ungulates.

26. The pharmaceutical composition of any one of claims 20 to 25, which is formulated for intramuscular, intradermal, intravenous or subcutaneous administration.

27. A recombinant LSDV wherein a gene encoding a SOD-homolog polypeptide has been inactivated by deletion from the viral genome for use in a method of preventing a disease in a subject.

28. The recombinant LSDV for use of claim 27, wherein the disease is caused by a microorganism.

29. The recombinant LSDV for use of claim 28, wherein the microorganism is a virus from the viral genus Capripoxviridae.

30. The recombinant LSDV for use of any one of claims 27 to 29, wherein the disease is selected from lumpy skin disease, sheep pox and goat pox.

31 . The recombinant LSDV for use of any one of claims 27 to 30, wherein the subject is a ruminant.

32. The recombinant LSDV for use of claim 31 , wherein the ruminant is selected from the group consisting of cattle, sheep, goats and wild ungulates.

Description:
STABILISED SUPEROXIDE DISMUTASE (SOD) HOMOLOG GENE AND USES

THEREOF

BACKGROUND OF THE INVENTION

The present invention relates to a stabilised SOD-homolog (SODis) gene of SEQ ID NO:2 and recombinant viruses comprising the gene, having improved immunogenicity. The invention further relates to a recombinant lumpy skin disease virus (LSDV) having a deleted gene encoding a SOD-homolog polypeptide. The present invention also relates to pharmaceutical compositions comprising the recombinant viruses, as well as the use of said recombinant viruses in methods of preventing a disease in a subject.

Lumpy skin disease virus (LSDV) is a member of the Capripoxvirus genus of the Poxviridae family of viruses and is the causative agent of lumpy skin disease (LSD) in cattle. Lactating cattle have been found to be the most susceptible to disease. The rate of morbidity is often higher during an outbreak and can rise to 85%. LSD in cattle ranges from subclinical to an acute infection and usually lasts for 2-5 weeks. The acute infection is characterised by fever and localised or disseminated nodules developing on the skin, where lesions are often found in the upper respiratory tract and a secondary bacterial infection often occurs. Nodules have been reported to occur on the skeletal muscles and the mucosa of the oral and upper respiratory tract. Systemic effects include pyrexia, anorexia, dysgalactia and pneumonia. Cows may lose reproductive ability and milk producing capacity for several months. Fever, anorexia and abortions are also common. The drop in milk production in lactating cows along with temporary or permanent infertility in cows and bulls and skin lesions caused by the virus all impact on the economic effects of the disease. Eradication campaigns and imposed trade restriction on live animals and animal products add to the financial toll of an outbreak. In Africa economic losses due to LSD are comparable to that of foot and mouth disease. This has resulted in the World Organization for Animal Heath (OIE) recognising LSD as a notifiable agricultural disease.

LSDV is only able to naturally cause disease in cattle despite a high level of sequence identity shared with sheeppox virus (SPV) and goatpox virus (GPV). Some isolates of sheep and goatpox virus are able to infect both sheep and goats; however most isolates display host preference for either sheep or goats. These host range characteristics are reflected in the independent distribution of the different Capripoxvirus species. Wild game animals such as giraffes and impala have been shown to be permissive for wild-type LSDV where, during experimental infection, both animal species displayed pathology resembling that of LSDV infection in cattle.

There is thus a need for vaccines against Lumpy Skin Disease (LSD), which affects cattle in most African countries as well as the Middle East and Gulf Peninsula. It is spreading at an alarming rate and has recently entered Europe. Vaccines which are commercially available include a vaccine based on attenuated Neethling LSDV isolate, Herbivac®LS (Herbivac) and MSD (Design Biologix product), all produced in South Africa. The Neethling LSDV isolate vaccine produced by Onderstepoort Biological Products is generally highly effective, but safety issues have been raised. The KS1 -O180 strain is used in Ethiopia for immunization of small ruminants and cattle, but gives poor protection. The RM-65 attenuated sheep pox vaccine has been used at ten times the dose in cattle, but is less effective than vaccination with homologous strains.

In addition to the varying degrees of virulence of commercial vaccines there is also variability in susceptibility of different breeds of cattle to the LSDV vaccines. The European high-producing cattle breeds have been observed to be more susceptible to adverse vaccine reactions. It may therefore be desirable to have vaccines of differing potency for different breeds of cattle.

Herbivac has been found to be more immunogenic than Neethling, its parent strain. The DNA sequence of Herbivac was recently published and one significant difference was noted between Herbivac and Neethling, at ORF 134, a VACV B22R homolog. The inventors of the present application have also sequenced Herbivac and found a different single significant change from the published Neethling strain, at ORF 131 , a gene potentially coding for a superoxide dismutase (SOD) homolog. Leporipoxviruses encode SOD homologs which act as decoy proteins. They are catalytically inert but bind the copper chaperone for SOD, thereby depleting the amount of copper available for use by the Cu, Zn SOD. This causes an intracellular increase in superoxide anion, which inhibits apoptosis.

In the present invention, a SOD-homolog ORF (SODis) was designed to resemble that of Herbivac, yet be more stable. The inventors have constructed recombinant LSDV viruses with and without the SOD-homolog gene. Assays to test the induction and inhibition of apoptosis indicated that the SOD-homolog causes inhibition of camptothecin induced apoptosis. The inventors have also shown that the presence or absence of the SOD-homolog influences immunogenicity of vaccine. In the present invention the SOD-homolog was modified to be more stable and less likely to mutate in the way that it has during its history of passaging, both in the generation of the Neethling vaccine as well as in the generation of Herbivac.

SUMMARY OF THE INVENTION

The present invention relates to the SODis gene, constructs containing said gene or having a knocked-out SOD-homolog gene and vaccines comprising the gene or constructs for use in treating diseases.

According to a first aspect of the present invention there is provided for a stabilised SOD-homolog (SODis) gene of SEQ ID NO:2.

In a second aspect of the invention there is provided for a recombinant virus comprising the SODis gene of of SEQ ID NO:2.

According to a third aspect of the present invention there is provided for a recombinant lumpy skin disease virus (LSDV), wherein a gene encoding a SOD- homolog polypeptide has been inactivated by deletion from the viral genome. The SOD-homolog polypeptide may be selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:17, SEQ ID NO:18, and SEQ ID NO:19.

In a first embodiment of the invention, the recombinant virus or recombinant LSDV of the invention further comprises a marker gene selected from the group consisting of a guanine phosphoribosyltransferase gene, a green fluorescent protein gene, an enhanced green fluorescent protein gene and a red fluorescence protein gene. For example, the marker gene may be GPT, eGFP, or mCherry. It will be appreciated by a person of skill in the art that any selectable marker may be used in the recombinant virus or recombinant LSDV.

According to a fourth aspect of the present invention there is provided for a pharmaceutical composition, comprising the recombinant virus or recombinant LSDV of the invention and a pharmaceutically acceptable diluent or excipient.

