WO2005030971A1 - Modified vaccinia virus ankara (mva) mutant and use thereof - Google Patents

Modified vaccinia virus ankara (mva) mutant and use thereof Download PDF

Info

Publication number
WO2005030971A1
WO2005030971A1 PCT/EP2004/010858 EP2004010858W WO2005030971A1 WO 2005030971 A1 WO2005030971 A1 WO 2005030971A1 EP 2004010858 W EP2004010858 W EP 2004010858W WO 2005030971 A1 WO2005030971 A1 WO 2005030971A1
Authority
WO
WIPO (PCT)
Prior art keywords
mva
mutant
cells
vimses
vaccinia
Prior art date
Application number
PCT/EP2004/010858
Other languages
French (fr)
Inventor
Caroline Staib
Gerd Sutter
Sigried Kiesling
Volker Erfle
Original Assignee
Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh filed Critical Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh
Priority to EP04765665A priority Critical patent/EP1668142A1/en
Priority to CN2004800244195A priority patent/CN1842602B/en
Priority to BRPI0414874-6A priority patent/BRPI0414874A/en
Priority to AU2004276486A priority patent/AU2004276486B2/en
Publication of WO2005030971A1 publication Critical patent/WO2005030971A1/en
Priority to US11/375,159 priority patent/US7767209B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/275Poxviridae, e.g. avipoxvirus
    • A61K39/285Vaccinia virus or variola virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention is directed to a MVA mutant and its use in the immunotherapy and vaccination against numerous diseases, in particular in the prevention and therapy of cancer and infectious diseases.
  • Vaccinia vims belongs to the genus Orthopoxvims of the family of poxvimses. Certain strains of vaccinia vims have been used for many years as live vaccine to immunize against smallpox, for example the Elstree strain of the Lister Institute in the UK. Because of the complications which may derive from the vaccination (Schar, Zeitschr. fur Praventiv Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff Kunststoff-39]), and since the declaration in 1980 by the WHO that smallpox had been eradicated nowadays only people at high risk are vaccinated against smallpox.
  • Vaccinia viruses have also been used as vectors for production and delivery of foreign antigens (Smith et al, Biotechnology and Genetic Engineering Reviews 2, 383-407 [1984]). This entails DNA sequences (genes) which code for foreign antigens being introduced, with the aid of DNA recombination techniques, into the genome of the vaccinia viruses. If the gene is integrated at a site in the viral DNA which is non-essential for the life cycle of the vims, it is possible for the newly produced recombinant vaccinia vims to be infectious, that is to say able to infect foreign cells and thus to express the integrated DNA sequence (EP Patent Applications No. 83,286 and No. 110,385). The recombinant vaccinia viruses prepared in this way can be used, on the one hand, as live vaccines for the prophylaxis of infections, on the other hand, for the preparation of heterologous proteins in eukaryotic cells.
  • Vaccinia vims is amongst the most extensively evaluated live vectors and has particular features in support of its use as recombinant vaccine: It is highly stable, cheap to manufacture, easy to administer, and it can accommodate large amounts of foreign DNA. It has the advantage of inducing both antibody and cytotoxic responses, and allows presentation of antigens to the immune system in a more natural way, and it was successfully used as vector vaccine protecting against infectious diseases in a broad variety of animal models. Additionally, vaccinia vectors are extremely valuable research tools to analyze structure-function relationships of recombinant proteins, determine targets of humoral and cell-mediated immune responses, and investigate the type of immune defense needed to protect against a specific disease.
  • vaccinia vims is infectious for humans and its use as expression vector in the laboratory has been affected by safety concerns and regulations. Furthermore, possible future applications of recombinant vaccinia vims e.g. to generate recombinant proteins or recombinant viral particles for novel therapeutic or prophylactic approaches in humans, are hindered by the productive replication of the recombinant vaccinia vector. Most of the recombinant vaccinia vimses described in the literature are based on the Western Reserve (WR) strain of vaccinia vims. On the other hand, it is known that this strain is highly neurovirulent and is thus poorly suited for use in humans and animals (Morita et al., Vaccine 5, 65-70 [1987]).
  • WR Western Reserve
  • the MVA vims was deposited in compliance with the requirements of the Budapest Treaty at CNCM (Institut Pasteur, Collectione Nationale de Cultures de Microorganisms, 25, me de Dondel Roux, 75724 Paris Cedex 15) on Dec. 15, 1987 under Depositary No. 1-721.
  • Modified vaccinia virus Ankara is a chicken cell adapted strain of vaccinia vims. Because of its avimlence found upon inoculation of animals and its striking deficiency to produce substantial amounts of new viral progeny in most cells of malian origin MVA can be used under laboratory conditions of biosafety level 1. MVA serves as an efficient vector virus for expression of recombinant genes (Sutter & Moss 1992) and as candidate recombinant vaccine (Moss et al 1996) with high safety profile since MVA has been tested for preimmunization in over 100.000 humans being vaccinated against smallpox without causing notable side effects. Several MVA vector vaccines have already entered clinical evaluation (McConkey et al.
  • MVA is reassessed as candidate second generation vaccine against smallpox in comparison to immunizations with conventional vaccinia virus strains (Drexler et al. 2003, Belyakov et al. 2003).
  • MVA was obtained by long-term serial passage in chicken embryo fibroblast tissue cultures, which resulted in great loss of genomic information including many genes regulating vims-host interactions (Meyer et al. 1991, Antoine et al 1998).
  • the MVA homologues of genes encoding recognized poxvims immune evasion molecules including the viral interferon type I and type II receptors, the interleukin converting enzyme inhibitor SPI-2, the vaccinia complement binding protein, the vaccinia semaphorin, the 35 kDa chemokine binding protein or the tumor necrosis factor ⁇ receptor are deleted or fragmented.
  • ILl ⁇ R viral interleukin 1 ⁇ receptor
  • Interleukin 1 is a cytokine that plays an important role in regulation of inflammatory processes and host innate immune response against infectious agents.
  • the soluble viral ILl ⁇ R has specific affinity only for LLl ⁇ (Alcami & Smith Cell 1992), the major endogenous pyrogen (Alcami & Smith 1996).
  • vaccinia vims infection of mice ILl ⁇ R was shown to prevent fever by interaction with LLl ⁇ . Furthermore, deletion of the ILl ⁇ R gene in vaccinia vims accelerated the appearance of symptoms of illness and mortality in intranasally infected mice, suggesting that the blockade of ILl ⁇ by vaccinia vims can diminish the systemic acute phase response to infection and modulate the severity of the disease (Alcami & Smith 1996).
  • one object underlying the present invention is to provide a MVA mutant, showing less unwanted immunoreactions and, at the same time, having superior immunogenicity in the long term treatment of several diseases.
  • the effects of the deletion of the ILl ⁇ R gene from the MVA genome are evaluated.
  • the constmction of MVA ILl ⁇ R deletion mutants allowed to analyze the significance of ILl ⁇ R synthesis upon in vitro and in vivo infection with MVA.
  • the present data show that inactivation of the ILl ⁇ R gene is beneficial for the development of MVA vaccines.
  • a MVA mutant lacking the ILl ⁇ R gene showed no signs of fever or other illness, also after a high dose intranasal infection of mice with MVA- ⁇ ILl ⁇ R. This fact was absolutely unexpected, since the deletion of the LLl ⁇ R gene in vaccinia vims (which was disclosed before, see above) accelerated the appearance of symptoms of illness and mortality in intranasally infected mice.
  • Interleukin- 1 is an important regulator of inflammatory and immune responses that contributes to host defense against infection.
  • Vaccinia vims encodes a viral soluble ILIB receptor (vLLl ⁇ R), which modulates acute phase host response to infection (induction of fever) and might influence induction of immune responses against vims-associated antigens.
  • the inventors obtained MVA mutant vimses defective in vLLl ⁇ R production through transient insertion of selectable marker gene sequences, which precisely deleted the vILl ⁇ R coding sequences from the MVA genome. Analysis of MVA mutants indicated that deletion of the vILlBR gene did not abrogate the formation of MVA progeny upon tissue culture propagation.
  • mice with MVA- ⁇ LLl ⁇ R After high dose intranasal infection of mice with MVA- ⁇ LLl ⁇ R, animals showed no signs of fever or other illness suggesting that the avimlent phenotype remains preserved for MVA- ⁇ vILl ⁇ R.
  • MVA- ⁇ JJ 1BR or non-mutated MVA induced similar levels of vaccinia vims-specific circulating antibodies.
  • Vaccination with MVA- ⁇ ILl ⁇ R elicited somewhat higher levels of vaccinia virus epitope-specif ⁇ c T cells.
  • the present invention is directed to a MVA mutant, wherein the ILl ⁇ R coding sequence or a functional part thereof has been inactivated, preferably by deletion or mutation, which mutant may be used in immunotherapy and/or vaccination.
  • the term "functional part thereof as used herein is to be understood as any part of the LLl ⁇ R sequence, the loss of which is leading to an inactivation of the ILl ⁇ R function as described herein.
  • the inactivation is preferably performed by mutation or deletion. This loss of function can, as mentioned above, be seen in the induction of immune responses against virus-associated antigens. Therefore, it does not require more than routine experimentation to determine for a skilled person, whether a certain deletion or mutation is capable of performing this or not.
  • the immunological effect of enhancing CD8+ T cell responses may, for example, be evaluated by the method indicated in the Examples (see, in particular, Fig. 1) using a methodology as described in Tatsis N, Sinnathamby G, Eisenlohr LC; Methods Mol. Biol. 2004;269:267-288.
  • any deletion or mutation of ORF184 will be regarded as being sufficient, which will lead to a memory response of CD8+ cells, which is enhanced by at least 10%, preferably at least 20%, more preferably 30 or 40% and most preferably more than 50% compared to unmodified, i.e. wild type, MVA.
  • the ILl ⁇ R gene or a functional part thereof can be inactivated by deletion from the viral genome.
  • a recombinant MVA defective in J l ⁇ R sequence function may be generated by sequence mutagenesis, e.g. insertional mutagenesis, leading to the inactivation of ILl ⁇ R.
  • the MVA mutant of the present invention may additionally comprise a foreign DNA sequence, which can be a gene coding for a therapeutic polypeptide, e.g secreted proteins, e.g. polypeptides of antibodies, chemokines, cytokines or interferons, or a polypeptide from a pathogenic agent which can be used preferably for vaccination purposes or for the production of therapeutic or scientific valuable polypeptides.
  • a pathogenic agent which can be used preferably for vaccination purposes or for the production of therapeutic or scientific valuable polypeptides.
  • Pathogenic agents are to be understood to be vimses, bacteria and parasites which may cause a disease, as well as tumor cells which multiply unrestrictedly in an organism and may thus lead to pathological growths. Examples of such pathogenic agents are described in Davis, B.D.
  • genes of pathogenic agents are those of influenza vimses, of measles and respiratory syncytial vimses, of dengue vimses, of human immunodeficiency vimses, for example HIV I and HJV II, of human hepatitis vimses, e.g. HCV and HBV, of herpes viruses, of papilloma vimses, of the malaria parasite Plasmodium falciparam, and of the tuberculosis-causing Mycobacteria.
  • the MVA mutant of the present invention may be used for vaccination against smallpox or other diseases caused by orthopoxvims infections.
  • tumor associated antigens are those of melanoma-associated differentiation antigens, e.g. tyrosinase, tyrosinase-related proteins 1 and 2, of cancer testes antigens, e.g. MAGE- 1,-2,-3, and BAGE, of non-mutated shared antigens overexpressed on tumors, e.g. Her-2/neu, MUC-1, andp53.
  • melanoma-associated differentiation antigens e.g. tyrosinase, tyrosinase-related proteins 1 and 2
  • cancer testes antigens e.g. MAGE- 1,-2,-3, and BAGE
  • promoters are known to those skilled in the art, for example a vaccinia vims specific promoter as that of the vaccinia 11 kDa gene as are described in EP-A-198,328, and those of the 7.5 kDa gene (EP-A-110,385) or a heterologous poxvims promoter which allows for vaccinia virus specific transcription, or a synthetic promoter which allows for vaccinia virus specific transcription.
  • the ingredients of the present invention are preferably used in form of a pharmaceutical composition where they are mixed with suitable carriers or excipients in doses to treat or ameliorate the disease.
  • a composition may also contain (in addition to the ingredient and the carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers and other materials well known in the art.
  • pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s).
  • the characteristics of the carrier will depend on the route of administration.
  • the pharmaceutical composition may further contain other agents which either enhance the activity of the activity or use in treatment. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect or to minimize side-effects.
  • compositions are to be used for medical purposes, they will contain a therapeutically effective dose of the respective ingredient.
  • a therapeutically effective dose further refers to that amount of the compound/ingredient sufficient to result in amelioration of symptoms, e.g., treatment, healing, prevention or amelioration of such conditions.
  • the MVA vaccinia viruses generated according to the invention are converted into a physiologically acceptable form. This can be done based on the many years of experience in the preparation of vaccines used for vaccination against smallpox (Kaplan, Br. Med. Bull. 25, 131-135 [1969]). Typically, about 10 6 -10 7 particles of the recombinant MVA are freeze-dried in 100ml of phosphate -buffered saline (PBS) in the presence of 2% peptone and 1% human albumin in an ampoule, preferably a glass ampoule.
  • PBS phosphate -buffered saline
  • the lyophilisate can contain extenders (such as mannitol, dextran, sugar, glycine, lactose or polyvinylpyrrolidone) or other aids (such as antioxidants, stabilizers, etc.) suitable for parenteral administration.
  • extenders such as mannitol, dextran, sugar, glycine, lactose or polyvinylpyrrolidone
  • other aids such as antioxidants, stabilizers, etc.
  • the lyophilisate can be dissolved in 0.1 to 0.2 ml of aqueous solution, preferably physiological saline, and administered parenterally, for example by intradermal inoculation.
  • the vaccine according to the invention is preferably injected intracutaneously. Slight swelling and redness, sometimes also itching, may be found at the injection site (Stickl et al., supra).
  • the mode of administration, the dose and the number of administrations can be optimized by those skilled in the art in a known manner. It is expedient where appropriate to administer the vaccine several times over a lengthy period in order to obtain a high level immune responses against the foreign antigen.
  • the MVA mutant of the present invention may be used in a method for treating a human patient in need of an immunotherapy and/or vaccination (which is, for example, suffering from cancer and/or infectious diseases), which is characterized in administering a therapeutically effective amount of a MVA mutant/vaccine of the invention to said patient.
  • the physician in any event will determine the actual dosage which will be most suitable for an individual patient and will vary with the age, weight and response of the particular patient. There can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • the pharmaceutical composition is adapted for in vivo use in a mammal, preferably a human patient.
  • a method of generating mutant MVA comprising the steps of: - Infecting host cells of MVA with a nucleic acid coding for the MVA mutants as defined above, - expressing said nucleic acid under suitable conditions in said host cells; and - isolating expressed mutant MVA.
  • the present invention provides a method for generating a pharmaceutical composition comprising the steps of: - Infecting host cells of MVA with a nucleic acid coding for the MVA mutants as defined above, - expressing said nucleic acid under suitable conditions in said host cells; - isolating expressed mutant MVA; - adding a pharmaceutically acceptable carrier and further ingredients in order to manufacture a pharmaceutical composition.
  • the host cells are CEF cells, chicken embiyo derived LSCC-H32 cells, chicken DF-1 cells or avian cells, e.g. quail fibroblasts QT6 or QT35 cells.
  • mutant MVA are selected by the KIL gene based host range selection protocol (Staib et al, "Transient Host Range Selection For Genetic Engineering Of Modified Vaccinia Virus Ankara” BioTechniques 28: 1137-1148 (June 2000).
  • MVA Compared to its parental strain, MVA has deletions that consist of about 15 percent (30,000 base pairs) of its former genome, including most of the KIL gene. Only a fragment of a length of 263 bp is still present in the MVA genome.
