AU5842099A - Immunogenic compositions and uses thereof - Google Patents

Immunogenic compositions and uses thereof Download PDF

Info

Publication number
AU5842099A
AU5842099A AU58420/99A AU5842099A AU5842099A AU 5842099 A AU5842099 A AU 5842099A AU 58420/99 A AU58420/99 A AU 58420/99A AU 5842099 A AU5842099 A AU 5842099A AU 5842099 A AU5842099 A AU 5842099A
Authority
AU
Australia
Prior art keywords
etn
gpi
domain
molecule
ma4g
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU58420/99A
Other versions
AU766837B2 (en
AU766837C (en
Inventor
Louis Schofield
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Walter and Eliza Hall Institute of Medical Research
Original Assignee
Walter and Eliza Hall Institute of Medical Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AUPP5893A external-priority patent/AUPP589398A0/en
Application filed by Walter and Eliza Hall Institute of Medical Research filed Critical Walter and Eliza Hall Institute of Medical Research
Priority to AU58420/99A priority Critical patent/AU766837C/en
Publication of AU5842099A publication Critical patent/AU5842099A/en
Publication of AU766837B2 publication Critical patent/AU766837B2/en
Application granted granted Critical
Publication of AU766837C publication Critical patent/AU766837C/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Classifications

    • 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

Description

WO 00/15254 PCT/AU99/00770 -1 IMMUNOGENIC COMPOSITIONS AND USES THEREOF FIELD OF THE INVENTION 5 The present invention relates generally to a method of eliciting or otherwise inducing an immune response to a micro-organism and compositions for use therein. More particularly, the present invention relates to a method of inducing an immune response to a parasite utilising an immunogenic composition comprising a glycosylphosphatidylinositol (referred to herein as "GPI") inositolglycan domain or its 10 derivative or equivalent. The present invention is useful, inter alia, as a prophylactic and/or therapeutic treatment for micro-organism infections of mammals such as, for example, parasite infections and in particular infection by Plasmodium species. BACKGROUND OF THE INVENTION 15 Bibliographic details of the publications numerically referred to in this specification are collected at the end of the description. The malaria parasite is considered to be one of the single most serious infectious agents 20 in the world, infecting 5 % of the global population and causing serious mortality and morbidity to sensitive populations and hampering socio-economic development. Severe malaria infection shares several clinical features in common with bacterial septic shock. In both conditions, the excess production by macrophages of pro-inflammatory 25 cytokines such as Tumor Necrosis Factor (TNF), Interleukin-1 (IL-1) and IL-6 occurs in response to malaria "toxins" and lipopolysaccharide (LPS), respectively, leading to complications such as fever and hyperpyrexia, leukopenia, thrombocytopenia, hypotension, disseminated intravascular coagulation, leukocyte infiltration, vascular permeability and multi-organ inflammation, which may lead eventually to death. Thus, 30 many signs, symptons and syndromes in acute and severe malaria infection result from WO 00/15254 PCT/AU99/00770 -2 the activity of a parasite "toxin" released into the circulation during the blood-stage developmental cycle of the infection. GPI has been identified as a candidate toxin of parasite origin (Schofield and Hackett, 5 1993 and Tachado et al, 1997). The structure of the molecule has been elucidated (Gerold et al, 1992 and Gerold et al, 1996) and it comprises a lipidic domain and a glycan domain. Intact GPI occurs in two closely related forms, Pfgle (NH-CH 2
-CH
2 PO 4 -(Mana l-2)-6Manal-2Mana l-6Manal 1-4GlcN-H 2 al-6(myristoyl)-myo-Inositol-1
PO
4 -dipalmitoylglycerol), and Pfglp (NH-CH 2 - CH 2
-PO
4 -6Manal-2Manal 10 6Manal 1,4GlcN-H 2 a 1-6(myristoyl)-myo-Inositol-1-PO 4 -dipalmitoylglycerol). The parasite derived GPI molecule regulates host cell function and gene expression in various tissues by activating endogenous GPI-based signal transduction pathways, involving hydrolysis into second messengers and the activation of both tyrosine and 15 serine/threonine kinases. This leads to the activation of the NFKB/c-rel family of transcription factors, which regulate the expression of numerous pro-inflammatory loci implicated in malarial pathology, such as TNF, IL-1, iNOS and ICAM-1. The toxin theory of malarial pathogenesis can be ascribed to Camillo Golgi, in 1886, 20 who hypothesized that the proximal cause of the febrile paroxysm was a released toxin of parasite origin (Golgi, 1886). Clark proposed that the systemic inflammation of malaria infection resulted from a functional malarial endotoxin, and suggested that this agent exerts systemic effects largely through the induction of endogenous pyrogens of host origin. Clark correctly identified TNF as a major host mediator of disease (Clark, 25 1978 and Clark et al, 1981). Consequently, the production of this and related pyrogenic cytokines (IL-1, IL-6) from monocyte/macrophages is often taken as a useful surrogate marker for the initiation of pathological processes in malaria infection. John Playfair and his colleagues extended this work to show that crude extracts of rodent malaria parasites could induce macrophages to secrete TNF in vitro (Bate et al, 1988 30 and and Bate et al, 1989) and inferred that the toxin included a phospholipid moiety.
WO 00/15254 PCT/AU99/00770 -3 Nonetheless, prior to the advent of the present invention, the specific biochemical identity of the parasite toxin, and its mechanism of action, have remained obscure. In work leading up to the present invention, the inventors investigated the use of 5 portions of GPI to induce protective immunity against malarial pathology. The inventors have surprisingly discovered that GPI portions which exclude the lipidic domain induce protective immunity whereas portions carrying the lipidic domain do not. 10 SUMMARY OF THE INVENTION Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or group of integers but not 15 the exclusion of any other integer or group of integers. One aspect of the present invention is directed to a method of eliciting or inducing, in a mammal, an immune response directed to a micro-organism said method comprising administering to said mammal an effective amount of an immunogenic composition 20 which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI but which molecule is substantially incapable of inducing an immune response directed to a lipidic domain of said GPI. Another aspect of the present invention provides a method of eliciting or inducing, in a 25 mammal, an immune response directed to a micro-organism said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain. 30 WO 00/15254 PCT/AU99/00770 -4 Still another aspect of the present invention is directed to a method of eliciting or inducing, in a mammal, an immune response directed to a parasite said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises the inositolglycan domain portion of a 5 parasite GPI or derivative or equivalent thereof and which inositolglycan domain portion comprises insufficient lipidic domain of said parasite GPI to induce or elicit an immune response directed to said lipidic domain. Still yet another aspect of the present invention contemplates a method of eliciting or 10 inducing, in a mammal, an immune response directed to P. falciparum said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises the structure 15 ethanolamine-phosphate-(Manal,2)-Manal 1,2Mana 1,6Manal 1,4GlcN-myo inositol phosphoglycerol or derivative or equivalent thereof. 20 Still yet another aspect of the present invention contemplates a method of eliciting or inducing, in a mammal, an immune response directed to P. falciparum said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises the structure 25 X, - X2 - X 3
-X
4 - ethanolamine-phosphate-(Manal, 2
)
Manal,2Manal,6Manal,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said 30 GPI inositolglycan domain.