In another embodiment of the invention, the pharmaceutical composition is capable of eliciting a protective immune response in a subject, particularly a ruminant, against a disease caused by a microorganism, particularly a virus from the viral genus Caphpoxvihdae.

According to one embodiment, the disease may be selected from the group consisting of lumpy skin disease, sheep pox, and goat pox.

In yet another embodiment, the ruminant is selected from the group consisting of cattle, sheep, goats, and wild ungulates. The pharmaceutical composition of the present invention may be formulated for intramuscular, intradermal, intravenous or subcutaneous administration.

According to a further aspect of the present invention there is provided for a recombinant virus or recombinant LSDV as described, for use in a method of preventing a disease in a subject, particularly a ruminant. Preferably, the disease is caused by a microorganism, particularly a virus from the viral genus Caphpoxvihdae.

In another embodiment, the disease may be selected from the group consisting of lumpy skin disease, sheep pox, and goat pox.

In yet another embodiment, the ruminant is selected from the group consisting of cattle, sheep, goats, and wild ungulates.

BRIEF DESCRIPTION OF THE FIGURES

Non-limiting embodiments of the invention will now be described by way of example only and with reference to the following figures:

Figure 1 : Schematic showing the deletion of SOD-homolog gene

(ORF131 ) from nLSDV and replacement with GPT and eGFP marker gene to product nLSDVASOD-M. Within the infected and transfected cell homologous recombination between the plasmid and nLSDV genomic DNA at the left flank (ORF 130) and right flank (ORF 132) resulted in ORF 131 being replaced by the GPT and eGFP genes present in plasmid pHM1 .

Figure 2: Diagram showing overlapping primers for assembly of fragments in the construction of pHM1 .

Figure 3: Transfer vector pHM1 . LSDV 130 and 132 are open reading frames that flank the LSDV ORF131 (SOD). GPT - xanthine-guanine phosphoribosyltransferase, GFP - green fluorescence protein, bla - B-lactamase.

Figure 4: Schematic showing the insertion of a synthetic SOD-homolog gene (SODis) into nLSDVASOD-M to construct nLSDVSODis-M.

Figure 5: Plasmid map of transfer vector pHM2v3. GPT and LSDV132 served as flanking sequences for homologous recombination, such that the eGFP gene of nLSDVASOD-M can be replaced with the synthetic SOD-homolog gene (SODis) under its native promoter and mCherry under the control of the mH5 promoter.

Figure 6: DNA sequence of pHM2v3 between the sites of insertion (Pvul and Notl). This sequence was synthesized and ligated into a pUC57 simple vector by Gene Script (USA). Restriction sites are underlined and the start codons of SODis and mCherry are italicized in bold. Also in bold italics are transcriptional stop signals. The native SOD-homolog gene promoter and the mH5 promoter upstream of the mCherry gene are depicted in lower case. Highlighted in bold and of larger font is the sequence that was altered to improve stability of the SOD-homolog gene by removing a series of AT repeats, yet retaining the translated amino acid sequence. An abbreviated form of the sequence is: Pvul-GPT-Clal-BamH1 - SODis -TTTTTCT - BamHI-Ascl- Xbal- PmH5 -Sall-mCherrv - TTTTTCT- Sacll- Smal-LSDV 132- Kpnl- Acll-Notl.

Figure 7: Confirmation of recombinant viruses nLSDVASOD-M and nLSDVSODis-M by PCR: Panel A - Schematic showing fragment sizes generated using a primer pair which bind in LSDV ORFs 129 and 132. Panel B - 1 % Agarose gel electrophoresis of PCR products amplified from WT nLSDV (lane 1), nLSDVASOD-M (lane 2) and nLSDVSODis-M (lane 3).

Figure 8: Transfer vector pHMI dSOD and strategy of construction of nLSDVASOD-UCT using nLSDVASOD-M as the parent virus. Homologous recombination can occur between the left (LSDV ORF 130) and right (LSDV ORF132) flanks, resulting in the removal of GPT and eGFP from the parent virus and the generation of a non-fluorescing recombinant lacking the LSDV SOD-homolog gene (ORF 131 ).

Figure 9: Confirmation of recombinant virus nLSDVASOD-UCT by PCR:

Diagram to show where primers bind and sizes of PCR products amplified from the parent virus nLSDVASOD-M, the original parent virus Neethling and the recombinant nLSDVASOD-UCT.

Figure 10: Confirmation of recombinant virus nLSDVASOD-UCT by PCR: Agarose gel electrophoresis of PCR products amplified from uninfected MDBK cells (lane 1 ), nLSDVASOD-M (lanes 2 and 3), nLSDV (lanes 4 and 5) and nLSDVASOD- UCT (lane 6). Primer sequences used were: LSDV 130 for: 5’- GG AGCACCATTT CCAT AT AC -3’ (SEQ ID NO:24) and LSDV 132 rev: 5’- AGT AGCTT AAAAGG AGGG AAG -3’ (SEQ ID NO:25).

Figure 11 : T ransfer vector pHM1 -SODis. This transfer vector has the novel SODis gene inserted between LSDV ORFs 130 and 132. It was used to remove the marker genes from nLSDVASOD-M in the construction of nLSDVSODis-UCT.

Figure 12: Confirmation of recombinant virus nLSDVSODis-UCT by PCR. Panel A - diagram to show sizes of PCR products amplified from the desired recombinant virus nLSDVDOSis-UCT, nLSDV and parent virus nLSDVASOD-M. Primers used were: forward primer 129F: 5’ - GAGCCCTGTAATTCACTTTT - 3’ (SEQ ID NO:26) and reverse primer 132R: 5’ - AT CGAT GG AAAAG AT CCG - 3’ (SEQ ID NO:27). Panel B - 1 % agarose gel electrophoresis of PCR products amplified from nLSDVSODis-UCT (lane 1), the parent virus, nLSDVASOD-M (lane 2) and nLSDV (lane 3).

Figure 13: Nucleotide alignment of the SOD homologs from the virulent Neethling NI-2490, the OBP Neethling nLSDV and Herbivac. Herbivac and nLSDV share a single base pair deletion at position 252. Herbivac has a unique deletion of 2bp at positions 324-325, which restores the SOD-homolog gene ORF. It is noteworthy that the sequence from positions 314-325 comprises repeated AT binucleotides which explains how the 2bp deletion could have arisen.

Figure 14: Amino acid alignment of the SOD homologs from the virulent Neethling NI-2490, Herbivac and the OBP Neethling nLSDV. The single base pair deletion common to nLSDV and Herbivac alters the amino acid sequence at position 84. The 2bp deletion unique to Herbivac restores the SOD-homolog gene ORF such that Herbivac resembles the virulent NI-2490 from position 106.