  • the MVA KIL gene sequences represent the first 263 bp of the ORF 022L in the MVA genome at position nt 20685-20981 as described in Antoine, G., F. Scheiflinger, F. Dorner, and F. G. Falkner. 1998.
  • the complete genomic sequence of the modified vaccinia Ankara strain and a comparison with other orthopoxvims es can be found in: Virology 244:365-396.
  • the present invention provides the use of a MVA mutant as defined hereinabove or of a pharmaceutical composition, see above, for the manufacture of a medicament for use in immunotherapy and/or vaccination. Furthermore, the present invention is directed to a method for treating a patient, comprising a therapeutically effective dose of mutant MVA or of a pharmaceutical composition as defined herein to an individual in need of said treatment. Regarding the way and form of administration, see also above.
  • the MVA mutant of the present invention may preferably used for vaccination against smallpox or other diseases caused by orthopoxvims infections.
  • FIG. 1 Constmction of Ll ⁇ R deficient MVA.
  • Upper panel schematic map of the MVA genome. Sites of the restriction endonuclease H dIII within the genome of MVA are indicated.
  • the position of the 184R ORF (ILl ⁇ R gene) is marked by an anOw.
  • MVA DNA sequences adjacent to the ILl ⁇ R coding sequence flank 184R-I, flank 184R-JJ
  • the final mutant vims MVA- ⁇ ILl ⁇ R was obtained after the deletion of the KIL marker gene during a second step of homologous recombination involving synthetic repetitive sequences (rep).
  • FIG. 2 In vitro characterization of MVA- ⁇ LLl ⁇ R.
  • A PCR analysis of viral DNA. Genomic template DNA was prepared from MVA- ⁇ ILl ⁇ R (lane 1) or MVA (lane 2) infected cells and incubated with oligonucleotides adjacent to the 184R gene locus to amplify specific DNA fragments. PCR products were separated by agarose gel electrophoresis. M, molecular weight marker.
  • B Southern blot analysis of viral DNA. Genomic DNA was prepared from MVA- ⁇ LLl ⁇ R (lane 1) or MVA (lane2) infected cells, digested with EcoRl, separated by agarose gel electrophoresis and transferred to a nylon membrane.
  • FIG. 4 Ex vivo analysis of vaccine induced CD8+ T cells.
  • HHD mice were vaccinated with a single dose of MVA- ⁇ ILl ⁇ R, MVA or a revertant vims. After 10 days, splenocytes were stimulated with the HLA-A*0201 resticted vaccinia specific VP35#1 (VP35#1) or influenza Ml 58-66 (inelevant control) peptide, then stained with EMA, PE-anti-CD8, APC-anti- CD62L and FITC-anti-IFN ⁇ or -anti-TNF ⁇ or the respective FITC-labelled isotype control.
  • FIG. 5 Analysis of vaccine-induced protection in a mouse model for respiratory poxvims infection.
  • Three weeks after vaccination animals were challenged intranasally with lxlO 6 PFU WR.
  • Individual animal weights (A, B) and signs of illness (C, D) were monitored daily and are expressed as means for each group.
  • Mock vaccinated ( ⁇ ) and mock challenged ( ⁇ ) mice served as control groups.
  • FIG. 6 Ex vivo analysis of vaccine induced memory CD8+ T cells and analysis of vaccine- induced long-term protection in HHD mice.
  • A Mice were vaccinated intraperitoneally with a single dose 10 8 IU of MVA- ⁇ ILl ⁇ R or MVA. After 6 month, splenocytes were either stimulated with the HLA-A*0201 resticted vaccinia specific VP35#1 peptide or infected with MVA for detection of vaccinia specific total CD8+ or CD4+ responses. Cells were stained with EMA, PE-anti-CD8, APC-anti-CD62L and FITC-anti-IFN ⁇ or the respective FITC-labelled isotype control.
  • mice were analyzed by flow cytometry for the presence of VP35#1 peptide or vaccinia-specific, activated (CD62L low ) CD8+ T cells or vaccinia specific CD4+ T cells.
  • mice were challenged with lxl 0 7 PFU WR. Survival was monitored daily and is expressed as percentage of surviving animals per group.
  • Mock vaccinated ( ⁇ ) and mock challenged ( ⁇ ) mice served as control groups.
  • FIG. 7 Ex vivo analysis of vaccine induced memoiy CD8+ T cells and analysis of vaccine- induced long-term protection in non transgenic mice.
  • A C57BL/6 mice were vaccinated intraperitoneally with a single dose 10 8 IU of MVA- ⁇ ILl ⁇ R or MVA. After 6 month, splenocytes were infected with MVA for detection of vaccinia specific total CD8+ or CD4+ responses. Cells were stained with EMA, PE-anti-CD8, APC-anti-CD62L and FITC-anti- IFN ⁇ or the respective FITC-labelled isotype control.
  • mice were analyzed by flow cytometry for the presence of vaccinia-specific, activated (CD62L low ) CD8+ T cells or vaccinia specific CD4+ T cells.
  • Vaccinia vims strains Western Reserve, CVA and MVA (cloned isolate F6, from the 582 nd passage on chicken embryo fibroblasts (CEF)) were used for this study. All vimses were propagated and titered following standard methodology. To generate vaccine preparations, viruses were routinely purified by ultra centrifugation through sucrose and reconstituted in 1 mM Tris pH 9.0. CEF and rabbit kidney RK-13 (ATCC CCL-37) cells were grown in minimal essential medium (MEM) supplemented with 10% fetal calf semm (FCS), and maintained at 37 °C and 5% CO 2 .
  • MEM minimal essential medium
  • FCS fetal calf semm
  • the transfer plasmid p ⁇ KlL-184R carries two DNA fragments that represent flanking sequences of MVA ORF 184R (nucleotide position 162021-163001, GenBank U94848) and which were inserted into multiple cloning sites 1 and 2 of plasmid p ⁇ KIL (Staib et al. 2000).
  • flank 184-1 consists of a 486-bp MVA-DNA sequence starting in the 5' intergenic region of ORF 184R and ending at the start codon for translation of ORF 184R, the other fragment, flank 184-2, is a 544-bp PCR-fragment of MVA-DNA extending from the codon for 184R translation termination into the 3'- intergenic region of the 184R gene.
  • Mutant MVA were obtained following the transient KlL-based host range selection protocol as described previously (Staib et al, 2000). Briefly, for generation of deletion mutant vimses, monolayers of lxlO 6 confluent CEF cells were infected with MVA at a multiplicity of infection (MOI) of 0.01 IU per cell. Ninety minutes after infection cells were transfected with 1.5 ⁇ g of plasmid p ⁇ KlL-184R DNA using FUGENETM (Roche, Mannheim, Germany) as recommended by the manufacturer. At 48 h after infection, transfected cells were harvested and plated on RK-13 cell monolayers for growth selection. Mutant viruses were isolated through plaque cloning on RK-13 cells and then passaged on CEF cells to remove the selectable marker gene KIL.
  • MOI multiplicity of infection
  • Genomic viral DNA was isolated from infected CEF cells and analyzed by PCR using oligonucleotides annealing within the flanking regions flankl84-l and -2, respectively (pair 1) or within flankl84-l and the coding region of 184R, respectively (pair 2) (Staib et al, 2000). Specific DNA fragments were amplified by 30 cycles of PCR at an annealing temperature of 52°C (pair 1) or 50°C (pair 2).
  • total DNA isolated from vims -infected cells was digested with EcoRI, separated by gel electrophoresis in 0.8% agarose, transferred to a Hybond TM-N membrane (Amersham, Freiburg, Germany), and hybridized to a DNA probe consisting of a PCR fragment from 184R-flankl sequences labeled with [ ⁇ - 32 P]CTP.
  • Prehybridization and hybridization was performed according to Sambrook et al. (Southern 1975, Sambrook et al, 1989). Blots were exposed to a Kodak BioMax film.
  • Radioimmunoprecipitation of virus-infected cell lysates C ⁇ F cells grown in 6-well tissue culture plates were infected with a multiplicity of 20 infectious units MVA. At 2h post infection, the virus inoculum was replaced with methionine-free minimal essential medium containing 5% dialyzed fetal calf serum and 50 ⁇ Ci of [ 35 S]methionine per ml and incubated overnight at 37 °C.
  • RTPA-buffer containing 0.15 M NaCl, 0.01 M Tris-HCl (pH 7.4), 1% Triton X-100, and incubated for 14 h with rabbit polyclonal antibody AcB15R (Alcami & Smith, 1992), followed by 50% protein A - sepharose suspension. Immune complexes were washed in RJJPA-buffer, resuspended in Laemmli buffer, and proteins were separated by electrophoresis in a 10% SDS-polyacrylamide gel. Analysis of virus growth.
  • confluent CEF monolayers grown on 6 well plates were infected with 0.05 infectious units (IU) or 10 IU MVA or mutant MVA per cell, respectively. After vims adsorption for 60 min at 37 °C, the inoculum was removed. Cells were washed twice with RPMI 1640 and incubated with fresh RPMI 1640 medium containing 10% FCS at 37 °C and 5% CO2- At multiple time points post infection (p.i.) infected cells were harvested and virus was released by freeze-thawing and brief sonication. Serial dilutions of the resulting lysates were plated on confluent CEF monolayers grown in 6-well plates as replicates of two.
  • Humoral vaccinia virus responses Serum samples from mice immunized with MVA, mutant MVA, or vaccinia vims CVA were assessed for antibodies to vaccinia virus proteins by ELISA and neutralizing-antibody assay.
  • Vaccinia antigen-specific binding titers were determined by an ELISA in which Maxisorp plates (Nunc, Germany) were coated with sucrose-gradient purified MVA (at a protein concentration of l ⁇ g/ml) for 3h at 37 °C and overnight at 4 °C. Plates were blocked with PBS / 0.05% Tween 20 / 10% fetal calf serum for 60 min at 37 °C.
  • Vaccinia vims-specific neutralizing antibodies were analyzed by a plaque reduction assay using recombinant MVA-LZ. Twofold serial dilutions of sera were mixed with 200 infectious units MVA-LZ in a total volume of 200 ⁇ l PBS and incubated for two hours at 37 °C. Afterwards, confluent CEF monolayers (grown on 24 well plates) were infected in duplicate, and foci of vims infected cells were visualized 48 hours after inoculation by staining with 5-bromo-4-chloro-3-indolyl- ⁇ -galactopyranoside substrate (X-Gal, Roche Molecular Biochemicals, Mannheim Germany) as described previously (Drexler et al., 1998). Blue-stained foci were counted and the number obtained with each serum was compared to controls with mouse preimmune sera or medium control. Antibody titers were calculated as the serial twofold dilution yielding a 50% reduction of foci numbers.
  • splenocytes from vaccinia virus-immunized HHD mice were prepared and incubated for 5 h with A*0201 -binding peptides at 10 "6 M. After 2 h, brefeldin A was added at a final concentration of 1 ⁇ g/ml (GolgiPlugTM; PharMingen Becton Dickinson).
  • Cells were then either stored ON on ice at 4°C, or directly live/dead stained in PBS/1%BSA/1 ⁇ g/ml ethidium monoazide bromide (EMA; Molecular Probes) and blocked for unspecific Fc ⁇ in and -II receptor-mediated binding with 5 ⁇ g/ml purified anti- CD 16/CD32 (Fc BlockTM; PharMingen Becton Dickinson) for 20 min at 4°C. Cell surface staining was performed with PE-anti-CD8 (53-6.7) and APC-anti-CD62L (Mel-14) for 30 min at 4°C.
  • EMA ethidium monoazide bromide
  • HHD mice Female six to eight week-old transgenic HHD +/+ ⁇ 2 m "/_ D " mice (HHD) (Pascolo et al, 1997) or female six to eight week old BALB/c or C57BL/6 mice were used for vaccination experiments. HHD mice were inoculated with 0.5 ml volumes of vims vaccine by the intraperitoneal route, and monitored for HLA-A*0201-restricted T cell responses at days 10 or 180 after immunization. For protection assays, animals were vaccinated once with 0.1 - 0.5 ml vims vaccine given by intramuscular or intraperitoneal route.
  • mice were anesthetized, infected intranasally with vaccinia vims Western Reserve diluted in 30 ⁇ l phosphate buffered saline, and monitored for at least further three weeks for morbidity and mortality with daily measurement of individual body weights and scoring of signs of illness as described previously. Animals suffering from severe systemic infection and having lost>30% of body weight were sacrificed. The mean change in body weight was calculated as the percentage of the mean weight for each group on the day of challenge. Body temperature was determined with a Electronic Laboratory Animal Monitoring System (BioMedic Data Systems, Marywood, NJ) using subcutaneously implanted microchip battery-free transponders and a DAS-5004 Pocket Scanner for data collection. Mean changes in body temperatures were calculated by subtracting the pre-challenge (days -3 to 0) baseline temperature of each group from each subsequent time point.
  • Furthemiore we digested viral DNAs with restiiction endonuclease EcoRI and revealed DNA fragments containing the ILl ⁇ R gene locus by Southern blot analysis. Confirming the PCR data, we detected an about 1.3-kb lower molecular weight EcoRI- fragment in the genomic DNA of deletion mutant MVA- ⁇ ILl ⁇ R (Fig. 2B) again confirming the proper deletion of the targeted ORF 184R sequences. In a second step we wanted to prove that ⁇ L l ⁇ R protein is produced during MVA infection and to demonstrate that the generated mutants fail to synthesize this polypeptide.
  • mice with subcutaneous microchip transponders to allow for computable readings, one week later infected the animals with 108 IU of MVA or MVA- ⁇ ILl ⁇ R or with 5xl0 5 PFU of replication competent vaccinia vims CVA or 3x10 4 PFU Western Reserve (WR) as control, and monitored animals daily over a period of three weeks (Fig. 3). Infection of mice with MVA or mutant MVA- ⁇ ILl ⁇ R did not result in any obvious disease.
  • MVA- ⁇ ILl ⁇ R vaccination improves T cell memory response and long-term protective capacity.
  • the mature form of the inflammatory cytokine ILl ⁇ has multiple effects in vivo as revealed by the study of mice deficient for different components of the EL-1 system (33).
  • An active area of ongoing research on IL1 function is to elucidate the likely importance of the cytokine in protective T cell immunity including the activation of professional antigen presenting cells and memory T cells (16, 17).
  • HHD mice HLA-A*0201 transgenic mice once with 10 8 IU MVA- ⁇ ILl ⁇ R or MVA and monitored for vims-specific T cells more than six months after this primary immunization.
  • VP35#1 -specific memoiy T cells can be detected using our standard protocol for ICS/FACS analysis (0.30-0.60%) (11).
  • An improved protocol using overnight incubation of peptide stimulated splenocytes allows to notice a more prominent population of antigen-specific CD8+ T cells often exceeding the number of activated T cells found with our conventional protocol during the acute phase response.
  • the HHD mouse model allows for convenient analysis of epitope specific HLA- A*0201 -restricted CD8+ T cell responses, yet, possibly because of their knock-out phenotype for mouse MHC class I, these mice develop unusually low numbers of total CD8+ T cells (but normal levels of CD4+ T cells) (11).
  • mice having received MVA- ⁇ ILl ⁇ R vaccine survived the challenge, and animals in the group vaccinated with 10 7 IU of MVA- ⁇ EL l ⁇ R demonstrated an average ⁇ 15% reduction of body weight and only mild signs of illness (data not shown).
  • the MVA ORF 184R encodes a vaccinia viral soluble receptor for IL-l ⁇ with proposed function to block inflammatory and febrile host response to infection.
  • the removal of putative immune evasion genes from viral genomes is a promising approach to further elucidate roles of these regulatory vims proteins in the in vivo viral life cycle (1).
  • application of this research to a vims such as MVA being suitable for use as (recombinant) live viral vaccine may directly lead to second generation vaccines with rationally improved properties.
  • Even by means of molecular engineering techniques allowing for precise mutagenesis (35, 36) the resulting phenotypes of mutant vimses are unpredictable, and the inactivation of the MVA ILl ⁇ R gene may serve as further example.
  • the H3L gene product-derived epitope VP35#1 is the target of an immunodominant HLA-A*0201 -restricted T cell specificity and can be used to examine the induction of vims-specific CD8+ T cells in an epitope-specific manner (10, 12).
  • bulk analysis of total vaccinia virus-specific responses offers a representative picture based on a multitude of different T cell specificities but might not allow to assess subtile changes in the activation of single T cell populations.
  • Suspecting a possible effect on T cell activation we opted to also assess VP35#1 -specific T cell memory responses which we had previously found detectable for more than 6 month after vaccination of HLA-A*0201 transgenic mice (12).
  • the capacity of the viral ILl ⁇ R to apparently specifically downmodulate anti-viral CD8+ T cell memoiy responses appears alluring in the view of recent data from the analysis of vaccinia vims-specific memoiy T cells in humans (after vaccination with conventional wildtype vaccinia virus) showing better persistence of CD4+ than CD8+ T cells (4).
  • our analysis recommends deletion of the viral ILl ⁇ R gene as a first step in approaching the development of a new generation of MVA-based vaccines. High virus titers could be obtained upon in vitro propagation of deletion mutant MVA- ⁇ ILl ⁇ R, and there was no evidence of MVA- ⁇ ILl ⁇ R being less well tolerated than wildtype vims upon high dose in vivo infection.
  • Nonreplicating vaccinia vector efficiently expresses recombinant genes. Proc Natl Acad Sci USA 89:10847-10851.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Biomedical Technology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Plant Pathology (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention is directed to a MVA mutant and its use in the immunotherapy and vaccination against numerous diseases, in particular in the prevention and therapy of cancer and infectious diseases. The construction of MVA IL 1βR deletion mutants allowed to analyze the significance of IL 1βR synthesis upon in vitro and in vivo infection with MVA. The present data show that inactivation of the IL 1βR gene is beneficial for the development of MVA vaccines.