WO 00/15254 PCT/AU99/00770 -5 Still yet another aspect of the present invention contemplates a method of eliciting or inducing, in a mammal, an immune response directed to P. falciparum said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a GPI inositolglycan domain wherein said 5 GPI inositolglycan domain comprises the structure EtN-P-[Ma2]Ma2 Mc6 Ma4Gac6Ino EtN-P-[Mca2][G]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][X]Ma2 Mac6 Ma4Ga6Ino 10 EtN-P-[Ma2][EtN-P]Ma2 Mu6 Ma4Ga6lno EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G EtN-P-Ma2 Ma6 M EtN-P-[Mu2][G]Ma2 Ma6 Ma4G 15 EtN-P-[Ma2][X]Ma2 Ma 6 Ma4G EtN-P-[Ma2] [EtN-P]Mac2 Ma6 Ma4G Ma2 [Ma2][G]Ma2 Ma6 Ma4G Ma2 [Ma2][X]Mac2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Ma 6 Ma4G 20 Ma6 Mu4Ga6Ino Ma2 Ma6 Ma4Ga6Ino Ma2 [Ma2]Ma6 Ma4Ga6Ino Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino 25 EtN-P-[Ma2][G]M2 Ma6 M EtN-P-[Ma2][X]Ma 2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M Ma2 [Ma2][G]Ma2 M(6 M Ma2 [Ma2][X]Ma2 Ma6 M 30 Ma2 [Ma2][EtN-P]Ma 6 M Ma2 Ma6 M WO 00/15254 PCT/AU99/00770 -6 Mca6 Ma4G EtN-P-[Ma2][G]Ma2 M EtN-P-[Mca2][X]Ma2 M EtN-P-[Ma2] [EtN-P]Ma2 M 5 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents ac linkages which may be substituted with P-linkages wherever required, and numeric 10 values represent positional linkages which may be substituted with any other positional linkages as required. A further aspect of the present invention contemplates a method of therapeutically or prophylactically treating a mammal for a micro-organism infection said method 15 comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI, but substantially incapable of inducing an immune response directed to the lipid domain of a GPI, for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise 20 alleviate any one or more symptoms associated with infection of said mammal by said micro-organism. Another further aspect of the present invention is directed to a method of therapeutically or prophylactically treating a mammal for a micro-organism infection 25 said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain for a time and under conditions sufficient for said immune response to 30 reduce, inhibit or otherwise alleviate any one or more symptoms associated with infection of said mammal by said micro-organism.
WO 00/15254 PCT/AU99/00770 -7 In a related aspect, the present invention provides a method for the treatment and/or prophylaxis of a mammalian disease condition characterised by a micro-organism infection, said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of 5 inducing an immune response directed to the inositolglycan domain of a GPI, but substantially incapable of inducing an immune response directed to the lipid domain of a GPI, for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with said micro organism infection. 10 Still another further aspect of the present invention is directed to a method for the treatment and/or prophylaxis of a mammalian disease condition characterised by a micro-organism infection said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a 15 modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with said micro-organism infection. 20 Still yet another aspect of the present invention relates to the use of a composition comprising a molecule capable of inducing an immune response directed to a micro organism GPI inositolglycan domain but substantially incapable of inducing an immune response directed to a lipidic domain of GPI in the manufacture of a medicament for the 25 therapeutic and/or prophylactic treatment of a mammalian disease condition characterised by infection with said micro-organism. Still yet another further aspect of the present invention relates to the use of an immunogenic composition comprising a Plasmodium GPI inositolglycan domain or 30 derivative or equivalent thereof which inositolglycan domain comprises insufficient lipidic domain of a Plasmodium GPI to elicit or induce an immune response directed to WO 00/15254 PCT/AU99/00770 -8 a GPI lipidic domain in the manufacture of a medicament for the therapeutic and/or prophylactic treatment of a mammalian disease condition characterised by infection with said Plasmodium. 5 Another aspect of the present invention is directed to a composition capable of inducing an immune response directed to a micro-organism, said composition comprising a molecule capable of inducing an immune response against a micro-organism GPI inositolglycan domain but substantially incapable of inducing an immune response to a lipidic domain of a GPI. 10 Still another aspect of the present invention is directed to a composition capable of inducing an immune response directed to a micro-organism said composition comprising a modified GPI molecule or derivative or equivalent thereof which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune 15 response directed to a GPI lipidic domain. Yet another aspect of the present invention relates to a vaccine composition comprising as the active component a molecule capable of inducing an immune response directed to a micro-organism GPI inositolglycan domain but substantially incapable of inducing an 20 immune response directed to a lipidic domain of a GPI, as broadly described above, together with one or more pharmaceutically acceptable carriers and/or diluents. Still yet another aspect of the present invention relates to a vaccine composition comprising as the active component a modified GPI molecule or derivative or 25 equivalent thereof which modified GPI molecule or derivative or equivalent thereof which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain. Still another aspect of the present invention is directed to a pharmaceutical composition 30 comprising a molecule capable of inducing an immune response directed to a micro organism GPI inositolglycan domain but substantially incapable of inducing an immune WO 00/15254 PCT/AU99/00770 -9 response directed to a lipidic domain of a GPI, as broadly described above, together with one or more pharmaceutically acceptable carriers and/or diluents. A further aspect of the present invention is directed to antibodies to GPI inositolglycan 5 domains but substantially incapable of interacting with the lipidic domain of a GPI. Yet another aspect of the present invention relates to a pharmaceutical composition comprising an antibody directed to a GPI inositolglycan domain together with one or more pharmaceutically acceptable carriers or diluents as hereinbefore described. 10 A further aspect of the present invention relates to the use of the antibodies of the present invention in relation to disease conditions. For example, the present invention is particularly useful but in no way limited to use in treating parasitic infections, their symptoms and pathologies. 15 Another aspect of the present invention relates to a method of inhibiting, halting or delaying the onset of progression of a mammalian disease condition characterised by a micro-organism infection said method comprising administering to said mammal an effective amount of an antibody has hereinbefore described. 20 In yet another aspect the present invention relates to the use of an antibody in the manufacture of a medicament for inhibiting, halting or delaying the onset or progression of a disease condition characterised by the infection of a mammal by a micro-organism.