Figure 15: Induction of apoptosis. MDBK cells were infected with nLSDV, Herbivac, nLSDVASOD-M and nLSDVSODis-M at an MOI of 1 . Uninfected cells were used as a control. Six hours post infection the cell medium was removed and the cells lysed. The Roche cell death detection ELISAPLUS kit was used to measure the number of nucleosomes released. The symbol *** represents a significance level of p<0.001 . Bars denote mean and error bars SEM.

Figure 16: Inhibition of camptothecin induced apoptosis. MDBK cells were infected with nLSDV, Herbivac, nLSDVASOD-M and nLSDVSODis-M at an MOI of 1 in duplicate sets of wells. Uninfected cells were used as a control. Apoptosis was induced with 0.0001 mg/ml camptothecin in one set of wells. The enrichment factor after induction of apoptosis was calculated and is expressed as fold decrease in apoptosis compared to uninfected cells.

Figure 17: Inflammatory changes associated with the SOD-homolog gene. Seven day old fertilized chick embryo chorioallantoic membranes (CAMs) were infected for 5 days, washed, fixed with 4% buffered formalin, embedded, sectioned and stained with haematoxylin and eosin. For each virus-infected membrane five sections were evaluated under the light microscope and scored for inflammation (oedema, haemorrhage and infiltration with white blood cells). One-way ANOVA and Tukey post hoc testing was used to test for significance. The symbols ** and *** represent significance levels of p<0.01 and p<0.001 respectively. Bars denote mean and error bars denote standard deviation.

Figure 18: Comparison of lesions produced by different LSDV vaccines in cows. A group of five friesian calves, 6-12 months old, and LSDV seronegative, were each inoculated at different sites on the neck with a dose of 104 TCID50 of each of nLSDV, Herbivac, nLSDVASOD-UCT and nLSDVSODis-UCT. The animals were monitored daily for two weeks for temperature and lesion size. Two of the five animals (Animals 166 and 82) developed lesions. The surface area (cm 2 ) of the lesions was measured and is graphically displayed. The black continuous line graph above represents rectal temperature. The neutralization titres at the end of the experiment were 1 :64 for Animal 166 and 1 :128 for Animal 82.

Figure 19: Amino acid sequence of the SOD-homolog polypeptide expressed from the nLSDVSODis-M and nLSDVSODis-UCT constructs (SEQ ID NO:1 ).

Figure 20: Nucleic acid sequence of the stabilised SOD-homolog (SODis) gene used in the nLSDVSODis-M and nLSDVSODis-UCT constructs (SEQ ID NO:2).

Figure 21 : Nucleic acid sequence of the pHM1 insertion cassette used to obtain the nLSDVASOD-M construct (SEQ ID NO:20).

Figure 22: Nucleic acid sequence of the pHM2v3 insertion cassette used to obtain the nLSDVSODis-M construct (SEQ ID NO:21 ).

Figure 23: Nucleic acid sequence of the pHMIASOD insertion cassette used to obtain the nLSDVASOD-UCT construct (SEQ ID NO:22).

Figure 24: Nucleic acid sequence of the pHM1 -SODis insertion cassette used to obtain the nLSDVSODis-UCT construct (SEQ ID NO:23).

SEQUENCE LISTING

The nucleic acid and amino acid sequences listed in the accompanying sequence listing are shown using standard letter abbreviations for nucleotide bases, and the standard three letter abbreviations for amino acids. It will be understood by those of skill in the art that only one strand of each nucleic acid sequence is shown, but that the complementary strand is included by any reference to the displayed strand. In the accompanying sequence listing:

SEQ ID NO:1 - Amino acid sequence of the SOD-homolog expressed from the SODis gene.

SEQ ID NO:2 - Nucleic acid sequence of SODis gene.

SEQ ID NO:3 - Full genome sequence of Herbivac vaccine strain.

SEQ ID NO:4 - Full genome sequence of nLSDVASOD-UCT vaccine construct.

SEQ ID NO:5 - Full genome sequence of nLSDVASODis-UCT vaccine construct. SEQ ID NO:6 - Nucleic acid sequence of LSDV 130 forward primer used to amplify flanking sequences of nLSDV ORF 130 from pZG1 .

SEQ ID NO:7 - Nucleic acid sequence of LSDV 130 reverse primer used to amplify flanking sequences of nLSDV ORF 130 from pZG1 .

SEQ ID NO:8 - Nucleic acid sequence of pRO-2-GPT forward primer used to amplify selection and marker genes from pRO-2.

SEQ ID NO:9 - Nucleic acid sequence of pRO-2-eGFP reverse primer used to amplify selection and marker genes from pRO-2.

SEQ ID NO:10 - Nucleic acid sequence of LSDV 132 forward primer used to amplify flanking sequences of nLSDV ORF 132 from pZG2.

SEQ ID NO:1 1 - Nucleic acid sequence of LSDV 132 reverse primer used to amplify flanking sequences of nLSDV ORF 132 from pZG2.

SEQ ID NO:12 - Nucleic acid sequence of pHM2v3 between the sites of insertion (Pvul and Notl).

SEQ ID NO:13 - Nucleic acid sequence encoding NI-2490 SOD-homolog.

SEQ ID NO:14 - Nucleic acid sequence encoding nLSDV SOD-homolog.

SEQ ID NO:15 - Nucleotide sequence encoding nLSDV SOD-homolog of truncated ORF

SEQ ID NO:16 - Nucleic acid sequence encoding Herbivac SOD-homolog.

SEQ ID NO:17 - Amino acid sequence of NI-2490 SOD-homolog.

SEQ ID NO:18 - Amino acid sequence of nLSDV SOD-homolog.

SEQ ID NO:19 - Amino acid sequence of Herbivac SOD-homolog.

SEQ ID NO:20 - Nucleic acid sequence of the pHM1 insertion cassette used to make the nLSDVASOD-M construct.

SEQ ID NO:21 - Nucleic acid sequence of the pHM2v3 insertion cassette used to obtain the nLSDVSODis-M construct.

SEQ ID NO:22 - Nucleic acid sequence of the pHMIASOD insertion cassette used to obtain the nLSDVASOD-UCT construct.

SEQ ID NO:23 - Nucleic acid sequence of the pHM1 -SODis insertion cassette used to obtain the nLSDVSODis-UCT construct.

DETAILED DESCRIPTION OF THE INVENTION

The present invention will now be described more fully hereinafter with reference to the accompanying drawings, in which some, but not all embodiments of the invention are shown. The invention as described should not be limited to the specific embodiments disclosed and modifications and other embodiments are intended to be included within the scope of the invention. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.

As used throughout this specification and in the claims which follow, the singular forms“a”,“an” and“the” include the plural form, unless the context clearly indicates otherwise.

The terminology and phraseology used herein is for the purpose of description and should not be regarded as limiting. The use of the terms“comprising”,“containing”, “having” and“including” and variations thereof used herein, are meant to encompass the items listed thereafter and equivalents thereof as well as additional items.

