AU728962B2 - Enhancement of immune response using targeting molecules - Google Patents

Enhancement of immune response using targeting molecules Download PDF

Info

Publication number
AU728962B2
AU728962B2 AU64902/98A AU6490298A AU728962B2 AU 728962 B2 AU728962 B2 AU 728962B2 AU 64902/98 A AU64902/98 A AU 64902/98A AU 6490298 A AU6490298 A AU 6490298A AU 728962 B2 AU728962 B2 AU 728962B2
Authority
AU
Australia
Prior art keywords
hig
mice
pci
antigen
dna
Prior art date
Legal status (The legal status 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 status listed.)
Expired
Application number
AU64902/98A
Other versions
AU6490298A (en
Inventor
Jefferey Stephen Boyle
Jamie Louise Brady
Andrew Mark Lew
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Commonwealth Scientific and Industrial Research Organization CSIRO
Walter and Eliza Hall Institute of Medical Research
CSL Ltd
University of Melbourne
QIMR Berghofer Medical Research Institute
Original Assignee
Commonwealth Scientific and Industrial Research Organization CSIRO
Walter and Eliza Hall Institute of Medical Research
Queensland Institute of Medical Research QIMR
CSL Ltd
University of Melbourne
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
Priority claimed from AUPO5891A external-priority patent/AUPO589197A0/en
Priority claimed from AUPP1830A external-priority patent/AUPP183098A0/en
Application filed by Commonwealth Scientific and Industrial Research Organization CSIRO, Walter and Eliza Hall Institute of Medical Research, Queensland Institute of Medical Research QIMR, CSL Ltd, University of Melbourne filed Critical Commonwealth Scientific and Industrial Research Organization CSIRO
Priority to AU64902/98A priority Critical patent/AU728962B2/en
Priority claimed from PCT/AU1998/000208 external-priority patent/WO1998044129A1/en
Publication of AU6490298A publication Critical patent/AU6490298A/en
Application granted granted Critical
Publication of AU728962B2 publication Critical patent/AU728962B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Landscapes

  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Steroid Compounds (AREA)