Description

MODIFIED VACCINIA VIRUS ANKARA (MVA) MUTANT AND USE THEREOF
The present invention is directed to a MVA mutant and its use in the immunotherapy and vaccination against numerous diseases, in particular in the prevention and therapy of cancer and infectious diseases.
Vaccinia vims ( W) belongs to the genus Orthopoxvims of the family of poxvimses. Certain strains of vaccinia vims have been used for many years as live vaccine to immunize against smallpox, for example the Elstree strain of the Lister Institute in the UK. Because of the complications which may derive from the vaccination (Schar, Zeitschr. fur Praventivmedizin 18, 41-44 [1973]), and since the declaration in 1980 by the WHO that smallpox had been eradicated nowadays only people at high risk are vaccinated against smallpox.
Vaccinia viruses have also been used as vectors for production and delivery of foreign antigens (Smith et al, Biotechnology and Genetic Engineering Reviews 2, 383-407 [1984]). This entails DNA sequences (genes) which code for foreign antigens being introduced, with the aid of DNA recombination techniques, into the genome of the vaccinia viruses. If the gene is integrated at a site in the viral DNA which is non-essential for the life cycle of the vims, it is possible for the newly produced recombinant vaccinia vims to be infectious, that is to say able to infect foreign cells and thus to express the integrated DNA sequence (EP Patent Applications No. 83,286 and No. 110,385). The recombinant vaccinia viruses prepared in this way can be used, on the one hand, as live vaccines for the prophylaxis of infections, on the other hand, for the preparation of heterologous proteins in eukaryotic cells.
Vaccinia vims is amongst the most extensively evaluated live vectors and has particular features in support of its use as recombinant vaccine: It is highly stable, cheap to manufacture, easy to administer, and it can accommodate large amounts of foreign DNA. It has the advantage of inducing both antibody and cytotoxic responses, and allows presentation of antigens to the immune system in a more natural way, and it was successfully used as vector vaccine protecting against infectious diseases in a broad variety of animal models. Additionally, vaccinia vectors are extremely valuable research tools to analyze structure-function relationships of recombinant proteins, determine targets of humoral and cell-mediated immune responses, and investigate the type of immune defense needed to protect against a specific disease.
However, vaccinia vims is infectious for humans and its use as expression vector in the laboratory has been affected by safety concerns and regulations. Furthermore, possible future applications of recombinant vaccinia vims e.g. to generate recombinant proteins or recombinant viral particles for novel therapeutic or prophylactic approaches in humans, are hindered by the productive replication of the recombinant vaccinia vector. Most of the recombinant vaccinia vimses described in the literature are based on the Western Reserve (WR) strain of vaccinia vims. On the other hand, it is known that this strain is highly neurovirulent and is thus poorly suited for use in humans and animals (Morita et al., Vaccine 5, 65-70 [1987]).
Concerns with the safety of standard strains of VV have been addressed by the development of vaccinia vectors from highly attenuated vims strains which are characterized by their restricted replicative capacity in vitro and their avimlence in vivo. Strains of vimses specially cultured to avoid undesired side effects have been known for a long time. Thus, it has been possible, by long-term serial passages of the Ankara strain of vaccinia vims (CVA) on chicken embryo fibroblasts, to culture a modified vaccinia vims Ankara (MVA) (for review see Mayr, A., Hochstein-Mintzel, V. and Stick!, H. (1975) Infection 3, 6-14; Swiss Patent No. 568 392). The MVA vims was deposited in compliance with the requirements of the Budapest Treaty at CNCM (Institut Pasteur, Collectione Nationale de Cultures de Microorganisms, 25, me de Docteur Roux, 75724 Paris Cedex 15) on Dec. 15, 1987 under Depositary No. 1-721.
Modified vaccinia virus Ankara (MVA) is a chicken cell adapted strain of vaccinia vims. Because of its avimlence found upon inoculation of animals and its striking deficiency to produce substantial amounts of new viral progeny in most cells of mamalian origin MVA can be used under laboratory conditions of biosafety level 1. MVA serves as an efficient vector virus for expression of recombinant genes (Sutter & Moss 1992) and as candidate recombinant vaccine (Moss et al 1996) with high safety profile since MVA has been tested for preimmunization in over 100.000 humans being vaccinated against smallpox without causing notable side effects. Several MVA vector vaccines have already entered clinical evaluation (McConkey et al. 2003, Cosma et al. 2003). Most recently MVA is reassessed as candidate second generation vaccine against smallpox in comparison to immunizations with conventional vaccinia virus strains (Drexler et al. 2003, Belyakov et al. 2003).
As indicated above, MVA was obtained by long-term serial passage in chicken embryo fibroblast tissue cultures, which resulted in great loss of genomic information including many genes regulating vims-host interactions (Meyer et al. 1991, Antoine et al 1998). The MVA homologues of genes encoding recognized poxvims immune evasion molecules (for review see Moss & Shisler 2001, Alcami 2003) including the viral interferon type I and type II receptors, the interleukin converting enzyme inhibitor SPI-2, the vaccinia complement binding protein, the vaccinia semaphorin, the 35 kDa chemokine binding protein or the tumor necrosis factor α receptor are deleted or fragmented. Interestingly, some viral genes with irnmunomodulatory function are maintained in the MVA genome and their possible relevance for the use of MVA-based vaccines remains to be determined. One such example is the coding sequence for the viral interleukin 1 β receptor (ILlβR) that is highly conserved in MVA. Interleukin 1 is a cytokine that plays an important role in regulation of inflammatory processes and host innate immune response against infectious agents. In contrast to its cellular counterpart, the soluble viral ILlβR has specific affinity only for LLlβ (Alcami & Smith Cell 1992), the major endogenous pyrogen (Alcami & Smith 1996). During vaccinia vims infection of mice ILlβR was shown to prevent fever by interaction with LLlβ. Furthermore, deletion of the ILlβR gene in vaccinia vims accelerated the appearance of symptoms of illness and mortality in intranasally infected mice, suggesting that the blockade of ILlβ by vaccinia vims can diminish the systemic acute phase response to infection and modulate the severity of the disease (Alcami & Smith 1996).
It is noted that a MVA mutant, in which the ILlβR gene has been inactivated, was already disclosed in Staib et al. in „Transient Host Range Selection for Genetic Engineering of Modified Vaccinia Virus Ankara", BioTechniques 28: 1137-1148 (June 2000). The deleted ILlβR gene sequence is termed and corcesponds to ORF 184R. The whole genome of the MVA was disclosed in Antoine et al, Journal of Virology, 1998, which is incoiporated herein by reference. However, no information was presented regarding the immunogenicity or further characteristics of the mutant, which could show their potential use in the prevention or therapy of numerous diseases.
As a summary, there remains a demand for an improvement of the already existing MVA strains in view of their immunogenicity and/or their protective capacities, when used as vaccines.
Therefore, one object underlying the present invention is to provide a MVA mutant, showing less unwanted immunoreactions and, at the same time, having superior immunogenicity in the long term treatment of several diseases.
This object is accomplished by the subject-matter of the independent claims. Preferred embodiments of the present invention are set forth in the dependent claims.
In this invention, the effects of the deletion of the ILlβR gene from the MVA genome are evaluated. The constmction of MVA ILlβR deletion mutants allowed to analyze the significance of ILlβR synthesis upon in vitro and in vivo infection with MVA. The present data show that inactivation of the ILlβR gene is beneficial for the development of MVA vaccines.
Suφrisingly, it turned out that an inactivation of the viral interleukin lβ receptor enhances CD8+ T cell responses elicited upon immunization with modified vaccinia vims Ankara.
Moreover, is could be shown herein that a MVA mutant lacking the ILlβR gene showed no signs of fever or other illness, also after a high dose intranasal infection of mice with MVA- ΔILlβR. This fact was absolutely unexpected, since the deletion of the LLlβR gene in vaccinia vims (which was disclosed before, see above) accelerated the appearance of symptoms of illness and mortality in intranasally infected mice.
Interleukin- 1 (ILl) is an important regulator of inflammatory and immune responses that contributes to host defense against infection. Vaccinia vims encodes a viral soluble ILIB receptor (vLLlβR), which modulates acute phase host response to infection (induction of fever) and might influence induction of immune responses against vims-associated antigens. The inventors obtained MVA mutant vimses defective in vLLlβR production through transient insertion of selectable marker gene sequences, which precisely deleted the vILlβR coding sequences from the MVA genome. Analysis of MVA mutants indicated that deletion of the vILlBR gene did not abrogate the formation of MVA progeny upon tissue culture propagation. After high dose intranasal infection of mice with MVA-ΔLLlβR, animals showed no signs of fever or other illness suggesting that the avimlent phenotype remains preserved for MVA-ΔvILlβR. Upon vaccination of mice MVA-ΔJJ 1BR or non-mutated MVA induced similar levels of vaccinia vims-specific circulating antibodies. Vaccination with MVA-ΔILlβR elicited somewhat higher levels of vaccinia virus epitope-specifϊc T cells. Yet, surprisingly a significantly superior immunogenicity of MVA-ΔLLlβR (p=0.01) was found when memory T cell responses were monitored at six months after vaccination. Moreover, while we found equal protective capacities for MVA-ΔLLlβR and wild-type MVA three weeks after immunization, at six months after vaccination MVA-ΔB lβR protected better (5/5 =100%, MVA 3/5 =60%) against the lethal respiratory challenge with virulent vaccinia vims strain Western Reserve. Therefore, the data presented herein suggest that deletion of vLLlβR gene sequences may be considered as first step towards obtaining genetically optimized MVA viruses for the development of vaccines with even improved immunogenicity.
The present invention is in particular directed to the following aspects and embodiments:
The present invention is directed to a MVA mutant, wherein the ILlβR coding sequence or a functional part thereof has been inactivated, preferably by deletion or mutation, which mutant may be used in immunotherapy and/or vaccination.
The term "functional part thereof as used herein is to be understood as any part of the LLlβR sequence, the loss of which is leading to an inactivation of the ILlβR function as described herein. The inactivation is preferably performed by mutation or deletion. This loss of function can, as mentioned above, be seen in the induction of immune responses against virus-associated antigens. Therefore, it does not require more than routine experimentation to determine for a skilled person, whether a certain deletion or mutation is capable of performing this or not. In particular, the immunological effect of enhancing CD8+ T cell responses may, for example, be evaluated by the method indicated in the Examples (see, in particular, Fig. 1) using a methodology as described in Tatsis N, Sinnathamby G, Eisenlohr LC; Methods Mol. Biol. 2004;269:267-288.
In particular, any deletion or mutation of ORF184 will be regarded as being sufficient, which will lead to a memory response of CD8+ cells, which is enhanced by at least 10%, preferably at least 20%, more preferably 30 or 40% and most preferably more than 50% compared to unmodified, i.e. wild type, MVA.
Generally, the ILlβR gene or a functional part thereof can be inactivated by deletion from the viral genome. Alternatively, a recombinant MVA defective in J lβR sequence function may be generated by sequence mutagenesis, e.g. insertional mutagenesis, leading to the inactivation of ILlβR.
The MVA mutant of the present invention may additionally comprise a foreign DNA sequence, which can be a gene coding for a therapeutic polypeptide, e.g secreted proteins, e.g. polypeptides of antibodies, chemokines, cytokines or interferons, or a polypeptide from a pathogenic agent which can be used preferably for vaccination purposes or for the production of therapeutic or scientific valuable polypeptides. Pathogenic agents are to be understood to be vimses, bacteria and parasites which may cause a disease, as well as tumor cells which multiply unrestrictedly in an organism and may thus lead to pathological growths. Examples of such pathogenic agents are described in Davis, B.D. et al., (Microbiology, 3rd ed., Harper International Edition). Preferred genes of pathogenic agents are those of influenza vimses, of measles and respiratory syncytial vimses, of dengue vimses, of human immunodeficiency vimses, for example HIV I and HJV II, of human hepatitis vimses, e.g. HCV and HBV, of herpes viruses, of papilloma vimses, of the malaria parasite Plasmodium falciparam, and of the tuberculosis-causing Mycobacteria.