The present invention relates to modified viral vaccine backbones. In particular, the inventors of the present invention have identified a gene (superoxide dismutase homolog (SOD)) which may influence immunogenicity of live virus vectors, in particular, poxviruses, which include Lumpy Skin Disease Virus (LSDV), other capripoxviruses, orthopoxviruses, avipoxviruses, as well as other viral vectors.

The Neethling vaccine strain of LSDV (nLSDV) was modified so as to delete the gene encoding the SOD-homolog completely and produce the vaccine nLSDVASOD-UCT (SEQ ID NO:4). In another embodiment, nLSDVSODis-UCT (SEQ ID NO:5) was produced, wherein the Neethling vaccine strain of LSDV SOD-homolog gene was replaced by a modified SOD-homolog gene, termed SODis, which has been modified so as to improve its stability. Two additional recombinants have been generated which contain marker genes. nLSDVASOD-M has the gene encoding the SOD-homolog deleted and replaced by the E.coli guanine phosphoribosyltransferase (GPT) gene and the enhanced Green Fluorescent Protein gene (eGFP). nLSDVSODis-M has the gene encoding the SOD-homolog replaced by the synthetic novel SODis gene, as well as GPT and mCherry (red fluorescence protein).

The SODis was constructed by the inventors by altering the nucleotide sequence of the Neethling vaccine strain SOD-homolog gene such that the repeat sequences of adenosine and thymine were removed, yet the amino acid sequence was identical to that of the Herbivac SOD homolog gene. Whole genome sequencing of the Herbivac vaccine indicated a mixed viral population where some viruses contained a restored SOD-homolog gene and some a truncated one. It is believed the mixed populated is due to the instability of the native SOD-homolog gene. When whole genome sequencing was performed on a population of nLSDVSODis-UCT all viruses contained the SODis gene and no truncated SOD-homolog sequences could be detected within the viral population.

The deletion or modification of the SOD-homolog gene in nLSDV has been engineered to improve immunogenicity of the vaccine or vaccine vector. The modified SOD homolog gene may be useful in other viral vectors to improve immunogenicity of the vaccine.

An improved LSDV vaccine would be of economic importance in the cattle industry. In addition the viral vector backbone could be used in the development of vaccines against other diseases, both veterinary and human. The SODis gene may be used in other viral vector backbones to improve immunogenicity.

Histological examination of infected chicken chorioallantoic membranes show that the presence and expression of the SODis gene increases mesodermal proliferation and immune cell infiltration at the site of infection. The expression of the SODis gene to produce a functional SOD-homolog also functions by inhibiting apoptosis.

Histologically, LSDV with the SOD-homolog gene deleted (nLSDVASOD-M and nLSDVASOD-UCT) and LSDV containing the modified SOD-homolog gene (nLSDVSODis-M and nLSDVSODis-UCT) differed from the Neethling LSDV. Herbivac resulted in increased immune cell infiltration at the site of infection compared to the parent Neethling vaccine strain and membranes infected with nLSDVSODis-M and nLSDVSODis-UCT showed marked proliferation of fibroblasts compared to nLSDVASOD-M and nLSDVASOD-UCT infected membranes.

It appears that inclusion of the SODis gene improves the LSDV vaccine in general. However, all vaccines including the SODis gene and those having the SOD- homolog gene deleted appear to induce different responses in vivo, as reflected by histological examination of chick CAMs and preliminary necrosis experiments.

It is known that cattle breeds differ in their responses to vaccination. More in- bred European cattle are more sensitive to vaccination and are more likely to have adverse reactions to vaccination compared to the "hardier" animals found in more rural settlements, such as those in Africa. It is possible that the presence of the SODis gene and expression of the full-length SOD-homolog or the deletion of the SOD gene would not only alter the immunogenicity but also the virulence of the vaccine. A vaccine with increased virulence would not be desirable for use in more sensitive inbred cattle, but, if more immunogenic, may be better for use in Africa, where the cattle are hardier. A less virulent vaccine would be more desirable in places like Europe, where the cattle are inbred. The Neethling strain of LSDV has a partial SOD-homolog gene. It would appear that this truncated protein product has some effect on virus-host interaction, making the recombinant virus (in which the SOD-homolog gene is deleted) different from its parent strain. The differences between Neethling, LSDV having a deleted SOD-homolog gene and LSDV with the modified SODis gene will be further tested in cattle.

The modified viral vaccine backbones, vaccine constructs and compositions of the present invention are useful as improved vaccines against LSDV or as vector backbones for additional vaccines, either multivalent veterinary vaccines, or vectors for vaccines such as, but not restricted to, HIV-1 .

The constructs according to the invention include, without limitation, a lumpy skin disease virus vaccine construct, wherein the gene or a functional part of the gene encoding a SOD-homolog has been deleted or inactivated in the viral genome. Alternatively, the constructs according to the present invention include a lumpy skin disease virus vaccine construct wherein the gene encoding the SOD-homolog has been stabilised.

The present invention targets the gene encoding a SOD-homolog of lumpy skin disease virus which is situated in open reading frame 131 (ORF131 ) of the LSDV Neethling strain. SEQ ID NO:1 provides a representative sequence of the SOD- homolog encoded by the stabilised SOD-homolog gene, termed SODis, having a nucleotide sequence of SEQ ID NO:2. SEQ ID NO:4 provides a representative sequence of the viral vaccine backbone of the present invention wherein the gene or a functional part of the gene encoding a SOD-homolog has been deleted from the viral genome. SEQ ID NO:5 provides a representative sequence of the viral vaccine backbone comprising a stabilised gene encoding the SOD-homolog.

By“lumpy skin disease” or“LSD” is meant an infectious disease in cattle caused by a virus of the family Poxviridae. The disease is characterized by fever, enlarged superficial lymph nodes and multiple nodules approximately 2-5 cm in diameter on the skin and mucous. Infected subjects also may develop edematous swelling in the limbs and exhibit lameness. LSD is caused by“lumpy skin disease virus” or“LSDV”. LSD has important economic implications since affected animals tend to have permanent damage to their skin, lowering the commercial value of their hide, as well as chronic debility, reduced milk production, poor growth, infertility, abortion, and sometimes death.

Onset of fever, which may exceed 41 °C, occurs almost one week after infection by the virus and persists for about one week. Around 1 week post-infection, all of the superficial lymph nodes become enlarged. Nodules appear seven to nineteen days after viral inoculation. The nodular lesions involve the dermis and the epidermis, but may extend to the underlying subcutis or even to the muscle. These lesions occur all over the body (but particularly on the head, neck, udder, scrotum, vulva and perineum).

LSDV is a double stranded deoxyribonucleic acid (dsDNA) virus. It is a member of the capripoxvirus genus of Poxviridae with an average virion size of 320 nm by 260 nm. Lumpy skin disease virus has a 151 kbp genome, consisting of a central coding region which is bounded by identical 2.4 kbp inverted terminal repeats and contains 156 genes. There are 146 conserved genes when comparing LSDV with chordopoxviruses of other genera. Within the central genomic region, LSDV genes share a high degree of collinearity and amino acid identity with the genes of other mammalian poxviruses. Examples of viruses with similar amino acid identity include suipoxvirus, yatapoxvirus, and lepohpoxvirus.