Description

WO 98/44129 PCT/AU98/00208 1 Enhancement of Immune Response Using Targeting Molecules FIELD OF THE INVENTION The present invention relates to methods of enhancing the immune response to an immunogen and to compositions for use in these methods. In particular the present invention relates to the use of targeting molecules in DNA and protein vaccination.
BACKGROUND OF THE INVENTION The ability of direct injection of non-replicating plasmid DNA coding for viral proteins to elicit protective immune responses in laboratory and preclinical models has created increasing interest in DNA immunisation.
A
useful review of DNA vaccination is provided in Donnelly et al, Journal of Immunological Methods 176 (1994) 145-152, the disclosure of which is incorporated herein by reference.
Intramuscular injection of DNA as a means of vaccination can induce both cellular and humoral responses Studies using reporter proteins demonstrated that muscle cells are the principal target for transfection after intramuscular DNA injection The mechanisms underlying the induction of immune responses after DNA immunisation are unclear. Since myocytes express MHC Class I at low levels and do not constitutively express Class II or costimulatory molecules such as B-7 they appear unlikely candidates for the induction of Ab or CTL responses. It is possible that low level transfection of antigen presenting cells (APCs) occurs at the injection site and these APCs then traffic to lymphoid organs and present the encoded antigen to B and T cells as has been shown after intradermal and biolistic DNA immunisation Alternatively the myocyte may act merely as a source of antigen and priming occurs in the draining lymph node. In the latter case, optimum immune induction would result if the antigen was released from the myocyte by secretion or subsequent to cell damage.
One strategy that has been shown to augment the response to polynucleotide, or DNA, vaccination is the use of sequences encoding cytokines or co-stimulatory molecules (Conry et al, (1996) Gene Therapy 3: 67-74). These investigators showed an increased response when the DNA administered encoded not only the antigen of interest but also for B7-1.
The present inventors investigated the effects of modifying the antigen such that it will be targeted to APC or sites of immune induction. This was shown to not only markedly enhance the immune response but also cause immune deviation.
SUMMARY OF THE INVENTION In a first aspect the present invention consists in a DNA molecule for use in raising an immune response to an antigen, the DNA molecule including a first sequence encoding a non-antibody targeting molecule in which the targeting molecule is a mammalian cell surface receptor counter receptor, a second sequence encoding the antigen or an epitope thereof, and optionally a third sequence encoding a polypeptide which promotes dimerisation or multimerisation of the product encoded by the DNA molecule.
As will be appreciated by those skilled in the art in a number of instances the antigen or epitope encoded by the second sequence will be a 20 polypeptide which promotes dimerisation or multimerisation of the encoded product. As will be understood in such instances the third sequence may be omitted.
In a second aspect the present invention consists in a polypeptide, the 00 polypeptide being encoded by the DNA molecule of the first aspect of the invention.
In a third aspect the present invention consists in a method of raising an immune response in an individual, the method comprising administering to the individual the DNA molecule of the first aspect of the present invention or the polypeptide of the second aspect of the present invention.
There are a wide range of molecules which could be used as targeting molecules. These include ligands which target lymphoid cells (which will either be at or take the Ag to sites of immune induction), lymphoid sites (eg.
spleen, lymph nodes, Peyers patches) or APCs directly. Examples of such ligands include, but are not limited to, CD40L, OX40, CD28, CTLA4 and L-selectin. It is presently preferred that the targeting molecule is CTLA4 or L-selectin.
WO 98/44129 PCT/AU98/00208 3 In a fourth aspect the present invention consists in a method of deviating the immune response to an antigen in an individual, the method comprising administering to the individual a DNA molecule including a first sequence encoding CTLA4, a second sequence encoding the antigen or an epitope thereof, and optionally a third sequence encoding a polypeptide which promotes dimerisation or multimerisation of the encoded product.
There are many ways of producing dimerisation or multimerisation including tandem duplication and the use of any molecule that normally forms multimers Immunoglobulins, CD8, TNF, glutathione s-transferase, zinc finger dimers etc). There are many references in the scientific literature regarding this area. These include Classon BJ et al (1992) "The hinge region of the CD8 alpha chain: structure, antigenicity, and utility in expression of immunoglobulin superfamily domains" Int Immunol 4:215-25; Yang J, Moyana T, Xiang J (1995) "A genetically engineered single-chain FV/TNF molecule possesses the anti-tumor immunoreactivity of FV as well as the cytotoxic activity of tumor necrosis factor." Mol Immunol.
32:873-81; Tudyka T, Skerra A (1997) "Glutathione s-transferase can be used as a c-terminal, enzymatically active dimerization module for a recombinant protease inhibitor, and functionally secreted into the periplasm of Escherichia coli." Protein Science. 6:2180-2187; Pomerantz JL, Wolfe SA, Pabo CO (1998) "Structure-based design of a dimeric zinc finger protein" Biochemistry 37:965-970; and Whiteheart SW, Rossnagel K, Buhrow SA, Brunner M, Jaenicke R, Rothman JE (1994) "N-ethylmaleimide-sensitive fusion protein: a trimeric ATPase whose hydrolysis of ATP is required for membrane fusion." J Cell Biol 126:945-54. The disclosure of these references and the other references referred to in this application are included herein by cross-reference.
As will be appreciated by those skilled in the art in the constructs of the present invention the first, second and third DNA sequences may be in any particular order. It is presently preferred that the order is first, third then second.
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
WO 98/44129 PCT/AU98/00208 4 DETAILED DESCRIPTION OF THE INVENTION In order that the nature of the present invention may be more clearly understood preferred forms thereof will now be described with reference to the following examples and Figures in which: Figure 1. Secretion of AIg, CTLA4Ig and L-SELIg proteins from NIT transfectants. NIT cells were transfected with the pRep7::mCTLA4-hlg and pRepl0::hL-SEL-hIg expression plasmids. Secreted protein was purified on immobilised protein A and samples run by SDS PAGE under reducing and non-reducing conditions.
Figure 2. hIg specific IgG responses in DNA immunized mice. Sera were obtained from BALB/c mice immunized with pRep7::mCTLA4-hlg and pReplO::hL-SEL-hIg at the indicated times post immunisation and stored at -20 0 C until assayed for hIg specific IgG in an ELISA. Titres were defined as the highest dilution to give a 0.2 OD at 450 nm. Results are expressed as the mean of the log titre ±SEM from 5 mice in each group. Normal mouse sera and hyperimmune mouse sera served as the negative and positive controls respectively.
Figure 3. hIg specific IgG subclass responses in DNA immunized mice. A. Sera were obtained from BALB/c mice immunized with pRep7::mCTLA4-hIg and pRepl0::hL-SEL-hIg at 8 weeks post immunisation and stored at -20°C until assayed for hIg specific IgG1, IgG2a or IgG2b in an ELISA. Titres were defined as the highest dilution to reach an OD of 0.2 at 450 nm. Results are expressed as the mean of the log titre ±SEM from 5 mice in each group. B. The log IgG1 titre for each mouse was divided by the corresponding log IgG2a titre to obtain a log IgGi: log IgG2a ratio. Results are expressed as the mean SEM from 5 mice in each group.
Figure 4. hlg specific IgG subclass responses in soluble protein immunized mice. Sera were obtained from BALB/c mice immunized with pig of hlg or 5 ptg of CTLA4Ig protein in 100 pl of PBS, 2 weeks post immunisation and assayed for hIg specific IgG in an ELISA. Titres were defined as the highest dilution to reach an OD of 0.2 at 450 nm. Results are expressed as the mean of the log titre ±SEM from 5 mice in each group.
WO 98/44129 PCT/AU98/00208 Figure 5. hIg specific IgG subclass responses in CTLA4Ig DNA immunized mice. Sera were obtained from BALB/c mice immunized with the indicated dose of pRep7::mCTLA4-hIg 2 weeks post immunisation and assayed for hlg specific IgG1, IgG2a or IgG2b in an ELISA. Titres were defined as the highest dilution to reach an OD of 0.2 at 450 nm. Results are expressed as the mean of the log titre ±SEM from 5 mice in each group.
Figure 6. OVA specific IgG and IgG subclass responses after co-injection of DNA. Sera were obtained from BALB/c mice immunized with pReplO::hL-SEL-hlg and pCI-OVA or pRep7::mCTLA4-hIg and pCI-OVA at 4 weeks post immunisation and assayed for OVA specific IgG or IgG1, IgG2a or IgG2b in an ELISA. Titres were defined as the highest dilution to reach an OD of 0.2 at 450 nm. Results are expressed as the mean of the log titre SEM from 5 mice in each group.
Figure 7. Shows stimulation index with hIg, Lsel-hIg and CTLA4-hIg HuIg lmg/ml; I HuIg lmg/ml; I Hulg 1mg/ml) Figure 8. Shows anti-ovalbumin IgG titres with various constructs 2 weeks; 4 weeks) Figure 9. Shows anti-OVA IgG titres with pCI::mCTLA4-g3h-OVA and pCI: :mCTLA4-hIg-OVA Figure 10. Groups of 5 Balb/c mice were vaccinated intramuscularly on days 0 and 28 with 0.1mg of pCI::mCTLA4-hIg-45W (black circles) or pCI::CD5L-hIg-45W (grey circles). Mice were bled on days 0, 7, 14, 28, and 42. Sera was assayed for anti-45w antibodies by ELISA using recombinant 45W(His)6. The Student's t-test was used to compared the two groups and the probability values for the two vaccines at each time point are shown at the top of the figure.
Figure 11. Groups of 5 Balb/c mice were vaccinated either intraperitoneally with 20pg of recombinant 45w(His)6 protein (grey circles in Freund's complete adjuvant or intramuscularly with 0.1mg pCI::mCTLA4-hIg-45W (black circles) in 0.lml of saline. Mice were bled on days 0, 2 ,5 ,8 14 and 28 post-vaccination and anti-45w antibodies measured by ELISA using 45W(His)6 protein. The responses were compared by Student's t-test and were different at day 8 (p<0.05).
Figure 12. Shows survival of mice following challenge with Plasmodium chabaudi adami DS (i pCI::CTLA4-hlg-AMA; pCI::CTLA4-hIg).
WO 98/44129 PCT/AU98/00208 6 Figure 13 shows antibody titres.
Figure 14 shows antibody titres at day 14 with varying immunisations Figure 15 shows antibody titres at day 54 with varying immunisations Figure 16 shows lung virus titres Figure 17. Kinetics of induction of the anti-hIg antibody titres post-immunisation. Filled symbols represent plasmids containing hIg: filled squares (pCI::bCTLA4-hIg-APLD), filled circles (pCI::CD5L-hlg-APLD), filled triangles (pCI::bCTLA4-hIg). Open symbols represent control animal groups not injected with hIg in any form: open circles (pCI::APLD), open squares (Unvaccinated controls) and open triangles (Glan-Vac).
Figure 18. Western Blot of PLD expressed by eukaryotic and prokaryotic cells. Lane 1-3. Supernatant from Cos-m6 cells transfected with pCI::PLD (lane pCI::APLD (lane 2) and pCI alone (lane Lane 4. Cell filtrate containing PLD expressed from Corynebacterium pseudotuberculosis Figure 19. Protection from challenge with Corynebacterium pseudotuberculosis. Percentage of the animals protected from challenge by 106 CFU of Corynebacterium pseudotuberculosis injected just above the coronet. Protection was defined as the animal not having abscesses in any of the following lymph nodes: popliteal, inguinal and prefemoral both left and right.
Figure 20 Shows number of mice with lesions at various time points after challenge with L. major (D PBS; A pCI::CD5L-hIg-PSA2; pCI::mCTLA4-hIg-PSA2; A pCI::PSA2) EXAMPLE 1 Materials and Methods Mice Female mice (BALB/c, CBA and C57B1/6) aged 6 to 8 weeks were used in all experiments. Mice were maintained in SPF conditions.
Plasmids and immunisations Expression plasmids were constructed to produce secreted forms of the Fc fragment of human IgG1 (AIg) by using the Cd5 leader sequence either alone or fused with murine CTLA4 (mCTLA4Ig) or human WO 98/44129 PCT/AU98/00208 7 L-selectin (hL-SELIg) under the control of the RSV promoter in the Rep7 or vectors (these vectors differ only in the direction of the multiple cloning site, Invitrogen, San Diego, CA, USA). The sequence of pREP7::CTLA4-hlg is shown in Sequence ID No. 1 Promoter RSV: 13-640, CTLA4-hIg: 703-2462. The constructs were obtained from plasmids given by Drs. P. Lane (Basel Institute, Switzerland), B. Seed (Massachusetts General Hospital, Boston, USA) and D. L. Simmons (Institute of Molecular Medicine, Oxford, UK). The following constructs were generated: pRep7::mCTLA4-hlg pRepl0::hLSEL-hlg Plasmids for injection were prepared fromE. coli by PEG precipitation as described except that volumes of Solution I, II and III were adjusted such that pellets were resuspended in 50 mL of Solution I for each Litre of broth media used. Endotoxin was removed from plasmid preparations by four Triton X-114 phase separations and DNA was stored at-20 0 C in normal saline until injected. The resultant plasmid preparations contained less than 10 IU endotoxin per mg of plasmid DNA as determined by the limulus amoebocyte lysate assay (QCL -1000 BioWhittaker, Walkersville, MD, USA). Mice received 100 gg of plasmid DNA in both quadriceps or intradermally at the base of the tail on day 0 and 14 of each experiment.
Antibody assays Microtitre plates (Dynatech, Chantilly, VI, USA) were coated with human Ig (hIg) protein (Intragam, CSL, Parkville, Australia; 10 Pg/ml in PBS) by overnight incubation at 4 0 C and washed four times with PBS to remove unbound antigen. Plates were incubated with serially diluted sera in blocking buffer milk powder in PBS) overnight at 4C. After washing times with PBS to remove unbound Ab, plates were incubated with peroxidase conjugated anti-mouse IgG, IgG1, IgG2a or IgG2b antibodies (Southern Biotechnology, Birmingham, AL, USA) diluted in blocking buffer.
After washing five times with PBS, the amount of bound Ab was determined by addition of substrate solution (0.1 mg/ml 3,3,5,5-tetra methylbenzidine (T2885, Sigma St. Louis, MO, USA) 0.03% H202 in 0.1M Na acetate pH The reaction was stopped with 1M H2S0 4 and the OD read at 450nm. Titres were defined as the highest dilution to reach an OD of 0.2.