Additionally, the MVA mutant of the present invention may be used for vaccination against smallpox or other diseases caused by orthopoxvims infections.
Preferred genes encoding tumor associated antigens are those of melanoma-associated differentiation antigens, e.g. tyrosinase, tyrosinase-related proteins 1 and 2, of cancer testes antigens, e.g. MAGE- 1,-2,-3, and BAGE, of non-mutated shared antigens overexpressed on tumors, e.g. Her-2/neu, MUC-1, andp53.
In order for it to be possible for the foreign DNA sequence or the gene to be expressed, it is necessary for regulatory sequences, which are required for the transcription of the gene, to be present on the DNA. Such regulatoiy sequences (called promoters) are known to those skilled in the art, for example a vaccinia vims specific promoter as that of the vaccinia 11 kDa gene as are described in EP-A-198,328, and those of the 7.5 kDa gene (EP-A-110,385) or a heterologous poxvims promoter which allows for vaccinia virus specific transcription, or a synthetic promoter which allows for vaccinia virus specific transcription.
The ingredients of the present invention are preferably used in form of a pharmaceutical composition where they are mixed with suitable carriers or excipients in doses to treat or ameliorate the disease. Such a composition may also contain (in addition to the ingredient and the carrier) diluents, fillers, salts, buffers, stabilizers, solubilizers and other materials well known in the art. The term "pharmaceutically acceptable" means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration. The pharmaceutical composition may further contain other agents which either enhance the activity of the activity or use in treatment. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect or to minimize side-effects.
Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences", Mack Publishing Co., Easton, PA, latest edition. Whenever the compositions are to be used for medical purposes, they will contain a therapeutically effective dose of the respective ingredient. A therapeutically effective dose further refers to that amount of the compound/ingredient sufficient to result in amelioration of symptoms, e.g., treatment, healing, prevention or amelioration of such conditions.
To prepare vaccines, the MVA vaccinia viruses generated according to the invention are converted into a physiologically acceptable form. This can be done based on the many years of experience in the preparation of vaccines used for vaccination against smallpox (Kaplan, Br. Med. Bull. 25, 131-135 [1969]). Typically, about 106-107 particles of the recombinant MVA are freeze-dried in 100ml of phosphate -buffered saline (PBS) in the presence of 2% peptone and 1% human albumin in an ampoule, preferably a glass ampoule. The lyophilisate can contain extenders (such as mannitol, dextran, sugar, glycine, lactose or polyvinylpyrrolidone) or other aids (such as antioxidants, stabilizers, etc.) suitable for parenteral administration. The glass ampoule is then sealed and can be stored, preferably at temperatures below -20°C, for several months.
For vaccination the lyophilisate can be dissolved in 0.1 to 0.2 ml of aqueous solution, preferably physiological saline, and administered parenterally, for example by intradermal inoculation. The vaccine according to the invention is preferably injected intracutaneously. Slight swelling and redness, sometimes also itching, may be found at the injection site (Stickl et al., supra). The mode of administration, the dose and the number of administrations can be optimized by those skilled in the art in a known manner. It is expedient where appropriate to administer the vaccine several times over a lengthy period in order to obtain a high level immune responses against the foreign antigen.
Thus, the MVA mutant of the present invention may be used in a method for treating a human patient in need of an immunotherapy and/or vaccination (which is, for example, suffering from cancer and/or infectious diseases), which is characterized in administering a therapeutically effective amount of a MVA mutant/vaccine of the invention to said patient. The physician in any event will determine the actual dosage which will be most suitable for an individual patient and will vary with the age, weight and response of the particular patient. There can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
Preferably, the pharmaceutical composition is adapted for in vivo use in a mammal, preferably a human patient.
According to a further aspect, a method of generating mutant MVA is provided, comprising the steps of: - Infecting host cells of MVA with a nucleic acid coding for the MVA mutants as defined above, - expressing said nucleic acid under suitable conditions in said host cells; and - isolating expressed mutant MVA.
Furthermore, the present invention provides a method for generating a pharmaceutical composition comprising the steps of: - Infecting host cells of MVA with a nucleic acid coding for the MVA mutants as defined above, - expressing said nucleic acid under suitable conditions in said host cells; - isolating expressed mutant MVA; - adding a pharmaceutically acceptable carrier and further ingredients in order to manufacture a pharmaceutical composition.
According to a preferred embodiment, the host cells are CEF cells, chicken embiyo derived LSCC-H32 cells, chicken DF-1 cells or avian cells, e.g. quail fibroblasts QT6 or QT35 cells.
Even more preferred, the mutant MVA are selected by the KIL gene based host range selection protocol (Staib et al, "Transient Host Range Selection For Genetic Engineering Of Modified Vaccinia Virus Ankara" BioTechniques 28: 1137-1148 (June 2000).
Compared to its parental strain, MVA has deletions that consist of about 15 percent (30,000 base pairs) of its former genome, including most of the KIL gene. Only a fragment of a length of 263 bp is still present in the MVA genome. The MVA KIL gene sequences represent the first 263 bp of the ORF 022L in the MVA genome at position nt 20685-20981 as described in Antoine, G., F. Scheiflinger, F. Dorner, and F. G. Falkner. 1998. The complete genomic sequence of the modified vaccinia Ankara strain and a comparison with other orthopoxvims es can be found in: Virology 244:365-396.
In Staib et al, an easy and highly efficient method for generation of recombinant MVA based on selection for transient expression of the vaccinia vims host range gene KIL is described. This method is based on selection of recombinant MVA by transient host range gene expression using the vaccinia vims KIL gene function as stringent marker to rescue MVA growth on rabbit kidney RK-13 cells. The constmction and use of new MVA vector plasmids was described which cany an expression cassette of the vaccinia virus host range gene KIL as transient selectable marker. These plasmids allow either stable insertion of additional recombinant genes into the MVA genome or precisely targeted mutagenesis of MVA genomic sequences. Repetitive DNA sequences flanking the KIL gene were designed to remove the marker gene from the viral genome by homologous recombination under non- selective growth conditions.
The publication of Straib et al., mentioned above, is incorporated herein in its entirety.
Additionally, the present invention provides the use of a MVA mutant as defined hereinabove or of a pharmaceutical composition, see above, for the manufacture of a medicament for use in immunotherapy and/or vaccination. Furthermore, the present invention is directed to a method for treating a patient, comprising a therapeutically effective dose of mutant MVA or of a pharmaceutical composition as defined herein to an individual in need of said treatment. Regarding the way and form of administration, see also above. The MVA mutant of the present invention may preferably used for vaccination against smallpox or other diseases caused by orthopoxvims infections.
All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The present invention is further illustrated by the following figures:
FIG. 1. Constmction of LlβR deficient MVA. Upper panel, schematic map of the MVA genome. Sites of the restriction endonuclease H dIII within the genome of MVA are indicated. The position of the 184R ORF (ILlβR gene) is marked by an anOw. MVA DNA sequences adjacent to the ILlβR coding sequence (flank 184R-I, flank 184R-JJ) were cloned into plasmid pΔKIL to allow for transient insertion of the KIL gene by homologous recombination at the site of ORF 184R, resulting in deletion of this gene sequence. The final mutant vims MVA-ΔILlβR was obtained after the deletion of the KIL marker gene during a second step of homologous recombination involving synthetic repetitive sequences (rep).
FIG. 2. In vitro characterization of MVA-ΔLLlβR. (A) PCR analysis of viral DNA. Genomic template DNA was prepared from MVA-ΔILlβR (lane 1) or MVA (lane 2) infected cells and incubated with oligonucleotides adjacent to the 184R gene locus to amplify specific DNA fragments. PCR products were separated by agarose gel electrophoresis. M, molecular weight marker. (B) Southern blot analysis of viral DNA. Genomic DNA was prepared from MVA-ΔLLlβR (lane 1) or MVA (lane2) infected cells, digested with EcoRl, separated by agarose gel electrophoresis and transferred to a nylon membrane. DNA fragments specific for the ILlβR gene locus were detected using a 32P- dCTP labeled specific probe. (C) Radioimmunoprecipitation of EL lβR proteins from lysates of MVA- ΔILlβR (lanes 1, 4), MVA (lanes 2, 5) or mock (lanes 3, 6) infected and radiolabeled CEF cells. Immunoprecipitation was performed using polyclonal anti-B15R antibody coupled to Protein A (lanes 1-3) or uncoupled sepharose (lanes 4-6). The arrow head indicates the ILlβR protein. (D, E) Analysis of virus growth in CEF after high (D) or a low (E) MOI infection with MVA-ΔILlβR (■) or MVA (A).
FIG. 3. Analysis of virus vimlence in a mouse model for respiratory poxvi s infection. Characterization of infections with MVA, MVA-ΔILlβR, CVA, and WR. BALB/c mice (n=10) were inoculated by the intranasal route with lxl 08 IU MVA (♦) and MVA- ΔILlβR (A), or 5xl05 PFU CVA (■) and 3xl04 PFU WR (-). Body weight (A) and body temperature (B) was monitored daily and is expressed as mean for each group.
FIG. 4 Ex vivo analysis of vaccine induced CD8+ T cells. HHD mice were vaccinated with a single dose of MVA-ΔILlβR, MVA or a revertant vims. After 10 days, splenocytes were stimulated with the HLA-A*0201 resticted vaccinia specific VP35#1 (VP35#1) or influenza Ml 58-66 (inelevant control) peptide, then stained with EMA, PE-anti-CD8, APC-anti- CD62L and FITC-anti-IFNγ or -anti-TNFα or the respective FITC-labelled isotype control. Cells were analyzed by flow cytometry for the presence of VP35#1 peptide-specific, activated (CD62Llo ) CD8+ T cells. The magnitude of the induced T cell response is depicted as percentages of cytokine secreting CD8+ T-cells within the live (EMAnegatIve) and CD8p0Sltlve cell population. Representation of mean of 12 mice per group vaccinated with either MVA-ΔILlβR (■), MVA (■), or MVA-ELlβR-Rev (If), error bars indicate standard error x 1,96. p-value (p=0.07) was determined using Student's t-test.
FIG. 5 Analysis of vaccine-induced protection in a mouse model for respiratory poxvims infection. BALB/c mice (n=10) were immunized intramuscularly with 104 ( A), 105 (-), 106 (D), 107 (Δ) IU of MVA-ΔILlβR (A, C) or MVA (B, D). Three weeks after vaccination animals were challenged intranasally with lxlO6 PFU WR. Individual animal weights (A, B) and signs of illness (C, D) were monitored daily and are expressed as means for each group. Mock vaccinated (♦) and mock challenged (■) mice served as control groups.
FIG. 6 Ex vivo analysis of vaccine induced memory CD8+ T cells and analysis of vaccine- induced long-term protection in HHD mice. (A) Mice were vaccinated intraperitoneally with a single dose 108 IU of MVA-ΔILlβR or MVA. After 6 month, splenocytes were either stimulated with the HLA-A*0201 resticted vaccinia specific VP35#1 peptide or infected with MVA for detection of vaccinia specific total CD8+ or CD4+ responses. Cells were stained with EMA, PE-anti-CD8, APC-anti-CD62L and FITC-anti-IFNγ or the respective FITC-labelled isotype control. Cells were analyzed by flow cytometry for the presence of VP35#1 peptide or vaccinia-specific, activated (CD62Llow) CD8+ T cells or vaccinia specific CD4+ T cells. The mean of 6 mice is depicted after vaccination with either MVA- ΔILlβR (■) or MVA (■), error bars indicate standard error x 1.96. p-value (p=0.01) was determined using Student's t-test. (B) HHD mice (n=5) were immunized intraperitoneally with 10s IU of MVA-ΔILlβR (Δ) or MVA (D ). At 6 month after vaccination mice were challenged with lxl 07 PFU WR. Survival was monitored daily and is expressed as percentage of surviving animals per group. Mock vaccinated (♦) and mock challenged (■) mice served as control groups.
FIG. 7 Ex vivo analysis of vaccine induced memoiy CD8+ T cells and analysis of vaccine- induced long-term protection in non transgenic mice. (A) C57BL/6 mice were vaccinated intraperitoneally with a single dose 108 IU of MVA-ΔILlβR or MVA. After 6 month, splenocytes were infected with MVA for detection of vaccinia specific total CD8+ or CD4+ responses. Cells were stained with EMA, PE-anti-CD8, APC-anti-CD62L and FITC-anti- IFNγ or the respective FITC-labelled isotype control. Cells were analyzed by flow cytometry for the presence of vaccinia-specific, activated (CD62Llow) CD8+ T cells or vaccinia specific CD4+ T cells. The mean of 6 mice is depicted after vaccination with either MVA-ΔILlβR (■) or MVA (■), error bars indicate standard error x 1.96. p-value (p=0.001) was determined using Student's t-test. (B) BALB/c mice (n=4) were immunized intranasally with 105 (A),106 (D) or 107 (Δ) IU of MVA-ΔILlβR or MVA. 4 month after vaccination animals were challenged intranasally with lxl 07 PFU WR. Individual animal weights were monitored daily and are expressed as means for each group. Mock vaccinated (♦) and mock challenged (■) mice served as control groups.