WO 00/15254 PCT/AU99/00770 - 10 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a graphical representation of the epitope specificity of anti-GPI antibodies determined by competition ELISA. Sera from mice immunized with free GPI were 5 screened for reactivity to malarial GPI in the presence or absence of defined competitors (Phosphatidylinositol or phosphatidylserine). Figure 2 is a graphical representation of the results of C57B1/6 mice immunized with free GPI in IFA and sham-immunized mice (IFA alone) which were challenged with P. 10 berghei ANKA and survival assessed over 15 days. Figure 3 is a graphical representation of the epitope mapping of anti-lipid monoclonal antibodies. Monoclonal antibody 1C7 to GPI derived from mice immunized with free GPI (1,5) were screened by competition ELISA for reactivity with GPI in the presence 15 or absence of PI and GPI glycan competitors. Figure 4 is a graphical representation of monoclonal antibody 1C7, to malarial GPI lipid domains, recognition of mammalian GPIs at the cell surface as determined by FACS analysis. Solid line, binding of 1C7 to macrophages; dotted line, no antibody; 20 dashed line, binding of 1C7 following PI-PLC treatment of macrophages. Figure 5 is a photographic representation of monoclonal 1C7, to lipid domain of the GPI, induction of rapid onset tyrosylphosphorylation in host cells. 25 Figure 6 is a graphical representation of monoclonal 1C7 synergizy with GPI, phorbol esters and parasite extracts in the induction of TNF output from murine C3H/HeJ macrophages. Figure 7 is a graphical representation of monoclonal 1C7 exacerbation of the P. 30 berghei ANKA cerebral malaria syndrome in C57B1/6 mice.
WO 00/15254 PCT/AU99/00770 - 11 Figure 8 is a graphical representation of polyclonal antisera from mice immunized with the purified P. falciparum GPI glycan covalently conjugated to a protein carrier inhibiting TNF output from macrophages in response to GPI or total parasite extracts. Values show absorbance at 450mM by anti-TNF ELISA (Pharmingen) and are 5 proportional to mass TNF. Figure 9 is a graphical representation of immunization of C57B1/6 mice with the purified P. falciparum GPI glycan covalently coupled to KLH providing a significant level of protection against the cerebral malaria syndrome induced by P. berghei ANKA. 10 Figure 10 is a graphical representation of GPI being the dominant TNF-inducing toxin of P. falciparum. Monoclonal antibodies 1G7 and 3G6 specific for the GPI glycan derived from OVA-TCR mice immunized with OVA-glycan inhibiting TNF output from macrophages in response to total parasite extracts. 15 Figure 11 is a graphical representation of monoclonal antibodies to the P. falciparum GPI inositolglycan, upon passive transfer, substantially protecting mice against cerebral malaria and other pathologies. 20 Figure 12 is a graphical representation of monoclonal antibodies to the P. falciparum inositolglycan, upon passive transfer, protecting mice against parasite-induced lethal toxic shock.
WO 00/15254 PCT/AU99/00770 - 12 DETAILED DESCRIPTION OF THE INVENTION The present invention is predicated, in part, on the surprising determination that a Plasmodium GPI molecule which excludes the lipidic portion will induce protective 5 immunity whereas a GPI molecule which comprises the lipidic domain will not. This determination has facilitated the development of compositions and methodology for application, inter alia, in the prophylactic or therapeutic treatment of micro-organism infection. 10 Accordingly, one aspect of the present invention is directed to a method of eliciting or inducing, in a mammal, an immune response directed to a micro-organism said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI but which molecule is 15 substantially incapable of inducing an immune response directed to a lipidic domain of said GPI. The present invention is predicated on the surprising observation that mice immunised with purified, intact, free GPI mount an IgM dominated response directed 20 predominantly to the lipidic domain of the molecule, which cross reacts with host GPI lipidic domains which are exposed at host cell surfaces. The antibodies are not protective clinically against subsequent parasite infection. In fact, passive transfer of these antibodies exacerbates disease severity. However, immunisation with the glycan domain of malarial GPI results in IgG antibodies interactive with the glycan domain of 25 GPI and mice thus immunised are substantially protected against pathology induced by subsequent malaria challenge. Passive transfer of these IgG antibodies is protective against pathology. The inventors have demonstrated, therefore, that IgM antibodies to the lipidic domain and IgG antibodies to the glycan domain of the malaria GPI differ in their effects, the former promoting disease and the latter preventing it. It should be 30 understood that in preventing or minimising the induction of an immune response directed to the GPI of a micro-organism, the onset of an immune response directed to WO 00/15254 PCT/AU99/00770 - 13 lipidic domain of the subject mammal (host) is thereby prevented or minimised by virtue of minimising the production of antibodies to a micro-organism GPI which would otherwise cross react with the host GPI. 5 GPIs are ubiquitous among eukaryotes, described from T. brucei, T. cruzi, Plasmodium, Leishmania, and Toxoplasma, as well as yeast, insect, fish and numerous mammalian sources (for recent reviews see McConville and Ferguson, 1993 and Stevens, 1995). GPIs consist of a conserved core glycan (Manal-2Mancal-6Mancl 4GlcNH 2 ) linked to the 6-position of the myo-inositol ring of phosphatidylinositol (PI). 10 GPIs are built up on the cytoplasmic face of the endoplasmic reticulum (ER) by the sequential addition of sugar residues to PI by the action of glycosyltransferases. The maturing GPI is then translocated across the membrane to the luminal side of the ER, whence it may be exported to the cell surface, free or in covalent association with proteins. The tetrasaccharide core glycan may be further substituted with sugars, 15 phosphates and ethanolamine groups in a species and tissue-specific manner. GPI fatty acid moieties can be either diacylglycerols, alkylacylglycerols, monoalkylglycerols or ceramides, with additional palmitoylations or myristoylations to the inositol ring. The overall picture is of a closely related family of glycolipids sharing certain core features but with a high level of variation in fatty acid composition and side-chain modifications 20 to the conserved core glycan. Accordingly, reference herein to "GPI inositolglycan domains" should be read as including reference to all forms of GPI inositolglycan domains and derivatives or equivalents thereof. The term "GPI inositolglycan" is used interchangeably with terms 25 such as but not limited to "inositolglycan" (IG), "inositophosphoglycan" (IPG), "phosphoinositolglycan" (PIG), "phosphooligosaccharide" (POS) and the molecules described by these terms should be understood as "GPI inositolglycan" molecules. It should also be understood that reference to "GPI inositolglycan domain" includes reference to a GPI inositolglycan domain