WO 98/44129 PCT/AU98/00208 8 To calibrate the IgG subclass ELISA, plates were coated with IgG1, IgG2a or IgG2b from mouse myelomas (10 4g/ml in 0.5 times PBS) overnight at 4 0 C, washed 3 times with PBS and then incubated with serially diluted anti-mouse IgG subclass HRP conjugated Ab. The dilution of each anti-mouse subclass Ab which gave identical absorbances in the ELISA were used subsequently.
Results and Discussion Expression plasmids were constructed to produce secreted forms of the human IgG1 heavy chain alone (pReplO::CD5L-hIg) or fused with CTLA4 (pRep7::mCTLA4-hIg) or L-selectin (pReplO::hLSEL-hIg). Cells transfected with these plasmids secreted the three molecules as disulphide linked dimers of expected size (Fig. Like others we were unable to detectin vivo protein expression by western blotting of muscle homogenates and or of protein A purified material from sera of B-cell deficient immunized mice (data not shown). However, the ability to detect immune responses in immunized mice is indicative of in vivo expression. No immune responses to human Ig were detected in unimmunised or mice receiving vector only (data not shown). However, mice immunized with pReplO::hL-SEL-hIgAIg or pRep7::mCTLA4-hlg had markedly different responses (Fig. Responses in pRep7::mCTLA4-hlg immunized mice were more rapid and of greater magnitude at all three time points: 2, 4 and 8 weeks (Fig. At 4 weeks both the pRep7::mCTLA4-hlg and pReplO::hLSEL-hIg immunized mice had 1000 and 100 fold higher IgG responses than pRepl0::CD5L-hIg controls respectively. The differences observed were not attributable to any adjuvant effects of endotoxin, because Triton X-114 was used to remove endotoxin so that the levels were 10IU/mg plasmid DNA. Similar results have been achieved in 3 experiments using BALB/c and CBA mice (data not shown).
The response in all mice to pReplO::CD5L-hIg was dominated by IgG2a (Fig. which mimics a viral infection, and has been reported for other antigens after DNA immunisation 10). The IgG subclass response in pReplO::hLSEL-hIg immunized mice was similar (although greater) to pReplO::CD5L-hIg controls whereas the pRep7::mCTLA4-hIg response was deviated to an IgG1 dominance (Fig. 3B). The possibility that the differences WO 98/44129 PCT/AU98/00208 9 in Ab responses was due to dose was unlikely since all constructs were made with identical plasmid backbone and mice immunized with soluble CTLA4Ig protein (Fig. 4) had higher Ab responses than those receiving an equivalent dose of hlg. Also, to determine if the IgG1 dominance of the response to pRep7::mCTLA4-hIg was due to dose we immunized mice with different amounts of pRep7::mCTLA4-hIg so that mice with total IgG antibody levels could be compared to that of pReplO::hLSEL-hlg (Figs. 2 and The IgG1 predominance was found at all doses of pRep::mCTLA4-hIg (Fig. Work with CTLA4 has demonstrated it can bind to B-7 and block co-stimulation which reduces the response to other immunogens
A
non-specific immunomodulatory effect of CTLA4 was unlikely for several reasons. Firstly, CTLA4Ig protein at least in high doses (and hence B-7 is blocked) has been ascribed immunosuppressive properties not immunostimulating ones (11) as we found for DNA and protein immunisations. Furthermore, mice co-injected with CTLA4Ig and DNA encoding ovalbumin (pCI-OVA) had similar ovalbumin specific IgG and IgG subclass titres to control mice (Fig. 6) indicating that there was not any immunosuppressive effect of CTLA4.
Example 2 Use of targeting ligand to augment T cell proliferative responses and requirement for dimerisation Introduction In Example 1 there is a demonstration that Ab levels to a model DNA vaccine could be enhanced when antigen was fused with the targeting ligands CTLA4 or L-selectin.
The hIg component would ensure dimerisation which we thought would be favourable because in general, binding of ligands to receptors is stronger when dimers are used. However, it was unclear in this system if dimerisation of the antigen targeting ligand fusion proteins was necessary for increased immune responses. To determine if the enhanced Ab response generated by antigen targeting vectors encoding proteins was dependent upon dimer formation, Ab responses were compared to immunisation with plasmid encoding monomeric antigen targeting ligand fusion proteins. The WO 98/44129 PCT/AU98/00208 hIg component of the vectors was replaced with coding sequence for another model antigen that would not form dimers (ovalbumin;
OVA).
Materials and Methods Female mice aged 6 to 8 weeks were used in all experiments and maintained in SPF conditions.
After PCR amplification to include an Mlu I restriction enzyme recognition sequence, the OVA cDNA (bp 470-1170) was inserted behind the human immunoglobulin Fc (hlg) gene via a 4 amino acid glycine linker at the Nsi I site. These vectors would form dimers due to the interchain disulfide bonds of hlg and are represented by an hIg-OVA suffix. A targeting vector that would not form dimers was obtained by direct fusion of the cDNA from OVA to the cDNA of CTLA4 (pCI::mCTLA4-OVA) or to the leader sequence of CD5 as a control (pCI::CD5L-OVA). After PCR amplification to include Hind III and Nsi I restriction sites the entire hig component of pCI::mCTLA4-hIg-OVA was replaced with the human IgG3 hinge region (a gift from Dr Y Akahori, Japan) to form pCI::mCTLA4-g3h-OVA. Plasmids for injection were prepared from E. coli with endofree QIAGEN maxi kits according to the manufacturer's instructions and stored at-20 0 C in normal saline until injected. Mice received 50 ug of plasmid DNA in 100 pl normal saline i.m. in both quadriceps at day 0 of each experiment.
The proliferation of 2 X 105 splenocytes was determined by a standard 5 day 3 H-thymidine uptake protocol at 6 weeks post initial immunisation.
The mean stimulation index was calculated as the cpm with antigen cpm splenocytes alone.
Microtitre plates (NUNC, Maxisorb) were coated with OVA protein (A-5503, Sigma, St. Louis, MO; 10 pg/ml in PBS) by overnight incubation at 4 0 C and washed four times with PBS to remove unbound antigen. Plates were incubated with serially diluted sera in blocking buffer casein in PBS) overnight at 4°C. After washing 5 times with PBS to remove unbound Ab, plates were incubated with peroxidase conjugated anti-mouse IgG (Southern Biotechnology, Birmingham, AL) diluted in blocking buffer. After washing five times with PBS, the amount of bound Ab was determined by addition of tetramethylbenzidine substrate solution. The reaction was WO 98/44129 PCT/AU98/00208 11 stopped with 1M H2S04 and the OD read at 450nm. Titres were defined as the highest dilution to reach an OD of 0.2.
Results and Discussion The proliferation splenocytes was determined by a standard 5 day 3H-thymidine uptake protocol at 6 weeks post initial immunisation (Fig. 7).
The stimulation index was calculated as the cpm with antigen cpm splenocytes alone. The mean ±SD from 3 mice in each group is shown after incubation with three different antigen concentrations. Mice immunized with the DNA constructs pCI::mCTLA4-hIg and pCI::Lsel-hlg had 8 and 3 fold higher T cell proliferative responses than controls respectively This data suggested that the targeting of antigen was having an enhancing effect on T cell activation.
Groups of 8 mice were immunized with DNAs expressing the monomeric targeting vector pCI::mCTLA4-OVA, the monomeric control or the dimeric vectors pCI::CD5L-hIg-OVA (control), pCI::Lsel-hIg-OVA or pCI::CTLA4-hlg-OVA on day 0 and bled 2 and 4 weeks post immunisation. The OVA specific IgG levels were determined by ELISA.
The results obtained at 2 and 4 weeks post immunisation are illustrated in Fig. 8 (2 weeks; hatched columns, 4 weeks; solid columns). There was no difference in Ab levels at 2 or 4 weeks between the mice immunized with or pCI::mCTLA4-OVA monomeric DNA vectors. The highest Ab responses were obtained with the pCI::mCTLA4-hlg-OVA vector, which forms dimers, compared to the monomeric (pCI::CD5L-OVA) or dimeric (pCI::CD5L-hIg-OVA) controls.
Surprisingly, the pCI::Lsel-hIg-OVA immunized mice had the poorest responses at both time points. This data is in contrast to the enhanced responses to hIg when fused with L-selectin alone. The observation that the responses obtained with pCI::Lsel-hlg-OVA were similar in magnitude to those obtained with the monomeric antigen fusions suggests that the fusion of OVA (or other antigens) to Lsel-hIg may interfere with the efficiency of binding of L-selectin to its ligand by interfering with dimerisation, by allosteric effects or by conformational changes to L-selectin). Alternative ways of fusion should be investigated.
WO 98/44129 PCT/AU98/00208 12 Overall, these results suggest that for effective antigen targeting the inclusion of a molecule such as hlg that facilitates dimerisation is essential unless the antigen itself facilitates dimerisation or multimerisation. This data was obtained using the hinge, CH2 and CH3 domains of human IgG1.
The dimerisation of hIg is facilitated by disulfide bonds between cysteine residues in the hinge domains. To determine if another molecule that would also facilitate dimerisation could replace the hIg component, the hinge region of human IgG3 was used to link mCTLA4 with OVA. Groups of 8 mice were immunized with DNAs expressing the targeting vectors pCI::mCTLA4-hIg-OVA or pCI::mCTLA4-g3h-OVA. At 2 weeks post immunisation sera was collected and shown to contain similar levels of anti-OVA antibodies (Fig. Therefore, this suggests that it may be possible to reduce the hIg component of the antigen targeting vectors to a hinge region alone or replace the hIgG1 component with another immunoglobulin hinge region, another molecule or part thereof such as the hIgG3 hinge to facilitate dimerisation. This would be of particular applicability when the targeting ligand is L-selectin or another molecule that does not dimerise or is structurally compromised by fusion of antigen via hIg.
Example 1 demonstrated an increased immune responses to hIg after DNA and protein immunisation was obtained with targeting ligand-hIg fusions. The following Examples were conducted to determine if antigens other that hIg could be used for increased immune responses. These data were obtained by the addition of antigens to the C-terminus of hIg which could facilitate dimer formation as was found with hIg alone. A gly-gly-gly-gly-thr spacer was introduced between hIg and the antigens.
Whilst these constructs have been used it will be appreciated that responses may be improved by routine optimisation. This optimisation may involve modification of the constructs as envisaged within the present invention eg different targeting molecules, different sequences which facilitate multimerisation, different linkers etc.
WO 98/44129 PCT/AU98/00208 13 EXAMPLE 3 Use of CTLA4 to accelerate immune responses against the host protective antigen of Taenia ovis known as Introduction The 45W antigen is a putative membrane glycoprotein present in, or underlying the tegument of, the Taenia ovis oncosphere. T. ovis is a pathogen of sheep which causes commercial losses of mutton and wool in New Zealand and other important sheep growing countries. Early immunisation studies using 45W protein partially purified from T. ovis revealed that it was a promising vaccine antigen. Subsequent field trials using recombinant forms of 45W, expressed in Escherichia coli, as a vaccine reported very high levels (about 95%) of protection The 45W antigen was used as a DNA vaccine in sheep and low levels of antibody, measured using a recombinant form of 45W, was observed 4 Materials and Methods Plasmids containing the CMV promoter and the genes encoding CTLA4, the Fc portion of human immunoglobulin (hIg) and the CD5 signal peptide were described above. The gene encoding 45W was obtained from Dr. Marshall Lightowlers (Dept. Veterinary Science, University of Melbourne).
Inbred Balb/c mice of 6-8 weeks of age were obtained from the Dept.
Microbiology and Immunology Animal House, University of Melbourne.
Standard DNA manipulation and CsCl purification techniques were used. The gene encoding 45W was ligated into two DNA vaccines.
Construct pCI::mCTLA4-hIg-45W expressed a fusion protein which comprised the CTLA4 signal peptide, mouse CTLA4 ectodomain, hIg and the antigen. Construct pCI::CD5L-hIg-45W expressed a fusion protein which contained the signal peptide from CD5, hIg and the 45W antigen.
DNA (100g) was injected into the quadriceps of mice on days 0 and day 28. Sera was obtained at appropriate intervals post vaccination and analysed for total antibodies specific for recombinant 45W antigen in a WO 98/44129 PCT/AU98/00208 14 titration ELISA" 5 using horseradish peroxidase conjugated anti-mouse immunoglobulins.
Purified recombinant 45W(His) 6 was obtained from E. coli according to Rothel et a.
14 using a polyhistidine 'tag' and nickel affinity chromatography.
Results Mice were immunised with the DNA vaccines pCI::mCTLA4-hIg-45W, or pCI::CD5L-hIg-45W (Figure 10). Mice received 100ug of DNA on days 0 and 28 and were bled at weekly intervals. Mice receiving the DNA vaccine which expressed CTLA4 fused to the hIg/45W vaccine developed a more rapid antibody response than the mice which received a similar plasmid vaccine construct ie. pCI::CD5L-hlg-45W which did not contain the CTLA4 gene. The mice receiving the vaccine with CTLA4 produced serum antibodies of high titre (ie.>10,000) on days 7, 14 and 28. In comparison, mice which received the construct lacking CTLA4 did not produce high titre antibodies (ie. titre >10,000) until after the second immunisation on day 28.
All mice(ie. 5/5) which received the DNA vaccine containing CTLA4 produced 45W-specific antibodies by 7 days post immunisation whereas only 1/5 animals which received the equivalent DNA vaccine lacking CTLA4 produced antibodies at day 7 post immunisation. The data was analysed using Student's t-test.
A second trial was undertaken where mice received either 204g of purified recombinant 45W(His) 6 protein in Complete Freund's Adjuvant, or the CTLA4 DNA vaccine (ie. pCI::mCTLA4-hIg-45)(Figure 11). The serum antibody response was examined on days 0, 2, 5, 8, 14 and 28. The serum antibody response specific for 45W was higher at day 8 in DNA vaccinated mice than in mice which received the 45W protein vaccine.
Discussion The murine serum antibody response to 45W DNA vaccination was accelerated by fusion of CTLA4 to the hIg-45W fusion protein. The antibody response to 45W correlates with protection in sheep against T. ovis disease.
Addition of CTLA4 led to a more rapid high titre response, with a shorter WO 98/44129 PCT/AU98/00208 unprotected period following immunisation. The effect of CTLA4 on the magnitude of the anti-45W response was not as dramatic as the effect on human Ig described above. This may have been due to the conformational restraints from the fusion of the various molecules or some inherent property of the 45W antigen. Furthermore, the immunisation protocol employed in this Example differed from Example 1 in that boosting occurred at 4 rather than two weeks. Due to the more rapid kinetics of the response via CTLA4 targeting boosting may not have been optimum and thus the magnitude was not effected.