The following example is intended to contribute to a better understanding of the present invention. However, it is not intended to give the impression that the invention is confined to the subject-matter of the example.
Materials and Methods
Viruses and cells. Vaccinia vims strains Western Reserve, CVA and MVA (cloned isolate F6, from the 582nd passage on chicken embryo fibroblasts (CEF)) were used for this study. All vimses were propagated and titered following standard methodology. To generate vaccine preparations, viruses were routinely purified by ultra centrifugation through sucrose and reconstituted in 1 mM Tris pH 9.0. CEF and rabbit kidney RK-13 (ATCC CCL-37) cells were grown in minimal essential medium (MEM) supplemented with 10% fetal calf semm (FCS), and maintained at 37 °C and 5% CO2.
Plasmids. The transfer plasmid pΔKlL-184R carries two DNA fragments that represent flanking sequences of MVA ORF 184R (nucleotide position 162021-163001, GenBank U94848) and which were inserted into multiple cloning sites 1 and 2 of plasmid pΔKIL (Staib et al. 2000). One fragment, designated flank 184-1, consists of a 486-bp MVA-DNA sequence starting in the 5' intergenic region of ORF 184R and ending at the start codon for translation of ORF 184R, the other fragment, flank 184-2, is a 544-bp PCR-fragment of MVA-DNA extending from the codon for 184R translation termination into the 3'- intergenic region of the 184R gene.
Genetical modification of vaccinia virus MVA. Mutant MVA were obtained following the transient KlL-based host range selection protocol as described previously (Staib et al, 2000). Briefly, for generation of deletion mutant vimses, monolayers of lxlO6 confluent CEF cells were infected with MVA at a multiplicity of infection (MOI) of 0.01 IU per cell. Ninety minutes after infection cells were transfected with 1.5 μg of plasmid pΔKlL-184R DNA using FUGENE™ (Roche, Mannheim, Germany) as recommended by the manufacturer. At 48 h after infection, transfected cells were harvested and plated on RK-13 cell monolayers for growth selection. Mutant viruses were isolated through plaque cloning on RK-13 cells and then passaged on CEF cells to remove the selectable marker gene KIL.
Analysis of viral DNA by PCR and Southern blot. Genomic viral DNA was isolated from infected CEF cells and analyzed by PCR using oligonucleotides annealing within the flanking regions flankl84-l and -2, respectively (pair 1) or within flankl84-l and the coding region of 184R, respectively (pair 2) (Staib et al, 2000). Specific DNA fragments were amplified by 30 cycles of PCR at an annealing temperature of 52°C (pair 1) or 50°C (pair 2).
Alternatively, total DNA isolated from vims -infected cells was digested with EcoRI, separated by gel electrophoresis in 0.8% agarose, transferred to a Hybond TM-N membrane (Amersham, Freiburg, Germany), and hybridized to a DNA probe consisting of a PCR fragment from 184R-flankl sequences labeled with [α-32P]CTP. Prehybridization and hybridization was performed according to Sambrook et al. (Southern 1975, Sambrook et al, 1989). Blots were exposed to a Kodak BioMax film.
Radioimmunoprecipitation of virus-infected cell lysates. CΕF cells grown in 6-well tissue culture plates were infected with a multiplicity of 20 infectious units MVA. At 2h post infection, the virus inoculum was replaced with methionine-free minimal essential medium containing 5% dialyzed fetal calf serum and 50 μCi of [35S]methionine per ml and incubated overnight at 37 °C. Cells were lysed in RTPA-buffer containing 0.15 M NaCl, 0.01 M Tris-HCl (pH 7.4), 1% Triton X-100, and incubated for 14 h with rabbit polyclonal antibody AcB15R (Alcami & Smith, 1992), followed by 50% protein A - sepharose suspension. Immune complexes were washed in RJJPA-buffer, resuspended in Laemmli buffer, and proteins were separated by electrophoresis in a 10% SDS-polyacrylamide gel. Analysis of virus growth. To determine low or high multiplicity growth profiles, confluent CEF monolayers (grown on 6 well plates) were infected with 0.05 infectious units (IU) or 10 IU MVA or mutant MVA per cell, respectively. After vims adsorption for 60 min at 37 °C, the inoculum was removed. Cells were washed twice with RPMI 1640 and incubated with fresh RPMI 1640 medium containing 10% FCS at 37 °C and 5% CO2- At multiple time points post infection (p.i.) infected cells were harvested and virus was released by freeze-thawing and brief sonication. Serial dilutions of the resulting lysates were plated on confluent CEF monolayers grown in 6-well plates as replicates of two. 48 hours p.i., monolayers were briefly fixed in acetone:methanol (1: 1), and cells were incubated for 60 min with polyclonal rabbit anti-vaccinia antibody (IgG fraction, Biogenesis Ltd, Poole, England, Cat.No. 9503-2057, diluted 1: 1000 in PBS-3% FCS), followed by an incubation for 45 min with horseradish-peroxidase-conjugated polyclonal goat anti-rabbit antibody (Dianova, Hamburg, Germany, dilution 1: 1000 in PBS-3% FCS). After washing with PBS, antibody-labeled cells were developed using o-dianisidine (Sigma, Taufkirchen, Germany) substrate solution, foci of stained cells were counted, and virus titers were calculated as IU/ml.
Humoral vaccinia virus responses. Serum samples from mice immunized with MVA, mutant MVA, or vaccinia vims CVA were assessed for antibodies to vaccinia virus proteins by ELISA and neutralizing-antibody assay. Vaccinia antigen-specific binding titers were determined by an ELISA in which Maxisorp plates (Nunc, Germany) were coated with sucrose-gradient purified MVA (at a protein concentration of lμg/ml) for 3h at 37 °C and overnight at 4 °C. Plates were blocked with PBS / 0.05% Tween 20 / 10% fetal calf serum for 60 min at 37 °C. Serial dilutions of serum samples were incubated for 60 min at 37 °C, washed five times with PBS, and incubated for 30 min with anti-mouse alkaline- phosphatase conjugate (diluted 1 : 1000 in PBS). Following five washes, plates were incubated with pNPP substrate (Sigma, Germany) at 37 °C, after 20 min, the optical density was measured on an ELISA reader at a wavelength of 405nm.
Vaccinia vims-specific neutralizing antibodies were analyzed by a plaque reduction assay using recombinant MVA-LZ. Twofold serial dilutions of sera were mixed with 200 infectious units MVA-LZ in a total volume of 200 μl PBS and incubated for two hours at 37 °C. Afterwards, confluent CEF monolayers (grown on 24 well plates) were infected in duplicate, and foci of vims infected cells were visualized 48 hours after inoculation by staining with 5-bromo-4-chloro-3-indolyl-β-galactopyranoside substrate (X-Gal, Roche Molecular Biochemicals, Mannheim Germany) as described previously (Drexler et al., 1998). Blue-stained foci were counted and the number obtained with each serum was compared to controls with mouse preimmune sera or medium control. Antibody titers were calculated as the serial twofold dilution yielding a 50% reduction of foci numbers.
Cellular vaccinia virus responses. For monitoring of peptide-specific acute and memory phase CD8+ T cell responses, splenocytes from vaccinia virus-immunized HHD mice were prepared and incubated for 5 h with A*0201 -binding peptides at 10"6 M. After 2 h, brefeldin A was added at a final concentration of 1 μg/ml (GolgiPlug™; PharMingen Becton Dickinson). Cells were then either stored ON on ice at 4°C, or directly live/dead stained in PBS/1%BSA/1 μg/ml ethidium monoazide bromide (EMA; Molecular Probes) and blocked for unspecific Fcγin and -II receptor-mediated binding with 5 μg/ml purified anti- CD 16/CD32 (Fc Block™; PharMingen Becton Dickinson) for 20 min at 4°C. Cell surface staining was performed with PE-anti-CD8 (53-6.7) and APC-anti-CD62L (Mel-14) for 30 min at 4°C. After permeabilization of cells (Cytofix/Cytoperm™ Kit, PharMingen Becton Dickinson), intracellular cytokine staining was performed for 30 min at 4°C applying FITC- anti-LFNγ (XMG1.2) or FITC-anti-TNFα (MP6-XT22) or the respective FITC-labelled IgGl isotype control (R3-34) (all PharMingen Becton Dickinson). Splenocytes were analyzed by four-color flow cytometry (FACSCalibur™) using CellQest® software (both Becton Dickinson).
Animal models. Female six to eight week-old transgenic HHD+/+ β2m"/_ D " mice (HHD) (Pascolo et al, 1997) or female six to eight week old BALB/c or C57BL/6 mice were used for vaccination experiments. HHD mice were inoculated with 0.5 ml volumes of vims vaccine by the intraperitoneal route, and monitored for HLA-A*0201-restricted T cell responses at days 10 or 180 after immunization. For protection assays, animals were vaccinated once with 0.1 - 0.5 ml vims vaccine given by intramuscular or intraperitoneal route. Three weeks or six months after immunization, animals were anesthetized, infected intranasally with vaccinia vims Western Reserve diluted in 30 μl phosphate buffered saline, and monitored for at least further three weeks for morbidity and mortality with daily measurement of individual body weights and scoring of signs of illness as described previously. Animals suffering from severe systemic infection and having lost>30% of body weight were sacrificed. The mean change in body weight was calculated as the percentage of the mean weight for each group on the day of challenge. Body temperature was determined with a Electronic Laboratory Animal Monitoring System (BioMedic Data Systems, Marywood, NJ) using subcutaneously implanted microchip battery-free transponders and a DAS-5004 Pocket Scanner for data collection. Mean changes in body temperatures were calculated by subtracting the pre-challenge (days -3 to 0) baseline temperature of each group from each subsequent time point.
Results
Deletion of ILlβR coding sequences from the MVA genome. In order to analyze the possible role of LLlβR gene expression during MVA infection, we constmcted MVA knockout mutants lacking the open reading frame (ORF) 184R (LLlβR). The coding sequences of the viral EL lβR together with its presumed promoter sequence are well conserved within the MVA genome. Equal to the previously characterized ILlβR of vaccinia vims strain Western Reserve the predicted MVA polypeptide consists of 326 amino acids at an identity level of 99% (2, 5, 34). Using PCR, we amplified DNA segments located up- and downstream of the 184R coding sequence and inserted these fragments into the deletion vector pΔKIL (Fig. 1), which contains the vaccinia vims KIL gene as selectable marker. Upon transfection of MVA infected cells with pΔKlL-184R, the 184R-flanking regions allowed for introduction of the KIL marker gene and simultaneous deletion of the ELlβR gene sequence in the MVA genome by homologous recombination. The resulting viruses were selected on RK-13 cells, where KIL function is essential for MVA growth. After isolation of clonally pure mutant viruses the KIL marker cassette was removed upon passage on CEF cells yielding the final mutant vimses MVA-ΔILlβR (Fig.1).
Molecular characterization and unimpaired in vitro replication of mutant virus MVA- ΔILlβR. After isolation of the MVA deletion mutants we first wished to confirm the correct removal of ILlβR coding sequences on a genetic level. We analyzed viral DANN extracted from CEF cells infected with wild-type or mutant MVA by PCR using oligonucleotide primers specific for MVA genomic sequences adjacent to the ELlβR gene locus. This PCR specifically amplified 2.1-kb DNA fragments from wild-type MVA templates, whereas the use of DNA from MVA-ΔILlβR-infected cells generated 1.1-kb PCR products corresponding to the expected reduction of molecular weights after deletion of ORF 184R (Fig. 2A). Furthemiore, we digested viral DNAs with restiiction endonuclease EcoRI and revealed DNA fragments containing the ILlβR gene locus by Southern blot analysis. Confirming the PCR data, we detected an about 1.3-kb lower molecular weight EcoRI- fragment in the genomic DNA of deletion mutant MVA-ΔILlβR (Fig. 2B) again confirming the proper deletion of the targeted ORF 184R sequences. In a second step we wanted to prove that ΕL lβR protein is produced during MVA infection and to demonstrate that the generated mutants fail to synthesize this polypeptide. Therefore, we performed immunoprecipitation experiments with polyclonal ΕL lβR-specific antibodies using lysates of metabolically labelled CΕF cells infected with MVA or MVA-ΔILlβR (Fig. 2C). The antiserum precipitated a specific protein of about 45 kDa from cell lysates obtained after infection with wild-type MVA corresponding in size to the glycosylated product of the ΕL- lβR polypeptide found in vaccinia vims WR infected cells (2). In contrast, this protein was not detected in lysates from mock infected or MVA- 16 ΔILlβR-infected cells demonstrating that the generated deletion mutant vims failed to make an LLlβR product. Furthermore, we wanted to asses the replicative capacity of mutant MVA-ΔΕ lβR in comparison to wild-type MVA. After infections of CΕF we found very comparable amounts of new viral progeny being formed at nearly identical kinetics during one step (Fig. 2D) and multiple step (Fig. 2Ε) vims growth. This data clearly suggested that inactivation of MVA ORF 184R does not affect the in vitro multiplication of the virus.