linked, bound or otherwise associated with 30 non-inositolglycan molecules such as, but not limited to, the glycerol linker sequence WO 00/15254 PCT/AU99/00770 - 14 which links the lipidic domain to the inositolglycan domain, a non-immunogenic portion of the lipidic domain or an amino acid peptide. Preferably the molecule is a portion of GPI which comprises the inositolglycan domain 5 or derivative or equivalent thereof but substantially does not contain a portion capable of inducing an immune response directed to a lipidic domain of said GPI. Accordingly, the present invention more particularly provides a method of eliciting or inducing, in a mammal, an immune response directed to a micro-organism said method 10 comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain. 15 Preferably, said modified GPI molecule is the inositolglycan domain portion of GPI or derivative or equivalent thereof. Still without limiting the present invention in any way, the administration of an immunogenic composition comprising an inositolglycan domain portion of GPI or 20 derivative or equivalent thereof substantially lacking the lipidic domain, as hereinbefore defined, is also thought to benefit the subject mammal by minimizing certain unwanted responses which may otherwise occur incidentially to immune response induction, but which enhance disease severity, if the subject immunogenic molecule comprised a lipid domain. Specifically; 25 (i) the intact GPI is a toxin and may induce non-immunological physiological sensitization in recipients such that the response to the natural GPI toxin is exacerbated upon malaria challenge. The inventors have shown that the lipidic portion of the intact GPI is necessary for full toxic activity by virtue of its' 30 ability to initiate lipid-dependent signaling in host cells, and act as a lipidic second messenger; WO 00/15254 PCT/AU99/00770 - 15 (ii) intact glycolipids may associate with host CD1 molecules for presentation to CD 1 -restricted NKT cells or other unusual T cell lineages. These T cells are known to produce high levels of cytokines such as interferon-y and IL-4 very rapidly in response to stimulation and are likely to be crucial regulators of 5 downstream TH1/TH2 differentiation. Immunization with purified, intact (i.e. lipidated), free GPI may result in priming of these T cells which subsequently respond with high levels of interferon-y upon parasite challenge, thereby exacerbating the disease syndromes. That is, immunological sensitization of unusual T cells may contribute to the phenomenon of exacerbated disease 10 severity. "Derivatives" and "equivalents" should be understood to include fragments, parts, portions, chemical equivalents, mutants, homologs and analogs. Chemical equivalents of a GPI inositolglycan domain can act as a functional analog of the GPI inositolglycan 15 domain. For example, a chemical equivalent of the GPI inositolglycan domain includes a GPI inositolglycan domain in which the phosphoglycerol component of the inositolglycan has been modified to increase hydrophobicity. This may be achieved by replacement with truncated, partial or modified fatty acids or other hydrophobic moieties and acts to improve the immunogenicity or stability of the molecule, without 20 generating an undesirable antibody response. Chemical equivalents may not necessarily be derived from a GPI inositolglycan domain but may share certain confirmational similarities. Alternatively, chemical equivalents may be specifically designed to mimic certain immunological and physiochemical properties of the GPI inositolglycan domain. Chemical equivalents may be chemically synthesised or may be detected following, for 25 example, natural product screening. Chemical equivalents also include synthetic carbohydrates and peptide mimics. Homologs of GPI inositolglycan domains contemplated herein include, but are not limited to, GPI inositolglycan domains from different species including, for example, Saccharomyces. Fragments, include portions such as the glycan component of the inositolglycan domain, which portions are effective 30 in achieving the object of the present invention.
WO 00/15254 PCT/AU99/00770 - 16 GPI inositolglycan domains suitable for use in the present invention may be derived from any natural, recombinant or synthetic source. This includes, for example, GPI inositolglycan domains derived by genetic manipulation of expression systems, and by manipulations of the GPI post-translational modifications of proteins via recombinant 5 DNA techniques such as glycosylation inhibitors. It also includes chemically synthetic or semi-synthetic inositolglycan domains and fragments thereof derived by any chemical process including the use of enzymes for the addition or removal of residues. The term "micro-organism" should be understood in its broadest sense and includes, for 10 example, the parasitic and fungal taxa Plasmodium, Trypanosoma, Leishmania, Toxoplasma and Candida. "Micro-organism" should also be understood to extend to molecules which are secreted by or shed from the subject organism. This would include for example, toxin molecules or molecules which are cleared from the surface of the micro-organism. Preferably, the GPI inositolglycan domain suitable for use in 15 the present invention is a parasite GPI inositolglycan domain and even more preferably a Plasmodium GPI inositolglycan domain. Accordingly, the present invention is preferably directed to a method of eliciting or inducing, in a mammal, an immune response directed to a parasite said method 20 comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises the inositolglycan domain portion of a parasite GPI or derivative or equivalent thereof and which inositolglycan domain portion comprises insufficient lipidic domain of said parasite GPI to induce or elicit an immune response directed to said lipidic domain. 25 Even more preferably said parasite GPI inositolglycan domain is a Plasmodium GPI inositolglycan domain or derivative or equivalent thereof. Most preferably, said Plasmodium is P. falciparum. 30 WO 00/15254 PCT/AU99/007704 - 17 Yet even more preferably, the present invention contemplates a method of eliciting or inducing, in a mammal, an immune response directed to P. falciparum said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a GPI inositolglycan domain wherein said 5 GPI inositolglycan domain comprises the structure ethanolamine-phosphate-(Manal 1,2)-Manal 1,2Mana 1,6Mancal 1,4GlcN-myo inositol phosphoglycerol 10 or derivative or equivalent thereof. In another most preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises the structure 15 X 1 - X2 - X 3
-X