Example 4 Use of CTLA4 with AMA1 to protect against Plasmodium chabaudi adami in mice Introduction AMA1' 6 is a candidate vaccine antigen against malaria. We have evidence that domain3 of AMA1 folds independently and as such may be a good candidate in producing a fusion protein with hlg. However, although AMA1 has been shown to confer protection in mouse malaria we are unaware of any work that has shown domain3 to be protective.
Materials and Methods Plasmids containing the CMV promoter and the genes encoding CTLA4, the Fc portion of human immunoglobulin (hIg) and the CD5 signal peptide were described above. Domain 3 of AMA-1 from the Plasmodium chabaudi adami DS strain 16 was fused to CTLA4Ig and (pCI::mCTLA4-hIg-AMA and pCI::CD5L-hlg-AMA). The plasmid pCI::mCTLA4-hlg was used as a negative control.
Inbred female Balb/c mice of 6-8 weeks of age were used.
DNA (100ug) was injected into the quadriceps of mice on day 0 only.
Mice were challenged with Plasmodium chabaudi adami DS and the number of deaths recorded.
WO 98/44129 PCT/AU98/00208 16 Antibody titres were measured by ELISA using refolded E.coli expressed entire ectodomain of AMA11 7 The titres were expressed as the log of the reciprocal of the last serum dilution to give an OD >0.1.
Results In the first trial, there were 8 mice per group. A single immunisation with the DNA vaccine pCI::mCTLA4-hlg-AMA afforded partial protection against an intraperitoneal challenge of 100,000 parasites (Figure 12).
Challenge was performed 14 days after immunisation. This was significant (log rank test; p<0.05) from the control pCI::mCTLA4-hIg group. There was a clear indication that the CTLA4 conferred better protection than the group, although this did not reach statistical significance. The antibody titres (Figure 13) show that the CTLA4 targeting ligand enhances the antibody response to AMA1 (p<0.005).
A second trial was undertaken with larger group (16/group) and with an intravenous challenge of 10,000 parasites. No protection was seen in this second trial.
Discussion In the first trial, CTLA4 conferred some protection against malaria by domain3 of antigen AMA1. This was not found in the second trial. We do not know why there was a difference between the two trials. Because there is not a full set of antibody data for comparison, we do not know whether the level of antibody achieved was sufficient in the mice that died in trial two, or whether the effect was due the different route of challenge. We also do not know how effective CTLA4IgAMA may be when booster doses are given.
Example Use of CTLA4 in influenza infection in mice Introduction As an additional model for testing protective efficacy we have used influenza infection of the murine respiratory tract. The influenza WO 98/44129 PCT/AU98/00208 17 haemagglutinin (HA) gene was cloned behind targeting molecules and the resulting DNA vaccines examined for their ability to generate higher anti-viral antibody titres and afford greater protection against live viral challenge compared to control vaccines not expressing the targeting molecule.
Materials and Methods Virus The type A influenza virus used in this study was PR8 A/Puerto Rico/8/34 (H1N1). Virus was grown in the allantoic cavity of embryonated hens' eggs for 2 days at 350C. The allantoic fluid was collected and clarified by centrifugation (2000g, 15 mins, 40C). Aliquots of allantoic fluid containing infectious virus were stored at -700C and used for immunisation and challenge of mice. Purified PR8 virus used in ELISA assays was obtained as zonally purified stocks from CSL Ltd, Parkville, Victoria, Australia. The haemagglutination assay 18 was used to quantitate virus and titres are expressed in haemagglutinating units (HAU) per ml.
Immunisation Groups of 10 BALB/c mice were immunised intramuscularly under anaesthesia on day 0 with 0.1 ml of DNA vaccine containing 504g of plasmid. The constructs used for immunisation were pCI::mCTLA4-hIg-HA, which were based on PR8 HA lacking the signal and transmembrane sequences, and pCI::CD5L-hIg-SIINFEKL (expressing an 8 amino acid sequence from chicken ovalbumin) as a negative control. As a positive control, a group of 10 BALB/c mice were infected intranasally with 50 plaque forming units (pfu) of infectious PR8 virus and another group immunised subcutaneously with 1ug of P-propiolactone (BPL)-inactivated and sodium taurodeoxycholate-disrupted PR8 virus (split virus). Serum samples were collected from all mice on days 7, 14 and 54 and mice were then challenged on day Intranasal challenge of mice and preparation of mouse lung extracts Penthrane anaesthetised mice were challenged with 50 pfu of infectious PR8 influenza virus Each mouse received 50 l of virus in the WO 98/44129 PCT/AU98/00208 18 form of allantoic fluid diluted in phosphate buffered saline (PBS). Five days after challenge, mice were killed by cervical dislocation and lungs were removed and transferred aseptically to bottles containing 1.5 ml Hanks Balanced Salt Solution (HBSS), supplemented with 100 U of penicillin per ml, 100 jAg of streptomycin per ml and 30 Ag of gentamicin per ml. Lung homogenates were prepared using a tissue homogeniser. Each lung suspension was then centrifuged at 300 x g for 5 minutes and the supernatants were removed, aliquoted and stored at-70 0 C prior to assay for infectious virus.
Enzyme-linked immunosorbent assay The enzyme-linked immunosorbent assay (ELISA) was performed as previously described by Jackson et al.19 using 96-well polyvinyl microtitre trays (Dynatech, Australia) coated with a solution containing 50 HAU of purified PR8 virus per well. Antibody titres are expressed as the reciprocal of the antibody dilution giving an absorbance of 0.2 units.
Plaque assay for infectious virus Virus titres were determined by plaque assay on monolayers of Madin-Darby canine kidney (MDCK) cells in 35 mm petri dishes (Nunc, Roskilde, Denmark). The culture medium was RPMI-1640 supplemented with 2 mM glutamine, 2 mM sodium pyruvate, 100 U of penicillin per ml, 100 Ag of streptomycin per ml, 30 Ag of gentamicin per ml and 10% (vol/vol) foetal calf serum (heat inactivated at 56 0 C for 30 min). Monolayers were washed with serum-free RPMI-1640 containing antibiotics and inoculated in duplicate with 100 .1 of dilutions of lung homogenates in the same medium.
After allowing 45 minutes in a humidified incubator (37 0 C, 5% C0 2 for virus adsorption, 3 ml of agarose overlay medium at 45 0 C was added.
Incubation was continued for a further 3 days and plaques counted. The agarose overlay medium was Leibovitz L-15 medium pH 6.8 (Gibco Laboratories, supplemented with 100 U penicillin per ml, 100 /g of streptomycin per ml, 0.01 M HEPES buffer pH 6.8 (Calbiochem, Australia), 0.1% trypsin-TPCK (Worthington, Biological Systems Inc., and 0.9% agarose (ICN Biomedicals Sydney, Australia).
WO 98/44129 PCT/AU98/00208 19 Statistical analysis The data were analysed using the nonparametric Mann-Whitney U test which compares two sets of unpaired samples. The null hypothesis is that the two population medians are equal and the resultant P value for particular comparisons is given.
Results and Discussion Serum antibody responses of mice Sera collected from mice immunised with the DNA constructs, split PR8 virus in PBS, or infectious PR8 virus were assayed for anti-viral antibody by ELISA. On day 7 after priming, only mice immunised with infectious PR8 virus exhibited anti-viral antibody titres significantly higher than the background titres detected in mice given the control DNA construct pCI::CD5L-hIg-SIINFEKL (p=0.0002). However by day 14, in addition to the high antiviral antibody titres detected in the virus infected mice, antibody titres of mice immunised with the pCI::mCTLA4-hlg-HA construct were significantly higher than those of mice given the control DNA constructs (p=0.0003) or pCI::CD5L-hIg-SIINFEKL (p=0.0004) (Fig.
14). Furthermore, mice immunised with the pCI::mCTLA4-hIg-HA construct had comparable levels of antiviral antibody to mice given the split virus vaccine The antiviral antibody titres of sera collected on day 54 post-priming were also determined (Fig 15). Overall, the day 54 titres were similar to those measured in sera collected on day 14, and the level of antibody in the day 54 sera of mice given the pCI::mCTLA4-hlg-HA construct remained significantly higher than that of mice given (p=0.009) or pCI::CD5L-hIg-SIINFEKL (p=0.0028), and comparable to split virus (p=0.85).
Ability of PR8 HA constructs to elicit protective immunity Protection of vaccinated mice from influenza infection was assessed by examining the ability of mice to clear a challenge dose of virus from their lungs by five days post-infection. It should be noted that both antibody and cytotoxic T cell-mediated responses can lead to viral titre reduction within this 5 day interval. Mice were challenged on day 65 post-priming and the titre of virus in their lung was determined by a plaque assay. Figure 16 WO 98/44129 PCT/AU98/00208 shows that all mice immunised with infectious virus were able to clear the challenge dose of virus. Of the mice given DNA constructs, the lung virus titres of mice immunised with the pCI::mCTLA4-hlg-HA construct were significantly lower than those of mice immunised with either pCI::CD5L-hIg-HA (p=0.0004) or pCI::CD5L-hlg-SIINFEKL (p=0.0002). Also the level of clearance observed in the pCI::mCTLA4-hIg-HA construct-immunised mice was almost as good as that seen in mice given the split virus vaccine.
Conclusions pCI::CTLA4-hIg-HA conferred higher antibody levels and better protection against challenge compared with the control vector demonstrating the immune enhancing effect of the incorporation of the targeting molecule.
Example Use of CTLA4 in Corynebacterium pseudotuberculosis in sheep.
Introduction Corynebacterium pseudotuberculosis is the causative agent of caseous lymphadenitis (CLA) in sheep. Established infection by these bacteria leads to the formation of abscesses in the lymph nodes, especially the draining lymph node of the site of infection. Phospholipase D (PLD) has been characterised as a virulence factor and a protective antigen for CLA. Indeed formalin-treated
PLD
20 or genetically toxoided PLD (APLD 2 2 has been shown to protect sheep from CLA.
We have used the genetically toxoided PLD as the basis for our DNA vaccination approach and investigated whether the addition of bovine (b)CTLA4-hIg or hIg alone to the APLD construct enhanced the immune response to PLD and to hIg.
WO 98/44129 PCT/AU98/00208 21 Materials and methods.
DNA constructs Using the known sequence of bCTLA4 (GenBank accession number X15070), the bCTLA4 gene was isolated from bovine peripheral blood mononuclear cells. A PCR product of bCTLA-4 (729 bp) was cloned into the Zeroblunt TM cloning vector according to the manufacturer's instructions (Invitrogen) and sequenced using the Applied Biosystems automated sequencer. The sequence of bCTLA4 was found to be identical to the published sequence.
The following constructs were generated in the pCI vector for DNA immunisation: pCI::bCTLA4-hIg-APLD pCI::APLD pCI::bCTLA4-hIg CD5L refers to the leader sequence of the CD5 molecule allowing the hulg-APLD protein to be secreted.
Experimental animals and immunisation regimen Cross-bred ewes aged 12 weeks were used in the challenge trial. animals were allocated randomly to each group. Animals were pre-screened for the presence of antibodies to PLD and to Corynebacterium pseudotuberculosis lysate. Positive animals were excluded from the trial.
Shearing, vaccination and tail docking of animals was avoided to minimise risk of infection with Corynebacterium pseudotuberculosis.
Animals were injected intra-muscularly with 500 gg LPS free pCI plasmid DNA (coding for either pCI::bCTLA4-hIg-APLD, pCI::APLD, pCI::bCTLA4-huIg or pCI::CD5L-hulg-APLD) in 5 ml of PBS. Control animals received either Glanvac or were left un-immunised. All animals received the same vaccine, at the same dose, 4 weeks later.
Challenge Bacterial cultures of wild type Corynebacterium pseudotuberculosis were grown at 370 C in Brain heart infusion broth (Difco Laboratories) containing 0.1% Tween 80 (BHI).
WO 98/44129 PCT/AU98/00208 22 All sheep were challenged 6 weeks after primary immunisation using a 1 ml dose of 106 CFU of Corynebacterium pseudotuberculosis injected just above the coronet of the right hind lateral claw.
Immunological assays Sera were collected from the sheep at weekly intervals and assayed for the presence of antibodies to genetically detoxified PLD (APLD) and hIg using an ELISA. Plates were coated with 1/50 of culture supernatant from APLD expressing Corynebacterium pseudotuberculosis or 5 g/ml hIg protein.
The sera were diluted in two fold steps starting at 1/100 and 1/10 for the detection of anti-APLD and anti-hIg antibodies respectively. Titres were calculated by linear regression on a double logarithmic scale in the linear part of the graph. The titre was defined as the dilution, which resulted in an O.D. 0.3 in the ELISA.
T cell proliferation assays were performed in triplicates using 2 concentrations of APLD (1/50 and 1/250) or hIg (5 g/ml and 25 g/ml) as an antigen. PBMC were purified by ficoll gradient and cultured in vitro for 3 days. The cultures were pulsed with 3H-methyl-thymidine for 18 hours before being harvested on glass fibre filters and radioactive incorporation assessed. The results are presented as stimulation indices ratio between counts obtained with antigen over counts obtained without antigen)..
Statistical analysis was performed using the Systat program. The non-parametric Mann-Whitney U test was used to calculate significance. p values below 0.05 were considered significant.
Results Anti-hIg antibody levels The antibody titres to human immunoglobulin (hlg) reflect the immune response to the DNA vaccination against the hIg part of the fusion protein in the case of bCTLA4-hIg-APLD, CD5L-hIg-APLD and bCTLA4-hlg.