It will be appreciated by those of skill in the art that the viral vaccine backbones, constructs and compositions of the present invention, in particular the viral vaccine backbone either comprising a stabilised SODis gene or having the SOD-homolog gene deleted may be useful in viral vaccines for microorganisms other than for LSDV, including viral vaccines for Human Immunodeficiency Virus-1 (HIV-1 ), Bovine Leukaemia Virus (BVL), East Coast Fever (ECF), rabies, Rift Valley Fever Virus (RVFV).

By “condition associated with poxvirus infection” is meant any condition, disease or disorder that has been correlated with the presence of an existing poxvirus infection, it includes secondary effects, such as reductions in milk production, temporary or permanent infertility in cows and bulls, abortions and damage to hides, weight loss and wool brake.

A“protein,”“peptide” or“polypeptide” is any chain of two or more amino acids, including naturally occurring or non-naturally occurring amino acids or amino acid analogues, irrespective of post-translational modification (e.g., glycosylation or phosphorylation).

The terms “nucleic acid” or “nucleic acid molecule” encompass both ribonucelotides (RNA) and deoxyribonucleotides (DIMA), including cDNA, genomic DNA, and synthetic DNA. The nucleic acid may be double-stranded or single-stranded. Where the nucleic acid is single-stranded, the nucleic acid may be the sense strand or the antisense strand. A nucleic acid molecule may be any chain of two or more covalently bonded nucleotides, including naturally occurring or non-naturally occurring nucleotides, or nucleotide analogs or derivatives. By“RNA” is meant a sequence of two or more covalently bonded, naturally occurring or modified ribonucleotides. The term“DNA” refers to a sequence of two or more covalently bonded, naturally occurring or modified deoxyribonucleotides.

The term“complementary” refers to two nucleic acids molecules, e.g., DNA or RNA, which are capable of forming Watson-Crick base pairs to produce a region of double-strandedness between the two nucleic acid molecules. It will be appreciated by those of skill in the art that each nucleotide in a nucleic acid molecule need not form a matched Watson-Crick base pair with a nucleotide in an opposing complementary strand to form a duplex. One nucleic acid molecule is thus“complementary” to a second nucleic acid molecule if it hybridizes, under conditions of high stringency, with the second nucleic acid molecule. A nucleic acid molecule according to the invention includes both complementary molecules.

In some embodiments, a viral vaccine backbone of the invention may include, without limitation, SODis (SEQ ID NO:1 ) encoded by the SODis gene (SEQ ID NO:2), the vaccine constructs of the invention may include, without limitation nLSDVASOD- UCT (SEQ ID NO:4), nLSDVSODis-UCT (SEQ ID NO:5), nLSDVASOD-M, or nLSDVSODis-M.

As used herein a “substantially identical” sequence is an amino acid or nucleotide sequence that differs from a reference sequence only by one or more conservative substitutions, or by one or more non-conservative substitutions, deletions, or insertions located at positions of the sequence that do not destroy or substantially reduce the antigenicity of one or more of the expressed polypeptides or of the polypeptides encoded by the nucleic acid molecules. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the knowledge of those with skill in the art. These include using, for instance, computer software such as ALIGN, Megalign (DNASTAR), CLUSTALW or BLAST software. Those skilled in the art can readily determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. In one embodiment of the invention there is provided for a polypeptide or polynucleotide sequence that has at least about 80% sequence identity, at least about 90% sequence identity, or even greater sequence identity, such as about 95%, about 96%, about 97%, about 98% or about 99% sequence identity to the sequences described herein.

Alternatively, or additionally, two nucleic acid sequences may be“substantially identical” if they hybridize under high stringency conditions. The“stringency" of a hybridisation reaction is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation which depends upon probe length, washing temperature, and salt concentration. In general, longer probes required higher temperatures for proper annealing, while shorter probes require lower temperatures. Hybridisation generally depends on the ability of denatured DNA to re-anneal when complementary strands are present in an environment below their melting temperature. A typical example of such“stringent” hybridisation conditions would be hybridisation carried out for 18 hours at 65°C with gentle shaking, a first wash for 12 min at 65°C in Wash Buffer A (0.5% SDS; 2XSSC), and a second wash for 10 min at 65°C in Wash Buffer B (0.1 % SDS; 0.5% SSC).

In some embodiments, the nucleic acid molecules of the invention may be operably linked to other sequences. By“operably linked” is meant that the nucleic acid molecules encoding the SODis gene of the invention and regulatory sequences are connected in such a way as to permit expression of the proteins when the appropriate molecules are bound to the regulatory sequences. Similarly, the nucleic acid molecules encoding the marker genes of the viral vaccine constructs of the present invention may also be operably linked to regulatory sequences. Such operably linked sequences may be contained in vectors or expression constructs which can be transformed or transfected into host cells for expression. It will be appreciated that any vector or vectors can be used for the purposes of expressing the SOD-homolog encoded by the SODis gene of the invention.

The term“recombinant” means that something has been recombined. When used with reference to a nucleic acid construct the term refers to a molecule that comprises nucleic acid sequences that are joined together or produced by means of molecular biological techniques. Recombinant nucleic acid constructs may include a nucleotide sequence which is ligated to, or is manipulated to become ligated to, a nucleic acid sequence to which it is not ligated in nature, or to which it is ligated at a different location in nature. Accordingly, a recombinant nucleic acid construct indicates that the nucleic acid molecule has been manipulated using genetic engineering, i.e. by human intervention. Recombinant nucleic acid constructs may be introduced into a host cell by infection or transfection. Such recombinant nucleic acid constructs may include sequences derived from the same host cell species or from different host cell species.

The term “homologous recombination” refers to a mechanism of genetic recombination in which two nucleic acid strands comprising similar nucleotide sequences exchange genetic material. Cells use homologous recombination during meiosis, where it serves to rearrange DNA to create an entirely unique set of haploid chromosomes, but also for the repair of damaged DNA, in particular for the repair of double strand breaks. The mechanism of homologous recombination is well known to the skilled person in the art.

The term “vector” refers to a means by which polynucleotides or gene sequences can be introduced into a cell. There are various types of vectors known in the art including plasmids, viruses, bacteriophages and cosmids. Generally, polynucleotides or gene sequences are introduced into a vector by means of a cassette. The term “cassette” refers to a polynucleotide or gene sequence that is expressed from a vector, for example, the SODis sequence encoding the SOD- homolog polypeptides of the invention. A cassette generally comprises a gene sequence inserted into a vector, which in some embodiments, provides regulatory sequences for expressing the polynucleotide or gene sequences. In other embodiments, the vector provides the regulatory sequences for the expression of the SOD-homolog polypeptide. In further embodiments, the vector provides some regulatory sequences and the nucleotide or gene sequence provides other regulatory sequences. “Regulatory sequences” include but are not limited to promoters, transcription termination sequences, enhancers, splice acceptors, donor sequences, introns, ribosome binding sequences, poly(A) addition sequences, and/or origins of replication.