4 - ethanolamine-phosphate-(Manal,2) Manal,2Mana l,6Manal,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain. 20 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: 25 EtN-P-[Ma2]M2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 M4Ga6Ino EtN-P-[Ma2][X]Ma2 Mca6 Mca4Ga6Ino EtN-P-[Mca2][EtN-P]Ma2 Ma6 Ma4Ga6Ino 30 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, WO 00/15254 PCT/AU99/00770 - 18 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 5 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: 10 EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G EtN-P-Ma2 Ma6 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 15 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 20 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: 25 EtN-P-[Ma2][G]Ma2 Ma6 Ma4G EtN-P-[Ma2][X]Ma2 Ma6 Ma4G EtN-P-[Ma2][EtN-P]Ma2 Ma6 Mca4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 30 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a- WO 00/15254 PCT/AU99/00770 - 19 linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 5 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: Ma2 [Ma2][G]Ma2 Ma6 Ma4G 10 Ma2 [Ma2][X]Ma2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 15 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 20 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: Ma6 Ma4Ga6Ino 25 Ma2 Ma6 Mac4Ga6Ino Ma2 [Ma2]Ma6 Ma4Ga6Ino Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino 30 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, WO 00/15254 PCT/AU99/00770 - 20 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 5 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: 10 EtN-P-[Ma2][G]Ma2 Ma6 M EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 15 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 20 In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure selected from: 25 Ma2 [Ma2][G]Ma2 Mac6 M Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G 30 WO 00/15254 PCT/AU99/00770 -21 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric 5 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment the immunogenic composition comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises a structure 10 selected from: EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M 15 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric 20 values represent positional linkages which may be substituted with any other positional linkages as required. Any of these structures may be further modified by substituents of positive, negative or neutral charge such as phosphates, phosphoglycerol, hexosamines, amino acids, thiols 25 etc in any position and with any type of linkage. These structures may be further modified by addition of any number of amino acids for the purpose of providing a linkage sequence. It should be understood that non-N-acetylated hexosamine includes glucosamine or any 30 other nitrous-acid labile substituent. It should be further understood that any of these structures may be further modified by substituents including, but not limited to, of WO 00/15254 PCT/AU99/00770 - 22 positive, negative or neutral charge such as phosphates, phosphoglycerol, hexosamines, amino acids or thiols in any position and with any type of linkage. The GPI inositolglycan domain of the present invention may be conjugated to another 5 molecule. Said conjugation may be performed for any one or more reasons such as, but not limited to: (i) The GPI inositolglycan domain may be too small to be antigenic. Accordingly, conjugation to a carrier molecule, such as a protein, may be required such that 10 said GPI inositolglycan domain, which forms part of the GPI inositolglycan domain-conjugate, acts as a hapten and immunity is induced to said GPI inositolglycan domain. The carrier protein may be selected from a range of antigenic proteins such as but not limited to recombinant proteins derived from Plasmodium gene sequences, tetanus toxoid, purified protein derivative, 15 hepatitis B or Key Hole Limpet Haemocyanin and Diptheria toxoid. (ii) The GPI inositolglycan domain when conjugated with specific anti-pathogen vaccine molecules (such as anti-malarial vaccine molecules) may result in the production of anti-inositolglycan domain antibodies which reverse the immune 20 suppression that otherwise may occur in response to exposure to the native form of the vaccine molecule where said molecule is itself GPI-anchored. For example, the GPI inositolglycan domain may be coupled to a malarial recombinant protein which can act as both a carrier protein and a vaccine in its own right. 25 Without intending to limit this aspect of the present invention to any one theory or mode of action, primary and secondary T lymphocyte responses to some GPI anchored surface protein antigens are inhibited by the GPI anchor. Examples of such protein antigens includes Circumsporozoite (CS) proteins of P. falciparum 30 and P. berghei and the membrane-form of Variant Surface Glycoprotein of T. brucei. Since immunisation against synthetic or recombinant peptides or WO 00/15254 PCT/AU99/00770 - 23 proteins of GPI-anchored surface molecules such as the CS protein, MSP-1, MSP-2 or MSP-4, for example, may be insufficient to allow MHC Class II anamnestic boosting when the native antigens are encountered during natural parastic challenge due to the induction of immunosuppresion, immunisation 5 against the GPI moiety provides a means to alleviate this immunosuppresion. (iii) The GPI inositolglycan domain may comprise only part of the target epitope. For example, peptide sequences, other carbohydrates (and any associated post translational modifications) corresponding to C-terminal domains of native GPI 10 anchored proteins or GPI-anchored glycosconjugates may also form part of the target GPI inositolglycan domain epitope. Removal of any part of the epitope (by removing the portion of the C-terminal domain which forms part of the GPI inositolglycan domain epitope) may lead to reduction or loss of binding of antibodies. Said peptide sequences or carbohydrates would therefore be 15 conjugated to said GPI inositolglycan domain. For example, some antibodies to malarial GPI, while specifically neutralising GPI function, recognise epitopes which predominantly include the inositolglycan but also include portions of the protein to which the GPIs are actually bound in nature, i.e. the adjacent C terminal portions of GPI-anchored proteins. The presence of peptide domains 20 can also improve the affinity of certain antibodies, for example by helping to stabilise the inositolglycan conformationally. Furthermore, such conjugation can render a relatively unimmunogenic inositolglycan domain sufficiently immunogenic. Specifically, the inclusion of a C-terminal peptide determinant, for example, may help increase the immunogenicity of the inositolglycan by 25 forming a composite antigen which is more immunologically foreign than inositolglycan alone. The resulting GPI inositolglycan domain-conjugate may be administered as a preparation formulated in or with an adjuvant. The adjuvant is selected from the range 30 of adjuvants known to induce high levels of antibody, including water in oil emulsions, oil in water emulsions, water in oil in water double emulsions, saponin, Quil A extracts WO 00/15254 PCT/AU99/00770 - 24 and other derivatives of saponin, DEAE-dextran, dextran sulphate, aluminium salts and nonionic block co-polymers. The adjuvant may include other immunomodulators, such as muramyl-dipeptide and derivatives, cytokines, and cell wall components from species of mycobacteria or corynebacteria. The adjuvant formulation may include a 5 combination of two or more of the adjuvants listed. These lists are not to be taken as exhaustive. The selection of adjuvant is in part dependent on the species being targeted and is based on the level and duration of the immune response required and on the lack of reactogenicity (ie tissue compatibility). The level of active component and adjuvant are chosen to achieve the desired level and duration of immune response. 