By comparing the response to hIg from the animals injected with pCI::bCTLA4-hlg-APLD to these injected with pCI::CD5L-hIg-APLD it is possible to specifically evaluate the effect of bCTLA4 targeting on the immunogenicity of hIg. Results shown in Fig 17 indicate that the antibody WO 98/44129 PCT/AU98/00208 23 response to hIg in animals injected with pCI::bCTLA4-hIg-APLD (filled squares) is both earlier and stronger than the anti-hIg response induced in animals immunised with pCI::CD5L-hlg-APLD (closed circles). The Mann-Whitney U test indicates a statistically significant difference for weeks 3 and 4. Corroborating these results the anti-hIg response in the group injected with pCI::bCTLA4-hIg (closed triangles) is also earlier and stronger than the response in animals injected with Anti-APLD antibody levels Immunisation with the detoxified-PLD protein antigen (Glan-Vac) resulted in little or no detectable antibodies during the first 7 weeks after immunisation. Two weeks after challenge antibody levels increased dramatically. This is consistent with previously reported results 21 All groups immunised with pCI encoding the APLD antigen either alone or as a fusion protein with bCTLA4-hIg or with CD5L-hIg, resulted in similar kinetics of antibody production. Indeed, no significant anti-APLD antibody levels were detected until 2 weeks post-challenge week At each time point there was no significant difference between the level of antibodies induced by the different pCI constructs, indicating that all constructs have a similar ability to induce immune memory to APLD.
However, this result is not surprising in light of the fact that detoxified-PLD is not a classical antigen in that two doses of the protein antigen (Glan-Vac group) failed to induce substantial antibody levels. This result has also been reported by others 22 Thus detoxified-PLD seems to result in the induction of immune memory without induction of high antibody titres. This immune memory is then activated during the early stage of the challenge by PLD expressed by Corynebacterium pseudotuberculosis. It is interesting to note that APLD expressed in eukaryotic cells (COSm6 cells) has a slightly larger size compared to APLD expressed in Corynebacterium pseudotuberculosis (Fig 18). This difference can be accounted for by the possible glycosylation of APLD in eukaryotic cells. As in the case with anti-hIg antibody levels it is noted that sheep, in contrast to mice, do not produce antibodies over prolonged periods of time following DNA vaccination.
WO 98/44129 PCT/AU98/00208 24 T cell responses to APLD and hIg.
T cell responses to APLD and hIg were analysed at 3 weeks, 6 weeks and 9 weeks. No significant antigen specific proliferation, to these two antigens could be demonstrated in peripheral blood mononuclear cells (PBMC) at any time point. This is most likely due to technical difficulties since the kinetics of the anti APLD response indicate the likely presence of memory T cells in the vaccinated animals.
Anti-APLD response subsequent to challenge with virulent C. pseudotuberculosis.
Challenge was performed at 6 weeks post-immunisation two weeks post-booster immunisation). Observation of the site of inoculation at week 10 did not reveal any correlation with immunisation regime. Except in the case of the unvaccinated control and pCI::bCTLA4-hlg immunised animals, the antibody response to APLD increased two weeks post-challenge (week This indicates that the immune memory induced by the immunisation with DNA can be boosted with APLD produced by Corynebacterium pseudotuberculosis. This indicates that although APLD produced by DNA vaccination is most likely glycosylated there is still cross-reaction with bacterial wild-type PLD.
In the un-immunised control animals the level of anti-APLD antibodies remains low until 3 to 4 weeks post-challenge (week This kinetics of antibody appearance has been described previously 22 Interestingly the antibody levels in the animals primed with APLD by DNA vaccination or detoxified-PLD by Glan-Vac injection, diminished after week 8 suggesting that the PLD antigen is no longer boosting the immune response. This may be due to a diminished level of PLD secretion due to the animals clearing Corynebacterium pseudotuberculosis.
Protection from challenge with virulent Corynebacterium pseudotuberculosis.
At week 12 post vaccination (6 weeks post challenge) necropsy of all animals was performed to evaluate the protective efficacy of the immunisation protocol. Both left and right popliteal, inguinal and prefemoral lymph nodes were dissected and visually assessed for the characteristic abscesses induced by Corynebacterium pseudotuberculosis.
The lungs were palpated to detect abscesses and no lung abscesses were WO 98/44129 PCT/AU98/00208 found in any of the animals. Protection was defined as the absence of the characteristic Corynebacterium pseudotuberculosis abscesses in any of the lymph nodes. In two sheep small dry lesions were observed in the draining popliteal lymph node, clearly distinct from the abscesses in the other animals. These lesions were most likely foci of Corynebacterium pseudotuberculosis which were regressing in the face of an effective immune response and these animals were also scored as "protected". From Fig 19 it can be seen that while about 10 of the unvaccinated animals did not develop lesions, 90 were protected by vaccination with Glan-Vac. Only 1 animal out of 10 injected with pCI::CTLA4-hIg did not have abscesses in the lymph nodes. All animals vaccinated with DNA encoding for APLD (pCI::bCTLA4-hIg-APLD, pCI::APLD and pCI::CD5L-hIg-APLD) were afforded some protection. The level of protection ranged from 40 to 70% with the highest degree of protection observed in the group injected with pCI::CTLA4-hIg-APLD.
Conclusion This study has examined the ability of bCTLA4 to increase the immune response to antigens during DNA immunisation in sheep. bCTLA4 has the ability to accelerate and increase the immune response to hIg.
Challenge with Corynebacterium pseudotuberculosis indicated that DNA vaccination could induce a protective immune response. When comparing the protection obtained with pCI::bCTLA4-hIg-APLD and pCI::CD5L-hIg-APLD a substantial difference was observed.
The protection induced by CD5L-hIg-APLD is lower than the protection induced by APLD on its own. This difference may be due to the fact that the APLD molecule is much smaller than the bCD5L-hIg-APLD and hence could be expressed in an acceptable conformation more easily. One could therefore expect that the bCTLA4-hIg-APLD molecule which is even larger would be even more difficult to express. Hence the fact that the bCTLA4-hIg-APLD molecule induced higher levels of protection indicates that the CTLA4 molecule effectively increases the immunogenicity of the fusion protein. It can be reasonably expected that by improving the level of expression and folding of the molecule even better protection could be WO 98/44129 PCT/AU98/00208 26 obtained. One way of achieving this would be to reduce the size of the hIg portion of the fusion protein.
Example 7 Vaccination of C3H/He mice with DNA encoding the Parasite Surface Antigen 2 (PSA2) of Leishmania major.
Introduction Protection against infection with this obligatory intracellular parasite is provided by CD4+ T cells of the macrophage activating, Thi type. We have shown that injection of plasmid DNA encoding full length PSA2 induced significant protection against a challenge infection in C3H/He mice.
This protection correlated with the induction of a very low, but consistent Thi type of immune response, ie induction of T cells secreting interferon gamma, but no IL-4. Here we aim to examine the ability of CTLA4-Ig to improve the level of protection.
Materials and methods PSA2 encoding residues 33-35 7 (missing the leader and gpi signal) was fused C-terminal to either pCI::CD5L-hIg or pCI::mCTLA4-hIg.
DNA
encoding a secreted form of PSA2 (residues 1-357) was also made (pCI::PSA2) Groups of 8 mice were injected with 100 4g DNA in 100 4l phosphate buffered saline (PBS) intramuscularly twice at two week intervals. Two weeks after each injection the mice were bled and the serum tested for antibodies to PSA-2. Two weeks after the second injection the mice were infected intradermally with 100.000 promastigotes. The development of lesions at the site of infection was monitored weekly and scored according to size. Parasite burdens in the lymph nodes draining the lesion were determined by limiting dilution analysis at 7 weeks after challenge infection.
WO 98/44129 PCT/AU98/00208 27 Results Antibody production Antibodies were measured only by ELISA OD at a single point Two weeks after the first immunisation, 4 of 8 mice immunised with pCI::mCTLA4-hlg-PSA2 and 3 of 8 mice given pCI::CD5L-hlg-PSA2 produced significant antibody at a dilution of 1:500. However, after the second injection of DNA mice immunised with pCI::mCTLA4-hg-PSA2, pCI::CD5L-hlg-PSA2 and our own secreted form of PSA-2 showed significant levels of antibody at this dilution. The PBS control had background antibody.
Protection from infection Mice immunised with DNA encoding pCI::CD5L-hlg-PSA2 and the controls PBS and vector DNA developed lesions at the site of infection 1 week after challenge. Mice immunised with pCI::mCTLA4-hlg-PSA2 or pCI::PSA2 developed lesions only 3 weeks after infection and the size of the lesions was smaller compared to the rest. Mice immunised with pCI::mCTLA4-hIg-PSA2 or pCI::PSA2 also had the smallest number of mice which developed lesions with only 5 of 8 mice showing any lesions at the peak of the disease curve (Fig 20). Notably, pCI::mCTLA4-hIg-PSA2 conferred better protection than pCI::CD5L-hIg-PSA2 (p=.0001; log rank test).
Summary The ability to overcome the problem of low or absent responsiveness in DNA immunisation by antigen targeting enhances the potential of genetic vaccines. The present inventors also show that intramuscular injection of DNA can also be employed to deviate immune responses to the same antigen allowing for the development of vaccines in which the response most likely to confer protection can be generated.
Intramuscular injection of expression plasmids shows great potential for genetic vaccination. The present inventors have shown that fusion proteins consisting of antigen and cell surface receptor ligands could deliver antigen to sites of immune induction which enhance the immune response and possibly the efficacy of genetic vaccines. As set out above mice were WO 98/44129 PCT/AU98/00208 28 immunized with plasmids encoding Fc fragment of human IgG1 as antigen.
This Ig fragment was fused with CTLA4 (CTLA4Ig) for delivery to antigen presenting cells (APC) expressing B-7, or with L-selectin (L-SELIg) for delivery to high endothelial venule cells of lymph nodes. L-selectin binds CD34 and MadCAM-1 and so could target any lymphoid organ with these receptors Enhanced antibody responses were shown in both the CTLA4Ig and L-SELIg immunized mice, 1000 and 100 fold respectively at 4 weeks. Moreover the response after CTLA4Ig immunisation was the most rapid. Immune deviation from an IgG2a to an IgG1 dominated response occurred in CTLA4Ig immunized mice and allows for the development of genetic vaccines in which the response most likely to confer protection can be generated.
It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodiments without departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
WO 98/44129 PCT/AU98/00208 29 References 1. Ulmer, J. J. J. Donnelly, S. E. Parker, G. H. Rhodes, P. L. Felgner, V. J.
Dwarki, S. H. Gromkowski, R. R. Deck, C. M. De Witt, A. Friedman, L. A.
Hawe, K. R. Laender, D. Martinez, H. C. Perry, J. W. Shiver, D. L.
Montgomery, and M. A. Liu. 1993. Heterologous protection against influenza by injection of DNA encoding a viral protein. Science 259:1745.
2. Wolff, J. P. Williams, G. Acsadi, S. Jiao, A. Jani, and W. Chong. 1991.
Conditions affecting direct gene transfer into rodent muscle in vivo.
Biotechniques 11:474.
3. Hohlfeld, and A. G. Engel. 1994. The immunobiology of muscle.
Immunol. Today 15:269.
4. Pardoll, D. and A. M. Beckerleg. 1995. Exposing the immunology of naked DNA vaccines. Immunity 3:165.
5. Raz, D. A. Carson, S. E. Parker, T. B. Parr, A. M. Abai, G. Aichinger, S.
H. Gromkowski, M. Singh, D. Lew, M. A. Yankauckas, S. M. Baird and G. H.
Rhodes. 1994. Intradermal gene immunization: the possible role of DNA uptake in the induction of cellular immunity to viruses. Proc. Natl. Acad.
Sci. 91:9519.
6. Condon, S. C. Watkins, C. M. Celluzzi, K. Thompson and L. D. Falo, Jr.
1996. DNA-based immunization by in vivo transfection of dendritic cells.
Nature Med. 2: 1122.
7. In Molecular cloning: A laboratory manual. 2nd ed. 1989. J. Sambrook, E. F. Fritsch, and T. Maniatis, eds. Cold Spring Harbor Press, Cold Spring Harbor, NY, pl.38.
8. Aida, and M. J. Pabst. 1990. Removal of endotoxin from protein solutions by phase separation using Triton X-114. J. Immunol. Methods.
132:191.
9. Major M. L. Vitvitski, M. A. Mink, M. Schleef, R. G. Whalen, C. Trepo and G. Inchauspe. 1995. DNA-based immunization with chimeric vectors for the induction of immune responses against the hepatitis C virus nucleocapsid. J. Virol 69:5798.
Manickan, R. J. Rouse, Z. Yu, W. S. Wire, and B. T. Rouse. 1995.
Genetic immunization against herpes simplex virus. Protection is mediated by CD4+ T lymphocytes. J. Immunol. 155:259.
WO 98/44129 PCT/AU98/00208 11. Linsley P. P. M. Wallace, J. Johnson, M. G. Gibson, J. L. Greene, J. A.
Ledbetter, C. Singh and M. A. Tepper. 1992. Immunosuppression in vivo by a soluble form of the CTLA-4 T cell activation molecule. Science 257:792.
12. Varki A. 1994. Selectin Ligands. PNAS 91:7390.
13. Rickard MD (1991). Cestode vaccines. Southeast Asian J Trop Med Public Health 22 Suppl:287-290.
14. Rothel JS, Waterkeyn JG, Strugnell RA, Wood PR, Seow HF, Vadolas J, Lightowlers MW. (1997) Nucleic acid vaccination of sheep: Use in combination with a conventional adjuvanted vaccine against Taenia ovis.
Immunol Cell Biol 75:41-46.
Rothel JS, Lightowlers MW, Seow HF, Wood PR, Rothel LJ, Heath DD, Harrison GB. (1996) Immune responses associated with protection in sheep vaccinated with a recombinant antigen from Taenia ovis. Parasite Immunol 18:201-208.
16. Hodder AN, Crewther PE, Matthew ML, Reid GE, Moritz RL, Simpson RJ, Anders RF (1996) The disulfide bond structure of Plasmodium apical membrane antigen-1. J Biol Chem 271:29446-52.
17. Crewther PE, Matthew ML, Flegg RH, Anders RF (1996) Protective immune responses to apical membrane antigen 1 of Plasmodium chabaudi involve recognition of strain-specific epitopes. Infection Immunity.
64:3310-7 18. Fazekas de St. Groth, Webster RG (1966) Disquisitions of Original Antigenic Sin. I. Evidence in man. J Exp Med 124:331-45 19. Jackson DC, Tang XL, Brown LE, Murray JM, White DO, Tregear GW (1986) Antigenic determinants of influenza virus hemagglutinin. XII. the epitopes of a synthetic peptide representing the C-terminus of HA1.
Virology 155:625-32 Burrell DH (1983) Caseous lymphadenitis vaccine NSW Vet. Proc. 19, 53-57 21. Hodgson AL, Krywult J, Corner LA, Rothel JS, Radford AJ (1992) Rational attenuation of Corynebacterium pseudotuberculosis: potential cheesy gland vaccine and live delivery vehicle. Infect Immun 60, 2900-2905 22. Hodgson AL, Tachedjian M, Corner LA, Radford AJ (1994) Protection of sheep against caseous lymphadenitis by use of a single oral dose of live recombinant Corynebacterium pseudotuberculosis. Infect Immun 62, 5275-5280 WO 98/44129 PCT/AU98/00208 31 SEQUENCE LISTING GENERAL INFORMATION:
APPLICANT:
NAME: The Council of the Queensland Institute of Medical Research STREET: 300 Herston Road CITY: Herston STATE: Queensland COUNTRY: Australia POSTAL CODE (ZIP): 4029 NAME: Commonwealth Scientific and Industrial Research Organisation STREET: Limestone Avenue CITY: Campbell STATE: ACT COUNTRY: Australia POSTAL CODE (ZIP): 2601 NAME: The University of Melbourne STREET: Royal Parade CITY: Parkville STATE: Victoria COUNTRY: Australia POSTAL CODE (ZIP): 3052 NAME: The Walter and Eliza Hall Institute of Medical Research STREET: Royal Melbourne Hospital, Royal Parade CITY: Parkville STATE: Victoria COUNTRY: Australia POSTAL CODE (ZIP): 3052 NAME: CSL Limited STREET: 45 Poplar Road CITY: Parkville STATE: Victoria COUNTRY: Australia POSTAL CODE (ZIP): 3052 (ii) TITLE OF INVENTION: Enhancement of Immune Response using Targeting Molecules (iii) NUMBER OF SEQUENCES: 1 (iv) COMPUTER READABLE FORM: MEDIUM TYPE: Floppy disk COMPUTER: IBM PC compatible OPERATING SYSTEM: PC-DOS/MS-DOS SOFTWARE: PatentIn Release Version #1.30 (EPO) (vi) PRIOR APPLICATION DATA: APPLICATION NUMBER: AU PO 5891 FILING DATE: 27-MAR-1997 (vi) PRIOR APPLICATION DATA: WO 98/44129 WO 9844129PCT/AU98/00208 32 APPLICATION NUMBER: AU PP 1830 FILING DATE: 13-FEB-1998 INFORMATION FOR SEQ ID NO: 1: SEQUENCE CHARACTERISTICS: LENGTH: 11265 base pairs TYPE: nucleic acid STRAN~DEDNESS: double TOPOLOGY: circular (ii) MOLECULE TYPE: DNA (genornic) (iii) HYPOTHETICAL:
NO
(iv) ANTI-SENSE: NO (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1: TCTAGAGTCG ACCAATTCTC GTGCACTCTC
AGTACAATCT
TTGTGTGTTG GAGGTCGCTG CTTGACCGAC AATTGCATGA TGTACGGGCC
AGATATACGC
GCTTCGGTTG TACGCGGTTA GAGGGGGAAA TGTAGTCTTA TAGCAACATG CCTTACAAGG TGGTACGATC GTGCCTTATT ACTGAATTCC GCATTGCAGA CATTTGACCA TTCACCACAT TTGCTAGCGG CCGCTCGAGC CGGAGGTACA AAGCTCAACT ACTCTTCTTT TCATCCCAGT TTGGCTAGCA GCCATGGTGT GATGAGGTCC
GGGTGACTGT
ACGACATTCA CAGAGAAGAA TTTAATGAAA
GCAGAGTGAA
TACCTCTGCA
AGGTGGAACT
ACGCAGATTT ATGTCATTGA CGCTCCTGCC TGGACGCATC CGTCTGCCTC TTCACCCGGA TGGCTTTTTC
CCAGGCTCTG
ACAAAGGGGC AGGTGCTGGG
ATGTTTGACA
GCTCTGCTGC
AGTAGTGCGC
AGAATCTGCT
GTATCTGAGG
GGAGTCCCCT
TGCAATACAC
AGAGAAAAAG
AGGAAGGCAA
GATAATTGTA
TGGTGTGCAC
TCAGCACATT
GCAGCTGCCT
CTTCTCTGAA
CGCCAGCTTT
GCTGCGGCAG
TACAGTGGGC
CCTCACCATC
CATGTACCCA
TCCAGAACCA
CCGGCTATGC
GCCTCTGCCC
GGCAGGCACA
CTCAGACCTG
GCTTATCATC
CGCATAGTTA
GAGCAAAATT
TAGGGTTAGG
GGACTAGGGT
CAGGATATAG
TTGTAGTCTT
CACCGTGCAT
CAGACAGGTC
TTTAAGTGCC
CTCCAAGCTG
T GC CC C CCAG
TCTAGGACTT
GCCATACAGG
CCATGTGAAT
ACAAATGACC
TTCCTAGATT
CAAGGACTGA
CCGCCATACT
TGCCCGGATT
AGCCCCAGTC
GCCCCACTCA
GGCTAGGTGC
CCAAGAGCCA
GCAGATCCTG
AGCCAGTATC
TAAGC TACAA
CGTTTTGCGC
GTGTTTAGGC
TAGTTTCGCT
GCAACATGGT
GCCGATTGGT
TGACATGGAT
TAGCTCGATA
GGTACCAGCT
CCATGGCTTG
GGCCTTTTGT
TGACCCAACC
ATTCACCATC
AAATGACTGA
ACCC CTTC TG
GAGCTGTTGA
TTGTGGGCAT
CTGGTAAGTA
AGCTTGTATG
TGCTCCCTGC
CAAGGCAAGG
TGCTTCGCGA
GCCCAGCGGG
TTTGCATAGG
AACGATGAGT
GGAAGTAAGG
TGGACGAAcC
CAATAAACGC
GCTAGCAAGC
TCTTGGACTC
AGCCCTGCTC
TTCAGTGGTG
ACACAACACT
GGTCTGTGCC
CAGTGGTACC
CACGGGACTG
GGGCAACGGG
TAAGCTTCAG
120 180 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 CAGGGCAGCA AGGCAGGCCC TGCTCAGGGA GAGGGTCTTC CCCTAACCCA GGCCCTGCAC TATCCGGGAG GACCCTGCCc WO 98/44129 WO 9844129PCT/AU98/00208 CTGACCTAAG CCCACCCCAA CCTCCCAGAT TCCAGTAACT CTCACACATG CCCACCGTGC GGGACAGGTG CCCTAGAGTA TCCACCTCCA TCTCTTCCTC CCCCCAAAAC CCAAGGACAC GTGGACGTGA GCCACGAAGA GTGCATAATG CCAAGACAAA AGCGTCCTCA CCGTCCTGCA TCCAACAAAG CCCTCCCAGC CGTGGGGTGC GAGGGCCACA GACCGCTGTA CCAACCTCTG CCCCCATCCC GGGATGAGCT TTCTATCCCA GCGACATCGC AAGACCACGC CTCCCGTGCT GTGGACAAGA GCAGGTGGCA CTGCACAACC ACTACACGCA CCGGATCCAG ACATGATAAG TGAAAAAAAT GCTTTATTTG AGCTGCAATA AACAAGTTAA GAGGTGGGGA GGTTTTTTAA ATCCGGCTGC CTCGCGCGTT GGAGACGGTC ACAGCTTGTC GTCAGCGGGT GTTGGCGGGT AAATCCGCGC GGTGGTTTTT CGTGTCGCGC CAGTACATGC CTCAGTCCAG TCGTGGACCA TAAACCATTC CCCATGGGGG CCTGCCGCCC CGACGTTGGC GGGGAAAAGG AAGAAACGCG GAGTGCCAGc CCTGGGACCG TTTATTCTGT CTTTTTATTG TGTTTCAGTT AGCCTCCCCC
AGGCCAAACT
CCCAATCTTC
CCAGGTAAGC
GCCTGCATCC
AGCACCTGAA
CCTCATGATC
CCC TGAGGT C
GCCGCGGGAG
CCAGGACTGG
CCCCATCGAG
TGGACAGAGG
TCCTACAGGG
GACCAAGAAC
CGTGGAGTGG
GGACTCCGAC
GCAGGGGAAC
GAAGAGCCTC
ATACATTGAT
TGAAATTTGT
CAACAACAAT
AGCAAGTAAA
TCGGTGATGA
TGTAAGCGGA
GTCGGGGCGC
GGGGGTCGGG
GGTCCATGCC
GACCCCACGC
ACC C CGT CCC
TGCGAGCCCT
GGCGTATTGG
AACCCCGCGT
CCGTCATAGC
ATC TCCC CTA
CTCCACTCCC
TCTCTGCAGA
CAGCCCAGGC
AGGGACAGGC
CTCCTGGGGG
TCCCGGACCC
AAGTTCAACT
GAGCAGTACA
CTGAATGGCA
AAAACCATCT
CCGGCTCGGC
CAGCCCC GAG
CAGGTCAGCC
GAGAGCAATG
GGCTCCTTCT
GTCTTCTCAT
TCCCTGTCTC
GAGTTTGGAC
GATGCTATTG
TGCATTCATT
ACCTCTACAA
CGGTGAAAAC
TGCCGGGAGC
AGCCATGACC
GGTGTTTGGC
CAGGCCATCC
AACGCCCAAA
TAACCCACGG
GGGCCTTCAC
CCCCAATGGG
TTATGAACAA
GCGGGTTCCT
TTCCTTTGCC
TCAGCTCGGA CACCTTCTCT
GCCCAAATCT
CTCGCCCTCC
CCCAGCCGGG
GACCGTCAGT
CTGAGGTCAC
GGTACGTGGA
ACAGCACGTA
AGGAGTACAA
CCAAAGCCAA
CCACCCTCTG
AACCACAGGT
TGACCTGCCT
GGCAGCCGGA
TCCTCTACAG
GCTCCGTGAT
CGGGTAAATG
AAACCACAAC
CTTTATTTGT
TTATGTTTCA
ATGTGGTATG
CTCTGACACA
AGACAAGCCC
GGTCGACCAC
AGCCACAGAC
AAAAACCATG
ATAATAACC C
GGCCAGTGGC
CCGAACTTGG
GTCTCGGTGG
ACGACCCAAC
TCCGGTATTG
CTCGGACGAG
TGTGACAAAA
AGCTCAAGGC
TGCTGACACG
CTTCCTCTTC
ATGCGTGGTG
CGGCGTGGAG
CCGGGTGGTC
GTGCAAGGTC
AGGTGGGACC
CC CTGAGAGT
GTACACCCTG
GGTCAAAGGC
GAACAACTAC
CAAGCTCACC
GCATGAGGCT
AGTGCGACGG
TAGAATGCAG
AACCATTATA
GGTTCAGGGG
GCTGATTATG
TGCAGCTCCC
GTCAGGGCGC
TGGGCGCCAG
GCCCGGTGTT
GGTCTGTCTG
CCACGAACCA
TATGGCAGGG
GGGGTGGGGT
GGTATCGACA
ACCCGTGCGT
TCTCCTTCCG
TGCTGGGGCG
1500 1560 1620 1680 1740 1800 1860 1920 1980 2040 2100 2160 2220 2280 2340 2400 2460 2520 2580 2640 2700 2760 2820 2880 2940 3000 3060 3120 3180 3240 3300 3360 3420 3480 TCGGTTTCCA CTATCGGCGA GTACTTCTAC ACAGCCATCG GTCCAGACGG CCGCGCTTCT WO 98/44129 WO 9844129PCT/AU98/00208 GCGGGCGATT TGTGTACGCC ACCCTGCGCC CAAGCTGCAT AAGACCAATG CGGAGCATAT CCGCTCGAAG TAGCGCGTCT GTTGGCGACC TCGTATTGGG TATGCGGCCA TTGTCCGTCA GACTTCGGGG CAGTCCTCGG ACTGACGGTG TCGTCCATCA TATGAAATCA CGCCATGTAG GCTCGTCTGG CTAAGATCGG AACAGCGGGC AGTTCGGTTT GATGCAATAG GTCAGGCTCT CGCGGCCGAT GCAAAGTGCC ATTTACCCGC AGGACATATC GCCCTCCGAG AGCTGCATCA GACAGACGTC GCGGTGAGTT CTGTTGACGC TGTTAAGCGG ACCTTAATAT GCGAAGTGGA TTCGCCAATG ACAAGACGCT TATATTTCTT CCGGGGACAC GGCATGGCGG CCGACGCGCT GCCTTCCCCA TTATGATTCT ATGCTGTCCA GGCAGGTAGA CTTACCAGCC TAACTTCGAT GCGACCcAT GGAACGGGTT CCCGCGTTGC GTCGCGGTGC ACCTCGCTAA CGGATTCACC TGAATGCGCA AACCAACCCT GCACGCGGCG CAGCAAAAGG TAGGCTCCGC CCCCCTGACG CCCGACAGGA CTATAAAGAT TGTTCCGACC CTGCCGCTTA GCTTTCTCAT AGCTCACGCT GGGCTGTGTG CACGAACCCC
CGACAGTCCC
CATCGAAATT
ACGCCCGGAG
GCTGCTCCAT
AAT CC C CGAA
GGACATTGTT
CCCAAAGCAT
CAGTTTGCCA
TGTATTGACC
CCGCAGCGAT
CAGGCAGGTC
CGCTGAATTC
GATAAACATA
CACGCCCTCC
GGTCGGAGAC
CAGGCTTTTT
GTCGCTGCAG
CCTGGGACCG
GGGCGGGGTT
CGCCAGCAAA
GGGCTACGTC
TCTCGCTTCC
TGACGACCAT
CACTGGACCG
GGCATGGATT
ATGGAGCCG
ACTCCAAGAA
TGGCAGAACA
CCAGGAACCG
AGCATCACAA
ACCAGGCGTT
CC GGATAC CT
GTAGGTATCT
CCGTTCAGcc
GGCTCCGGAT
GCCGTCAACC
CCGCGGCGAT
ACAAGCCAAC
CATCGCCTCG
GGAGCCGAAA
CAGCTCATCG
GTGATACACA
GATTCCTTGC
CGCATCCATG
TTGCAACGTG
CCCAATGTCA
ACGATCTTTG
TACATCGAAG
GCTGTCGAAC
CATATCTCAT
GGTCGCTCGG
CGCCGCCCCG
TGTGTCATCA
CGCGAGCAAC
TTGCTGGCGT
GGCGGCATCG
CAGGGACAGC
CTGATCGTCA
GTAGGCGCCG
GCCACCTCGA
TTGGAGCCAA
TATCCATCGC
TAAAAAGGCC
AAATCGACGC
TCCCCCTGGA
GTCCGCCTTT
CAGTTCGGTG
CGACCGCTGC
CGGACGATTG
AAGCTCTGAT
CCTGCAAGCT
CACGGCCTCC
CTCCAGTCAA
TCCGCGTGCA
AGAGCCTGCG
TGGGGATCAG
GGTCCGAATG
GCCTCCGCGA
ACACCCTGTG
AGCACTTCCG
TAGAAACCAT
CTGAAAGCAC
TTTTCGATCA
TGCCCGGGAT
TGTTCGAGGC
ACTGCATCTG
TAGAACTAAA
GGGCCACGG.G
TCGCGACGCG
GGATGCCCGC
TTCAAGGATC
CGGCGATTTA
CGTCGCATCG
AGAGTTGGTC
CCGGATGCCT
AGAAGAAGAT
TGACCGCTGT
CGAGGTGCCG
CGACGGACGC
CAATCGCGCA
GGCCGAACCC
CCGGCTGCAG
CACGGCGGGA
GAATCGGGAG
CGGCGCAGCT
GAGATTCTTC
GAAACTTCTC
CTGCGGCACG
CACACGCGTC
CGTGTTCGAA
GACATGCAAA
GATGAAGCAG
AGGCTGGATG
GTTGCAGGCC
GCTCGCGGCT
TGCCGCCTCG
~3540 3600 3660 3720 3780 3840 3900 3960 4020 4080 4140 4200 4260 4320 4380 4440 4500 4560 4620 4680 4740 4800 4860 4920 4980 5040 5100 5160 5220 5280 5340 5400 5460 5520 CCCTATACCT TGTCTGCCTC CCTGAATGGA AGCCGGCGGC TCAATTCTTG CGGAGAACTG GTCCGCCATC TCCAGCAGCC GCGTTGCTGG CGTTTTTCCA TCAAGTCAGA GGTGGCGAA AGCTCCCTCG TGCGCTCTCC CTCCCTTCGG GAAGCGTGGC TAGGTCGTTC GCTCCAAGCT GCCTTATCCG GTAACTATCG WO 98/44129 PTA9/00 PCT/AU98/00208 TCTTGAGTCC AACCCGGTAA GATTAGCAGA GCGAGGTATG CGGCTACACT AGAAGGACAG AAAAAGAGTT GGTAGCTCTT TGTTTGCAAG CAGCAGATTA TTCTACGGGG TCTGACGCTC ATTATCAAAA AGGATCTTCA CTAAAGTATA TATGAGTAAA TATCTCAGCG ATCTGTCTAT AACTACGATA CGGGAGGGCT ACGCTCACCG
GCTCCAGATT
AAGTGGTCCT GCAACTTTAT AGTAAGTAGT TCGCCAGTTA GGTGTCACGC TCGTCGTTTG AGTTACATGA
TCCCCCATGT
TGTCAGAAGT
AAGTTGGCCG
TCTTACTGTC
ATGCCATCCG
ATTCTGAGAA
TAGTGTATGC
TACCGCGCCA CATAGCAGAA AAAACTCTCA AGGATCTTAC CAACTGATCT
TCAGCATCTT
GCAAAATGCC GCAAAAAAGG CCTTTTTCAA
TATTATTGAA
TGAATGTATT TAGAAAAATA ACCTGACGTC TAAGAzAACCA GAGGCCCTTT
CGTCTTCAAG
TCACAGGCCG
CACCCAGCTT
GAAGAGGAAG AGGAGGGGTC GAGGAATTTG AGGCCTGGCT TTTTCCGGGC
TGCGAGTAAT
TCAGACCTGG ATCTGTCTGA GGGGGCAGGA
GTCTGCACTC
ATCTCTTTTA GTGTGAATCA
*GACACGACTT
*TAGGCGGTGC
TATTTGGTAT
GATCCGGCAA
CGCGCAGAAA
AGTGGAACGA
CCTAGATCCT
CTTGGTCTGA
TTCGTTCATC
TACCATCTGG
TATCAGCAAT
CCGCCTCCAT
ATAGTTTGCG
GTATGGCTTC
TGTGCAAAAA
CAGTGTTATC
TAAGATGCTT
GGCGACCGAG
CTTTAAAAGT
CGCTGTTGAG
TTACTTTCAC
GAATAAGGGC
GCATTTATCA
AACAAATAGG
TTATTATCAT
AATTCTCATG
TTCTTCCGTT
CCGAGAATCC
TGAGGCTCAG
TGGTGATGAG
CAGCGAC CAT
CCTGTATTCA
TGTCTGACGA
ATCGCCACTG
TACAGAGTTC
CTGCGCTCTG
ACAAACCAcC
AAAAGGATCT
AAACTCACGT
TTTAAATTAA
CAGTTACCAA
CATAGTTGcC
CCCCAGTGCT
AAACCAGCCA
CCAGTCTATT
CAACGTTGTT
ATTCAGCTCC
AGCGGTTAGC
ACTCATGGTT
TTCTGTGACT
TTGCTCTTGC
GCTCATCATT
ATCCAGTTCG
CAGCGTTTCT
GACACGGAAA
GGGTTATTGT
GGTTCCGCGC
GACATTAACC
TTTGACAGCT
GCCCCAGTAG
CCATCCCTAC
GACGCAAATC
GACGAGGATG
GAAGGGGATG
CTGAGCGTCG
GCAGCAGCCA
TTGAAGTGGT
CTGAAGCCAG
GCTGGTAGCG
CAAGAAGATC
TAAGGGATTT
AAATGAAGTT
TGCTTAATCA
TGACTCCCCG
GCAATGATAC
GCCGGAAGGG
AATTGTTGCC
GCCATTGCTG
GGTTCCCAAC
TCCTTCGGTC
ATGGCAGCAC
GGTGAGTACT
CCGGCGTCAA
GGAAAACGTT
ATGTAACCCA
GGGTGAGCAA.
TGTTGAATAC
CTCATGAGCG
ACATTTC CCC
T)TAAAAATA
TAT CAT CGAT
CATCTCTGTC
CGTCCAGCAA
TTGAGGATGT
GTTCGGAGGA
AGGGTGGGGG
TCTAATAAAG
CTGGTAACAG
GGCCTAACTA
TTACCTTCGG
GTGGTTTTTT
CTTTGATCTT
TGGTCATGAG
TTAAATCAAT
GTGAGGCACC
TCGTGTAGAT
CGCGAGACCC
CCGAGCGCAG
GGGAAGCTAG
CAGGCATCGT
GATCAAGGCG
CTCCGATCGT
TGCATAATTC
CAACCAAGTC
CACGGGATAA
CTTCGGGGCG
CTCGTGCACC
AAACAGGAAG
TCATACTCTT
GATACATATT
GAAAPAGTGCC
GGCGTATCAC
AAGCTGATCC
TGGTGACCTT
kAAGGGGGAC
TCAGCGGGAG
TGGGGAATTT
GGCTGTTGGA
kTGTCTATTG
GAAATGGCCT
5580 5640 5700 5760 5820 5880 5940 6000 6060 6120 6180 6240 6300 6360 6420 6480 6540 6600 6660 6720 6780 6840 6900 6960 7020 7080 7140 7200 7260 7320 7380 7440 7500 7560 GGGGCCAGGT ACAGGACCTG AGGAGAGAAG GGAGACACAT CTGGACCAGA AGGCTCCGGC GGCAGTGGAC CTCAAAGAAG WO 98/44129 WO 9844129PCT/AU98/00208 AGGGGGTGAT AACCATGGAC GAGGACGGGG AAGAGGACGA AGGAGCCCCG GGCGGCTCAG AAAACGTCCA AGTTGCATTG AGCGGGAGGG GCAGGAGCAG GCCGGGGTCG AGGAGGTAGT GAAGTCGTGA AAGAGCCAGG CCAGTAGTCA GTCATCATCA CATTTTTCCA CCCTGTAGGG ATGGTGAGCC TGACGTGCCC AAGGCCCAAG CACTGGACCC GGTTTGGAAA GCATCGTGGT GTTTAAGAGC TCTCCTGGCT TCGCCGGTGT GTTCGTATAT CTGCCCTTGC TATTCCACAA CCCCTGGACC CGGCCCACAA TCTTTTTACA AACTCATATA TGACAAAGCC CGCTCCTACC TAGATTTGCC TCCCTGGTTT GAGATGACGG AGATGAAGGA ACTTGTTAGG AGACGCCCTC AAATCCCCAG TAGACATCAT ATTTTCGTCC TCCCAACATG CGCGCTCAAC ACCTTCTCGC ACATGCGACG GCTTTAGCCT GCAGGAAAAG GACAAGCAGC GGATAGCACT CCCACTCTAc CATATGCTAC CCGGATACAG CATATGCTAc CCAGATATAG CATATACTAC CCAGATATAG CCTATGCTAC CCAGATATAG CATATGCTAT CCAGATATTT TACTACCCTA ATCTCTATTA TACTACCCAG ATATAGATTA
GATCAGGGCC
GCTGCAAAGG
GAGGGGCAGG
GGAGGCCGCC
GGGAGAGGTC
TCCGGGTCTC
GAAGCCGATT
CCGGGAGCGA
CGGGGTCAGG
CAAGGAGGTT
AGGAGTCACG
GGAGGTAGTA
TGTCGTCTTA
CCTGGCCCGC
TTTGCTGAGG
TGCAATATCA
CCACCTATGG
GGTGATGGAG
AATCGTATTA
GCGTGCTGTT
GGGCAATTGG
GTTGGAAAAC
GGCCTCCTTA
GAAAATTCAC
TACTGGGTAT
ATTAGGATAG
ATTAGGATAG
ATTAGGATAG
ATTAGGATAG
GGGTAGTATA
GGATAGCATA
GGATAGCATA
AAGACATAGA
GACCCACGGT
AGGGAGGCCG
GGGGTAGAGG
GTGGACGTGG
CACCGCGCAG
ATTTTGAATA
TAGAGCAGGG
GTGATGGAGG
C CAAC C CGAA
TAGAAAGGAC
AGACCTCCCT
CACCATTGAG
TAAGGGAGTC
TTTTGAAGGA
GGGTGACTGT
TGGAAGGGGC
ATGAGGGTGA
AAAGCCGTGT
GGTGTATTTC
GCATAC CCAT
ATTAGCGACA
AATTCACCTA
GCCCCCTTGG
CATATGCTGA
CATATACTAC
CCTATGCTAC
CATATGCTAC
CATATGCTAC
TGCTAcccAG TGCTAcccGG
TGCTACCCAG
GGACGAGGAC
GATGGTGTCC
GGAACAGGAG
GGGTCGAGG.A
ACGTGAAAGA
AGAAAAGAGG
GCCCCCTCCA
CCACCAAGAA
CC CCGCAGAT
CAGGCGCAAA
ATTTGAGAAC
TACCGACGAA
TTACAACCTA
TCGTCTCCCC
CATTGTCTGT
TGCGATTAAG
GTGCAGCTTT
TGCCGCGGAG
GGAAGGGCAG
ATTCCCCCGC
TGGCCATCTG
GTTGTCACGT
TTTACCTGGT
AGAATGGGAG
GAGGTGGCGG
CTGTATATGC
CCAGATATAG
CCAGATATAA
CCAGATATAG
CCAGATATAG
ATATAAATTA
ATACAGATTA
ATATAGATTA
GCGGAAkGACC GGAGAC C CCA
CAGGAGCAGG
GGTAGTGGAG
GCCAGGGGGG
CCCAGGAGTC
GGTAGAAGGC
GGTGGCCCAG
GACCCAGGAG
AAAGGAGGGT
ATTGCAGAAG
GGAACTTGGG
AGGCGAGGAA
TTTGGAATGG
TATTTCATGG
GACCTTGTTA
GACGATGGAG
GGTGATGACG
GAGTGATGTA
ACTAAAGAAT
TCTTGTCACC
CACTCAGCTC
GAGCAATCAG
CAACCAGCAT
CATATGCAAA
ATGAGGATAG
ATTAGGATAG
ATTAGGATAG
ATTAGGATAG
ATTAGGATAG
GGATAGCATA
GGATAGCATA
GGATAGCCTA
7620 7680 7740 7800 7860 7920 7980 8040 8100 8160 8220 8280 8340 8400 8460 8520 8580 8640 8700 8760 8820 8880 8940 9000 9060 9120 9180 9240 9300 9360 9420 9480 9540 9600 TGCTACCCAG ATATAAATTA GGATAGCATA TACTACCCAG ATATAGATTA GGATAGCATA WO 98/44129 WO 9844129PCT/AU98/00208 TGCTACCCAG ATATAGATTA GGATAGCCTA TGCTACCCAG TGCTATCCAG ATATTTGGGT CAGCGACCTC GTGAATATGA TGTAATTTGT CCTCCAGATC TGCAGGTATT CCCCGGGGTG TTAGCGGGGT TACAATCAGC GCGGCGTGTG GGGGCTGACG TTGTCTTTGT TTATGGGCCC GACAACCAGT GGAGTCCGCT GTGTAACAAC ATGGTTCAC TATCATATTG CACTAGGATT CCAGTCTTTA CGGCTTGTCC TTCATTCCTA CACTAGTATT GCACAATGCC ACCACTGAAC TTGTTTTCGA
GCACCTCACA
TAAACGAAGG AGAATGAAGA ATCTTCAGCC
ACTGCCCTTG
ATGTAAATAA AACCGTGACA TTACTAACCC TAATTCGATA GGTTAGTCTG GATAGTATAT CAAGGGGGCC TTATAAACAc ACACAGGCCC CTCTGATTGA GGTACATGTC CCCCAGCATT GTTGTGTTGC AGTCCACAGA *CAAATGTGCA
CATCCATTTA
GTCCCCCCCC GTGTCACATG AGGGATTACA TGCACTGCCC
AGTATATGCT
GGACCAACAA
GCAGCAATCG
CCATTAGTGG
CAAGTTATTA
CGTGCCCCCA
CATTGGCGTG
GCTGTCGGCG
TGTCTTGGTC
ATGTGTTGCC
CCACCCCATG
TATTGCCCAA
C CC CCGTC CA
TACACCTTAC
AGCAGGCGAA
TGACTAAAAT
GCTCATGGGG
GCATATGCTT
AC TAC TACC C
TATTGCTAAT
CGTTGGTGTA
GGTGTAAGAG
CTGCAAAGTC
TAAGGATGTC
TGGAACAGGG
CGAATACAAA
ACCCATGGCA
CCCTGTGCTT
CGCCCCTATC
TTTTGTGGGC
CACCCTTATT
CTCCACAATT
GAGCCCCGTT
TCCACTCTCT
CCTGCCTGGG
CATAGCCATA
GATTTCTATT
GGGGTTTGTG
AATTTTATTC
TGTTCACAAC
GATTCAGGAG
GGTTCACTAC
TGGGAGATAT
CCCGTTGGGT
GGGAAGCATA
GCCCTCTTGA
GCCTCCCGTA
CTTCAGCCAA
TGCTCCAGGA
AACTACAGTC
CCCAGTTGGC
ACAAAAGCGC
ATATAGATTA
ACATTAGCCC
GGCGCTCAGG
TTGGCCCGCC
AAGTGGTTTG
TTACAGTCCA
TCAAAAAAAA
TAATTTTCGG
TTCCCCTTGT
ACACATCTTA
AATTCGTGTG
GTTAAAGATA
AGGGTTATAT
TGGGGGCGTC
TCAGCAGTTA
AGTTCACTGC
CCTCGTGGAA
CGCTGTTCCT
AACATATGCT
TGCTACCCGT
GGGTCCGCTT
GTCTTCCTGG
GAGTTACACA
TGAAAGCCAC
AXGAGAACCCC
AAGTTGTAcC rCCTCGTACC
GGATAGCATA
ACCGTGCTCT
CGCAAGTGTG
CACCTACTTA
ACCGCAGTGG
AAACCGCAGG
GAGTGGCCAC
GGGTGTTAGA
TACAAATAGA
ATAACCCCAG
AGATGGACAT
TTCAGAATGT
TGGTGTCATA
AC CTGAAAcCC
TTCTATTAGC
CCGCTCCTTG
TCCTGAC CCC
TAGGACCCTT
ATTGAATTAG
TTAGGGTTAA
PTCGGTAGCT
GCCCCTGGGA
rAAAGGCAAT rCAGTGTTGG rTTGTGTTTG kACCAACTGA kGCGAAGAAG 9660 9720 9780 9840 9900 9960 10020 10080 10140 10200 10260 10320 10380 10440 10500 10560 10620 10680 10740 10800 10860 10920 10980 11040 11100 11160 11220 11265 GGGCAGAGAT GCCGTAGTCA GGTTTAGTTC GTCCGGCGGC GGGGC