As used herein the term“homolog” refers to a gene or polypeptide related to another gene or polypeptide by descent from a common ancestral nucleic acid or amino acid sequence. The term homolog may apply to the relationship between genes or polypeptides separated by an event of speciation or to the relationship between genes or polypeptides separated by an event of genetic duplication.

As used herein a“pharmaceutically acceptable carrier” or“excipient” includes any and all antibacterial and antifungal agents, coatings, dispersion media, solvents, isotonic and absorption delaying agents, and the like that are physiologically compatible. A“pharmaceutically acceptable carrier” may include a solid or liquid filler, diluent or encapsulating substance which may be safely used for the administration of the viral vaccine backbones, constructs and/or compositions to a subject. The pharmaceutically acceptable carrier can be suitable for intramuscular, intradermal, intravenous, intraperitoneal, subcutaneous, oral or sublingual administration. Pharmaceutically acceptable carriers include sterile aqueous solutions, dispersions and sterile powders for the preparation of sterile solutions. The use of media and agents for the preparation of pharmaceutically active substances is well known in the art. Where any conventional media or agent is incompatible with the viral vaccine backbone and/or construct, use thereof in the pharmaceutical compositions of the invention is not contemplated. Supplementary active compounds can also be incorporated into the compositions.

Suitable formulations or compositions to administer the viral vaccine backbones, constructs and/or compositions to subjects suffering from viral infection, particularly poxvirus infection, or subjects which are presymptomatic for a condition associated with viral infection fall within the scope of the invention. Any appropriate route of administration may be employed, such as, parenteral, intravenous, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intrathecal, intracistemal, intraperitoneal, intranasal, aerosol, topical, or oral administration.

As used herein the term“subject” includes mammals. Preferably the subjects are ruminants, including both wild and domestic ruminants. Most preferably, the subjects are cattle, sheep and/or goats. In an alternative embodiment, the subject is a human.

For vaccine formulations and pharmaceutical compositions, an effective amount of the viral vaccine backbones, constructs and/or compositions of the invention can be provided, either alone or in combination with other compounds (for example nucleic acid molecules, small molecules, peptides, or peptide analogues), with immunological adjuvants (for example, aluminium hydroxide dimethyldioctadecyl- ammonium hydroxide or Freund’s incomplete adjuvant). The viral vaccine backbones, constructs and/or compositions of the invention may also be linked with a pharmaceutically acceptable carrier and/or other molecules, such as bovine serum albumin or keyhole limpet haemocyanin in order to enhance immunogenicity. The viral vaccine backbones, constructs and compositions of the invention can also be provided in the presence of a liposome, an adjuvant, or any carrier, such as a and in a form suitable for administration to mammals, for example, humans, cattle, sheep, etc.

In some embodiments, the viral vaccine backbones, constructs and/or compositions according to the invention may be provided in a kit, optionally with a carrier and/or an adjuvant, together with instructions for use.

An “effective amount” of a viral vaccine backbone, construct(s) and/or composition(s) according to the invention includes a therapeutically effective amount, immunologically effective amount, or a prophylactically effective amount. A “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as treatment of an infection or a condition associated with such infection. The outcome of the treatment may for example be measured by a decrease in viraemia, inhibition of viral gene expression, delay in development of a pathology associated with poxvirus infection, stimulation of the immune system, or any other method of determining a therapeutic benefit. A therapeutically effective amount of a viral vaccine backbone, construct and/or composition may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the viral vaccine backbone, construct and/or composition to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the viral vaccine backbone, construct and/or composition are outweighed by the therapeutically beneficial effects.

The dosage of any of the viral vaccine backbones, constructs and/or compositions of the present invention will vary depending on the symptoms, age and body weight of the subject, the nature and severity of the disorder to be treated or prevented, the route of administration, and the form of the composition. Any of the compositions of the invention may be administered in a single dose or in multiple doses. The dosages of the compositions of the invention may be readily determined by techniques known to those of skill in the art or as taught herein.

By “immunogenically effective amount” is meant an amount effective, at dosages and for periods of time necessary, to achieve a desired immune response. The desired immune response is preferably a“protective immune response” and may include stimulation or elicitation of an immune response, for instance a T -cell response, particularly in order to prevent a disease caused by a virus.

A“prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired prophylactic result, such as prevention of onset of a condition associated with viral infection, particularly poxvirus infection. Typically, a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount.

Dosage values may vary and be adjusted over time according to the individual need and the judgment of the person administering or supervising the administration of the lumpy skin disease virus knock-out mutants or compositions of the invention. Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected. The amount of viral vaccine backbone or construct in the composition may vary according to factors such as the disease state, age, sex, and weight of the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single dose may be administered, or multiple doses may be administered over time. It may be advantageous to formulate the compositions in dosage unit forms for ease of administration and uniformity of dosage.

The term "preventing", when used in relation to an infectious disease, or other medical disease or condition, is well understood in the art, and includes administration of a composition which reduces the frequency of or delays the onset of symptoms of a condition in a subject relative to a subject which does not receive the composition. Prevention of a disease includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population.

The term "prophylactic or therapeutic" treatment is well known to those of skill in the art and includes administration to a subject of one or more of the compositions of the invention. If the composition is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the subject) then the treatment is prophylactic, i.e., it protects the host against developing the unwanted condition, whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate, or stabilise the existing unwanted condition or side effects thereof).

Toxicity and therapeutic efficacy of compositions of the invention may be determined by standard pharmaceutical procedures in cell culture or using experimental animals, such as by determining the LD 5 o and the ED 5 o. Data obtained from the cell cultures and/or animal studies may be used to formulate a dosage range for use in a subject. The dosage of any composition of the invention lies preferably within a range of circulating concentrations that include the ED 5 o but which has little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilised. For compositions of the present invention, the therapeutically effective dose may be estimated initially from cell culture assays.

The invention also relates in part to a method of prophylactically treating a subject, comprising administering to the subject in need thereof a prophylactically effective amount of the viral vaccine backbones, constructs and/or compositions of the present invention.

The following examples are offered by way of illustration and not by way of limitation. EXAMPLE 1

Construction of recombinant viruses

All recombinant LSDV viruses were made in a similar fashion. Primary lamb testes (LT) cells were infected with a parent LSDV virus (see sections below for specific parent virus used) at an MOI of approximately 0.05 in a 12-well plate which was approximately 70-80% confluent with 10 6 cells per well. After a two hour adsorption period the virus was removed and cells were transfected with 3 or 6pg of the transfer vector using X-tremeGENE™ Hp transfection reagent (Roche, Switzerland) (see different transfer vectors in each section below). Three days later the cells were lysed by three cycles of freezing and thawing, or by the addition of 1 mM Tris, and the lysate used to infect MDBK cells. Individual foci, either fluorescent or not, depending on the recombinant being isolated, were picked and passaged repeatedly. Once the virus population comprised the desired fluorescing or colourless foci, the virus was diluted and single foci were selected in 96 well plates. Single foci were expanded and confirmed to be correct by PCR.