10 Host GPIs play a significant role in the normal physiological regulation of various cellular responses in higher eukaryotes. Foreign GPIs such as GPIs of parasite origin exert pathophysiological effects, and specifically regulate host cell function, by acting as a mimic of endogenous host GPI signalling pathways. Signal transduction induced in 15 host cells by GPI's of P. falciparum, T. brucei, and L.mexicana, for example, activate the macrophage lineage-specific hck member of the src-family of protein tyrosine kinases within 30 seconds of addition to cells (Tachado et al, 1997). Protein tyrosine kinase (PTK) activation is required for downstream gene expression resulting in phosphorylation, cell signalling and TNF, IL-1, iNOS, ICAM-1 and VCAM expression 20 (Schofield et al, 1996, Tachado et al, 1996 and Tachado et al, 1997). PTK activation maps to the inositolglycan moiety of GPI and follows binding of the core glycan to a receptor on the surface of cells (Tachado et al, 1997). Parasite GPIs appear to activate similar kinases as those activated upon perturbation of endogenous GPI-anchored proteins at the cell surface. 25 The toxic nature of foreign GPIs such as parasite GPIs can be exemplified with respect to malarial GPIs. When inoculated in vivo, the malarial GPI induces pyrexia and symptoms of acute malaria and causes the death of the recipient in a standard assay of TNF driven lethality (Schofield and Hackett, 1993). In addition to inducing TNF and 30 IL-1 expression in macrophages, the GPI exerts several other TNF independent effects on host tissues that may contribute to pathological processes in malaria infections. GPI WO 00/15254 PCT/AU99/00770 - 25 directly increases expression of E-selectin, ICAM and VCAM in vascular endothelial cells (Schofield et al, 1996). GPI also induces de novo protein synthesis of inducable nitric oxide synthase and the production of NO in a time and dose dependent manner, from macrophages and synergises with interferon-y in this activity (Shenoy-Scaria et al, 5 1993). In the hypoxic or ischaemic model, cerebral malaria is proposed to result from a blockage of the post capillary venules of the brain by sequestered parasite infected erythrocytes binding to the adhesion molecules ICAM, VCAM and E-selectin (Berendt et al, 1994). GPI can therefore be lethal in vivo and induce malarial symptomology encompassing both systemic inflammation and organ-specific pathology such as the 10 cerebral syndrome. Foreign GPIs may also induce immunosuppression. GPIs isolated from P. falciparum and T. brucei, for example, when added at low concentrations to cultures of CD4 + and CD8 + a P TCR+ T cells block cell cycle progression and cellular proliferation, 15 inhibiting the upregulation of IL-2 R/CD25 and CD28 expression and blocking expression of IL-2, interferon y, and IL-4. The GPIs also inhibit the T cell proliferative response to IL-2. In vivo, GPI anchored surface proteins such as malaria CS protein, MSP-1, MSP-2, and the membrane form variant surface glycoprotein of T. brucei inhibit, via the covalently associated GPI anchor, primary and secondary T 20 lymphocyte responses to said antigens. While not intending to limit the present invention to any one theory or mode of action, immunisation with a GPI molecule lacking the lipid domain induces an IgG response to the inositolglycan domain which blocks subsequent parasitic GPI action. Both toxicity 25 and immunosuppression, as described above, are significantly reduced. A further aspect of the present invention relates to the use of the invention in relation to disease conditions. For example, the present invention is particularly useful, but in no way limited to use in therapeutically or prophylactically treating parasitic infections 30 such as by immunizing a mammal against a parasitic infection. In this regard, it should be understood that the method of the present invention is directed to inducing an WO 00/15254 PCT/AU99/00770 - 26 immune response for the purpose of alleviating or preventing the onset of symptoms associated with a parasitic infection (such as toxicity and immunosuppression) and/or where the GPI domain is conjugated to a suitable antipathogen molecule, reducing or preventing parasitic infection. Reference herein to "symptoms" associated with a 5 micro-organism infection should be understood to extend to both the infection itself as well as the physical and/or physiological consequences (such as toxicity or immunosuppresion) of such an infection. Accordingly, another aspect of the present invention contemplates a method of 10 therapeutically or prophylactically treating a mammal for a micro-organism infection said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI, but substantially incapable of inducing an immune response directed to the lipid domain of a GPI, for a 15 time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with infection of said mammal by said micro-organism. More particularly, the present invention is directed to a method of therapeutically or 20 prophylactically treating a mammal for a micro-organism infection said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain for a 25 time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with infection of said mammal by said micro-organism. Preferably, said micro-organism is a parasite and even more preferably Plasmodium 30 falciparum.
WO 00/15254 PCT/AU99/00770 - 27 In accordance with this preferred aspect of the present invention, the immunogenic composition preferably comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises the structure 5 ethanolamine-phosphate-(Man 1,2)-Manal 1,2Manal 1,6Mana 1l,4GlcN-myo inositol phosphoglycerol or derivative or equivalent thereof. 10 In another preferred embodiment, the subject inositolglycan domain comprises the structure X1 - X 2 - X 3
-X
4 - ethanolamine-phosphate-(Mancl,2) Manal 1,2Manal ,6Mancal 1,4GlcN-myo-inositol phosphoglycerol 15 wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain. In still another preferred embodiment, the subject inositolglycan domain comprises a 20 structure selected from: EtN-P-[Ma2]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Mca2 Ma6 Ma4Gca6Ino EtN-P-[M2][X]Ma2 Ma6 Ma4Ga6Ino 25 EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4Ga6Ino or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a 30 linkages which may be substituted with P-linkages wherever required, and numeric WO 00/15254 PCT/AU99/00770 - 28 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a 5 structure selected from: EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G EtN-P-Ma2 Ma6 M 10 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric 15 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 20 EtN-P-[Ma2][G]Ma2 Ma6 Ma4G EtN-P-[Ma2][X]Ma2 Ma6 Ma4G EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4G 25 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 30 linkages as required.