Avirulence of MVA-ΔILlβR upon high dose respiratory infection of mice. An important question was whether the inability to produce the viral IL-lβR protein would influence the outcome of MVA infection in vivo. Previous work in mice with vaccinia vims WR deletion mutants had revealed either enhancement of respiratory disease after intranasal infection (2) or reduced virulence after intracranial infection (34). The more severe respiratory infection appears to be linked to induction of fever response and the functional activity of the viral EL- lβR neutralizing EL-lβ as major endogenous pyrogen (3). Therefore, we tested mutant virus MVA-ΔELlβR upon intranasal infection of mice. Severity of disease in this mouse model is well reflected by changes in body weight and appearance of characteristic signs of illness (3, 12, 28, 29, 43). Additionally, we wished to monitor for changes in body temperatures because of the possible onset of febrile reactions. We transplanted BALB/c mice with subcutaneous microchip transponders to allow for computable readings, one week later infected the animals with 108 IU of MVA or MVA- ΔILlβR or with 5xl05 PFU of replication competent vaccinia vims CVA or 3x104 PFU Western Reserve (WR) as control, and monitored animals daily over a period of three weeks (Fig. 3). Infection of mice with MVA or mutant MVA-ΔILlβR did not result in any obvious disease. In contrast infection with replication competent vimses CVA and WR caused drastic loss of body weight (Fig. 3A) and severe signs of illness being also well reflected by reduced body temperature (Fig. 3B). In MVA infected animals, body temperature remained stable over the observation period. Taken together, these data suggested preservation of the attenuated phenotype of MVA after deletion of the EL lβR gene from its genome.
Early immune response induced by vaccination with MVA-ΔELlβR. It was of particular interest to assess, what possible influence deletion of the immunomodulatory ILlβR gene may have on MVA immunogenicity. First, we vaccinated HLA-A*0201 transgenic mice with a single dose of MVA or deletion mutant and monitored directly ex vivo in the acute phase of the immune response for induction of vaccinia vims-specific CD8+ T-cell responses using the orthopoxvims specific HLA-A*0201 restricted peptide epitope VP35#1 (12). By FACS analysis of freshly prepared splenocytes from vaccinated animals we were able to detect 0.4 to 2.45% of activated CD8+ T cells after immunization with MVA- ΔILlβR , whereas in animals inoculated with MVA levels of IFNγ releasing CD8+ T cells range from 0.16 to 0.82%. We analyzed 12 individual mice per group for VP35#1 specific T cell induction, including also a group vaccinated with revertant virus MVA-fLlβRev. Figure 4 depicts a difference between the groups of vaccinees, which albeit not statistically significant showed a tendency towards significance (p=0.07) with regard to higher levels of T cell immunogenicity elicited by MVA-ΔILlβR. Importantly, vaccinations with the revertant vims MVA-ILlβRev resulted in T cell responses that were very comparable to those induced by MVA wildtype.
Protective capacity of MVA-ΔILlβR immunization. Having found slightly higher VP35#1 epitope-specific CD8+ T cell responses after immunization with MVA-ΔILlβR mutant vims, we wished to ascertain possible differences in protective capacity of MVA and the MVA deletion mutant. For this we used a recently established mouse model, where groups of mice are vaccinated once intramuscularly with different doses of MVA vaccine, followed by a lethal intranasal challenge with vaccinia vims WR at three weeks after immunization (Fig. 5) (12). Body weight (Fig. 5A, B) and signs of illness (Fig. 5C, D) of animals were monitored daily for a period of three weeks after the challenge. Vaccination with both MVA vaccines given at 105 or higher doses fully protected all mice, whereas 104 or less resulted in death of all animals. This model has the advantage, that the observed vaccine protection titrates with the dose administered being depicted by the changes in average body weights of the various groups. Here by, we determined very similar weight curves for the different groups of mice immunized with MVA or MVA- ΔILlβR vaccines (Fig. 5A, B). This result was further confirmed when we monitored for the typical signs of illness after infection (Fig. 5C, D), as with increasing vaccine dose, signs of illness decreased alike among con-esponding groups. Thus, MVA and MVA-ΔILlβR showed a very comparable capacity to elicit protective responses within three weeks after vaccination being in agreement with our finding of similar acute phase immune responses.
MVA-ΔILlβR vaccination improves T cell memory response and long-term protective capacity. The mature form of the inflammatory cytokine ILlβ has multiple effects in vivo as revealed by the study of mice deficient for different components of the EL-1 system (33). An active area of ongoing research on IL1 function is to elucidate the likely importance of the cytokine in protective T cell immunity including the activation of professional antigen presenting cells and memory T cells (16, 17). To investigate whether the inactivation of viral ELlβR influences the formation of memory T cell responses, we vaccinated groups of HLA-A*0201 transgenic (HHD) mice once with 108 IU MVA-ΔILlβR or MVA and monitored for vims-specific T cells more than six months after this primary immunization. At late times after vaccination relatively low levels of VP35#1 -specific memoiy T cells can be detected using our standard protocol for ICS/FACS analysis (0.30-0.60%) (11). An improved protocol using overnight incubation of peptide stimulated splenocytes allows to notice a more prominent population of antigen-specific CD8+ T cells often exceeding the number of activated T cells found with our conventional protocol during the acute phase response. Here by, we detected clearly higher levels (up to 5.8%) of VP35#1 -reactive and IFN-γ releasing splenic CD8+ memory T cells in MVA-ΔELlβR immunized animals as compared to vaccination with non recombinant MVA resulting in up to 1.6% epitope- specific IFN-γ-secreting CD8+ T cells (Fig. 6A). This difference in favour of MVA-ΔILlβR vaccination was statistically significant (p=0.01), and, interestingly, splenocytes from vaccinees of this group also contained higher amounts of total vaccinia-specific CD8+ memory T cells, while we found comparable average levels of CD4+ T cells for both MVA- ΔILlβR and MVA. To monitor if the different levels of memory T cell responses would go along with alterations in protection, we challenged HHD mice by intranasal inoculation of 107 PFU vaccinia vims Western Reserve after being vaccinated with a single intraperitoneal inoculation of 108 EU MVA-ΔELlβR or MVA more than six months earlier (Fig. 6B). The infection resulted in mock vaccinated control animals in the onset of respiratory disease, weight loss, and death within 8 days after challenge. Mice inoculated with wildtype MVA were also affected by respiratory illness and substantial loss of body weight. Yet, the animals were partially protected because 3 out of 5 mice in this group survived the challenge infection. Notably, all animals (5/5) receiving the MVA- ΔILlβR vaccine were protected, an outcome that was also well reflected with regard to the lesser extent of weight loss and illness observed in this group (data not shown). This result implied that vaccination with MVA-ΔILlβR could indeed have an influence on the durability of protective immunity. The HHD mouse model allows for convenient analysis of epitope specific HLA- A*0201 -restricted CD8+ T cell responses, yet, possibly because of their knock-out phenotype for mouse MHC class I, these mice develop unusually low numbers of total CD8+ T cells (but normal levels of CD4+ T cells) (11). As this phenotype might influence the analysis of total vaccinia-specific T cell responses, we assessed total CD8+ memoiy T cells induced by MVA-ΔILlβR or MVA also after vaccination of normal C57BL/6 mice (Fig. 7A). Again in comparison to conventional MVA vaccination we found significantly (p=0.001) higher numbers of vaccinia-specific CD8+ T cells in animals immunized with MVA-ΔILlβR. This data strongly suggested an improved capacity of MVA-ΔELlβR to elicit or maintain vaccinia vims-specific CD8+ T cell memory. To investigate the longterm efficacy of MVA- ΔEL lβR immunizaton in more detail, we decided to test the vaccines also in the well established challenge model using non transgenic BALB/c mice (5, 11). We chose intranasal vaccination as this route of MVA immunization of mice results in comparison to intramuscular or intraperitoneal vaccination in lower levels of circulating virus-specific antibodies, which might be an advantage when assessing the potential protective capacity of T cell immunity. We inoculated groups of BALB/c mice with 105 to 107 IU of MVA-ΔELlβR or MVA. Four months after immunization, we submitted animals again to a respiratory infection with 10 PFU of vaccinia virus Western Reserve (Fig. 7B). Importantly, all mice having received MVA-ΔILlβR vaccine survived the challenge, and animals in the group vaccinated with 107 IU of MVA- ΔEL lβR demonstrated an average <15% reduction of body weight and only mild signs of illness (data not shown). In contrast, no protection from severe disease resulting in death of all animals was seen after inoculation with 105 IU MVA vaccine (Fisher exact test p=0.029), and while vaccination with higher doses (106, 107 IU) of MVA prevented death, animals in these groups showed a > 20% average weight loss and enhanced signs of disease (data not shown).
The MVA ORF 184R encodes a vaccinia viral soluble receptor for IL-lβ with proposed function to block inflammatory and febrile host response to infection. The removal of putative immune evasion genes from viral genomes is a promising approach to further elucidate roles of these regulatory vims proteins in the in vivo viral life cycle (1). Moreover, application of this research to a vims such as MVA being suitable for use as (recombinant) live viral vaccine may directly lead to second generation vaccines with rationally improved properties. Even by means of molecular engineering techniques allowing for precise mutagenesis (35, 36) the resulting phenotypes of mutant vimses are unpredictable, and the inactivation of the MVA ILlβR gene may serve as further example. Our finding that inactivation of the 184R ORF had no impact on the in vitro replicative capacity of the MVA mutant vims can be considered as quite unsurprising because no growth deficiencies have been reported with corresponding mutants derived from vaccinia vims WR (2). Yet, capacity for high level amplification is of utmost importance for a virus possibly serving in vaccine production. Additionally, it should be noted that with another MVA mutant defective in expression of the viral interferon response gene E3L we had recently found a very unexpected host range phenotype in CEF the preferred cell culture for MVA vaccine production (15). Of course, it was even more interesting to investigate the in vivo properties of MVA-ΔILlβR. It was possible that an unhampered ILlβ activity elicited by infection with MVA-ΔILlβR would trigger strong inflammation reactions and febrile responses that result in adverse effects of vaccination. In a more optimistic scenario we speculated that ILlβ action could be locally restricted and might influence the potency of MVA immunization in an adjuvant-like manner. Upon intranasal infection of BALB/c mice with MVA-ΔILlβR we did not detect signs of respiratory illness despite using high dose inoculations and despite the fact that this mouse model system appears to be particularly suitable to assess potential pathogenic consequences of inflammatory responses to viral infection (3, 28, 30). Our inability to detect pathogenic effects after infection with MVA- ΔILlβR might be a consequence of the particular MVA genotype with other vaccinia virus regulatory or immunomodulatory genes being fragmented or deleted (5). Yet, alternatively it could likely be that the disease enhancement observed with vaccinia virus ILlβR-deletion mutants requires active in vivo replication of the vims after intranasal infection. As there is good recent evidence confirming that MVA is incapable to productively replicate in vivo in mice (27) or macaques (39), our data might suggest that transient one-step infection with MVA-ΔILlβR is just not sufficient to result in ILlβ activities inducing adverse systemic fever or inflammation reactions. In first vaccination experiments we found equal properties of MVA and MVA-ΔILlβR with regard to vaccine immunogenicty monitoring for early total anti-vaccinia vims antibody or T cell responses. Data that was well confirmed by the close to identical capacity of both viruses to protect animals from lethal vaccinia virus challenge given three weeks after vaccination. Of particular interest is our demonstration of the benefit of MVA-ΔELlβR immunization when monitoring for vaccinia vims epitope- specific T cell responses. The H3L gene product-derived epitope VP35#1 is the target of an immunodominant HLA-A*0201 -restricted T cell specificity and can be used to examine the induction of vims-specific CD8+ T cells in an epitope-specific manner (10, 12). In contrast, bulk analysis of total vaccinia virus-specific responses offers a representative picture based on a multitude of different T cell specificities but might not allow to assess subtile changes in the activation of single T cell populations. Suspecting a possible effect on T cell activation we opted to also assess VP35#1 -specific T cell memory responses which we had previously found detectable for more than 6 month after vaccination of HLA-A*0201 transgenic mice (12). Indeed, we observed a clearly more prominent enhancement of VP35#1 -specific T cell responses after vaccination with MVA-ΔILlβR (p=0.01). Moreover, our additional findings appeared to solidly confirm a longterm beneficial effect of MVA- ΔILlβR