Construction of nLSDVASOD-M

Figure 1 shows how homologous recombination takes place between the transfer vector pHM1 and the nLSDV genome (Accession no: Genbank: AF409138) to generate nLSDVASOD-M. The transfer vector was assembled by overlapping PCR (Figure 2) using primers listed in table 1 . The flanking sequences were PCR amplified from pZG1 and pZG2 respectively (plasmids which contain the LSDV130 and LSDV132 flanks respectively cloned into pGEM®-T easy). The GPT and eGFP genes were PCR amplified from plasmid pZG4 (plasmid containing GPT and eGFP amplified from pRO-2 and cloned into pGEM®-T easy). Three PCR reactions were performed using the KAPA Hifi Polymerase to amplify the left flank, right flank and GPT-eGFP fragments. The cycling conditions comprised an initial denaturation step at 95 °C for 5 minutes, 25 cycles of: denaturation at 95 °C for 30 seconds, annealing at 55°C for 30 seconds for the left and right flank, and at 60°C for the marker genes, and extension at 70°C for 30 seconds for the left and right flank and 2 minutes for the marker genes. The final extension time was 7 minutes. After amplification, the PCR products were gel purified, quantified and used in an assembly PCR reaction. Equimolar amounts of DNA were added into the same tube for the overlapping PCR. The primers used for the assembly reaction were LSDV 130 forward and LSDV 132 reverse. The conditions for the overlapping PCR included an initial denaturation step at 95 °C for 5 minutes, 25 cycles of: denaturation at 95 °C for 30 seconds, annealing at 55°C for 30 seconds and extension at 70°C for 3 minutes. A final extension time of 7 minutes was used. The product from the overlapping assembly PCR was cloned into pJET (ThermoScientific) to produce pHM1 (Figure 3).

For the construction of nLSDVASOD-M primary LT cells were infected with the Neethling vaccine strain of LSDV (OBP, SA) at an MOI of 0.04 in a 12-well plate and transfected with 3 pg of pFIM1 . The virus was passaged in MDBK cells in the presence of 2.5 gg/ml mycophenolic acid (MPA), supplemented with hypoxanthine and xanthine (Sigma-Aldrich, USA) seven times, followed by serial dilution in a 96-well plate. A single green fluorescent focus was expanded and confirmed to be the desired recombinant nLSDVASOD-M by PCR (Figure 7).

TABLE 1 : Primers used to amplify flanking sequences of LSDV 130 and LSDV 132 from pZG1 and pZG2 respectively, and the selection and marker genes from pRO-2.

Construction of nLSDVSODis-M

nLSDVSODis-M was constructed using nLSDVASOD-M as the parent virus and pFIM2v3 as the transfer vector (Figure 4). The eGFP gene was replaced by mCherry, which encodes a red fluorescing protein, and a synthetic SOD-homolog gene, which was designed to have improved stability by removal of repeated nucleotide sequences. Specifically, a series of AT repeats (from position 313 to position 322 of SEQ ID NO:14) was replaced with ATCTACATAC (see position 313 to position 322 of SEQ ID NO:2) (Figure 5 and Figure 6). pFIM2v3 was ordered from GenScript.

For the construction of nLSDVSODis-M, primary LT cells were infected with nLSDVASOD-M and and transfected with 3 pg of pFIM2v3. Forty-eight hours later cells were lysed in 1 mM Tris and the lysate passaged in MDBK cells with no selection. Red fluorescing foci were picked over three successive passages. On the fourth passage, in a 96 well plate, a single red focus was isolated and confirmed upon further passage to be the desired recombinant nLSDVSODis-M (Figure 7). Construction of nLSDV ASOD-UCT

nLSDVASOD-UCT was constructed so as to remove the marker genes and make a recombinant virus which resembles nLSDV in all ways except for the complete deletion of the SOD-homolog gene in nLSDVASOD-UCT when compared to nLSDV. nLSDVASOD-M was the parent virus used to infect cells which were transfected with transfer vector pHMI dSOD (Figure 8).

Primary lamb testes cells were seeded into a 6-well plate at a total of 5x10 5 cells per well. Cells were infected with nLSDVASOD-M at an MOI of 0.05 within two hours of seeding. 24 hours post seeding and infecting, cells were transfected with 3 mI XtremeGeneHP + 6 pg pHM1 -sel-pVV. Six days post transfection a lysate was prepared by two rounds of freeze-thawing (37 °C / -80 °C). Virus from this lysate was passaged in MDBK cells and single non-fluorescing foci were picked. This was done five times before a serial dilution was made in a 96-well plate and a single non fluorescing focus was isolated for large scale preparation of the virus (Figure 9 and Figure 10).

Construction of nLSDV SODis-UCT

nLSDVSODis-UCT is a recombinant virus based on the Neethling vaccine strain of LSDV (OBP, SA), with the SOD-homolog gene completely replaced by the novel SODis gene (SEQ ID NO:2), designed to be more stable than other poxvirus SOD-homolog genes by deleting a series of AT repeated nucleotides as described in the construction of nLSDVSODis-M. The SODis sequence (SEQ ID NO:2) is shown in Figure 6 and was used in the construction of nLSDVSODis-M. No marker genes are present in the recombinant virus nLSDVSODis-UCT.

For the construction of nLSDVSODis-UCT LT cells were infected with nLSDVASOD-M (expressing eGFP) at an MOI of 0.0001 and transfected with 6 pg of transfer vector pHM1 -SODis (Figure 1 1 ). Cells were lysed by three cycles of freezing and thawing 48 hours post infection and transfection. The lysate was passaged in MDBK cells and three rounds of picking of non-fluorescing foci were performed. A single non-fluorescing focus was isolated on a 96-well plate and expanded in MDBK cells. PCR analysis confirmed the recombinant to be nLSDVSODis-UCT (Figure 12). EXAMPLE 2

Genomic Sequencing of Herbivac, nLSDVASOD-UCT and nLSDVSODis-

UCT

Full genomic sequencing has been performed on three LSDV genomes. Herbivac was provided by Deltamune® (batch 008), which had been freeze-dried after growth in primary LT cells. nLSDVASOD-UCT and nLSDVSODis-UCT were constructed and grown in MDBK cells at UCT as described in Example 1 . Virus was purified by centrifugation through a dextran (10 %) sucrose (36 %) gradient, DNA was extracted and sent to the Central Analytical Facility in Stellenbosch for whole genome sequencing using the Ion Torrent method. The genomic sequences for Herbivac, nLSDVASOD-UCT and nLSDVSODis-UCT are provided as SEQ ID NOs: 3, 4 and 5, respectively. Differences between the sequences are listed in Table 2.