WO 00/15254 PCT/AU99/00770 - 29 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: Ma2 [Ma2][G]Ma2 Ma6 Ma4G 5 Ma2 [Ma2][X]Mca2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 10 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 15 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: Ma6 Ma4Ga6Ino Ma2 Ma6 Ma4Ga6Ino 20 Ma2 [Ma2]Mac6 Ma4Ga6Ino Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 25 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 30 WO 00/15254 PCT/AU99/0077(T - 30 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Ma2][G]Ma2 Ma6 M 5 EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 10 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 15 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: Ma2 [Ma2][G]Ma2 Ma6 M Ma2 [Ma2][X]Ma2 Ma6 M 20 Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 25 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 30 WO 00/15254 PCT/AU99/00770 -31 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Ma2][G]Ma2 M 5 EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 10 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 15 The term "mammal" includes humans, primates, livestock animals (eg. horses, cattle, sheep, pigs, donkeys), laboratory test animals (eg. mice, rats, rabbits, guinea pigs), companion animals (eg. dogs, cats) and captive wild animals (eg. kangaroos, deer, foxes). Preferably, the mammal is a human or laboratory test animal. Even more preferably, the mammal is a human. 20 The mammal undergoing treatment may be a human or animal in need of therapeutic or prophylactic treatment for a disease condition or a potential disease condition. Without limiting this aspect of the present invention, administration of said 25 immunogenic composition may act to result in production of antibodies which either prevent manifestation of symptoms such as toxicity and immunosuppression or which affect the parasite directly, for example, by killing the parasite via binding to its surface and inhibiting its growth, development or the onward progression of the overall infection. 30 WO 00/15254 PCT/AU99/00770 ~ - 32 An "effective amount" means an amount necessary at least partly to attain the desired immune response, or to prevent or to delay the onset or inhibit progression or halt altogether, the onset or progression of a particular condition being treated. This amount varies depending upon the health and physical condition of the individual to be 5 treated, the taxonomic group of individual to be treated, the capacity of the individual's immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials. 10 Reference herein to "treatment" and "prophylaxis" is to be considered in its broadest context. The term "treatment" does not necessarily imply that a mammal is treated until total recovery. Similarly, "prophylaxis" does not necessarily mean that the subject will not eventually contract a disease condition. Accordingly, treatment and 15 prophylaxis include amelioration of the symptoms of a particular condition or preventing or otherwise reducing the risk of developing a particular condition. The term "prophylaxis" may be considered as reducing the severity of onset of a particular condition. "Treatment" may also reduce the severity of an existing condition or the frequency of acute attacks (for example, reducing the severity of initial infection). 20 In accordance with these methods, the modulatory agent defined in accordance with the present invention may be coadministered with one or more other compounds or molecules. By "coadministered" is meant simultaneous administration in the same formulation or in two different formulations via the same or different routes or 25 sequential administration by the same or different routes. By "sequential" administration is meant a time difference of from seconds, minutes, hours or days between the administration of the two types of molecules, These molecules may be administered in any order. 30 In a related aspect, the present invention provides a method for the treatment and/or prophylaxis of a mammalian disease condition characterised by a micro-organism WO 00/15254 PCT/AU99/00770 - 33 infection, said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI, but substantially incapable of inducing an immune response directed to the lipid domain of 5 a GPI, for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with said micro organism infection. More particularly, the present invention is directed to a method for the treatment and/or 10 prophylaxis of a mammalian disease condition characterised by a micro-organism infection said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI 15 lipidic domain for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with said micro-organism infection. Preferably, said disease condition is malaria and said micro-organism is Plasmodium 20 falciparum. In accordance with this preferred aspect of the present invention, the immunogenic composition preferably comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises the structure 25 ethanolamine-phosphate-(Manal,2)-Manal,2Mana l,6Manal,4GlcN-myo inositol phosphoglycerol or derivative or equivalent thereof. 30 WO 00/15254 PCT/AU99/00770~ - 34 In another preferred embodiment, the subject inositolglycan domain comprises the structure X1 - X 2 - X 3
-X
4 - ethanolamine-phosphate-(Manal,2) 5 Manxal,2Manal,6Manal,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain. 10 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Ma2]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 Ma4Ga6Ino 15 EtN-P-[Ma2][X]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4Gc6Ino or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 20 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 25 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G 30 EtN-P-Ma2 Ma6 M WO 00/15254 PCT/AU99/00770 - 35 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents Ca linkages which may be substituted with P-linkages wherever required, and numeric 5 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 10 EtN-P-[Ma2][G]Ma2 Ma6 Ma4G EtN-P-[Mc2][X]Ma2 Ma6 Ma4G EtN-P-[Mca2][EtN-P]Ma2 Ma6 Ma4G 15 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 20 linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 25 Ma2 [Ma2][G]Ma2 Ma6 Ma4G Ma2 [Ma2][X]Ma2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 30 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a- WO 00/15254 PCT/AU99/00770~ - 36 linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 5 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: Ma6 Ma4Ga6Ino Ma2 Ma6 Ma4Ga6Ino 10 Ma2 [Ma2]Ma6 Mu4Ga6Ino Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 15 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 20 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Ma2][G]Ma2 Mu6 M 25 EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Mu6 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 30 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric WO 00/15254 PCT/AU99/0077(Tff - 37 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a 5 structure selected from: Mca2 [Mu2][G]Ma2 Ma6 M Mca2 [Mca2][X]Mca2 Ma6 M Ma2 [Ma2][EtN-P]Mc6 M 10 Ma2 Mc6 M Mca6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 15 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 20 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2][X]Ma2 M 25 EtN-P-[Ma2][EtN-P]Ma2 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a 30 linkages which may be substituted with P-linkages wherever required, and numeric WO 00/15254 PCT/AU99/00770 - 38 values represent positional linkages which may be substituted with any other positional linkages as required. In yet another aspect the present invention relates to the use of a composition 5 comprising a molecule capable of inducing an immune response directed to a micro organism GPI inositolglycan domain but substantially incapable of inducing an immune response directed to a lipidic domain of GPI in the manufacture of a medicament for the therapeutic and/or prophylactic treatment of a mammalian disease condition characterised by infection with said micro-organism. 10 Accordingly, another aspect the present invention relates to the use of an immunogenic composition comprising a Plasmodium GPI inositolglycan domain or derivative or equivalent thereof which inositolglycan domain comprises insufficient lipidic domain of a Plasmodium GPI to elicit or induce an immune response directed to a GPI lipidic 15 domain in the manufacture of a medicament for the therapeutic and/or prophylactic treatment of a mammalian disease condition characterised by infection with said Plasmodium. Preferably said disease condition is malaria. 20 The present invention should also be understood to extend to immunogenic compositions for use in the methods as hereinbefore defined. Accordingly, in a related aspect, the present invention is directed to a composition 25 capable of inducing an immune response directed to a micro-organism, said composition comprising a molecule capable of inducing an immune response against a micro-organism GPI inositolglycan domain but substantially incapable of inducing an immune response to a lipidic domain of a GPI. 30 More particularly, the present invention is directed to a composition capable of inducing an immune response directed to a micro-organism said composition WO 00/15254 PCT/AU99/00770 - 39 comprising a modified GPI molecule or derivative or equivalent thereof which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain. 5 Preferably, said modified GPI molecule is the inositolglycan domain portion of GPI. Even more preferably, said micro-organism is a parasite and said parasite is Plasmodium. 10 In accordance with this preferred aspect of the present invention, the immunogenic composition preferably comprises a GPI inositolglycan domain wherein said GPI inositolglycan domain comprises the structure ethanolamine-phosphate-(Manal 1,2)-Mana 1,2Manc 1,6Mana 1,4GlcN-myo 15 inositol phosphoglycerol or derivative or equivalent thereof. In another preferred embodiment, the subject inositolglycan domain comprises the 20 structure