immunization. We had noticed for the first time an increased total CD8+ T cell responses in transgenic HHD mice, and more importantly, we observed significantly enhanced total CD8+ T cells after vaccination of non-transgenic C57BL/6 mice (p=0.001). In addition, we found higher protective capacities against lethal respiratory challenge with vaccinia vims Western Reserve in both HLA-A*0201 transgenic mice and normal BALB/c mice. How to explain an enhanced vaccine efficacy in the context of ELI β function? Interestingly, results from two recent studies investigating the Leishmania major infection of susceptible or resistant mice suggest that the ability of dendritic cells (DC) to secrete ILlα or ELlβ is specifically associated with the induction of protective Thl immunity (13, 42). In addition, there is recent evidence for ELlβ being an essential mediator of Fas-ligation induced maturation of murine DC (14). The same work demonstrated that maturation of murine DC could be completely abrogated by the use of ILlβ neutralizing antibodies which may function in a similar manner as one could expect with the soluble vaccinia virus EL lβR molecule. Therefore, the lack of ILlβ neutralization upon vaccination with MVA-ΔILlβR may lead to improved functionality of DC to serve as antigen-presenting cells, which might result in better T cell memoiy responses. Similarly, it has been shown that stimulation of endothelial cells with ILlβ resulted in ICOS-L mediated activation of memoiy T cells (17). Such activity of ILlβ might be the functional basis for our finding that vaccination with MVA-ΔlLlβR appeared to predominantly improve memoiy T cell responses. In addition, at early times after immunization, we did not find differences in the in vivo protective capacity of MVA wildtype or mutant vaccines. This may also indicate that the viral ILlβR could have a specific role in abrogating anti-viral memory T cell responses. Alternatively, we might have been unable to detect a more general effect of the viral EL lβR expression on virus-specific T cell immunity upon early ex vivo analysis at what likely is the peak level of vaccine induced T cell response, and our inability to demonstrate differences in protective capacities could be due to the impact of high level antibody mediated immunity during early weeks after immunization. The important role of vims-specific antibodies to protect against a lethal respiratory challenge with virulent vaccinia virus has been clearly demonstrated (6), yet the completely protective MVA vaccination in B cell deficient mice suggests that T cell immunity may complement for the lack of vaccinia vims-specific antibody responses (44). The capacity of the viral ILlβR to apparently specifically downmodulate anti-viral CD8+ T cell memoiy responses appears alluring in the view of recent data from the analysis of vaccinia vims-specific memoiy T cells in humans (after vaccination with conventional wildtype vaccinia virus) showing better persistence of CD4+ than CD8+ T cells (4). In summary, our analysis recommends deletion of the viral ILlβR gene as a first step in approaching the development of a new generation of MVA-based vaccines. High virus titers could be obtained upon in vitro propagation of deletion mutant MVA-ΔILlβR, and there was no evidence of MVA-ΔILlβR being less well tolerated than wildtype vims upon high dose in vivo infection. Our finding of improved vaccine properties of MVA-ΔILlβR is particularly promising because it provides first evidence for the possibility of obtaining more efficacious MVA vaccines through rational genetical engineering. References: 1. Alcami, A. 2003. Viral mimicry of cytokines, chemokines and their receptors. Nat Rev Immunol 3:36-50.
2. Alcami, A., and G. Smith. 1992. A soluble receptor for Interleukin- lb encoded by vaccinia vims: a novel mechanism of virus modulation of the host response to infection. Cell 71: 153-167.
3. Alcami, A., and G. L. Smith. 1996. A mechanism for the inhibition of fever by a vims. Proc Natl Acad Sci USA 93: 11029-11034.
4. Amara, R., P. Nigam, S. Sharma, J. Liu, and V. Bostik. 2004. Long-lived poxvims immunity, robust CD4 help, and better persistence of CD4 than CD8 T cells. J Virol 78:3811-6.
5. Antoine, G., F. Scheiflinger, F. Dorner, and F. G. Falkner. 1998. The complete genomic sequence of the modified vaccinia Ankara strain: comparison with other orthopoxvims es. Virology 244:365-96.
6. Belyakov, I. M., P. Earl, A. Dzutsev, V. A. Kuznetsov, M. Lemon, L. S. Wyatt, J. T. Snyder, J. D. Ahlers, G. Franchini, B. Moss, and J. A. Berzofsky. 2003.
Shared modes of protection against poxvims infection by attenuated and conventional smallpox vaccine vimses. Proc Natl Acad Sci U S A 100:9458-63.
7. Carroll, M. ., and B. Moss. 1997. Host range and cytopathogenicity of the highly attenuated MVA strain of vaccinia virus: propagation and generation of recombinant vimses in a nonhuman mammalian cell line. Virology 238: 198-211.
8. Corona Gutierrez, C. M., A. Tinoco, M. Lopez Contreras, T. Navarro, P. Calzado, L. Vargas, L. Reyes, R. Posternak, and R. Rosales. 2002. Clinical protocol. A phase II study: efficacy of the gene therapy of the MVA E2 recombinant virus in the treatment of precancerous lesions (NIC I and NIC U) associated with infection of oncogenic human papillomavirus. Hum Gene Ther 13:1127-40. 3
9. Cosma, A., R. Nagaraj, S. Buhler, J. Hinkula, D. H. Busch, G. Sutter, F. D. Goebel, and V. Erfle. 2003. Therapeutic vaccination with MVA-HTV-1 nef elicits Nef-specific T- helper cell responses in chronically HEV-1 infected individuals. Vaccine 22:21-9.
10. Di Nicola, M., C. Carlo-Stella, A. Anichini, R. Mortarini, A. Guidetti, G. Tragni, F. Gallino, M. Del Vecchio, F. Ravagnani, D. Morelli, P. Chaplin, N. Arndtz, G. Sutter, I. Drexler, G. Parmiani, N. Cascinelli, and A. M. Gianni. 2003. Clinical protocol. Immunization of patients with malignant melanoma with autologous CD34(+) cell-derived dendritic cells transduced ex vivo with a recombinant replication-deficient vaccinia vector encoding the human tyrosinase gene: a phase I trial. Hum Gene Ther 14: 1347-60.
11. Drexler, I., K. Heller, B. Wahren, V. Erfle, and G. Sutter. 1998. Highly attenuated modified vaccinia vims Ankara replicates in baby hamster kidney cells, a potential host for virus propagation, but not in various human transformed and primary cells. J Gen Virol 79:347-352.
12. Drexler, L, C. Staib, W. Kastenmuller, S. Stevanovic, B. Schmidt, F. A. Lemonnier, H. G. Rammensee, D. H. Busch, H. Bernhard, V. Erfle, and G. Sutter. 2003 Identification of vaccinia virus epitope-specific HLA-A*0201- restricted T cells and comparative analysis of smallpox vaccines. Proc Natl Acad Sci U S A 100:217-222.
13. Filippi, C, S. Hugues, J. Cazareth, V. Julia, N. Glaichenhaus, and S. Ugolini. 2003. CD4+ T cell polarization in mice is modulated by strain-specific major histocompatibility complex-independent differences within dendritic cells. J Exp Med 198:201-9.
14. Guo, Z., M. Zhang, H. An, W. Chen, S. Liu, J. Guo, Y. Yu, and X. Cao. 2003. Fas ligation induces IL-1 beta-dependent maturation and' EL- 1 beta-independent survival of dendritic cells: different roles ofERK and NF-kappaB signaling pathways. Blood 102:4441- 7.
15. Hornemann, S., O. Harlin, C. Staib, S. Kisling, V. Erfle, B. Kaspers, G. Hacker, and G. Sutter. 2003. Replication of modified vaccinia vims Ankara in primary chicken embryo fibroblasts requires expression of the interferon resistance gene E3L. J Virol 77:8394-407.
16. Iwasaki, A. 2003. The importance of CD 11 b+ dendritic cells in CD4+ T cell activation in vivo: with help from interleukin 1. J Exp Med 198: 185-90.
17. Khayyamian, S., A. Hutloff, K. Buhner, M. Grafe, V. Henn, R. A. Kroczek, and H. W. Mages. 2002. ICOS-ligand, expressed on human endothelial cells, costimulates Thl and Th2 cytokine secretion by memory CD4+ T cells. Proc Natl Acad Sci U S A 99:6198-203.
18. Mayr, A., V. Hochstein-Mintzel, and H. Stickl. 1975. Abstammung, Eigenschaften und Verwendung des attenuierten Vaccinia-Stammes MVA. Infection 3:6-14.
19. Mayr, A., and E. Munz. 1964. Veranderungen von Vaccinevirus durch Daueipassagen in Hϋhnerfibroblastenkulturen. Zbl. Bakt. I. Abt. Orig. 195:24.
20. Mayr, A., H. Stickl, H. Miiller, K. Danner, and H. Singer. 1978. Der Pockenimpfstamm MVA: Marker, genetische Struktur, Erfahrungen mit der parenteralen Schutzimpfung und Verhalten im abwehrgeschwachten Organismus. Zbl. Bakt. Hyg., I. Abt. Orig. B 167:375-390. 21. McConkey, S. J., W. H. Reece, V. S. Moorthy, D. Webster, S. Dunachie, G. Butcher, J. M. Vuola, T. J. Blanchard, P. Gothard, K. Watkins, C. M. Hannan, S. Everaere, K. Brown, K. E. Kester, J. Cummings, J. Williams, D. G. Heppner, A. Pathan, K. Flanagan, N. Arulanantham, M. T. Roberts, M. Roy, G. L. Smith, J. Schneider, T. Peto, R. E. S nden, S. C. Gilbert, and A. V. Hill. 2003. Enhanced T-cell immunogenicity of plasmid DNA vaccines boosted by recombinant modified vaccinia virus Ankara in humans. Nat Med 9:729-35.
22. Meyer, H., G. Sutter, and A. Mayr. 1991. Mapping of deletions in the genome of the highly attenuated vaccinia vims MVA and their influence on vimlence. J Gen Virol 72: 1031-1038.
23. Moss, B., M. W. Carroll, L. S. Wyatt, J. R. Bennink, V. M. Hirsch, S. Goldstein, W. R. Elkins, T. R. Fuerst, J. D. Lifson, M. Piatak, N. P. Restifo, W. Overwijk, R. Chamberlain, S. A. Rosenberg, and G. Sutter. 1996. Host range restricted, non- replicating vaccinia vims vectors as vaccine candidates. Adv Exp Med Biol 397:7-13.
24. Moss, B., and P. L. Earl. 1991. Preparation of cell cultures and vaccinia vims stocks, p. 16.16.1-16.16.7. In F. M. Ausubel, R. Brent, R. Kingston, D. Moore, J. Seidman, J. Smith, and K. Stmhl (ed.), Current Protocols in Molecular Biology. John Wiley & Sons, New York.
25. Moss, B., and J. L. Shisler. 2001. Immunology 101 at poxvims U: Immune evasion genes. Semin Immunol 13:59-66.
26. Pascolo, S., N. Bervas, J. M. Ure, A. G. Smith, F. A. Lemonnier, and B. Perarnau. 1997. HLA-A2.1 -restricted education and cytolytic activity of CD8(+) T lymphocytes from beta2 microglobulin (beta2m) HLA-A2.1 monochain transgenic H-2Db beta2m double knockout mice. J Exp Med 185:2043-51.
27. Ramirez, J. C, M. M. Gherardi, and M. Esteban. 2000. Biology of attenuated modified vaccinia vims Ankara recombinant vector in mice: virus fate and activation of B- and T-cell immune responses in comparison with the Western Reserve strain and advantages as a vaccine. J Virol 74:923-33.
28. Reading, P. C, J. B. Moore, and G. L. Smith. 2003. Steroid hormone synthesis by vaccinia vims suppresses the inflammatory response to infection. J Exp Med 197: 1269-78.
29. Reading, P. C, and G. L. Smith. 2003. A kinetic analysis of immune mediators in the lungs of mice infected with vaccinia vims and comparison with intradermal infection. J Gen Virol 84:1973-83. 30. Reading, P. C, and G. L. Smith. 2003. Vaccinia vims interleukin- 18-binding protein promotes virulence by reducing gamma interferon production and natural killer and T-cell activity. J Virol 77:9960-8. 31. Rochlitz, C, R. Figlin, P. Squiban, M. Salzberg, M. Pless, R. Herrmann, E. Tartour, Y. Zhao, N. Bizouarne, M. Baudin, and B. Acres. 2003. Phase I immunotherapy with a modified vaccinia vims (MVA) expressing human MUCl as antigen-specific immunotherapy in patients with MUCl -positive advanced cancer. J Gene Med 5:690-9. 32. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Molecular cloning: A laboratory manual, 2 ed. Cold Spring Harbor Laboratoiy Press, Cold Spring Harbor New York. 33. Sims, J. E. 2002. IL-1 and EL- 18 receptors, and their extended family. Cuιτ Opin Immunol 14: 117-22. 34. Spriggs, M., D. Hruby, C. Maliszewski, D. Pickup, J. Sims, R. Buller, and J. VanSlyke. 1992. Vaccinia and cowpox vimses encode a novel secreted interleukin- 1- binding protein. Cell 71:145-152. 35. Staib, C, I. Drexler, M. Ohlmann, S. Winter sperger, V. Erfle, and G. Sutter. 2000 Transient host range selection for genetic engineering of modified vaccinia vims Ankara [In Process Citation]. Biotechniques 28:1137-42, 1144-6, 1148. 36. Staib, C, M. Lowel, V. Erfle, and G. Sutter. 2003. Improved host range selection for recombinant modified vaccinia vims Ankara. Biotechniques 34:694-6, 698, 700. 37. Staib, C, and G. Sutter. 2003. Live viral vectors: vaccinia virus. Methods Mol Med 87:51-68. 38. Stickl, H., V. Hochstein-Mintzel, A. Mayr, H. Huber, H. Schafer, and A. Holzner. 1974. MVA-Stufenimpfung gegen Pocken. Dtsch. med. Wschr. 99:2386-2392. 39. Stittelaar, K. J., T. Kuiken, R. L. de Swart, G. van Amerongen, H. W. Vos, H. G. Niesters, P. van Schalkwijk, T. van der Kwast, L. S. Wyatt, B. Moss, and A. D. Osterhaus. 2001. Safety of modified vaccinia vims Ankara (MVA) in immune- suppressed macaques. Vaccine 19:3700-9.
40. Sutter, G., and B. Moss. 1992. Nonreplicating vaccinia vector efficiently expresses recombinant genes. Proc Natl Acad Sci USA 89:10847-10851.
41. Sutter, G., and B. Moss. 1995. Novel vaccinia vector derived from the host range restricted and highly attenuated MV strain of vaccinia vims. Dev Biol Stand 84: 195-200.
42. Von Stebut, E., J. M. Ehrchen, Y. Belkaid, S. L. Kostka, K. Molle, J. Knop, C.
Sunderkotter, and M. C. Udey. 2003. Interleukin 1 alpha promotes Thl differentiation and inhibits disease progression in Leishmania major-susceptible BALB/c mice. J Exp Med 198:191-9.
43. Williamson, J. D., R. W. Reith, L. J. Jeffrey, J. R. Arrand, and M. Mackett. 1990. Biological characterization of recombinant vaccinia vimses in mice infected by the respiratory route. J Gen Virol 71:2761-7.
44. Wyatt, L. S., P. L. Earl, L. A. Eller, and B. Moss. 2004. Highly attenuated smallpox vaccine protects mice with and without immune deficiencies against pathogenic vaccinia vims challenge. Proc Natl Acad Sci U S A 101:4590-5.