Initially, Herbivac was sequenced with the aim of determining what difference in the genome could account for the improved immunogenicity over the Neethling vaccine, observed by Deltamune®. One hundred percent of the genome was sequenced with 400x coverage. When assembled against the South African Neethling strain LSDV LW (Kara et al., 2003) only two single nucleotide changes (SNPs) and one deletion of 2bp were found. The SNPs are located in LSDV genes LW056 (hypothetical protein) and LW1 16 (RNA Polymerase subunit) (Figure 13). Both these SNPs result in conservative amino acid changes which are unlikely to affect the function of the expressed genes. The 2bp deletion is located in ORF131 , encoding a SOD-homolog. In the South African Neethling strain LSDV LW this SOD-homolog ORF (SEQ ID NO:15) is truncated relative to the more virulent LSDV Neethling 2490 strain (SEQ ID NO:13). The South African Neethling strain LSDV LW putative SOD-homolog protein is only 108 aa in length (SEQ ID NO:18) as opposed to 160 aa for the Herbivac SOD-homolog (SEQ ID NO:19) (Figure 14). The 2bp deletion in Herbivac (SEQ ID NO:16) relative to the South African Neethling strain LSDV LW (SEQ ID NO:15) appears to have created a“functional” SOD-homolog gene and this gene is now more similar to that of SOD-homolog from the more virulent Neethling 2490 strain which was sequenced by T ulman in 2001 (SEQ ID NO:13).

Based on this sequence analysis, a novel SOD-homolog gene was designed so as to remove the repeated AT binucleotides which render the gene unstable (SEQ ID NO:2). The novel gene is designated SODis (Superoxide dismutase homolog with improved stability). Figure 6 shows the SODis sequence within the transfer vector pHM2v3. The altered sequence can be seen in enlarged bold lettering. nLSDV was engineered to remove the SOD-homolog completely (nLSDVASOD-UCT) and to replace the SOD-homolog with SODis (SEQ ID NO:2) (nLSDVSODis-UCT). In the process of making these potential LSDV vaccines/vaccine vectors, recombinant viruses were made containing marker genes (nLSDVASOD-M and nLSDVSODis-M). Whole genomic sequences have been determined for nLSDVASOD-UCT (SEQ ID NO:4) and nLSDVSODis-UCT (SEQ ID NO:5). The only significant differences noted between these sequences, Herbivac (batch 008) and the published NI-2490 Neethling vaccine sequence are in the open reading frame encoding the SOD-homolog. Table 2 lists the differences found between the genomic sequences.

Table 2. Differences between LSDV genomic sequences determined (Herbivac, nLSDVASOD- UCT and nLSDVSODis-UCT) and the published sequences for the virulent NI-2490 LSDV and nLSDV LW vaccine strain.

EXAMPLE 3

Induction and inhibition of apoptosis by the SOD-homolog

The effect of the SOD-homolog on apoptosis was investigated. A monolayer of MDBK cells, in a 96-well plate, was infected, at an MOI of 1 , with nLSDV, Herbivac, nLSDVASOD-M or nLSDVSODis-M. 24 hours post infection, apoptotic cell death was measured in the cell lysate using the Cell death detection ELISAPLUS assay (Roche Switzerland). All four LSDVs induced apoptosis in MDBK cells compared to uninifected cells. Herbivac and LSDVSODis-M showed a significant (p>0.001 ) increase in induction of apoptosis as compared to nLSDV and LSDVASOD-M (Figures 15). The SOD-homolog of leporipoxviruses has been shown to inhibit apoptosis. It was hypothesized that the SODis gene would inhibit apoptosis in LSDV-infected cells. MDBK cells were infected with nLSDV, Herbivac, nLSDVASOD-M and nLSDVSODis- M and assayed for apoptosis in cells which were untreated and cells which were treated with camptothecin at 0.0001 mg/ml. Assays were performed 24 hours post infection using the Cell death detection ELISAPLUS assay (Roche, Switzerland) and results expressed as inhibition of camptothecin-induced apoptosis (Figure 16). Herbivac and LSDVSODis-M showed greater inhibition of apoptosis compared to nLSDV and LSDVASOD-M (p>0.001 ).

These experiments indicate that the full-length SOD homolog causes both activation of the extrinsic apoptotic pathway (figure 15) as well as inhibition of the intrinsic apoptotic pathway (Figure 16).

EXAMPLE 4

Inflammatory changes associated with the SOD-homolog gene on chick chorioallantoic membranes (CAMs)

The CAMs of seven day old fertilized chick embryos were either inoculated with PBS (negative control), or infected with nLSDV, nLSDVASOD-UCT or nLSDVSODis- UCT Five days post infection the membranes were harvested, washed, fixed with 4% buffered formalin, embedded, sectioned and stained with haematoxylin and eosin. For each virus-infected membrane five sections were evaluated under the light microscope and scored on a scale from 0 to 5 for inflammation (oedema, haemorrhage and infiltration with white blood cells) compared to the negative control (Figure 17). nLSDVASOD-UCT showed a significant (p>0.01 ) reduction in inflammatory changes compared to both nLSDV and nLSDVSODis-UCT.

EXAMPLE 5

Comparison of lesions produced by different LSDV vaccines in cows

A group of five Friesian calves, 6-12 months old, and LSDV seronegative, were each inoculated at different sites on the neck with a dose of 104 TCID50 of each of nLSDV, Herbivac, nLSDVASOD-UCT and nLSDVSODis-UCT. The animals were monitored daily for two weeks for temperature and lesion size. None of the animals developed serious complications, indicating the two vaccines nLSDVASOD-UCT and nLSDVSODis-UCT are safe for further field trials. Two of the five animals (Animals 166 and 82) developed lesions. Animal 166 developed a larger lesion to nLSDVASOD-UCT compared to the other vaccines. In both of the animals which formed lesions nLSDVSODis-UCT gave lesions smaller than those of nLSDV. This pilot experiment suggests that the SOD-homolog may contribute towards the restriction of LSDV infection in the bovine host, possibly by inducing greater immune cell infiltration to the site of infection.

REFERENCES

Kara, P.D., Afonso, C.L., Wallace, D.B., Kutish, G.F., Abolnik,C., Lu, Z., Vreede, F.T., Taljaard, L.C., Zsak, A., Viljoen, G.J., Rock, D.L. 2003. Comparative sequence analysis of the South African vaccine strain and two virulent field isolates of lumpy skin disease virus. Arch. Virol. 148: 1335-1356.

Tulman, ER, Afonso, CL, Zsak, L, Kutish, GF & Rock, DL, 2001. Genome of Lumpy Skin Disease Virus. J Virol. 2001 Aug;75(15):7122-30