Claims (14)

1. A DNA molecule for use in raising an immune response to an antigen, the DNA molecule including a first sequence encoding a non-antibody targeting molecule in which the targeting molecule is a mammalian cell surface receptor counter receptor, a second sequence encoding the antigen or an epitope thereof, and optionally a third sequence encoding a polypeptide which promotes dimerisation or multimerisation of the product encoded by the DNA molecule.
2. A DNA molecule as claimed in claim 1 in which the antigen or epitope thereof encoded by the second sequence is a polypeptide which promotes dimerisation or multimerisation of the product encoded by the DNA molecule and the third sequence is absent.
3. A DNA molecule as claimed in claim 1 in which the third sequence is present.
4. A DNA molecule as claimed in any one of claims 1 to 3 in which the targeting molecule encoded by the first sequence is a ligand which targets lymphoid cells, lymphoid sites or antigen presenting cells.
A DNA molecule as claimed in any one of claims 1 to 4 in which the targeting molecule encoded by the first sequence is selected from the group consisting of CTLA4, L-selectin, CD40L, OX40 and CD28.
6. A DNA molecule as claimed in any one of claims 1 to 5 in which the targeting molecule encoded by the first sequence is CTLA4.
7. A polypeptide, the polypeptide being encoded by the DNA molecule as claimed in any one of claims 1 to 6.
8. A vector including the DNA molecule as claimed in any one of claims ito 6.
9. A composition for use in raising an immune response in an animal the composition including the DNA molecule as claimed in any one of claims 1 to 6 or the vector as claimed in claim 8 and an acceptable diluent or excipient. A composition for use in raising an immune response in an animal the composition including the polypeptide as claimed in claim 7 and an acceptable diluent or excipient.
AMENDED SHEET IPEA/AU WO 98/44129 PCT/AU98/00208 39
11. A method of raising an immune response in an animal, the method comprising administering to the animal the composition as claimed in claim 9 or claim
12. A method of deviating the immune response to an antigen in an individual, the method comprising administering to the individual a DNA molecule including a first sequence encoding CTLA4, a second sequence encoding the antigen or an epitope thereof, and optionally a third sequence encoding a polypeptide which promotes dimerisation or multimerisation of the product encoded by the DNA molecule.
13. A method as claimed in claim 12 in which the antigen or epitope thereof encoded by the second sequence is a polypeptide which promotes dimerisation or multimerisation of the product encoded by the DNA molecule and the third sequence is absent.
14. A method as claimed in claim 12 in which the third sequence is present.
AU64902/98A 1997-03-27 1998-03-26 Enhancement of immune response using targeting molecules Expired AU728962B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU64902/98A AU728962B2 (en) 1997-03-27 1998-03-26 Enhancement of immune response using targeting molecules