Claims

C L A I M S
1. A MVA mutant, wherein the ELlβR coding sequence or a functional part thereof has been inactivated, preferably by deletion or mutation, for use in immunotherapy and/or vaccination.
2. The MVA mutant of claim 1, wherein the MVA mutant further comprises DNA sequences coding for a foreign protein or a functional part thereof.
3. The MVA mutant of claim 2, wherein the foreign protein is a heterologous protein derived from the group consisting of therapeutic polypeptides and polypeptides of pathogenic agents and functional parts thereof
4. The MVA mutant of claim 3, wherein the therapeutic polypeptide is derived from the group consisting of secreted proteins, e.g. polypeptides of antibodies, chemokines, cytokines or interferons.
5. The MVA mutant of claim 3, wherein the pathogenic agent is derived from the group consisting of vimses, bacteria, protozoa and parasites as well as tumor cells or tumor cell associated antigens and functional parts thereof.
6. The MVA mutant of claim 5, wherein the vimses are selected from the group consisting of influenza vimses, measles and respiratory syncytial vimses, dengue vimses, human immunodeficiency vimses, human hepatitis vimses, heroes vimses, or papilloma vimses.
7. The MVA mutant of claim 5, wherein the protozoa is Plasmodium falcipamm.
8. The MVA mutant of claim 5, wherein the bacteria is tuberculosis -causing Mycobacteria.
9. The MVA mutant of claim 5, wherein the tumor cell associated antigen is selected from the group consisting of melanoma-associated differentiation antigens, e.g. tyrosinase, tyrosinase-related proteins 1 and 2, of cancer testes antigens, e.g. MAGE- 1,-2,-3, and BAGE, and of non-mutated shared antigens overexpressed on tumors, e.g. Her-2/neu, MUC-1, and p53.
10. The MVA mutant as defined in one or more of claims 1-9 for use in the anti-cancer therapy or in the prevention of infectious diseases.
11. A pharmaceutical composition comprising one or more of the MVA mutants of claims 1-9 and a pharmaceutically acceptable carrier.
12. The pharmaceutical composition of claim 11, which is adapted for in vivo use in a mammal, preferably a human.
13. A method of generating mutant MVA, comprising the steps of: - Infecting host cells of MVA with a nucleic acid coding for the MVA mutants of one or more of claims 2-9, - expressing said nucleic acid under suitable conditions in said host cells; and - isolating expressed mutant MVA.
14. A method for generating a pharmaceutical composition comprising the steps of: - Infecting host cells of MVA with a nucleic acid coding for the MVA mutants of one or more of claims 1-9, - expressing said nucleic acid under suitable conditions in said host cells; - isolating expressed mutant MVA; - adding a pharmaceutically acceptable earner and further ingredients in order to manufacture a pharmaceutical composition.
15. The method of claim 13 or 14, wherein the host cells are CEF cells, chicken embryo derived LSCC-H32 cells, chicken DF-1 cells or other avian cells, e.g. quail fibroblasts QT6 or QT35 cells.
16. The method of one or more of claims 13-15, wherein the mutant MVA are selected by the KIL gene based host range selection protocol.
17. Use of a MVA mutant as defined in one or more of claims 1-9 or of a pharmaceutical composition of claim 11 or 12 in immunotherapy and/or vaccination.
18. The use of claim 17 for vaccination against smallpox or other diseases caused by orthopoxvims infections.
PCT/EP2004/010858 2003-09-29 2004-09-28 Modified vaccinia virus ankara (mva) mutant and use thereof WO2005030971A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP04765665A EP1668142A1 (en) 2003-09-29 2004-09-28 Modified vaccinia virus ankara (mva) mutant and use thereof
CN2004800244195A CN1842602B (en) 2003-09-29 2004-09-28 Modified vaccinia virus ankara (MVA) mutant and use thereof
BRPI0414874-6A BRPI0414874A (en) 2003-09-29 2004-09-28 modified ankara vaccinia virus mutant (mva) and use thereof
AU2004276486A AU2004276486B2 (en) 2003-09-29 2004-09-28 Modified vaccinia virus ankara (MVA) mutant and use thereof
US11/375,159 US7767209B2 (en) 2003-09-29 2006-03-14 Modified vaccinia virus Ankara (MVA) mutant and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP03022112A EP1518932A1 (en) 2003-09-29 2003-09-29 Modified vaccinia virus Ankara (MVA) mutant and use thereof
EP03022112.1 2003-09-29

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/375,159 Continuation US7767209B2 (en) 2003-09-29 2006-03-14 Modified vaccinia virus Ankara (MVA) mutant and use thereof

Publications (1)

Publication Number Publication Date
WO2005030971A1 true WO2005030971A1 (en) 2005-04-07

Family

ID=34178523

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/010858 WO2005030971A1 (en) 2003-09-29 2004-09-28 Modified vaccinia virus ankara (mva) mutant and use thereof

Country Status (6)

Country Link
US (1) US7767209B2 (en)
EP (2) EP1518932A1 (en)
CN (1) CN1842602B (en)
AU (1) AU2004276486B2 (en)
BR (1) BRPI0414874A (en)
WO (1) WO2005030971A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7767209B2 (en) 2003-09-29 2010-08-03 Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh Modified vaccinia virus Ankara (MVA) mutant and use thereof
WO2017209053A1 (en) 2016-05-30 2017-12-07 アステラス製薬株式会社 New genetically-modified vaccinia virus
WO2020067085A1 (en) 2018-09-26 2020-04-02 アステラス製薬株式会社 Cancer therapy where oncolytic vaccinia virus and immune checkpoint inhibitor are used in combination, and pharmaceutical composition and combination drug used therein
WO2020148612A1 (en) 2019-01-14 2020-07-23 Ignite Immunotherapy, Inc. Recombinant vaccinia virus and methods of use thereof
WO2020165730A1 (en) 2019-02-14 2020-08-20 Ignite Immunotherapy, Inc. Recombinant vaccinia virus and methods of use thereof
WO2021040056A1 (en) 2019-08-29 2021-03-04 Astellas Pharma Inc. Genetically engineered oncolytic vaccinia viruses and methods of uses thereof
WO2021116943A1 (en) 2019-12-12 2021-06-17 Ignite Immunotherapy, Inc. Variant oncolytic vaccinia virus and methods of use thereof
WO2021140435A1 (en) 2020-01-09 2021-07-15 Pfizer Inc. Recombinant vaccinia virus
WO2022013696A2 (en) 2020-07-14 2022-01-20 Pfizer Inc. Recombinant vaccinia virus

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10143490C2 (en) * 2001-09-05 2003-12-11 Gsf Forschungszentrum Umwelt Recombinant MVA with the ability to express HCV structural antigens
EP1835031A1 (en) 2006-03-14 2007-09-19 Paul-Ehrlich-Institut Bundesamt für Sera und Impfstoffe Use of a recombinant modified vaccinia virus Ankara (MVA) for the treatment of type I hypersensitivity in a living animal including humans
EP2397852B1 (en) 2006-03-14 2013-12-04 Oregon Health and Science University Methods for detecting a mycobacterium tuberculosis infection
CN101209351B (en) * 2006-12-31 2011-05-18 武昌船舶重工有限责任公司 Ankara vaccinia virus genetic engineering vaccine for pig replication and respiration complex
WO2010005474A1 (en) * 2008-06-16 2010-01-14 Emergent Product Development Gaithersburg Inc. Recombinant modified vaccinia virus ankara (mva) expressing chlamydia polypeptide antigens
JP6535337B2 (en) * 2013-11-05 2019-06-26 バヴァリアン・ノルディック・アクティーゼルスカブ Combination therapy for treating cancer with poxvirus expressing a tumor antigen and an antagonist and / or agonist of an immune checkpoint inhibitor
JP2018510143A (en) 2015-02-25 2018-04-12 メモリアル スローン ケタリング キャンサー センター Use of inactivated non-replicating modified vaccinia virus Ankara (MVA) in monotherapy for solid tumors or in combination with immune checkpoint blockers
KR20180006916A (en) * 2015-04-17 2018-01-19 메모리얼 슬로안-케터링 캔서 센터 Use of MVA or MVA delta E3L as an immunotherapeutic agent for solid tumors
BR102015032903A2 (en) * 2015-12-29 2017-09-19 Universidade Federal Do Rio Grande Do Sul RECOMBINANT CEPA OF A PATHOGENIC MICROORGANISM, AND A DOUBLE VACCINE
CA3011014A1 (en) 2016-01-08 2017-07-13 Harriet Robinson Compositions and methods for generating an immune response to a tumor associated antigen
JP7025339B2 (en) 2016-02-25 2022-02-24 メモリアル スローン ケタリング キャンサー センター Replicable attenuated vaccinia virus with or without expression of human FLT3L or GM-CSF with thymidine kinase deletion for cancer immunotherapy
WO2018032057A1 (en) * 2016-08-19 2018-02-22 Sementis Limited Viral vaccines
WO2018209315A1 (en) 2017-05-12 2018-11-15 Memorial Sloan Kettering Cancer Center Vaccinia virus mutants useful for cancer immunotherapy
WO2019040846A1 (en) * 2017-08-25 2019-02-28 Geovax Inc. Immuno-oncology compositions and methods for use therof
US11311612B2 (en) 2017-09-19 2022-04-26 Geovax, Inc. Compositions and methods for generating an immune response to treat or prevent malaria

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993018153A1 (en) * 1992-03-05 1993-09-16 Geoffrey Lilley Smith Vaccinia virus b15r used in interleukin b1-involving condition
WO1997002355A1 (en) * 1995-07-04 1997-01-23 GSF-Forschungszentrum für Umwelt und Gesundheit GmbH Recombinant mva virus, and the use thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE412762T1 (en) * 1999-05-28 2008-11-15 Helmholtz Zentrum Muenchen VECTOR FOR INTEGRATION OF HETEROLOGUESE SEQUENCES INTO POXVIRUS GENOMES
ES2249430T3 (en) * 2000-03-14 2006-04-01 Bavarian Nordic A/S ALTERADA CEPA OF THE ANKARA VACCINE OF THE MODIFIED VACCINE (MVA)
DE10143490C2 (en) * 2001-09-05 2003-12-11 Gsf Forschungszentrum Umwelt Recombinant MVA with the ability to express HCV structural antigens
EP1518932A1 (en) 2003-09-29 2005-03-30 GSF-Forschungszentrum für Umwelt und Gesundheit GmbH Modified vaccinia virus Ankara (MVA) mutant and use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993018153A1 (en) * 1992-03-05 1993-09-16 Geoffrey Lilley Smith Vaccinia virus b15r used in interleukin b1-involving condition
WO1997002355A1 (en) * 1995-07-04 1997-01-23 GSF-Forschungszentrum für Umwelt und Gesundheit GmbH Recombinant mva virus, and the use thereof

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ALCAMI A. ET AL.: "A soluble receptor for interleukin-1-beta encoded by vaccinia virus: A novel mechanism of virus modulation of the host response to infection", CELL, vol. 71, no. 1, 2 October 1992 (1992-10-02), pages 153 - 167, XP001156874 *
C. STAIB ET AL.: "TRANSIENT HOST RANGE SELECTION FOR GENETIC ENGINEERING OF MODIFIED VACCINIA VIRUS ANKARA", BIOTECHNIQUES, EATON PUBLISHING, NATICK, US, vol. 28, no. 6, June 2000 (2000-06-01), pages 1137 - 1148, XP001026101, ISSN: 0736-6205 *
KETTLE SUSAN ET AL: "Vaccinia virus serpin B1 3R (SPI-2) inhibits interleukin-1-beta-converting enzyme and protects virus- infected cells from TNF- and Fas-mediated apoptosis, but does not prevent IL-1-beta-induced fever", JOURNAL OF GENERAL VIROLOGY, vol. 78, no. 3, 1997, pages 677 - 685, XP002266345, ISSN: 0022-1317 *
See also references of EP1668142A1 *
SPRIGGS MELANIE K ET AL: "Vaccinia and cowpox viruses encode a novel secreted interleukin-1 binding protein", CELL, vol. 71, no. 1, 1992, pages 145 - 152, XP001157012, ISSN: 0092-8674 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7767209B2 (en) 2003-09-29 2010-08-03 Gsf-Forschungszentrum Fuer Umwelt Und Gesundheit Gmbh Modified vaccinia virus Ankara (MVA) mutant and use thereof
WO2017209053A1 (en) 2016-05-30 2017-12-07 アステラス製薬株式会社 New genetically-modified vaccinia virus
WO2020067085A1 (en) 2018-09-26 2020-04-02 アステラス製薬株式会社 Cancer therapy where oncolytic vaccinia virus and immune checkpoint inhibitor are used in combination, and pharmaceutical composition and combination drug used therein
WO2020148612A1 (en) 2019-01-14 2020-07-23 Ignite Immunotherapy, Inc. Recombinant vaccinia virus and methods of use thereof
WO2020165730A1 (en) 2019-02-14 2020-08-20 Ignite Immunotherapy, Inc. Recombinant vaccinia virus and methods of use thereof
WO2021040056A1 (en) 2019-08-29 2021-03-04 Astellas Pharma Inc. Genetically engineered oncolytic vaccinia viruses and methods of uses thereof
WO2021116943A1 (en) 2019-12-12 2021-06-17 Ignite Immunotherapy, Inc. Variant oncolytic vaccinia virus and methods of use thereof
WO2021140435A1 (en) 2020-01-09 2021-07-15 Pfizer Inc. Recombinant vaccinia virus
WO2022013696A2 (en) 2020-07-14 2022-01-20 Pfizer Inc. Recombinant vaccinia virus

Also Published As

Publication number Publication date
US20070160627A1 (en) 2007-07-12
BRPI0414874A (en) 2006-12-12
AU2004276486B2 (en) 2010-01-21
CN1842602B (en) 2011-06-29
CN1842602A (en) 2006-10-04
AU2004276486A1 (en) 2005-04-07
EP1668142A1 (en) 2006-06-14
EP1518932A1 (en) 2005-03-30
US7767209B2 (en) 2010-08-03

Similar Documents

Publication Publication Date Title
US7767209B2 (en) Modified vaccinia virus Ankara (MVA) mutant and use thereof
JP6130876B2 (en) Optimized early-late promoter combined with repeated vaccination favors cytotoxic T cell responses to antigens in replication-deficient recombinant virus vaccines
US9416371B2 (en) T-cell vaccination with viral vectors via mechanical epidermal disruption
JP4421188B2 (en) Denatured vaccinia Ankara virus mutant
AU2011316164B2 (en) Recombinant modified vaccinia virus Ankara (MVA) influenza vaccine
US20060216312A1 (en) Mutants of replication competent vaccinia virus
US9163237B2 (en) Replication deficient recombinant viruses expressing antigens regulated by transcriptional control elements comprising multiple elements
JP2013014588A (en) Vaccine based on use of mva
WO2012151272A2 (en) T-cell vaccination with viral vectors via mechanical epidermal disruption
Melamed et al. Attenuation and immunogenicity of host-range extended modified vaccinia virus Ankara recombinants
US20110064769A1 (en) Vv promoter-driven overexpression of recombinant antigens
DK2627774T3 (en) INFLUENZAVACCINE BASED ON RECOMBINANT MODIFIED VACCINIAVIRUS ANKARA (VAT)
NZ711569A (en) Single high dose of mva induces a protective immune response in neonates and infants

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480024419.5

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004276486

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 11375159

Country of ref document: US

Ref document number: 1402/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2004765665

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2004276486

Country of ref document: AU

Date of ref document: 20040928

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004276486

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2004765665

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0414874

Country of ref document: BR

WWP Wipo information: published in national office

Ref document number: 11375159

Country of ref document: US