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
AUPO5891A AUPO589197A0 (en) 1997-03-27 1997-03-27 Enhancement of immune response using targeting molecules
AUPO5891 1997-03-27
AUPP1830 1998-02-13
AUPP1830A AUPP183098A0 (en) 1998-02-13 1998-02-13 Enhancement of immune response using targeting molecules
PCT/AU1998/000208 WO1998044129A1 (en) 1997-03-27 1998-03-26 Enhancement of immune response using targeting molecules
AU64902/98A AU728962B2 (en) 1997-03-27 1998-03-26 Enhancement of immune response using targeting molecules

Publications (2)

Publication Number Publication Date
AU6490298A AU6490298A (en) 1998-10-22
AU728962B2 true AU728962B2 (en) 2001-01-25

Family

ID=27155570

Family Applications (1)

Application Number Title Priority Date Filing Date
AU64902/98A Expired AU728962B2 (en) 1997-03-27 1998-03-26 Enhancement of immune response using targeting molecules

Country Status (1)

Country Link
AU (1) AU728962B2 (en)

Also Published As

Publication number Publication date
AU6490298A (en) 1998-10-22

Similar Documents

Publication Publication Date Title
Raz et al. Intradermal gene immunization: the possible role of DNA uptake in the induction of cellular immunity to viruses.
Kodihalli et al. Cross-protection among lethal H5N2 influenza viruses induced by DNA vaccine to the hemagglutinin
Robinson et al. Protection against a lethal influenza virus challenge by immunization with a haemagglutinin-expressing plasmid DNA
Chambers et al. Protection of chickens from lethal influenza infection by vaccinia-expressed hemagglutinin
JP3814828B2 (en) Peptide capable of inducing immune response against HIV and anti-AIDS prophylactic / therapeutic agent containing the peptide
Mitchell et al. Induction of heterosubtypic immunity to influenza A virus using a DNA vaccine expressing hemagglutinin-C3d fusion proteins
US5916879A (en) DNA transcription unit vaccines that protect against avian influenza viruses and methods of use thereof
Justewicz et al. Antibody-forming cell response to virus challenge in mice immunized with DNA encoding the influenza virus hemagglutinin
US9505806B2 (en) DNA vaccine, method of inducing the immune response, method of immunisation, antibodies specifically recognising the H5 haemagglutinin of an influenza virus and use of the DNA vaccine
JP2002531415A (en) Peptide-based vaccine for influenza
PT1670828E (en) Fully human antibodies against human 4-1bb (cd137)
Saha et al. A fused gene of nucleoprotein (NP) and herpes simplex virus genes (VP22) induces highly protective immunity against different subtypes of influenza virus
PT1968632E (en) Improved influenza vaccine
US20030035793A1 (en) Novel immune response targeting molecules
CA2626452A1 (en) Canine influenza virus and related compositions and methods of use
JP2001504496A (en) Immunization of infants
Yoon et al. Comparison of responses elicited by immunization with a Legionella species common lipoprotein delivered as naked DNA or recombinant protein
AU728962B2 (en) Enhancement of immune response using targeting molecules
WO1993020846A1 (en) Recombinant influenza vaccines
DK2948157T3 (en) APPLICATION OF A RE-MODIFIED INFECTIOUS MEASURED VIRUS WITH IMPROVED PROAPOPTOTIC PROPERTIES (MV DELTAC VIRUS) FOR CANCER THERAPY
US20230137174A1 (en) Novel salmonella-based coronavirus vaccine
US20030099670A1 (en) Influenza viruses with enhanced transcriptional and replicational capacities
AU2009319667A1 (en) Peptide adjuvants
Arnon et al. Synthetic Recombinant Vaccine Induces Anti-Influenza Long-Term Immunity and Cross-Strain Protection
CA2090005C (en) Cross-reactive influenza a immunization

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)