X
1 - X 2 - X 3
-X
4 - ethanolamine-phosphate-(Manal,2) Man al,2Man al, 6Mana l ,4GlcN-myo-inositol phosphoglycerol 25 wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 30 EtN-P-[Mca2]Ma2 Ma6 Ma4Ga6Ino WO 00/15254 PCT/AU99/0077ff - 40 EtN-P-[Ma2][G]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Mc2][X]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][EtN-P]Ma2 M(6 Ma4Ga6Ino 5 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 10 linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 15 EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G EtN-P-Ma2 Ma6 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 20 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 25 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Ma2][G]Ma2 Ma6 Ma4G 30 EtN-P-[Ma2][X]Ma2 Ma6 Ma4G EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4G WO 00/15254 PCT/AU99/0077(f -41 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric 5 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 10 Ma2 [Ma2][G]Ma2 Ma6 Ma4G Ma2 [Ma2][X]Ma2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Ma6 Ma4G 15 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 20 linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 25 Ma6 Ma4Ga6Ino Ma2 Ma6 Ma4Ga6Ino Ma2 [Ma2]Mu6 Ma4Ga6Ino Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Mu4Ga6Ino 30 WO 00/15254 PCT/AU99/00770 - 42 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric 5 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 10 EtN-P-[Ma2][G]Ma2 Ma6 M EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M 15 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 20 linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 25 Ma2 [Ma2][G]Ma2 Ma6 M Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G 30 WO 00/15254 PCT/AU99/00770 - 43 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric 5 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 10 EtN-P-[Ma2][G]Max2 M EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M 15 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 20 linkages as required. Yet another aspect of the present invention relates to a vaccine composition comprising as the active component a molecule capable of inducing an immune response directed to a micro-organism GPI inositolglycan domain but substantially incapable of inducing an 25 immune response directed to a lipidic domain of a GPI, as broadly described above, together with one or more pharmaceutically acceptable carriers and/or diluents. More particularly, the present invention relates to a vaccine composition comprising as the active component a modified GPI molecule or derivative or equivalent thereof 30 which modified GPI molecule or derivative or equivalent thereof which modified GPI WO 00/15254 PCT/AU99/00770 - 44 molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain. Preferably said modified GPI molecule is a GPI inositoglycan domain. 5 More preferably, said GPI inositolglycan domain is a parasite GPI inositolglycan domain and even more preferably a Plasmodium GPI inositolglycan domain. Most preferably, said Plasmodium is P. falciparum. 10 In a most preferred embodiment, said molecule is a GPI inositolglycan domain comprising the structure ethanolamine-phosphate-(Man 1,2)-Manal 1,2Manal 1,6Manal 1,4GlcN 15 phosphatidyl-myo-inositol phosphoglycerol. In another most preferred embodiment said molecule is a GPI inositolglycan domain comprising the structure 20 X 1 - X2 - X3 - X4 - ethanolamine-phosphate-(Manl,2) Manal 1,2Manl 1,6Manl 1,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 , X 4 , are any 4 amino acids. 25 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: EtN-P-[Mca2]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 Ma4Ga6Ino 30 EtN-P-[Mca2][X]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Mc2] [EtN-P]Ma2 Ma6 Ma4Ga6Ino WO 00/15254 PCT/AU99/00770 - 45 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric 5 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 10 EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G EtN-P-Ma2 M(6 M 15 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with p-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional 20 linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 25 EtN-P-[Ma2][G]Ma2 Ma6 Ma4G EtN-P-[Ma2][X]Ma2 Ma6 Ma4G EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is 30 mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a- WO 00/15254 PCT/AU99/00770 - 46 linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. 5 In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: Ma2 [Ma2][G]Ma2 Ma6 Ma4G Ma2 [Ma2][X]Ma2 Ma6 Ma4G 10 Ma2 [Mc2][EtN-P]Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a 15 linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a 20 structure selected from: Ma6 Ma4Ga6Ino Ma2 Ma6 Ma4Ga6Ino Ma2 [Ma2]Ma6 Ma4Ga6Ino 25 Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, 30 Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric WO 00/15254 PCT/AU99/00770 - 47 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a 5 structure selected from: EtN-P-[Ma2][G]Ma2 Ma6 M EtN-P-[Mu2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M 10 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a linkages which may be substituted with P-linkages wherever required, and numeric 15 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a structure selected from: 20 Ma2 [Ma2][G]Ma2 Ma6 M Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M 25 Ma6 Ma4G or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a 30 linkages which may be substituted with P-linkages wherever required, and numeric WO 00/15254 PCT/AU99/00770 - 48 values represent positional linkages which may be substituted with any other positional linkages as required. In still another preferred embodiment, the subject inositolglycan domain comprises a 5 structure selected from: EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2] [X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M 10 or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, c represents a linkages which may be substituted with P-linkages wherever required, and numeric 15 values represent positional linkages which may be substituted with any other positional linkages as required. Still another aspect of the present invention is directed to a pharmaceutical composition comprising a molecule capable of inducing an immune response directed to a micro 20 organism GPI inositolglycan domain but substantially incapable of inducing an immune response directed to a lipidic domain of a GPI, as broadly described above, together with one or more pharmaceutically acceptable carriers and/or diluents. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions 25 (where water soluble) and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion or may be in the form of a cream or other form suitable for topical application. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of micro-organisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium 30 containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof, and WO 00/15254 PCT/AU99/00770 - 49 vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of superfactants. The preventions of the action of micro organisms can be brought about by various antibacterial and antifungal agents, for 5 example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. 10 Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilisation. Generally, dispersions are prepared by incorporating the various sterilised active ingredient into a 15 sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from previously sterile-filtered solution 20 thereof. When the active ingredients are suitably protected they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets, or it 25 may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 1% by weight of active compound. The percentage of the compositions and 30 preparations may, of course, be varied and may conveniently be between about 5 to about 80% of the weight of the unit. The amount of active compound in such WO 00/15254 PCT/AU99/00770 - 50 therapeutically useful compositions is such that a suitable dosage will be obtained. Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 0.1pg and 2000 mg of active compound. 5 The tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: a binder such as gum, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such 10 as sucrose, lactose or saccharin may be added or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar 15 or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound(s) may be incorporated into sustained-release 20 preparations and formulations. Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical 25 active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, use thereof in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. 30 It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein WO 00/15254 PCT/AU99/00770 -51 refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the novel dosage unit forms of the 5 invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of disease in living subjects having a diseased condition in which bodily health is impaired as herein disclosed in detail. 10 The principal active ingredient is compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in dosage unit form as hereinbefore disclosed. A unit dosage form can, for example, contain the principal active compound in amounts ranging from 0.5 pg to about 2000 15 mg. Expressed in proportions, the active compound is generally present in from about 0.5 Ig to about 2000 mg/ml of carrier. In the case of compositions containing supplementary active ingredients, the dosages are determined by reference to the usual dose and manner of administration of the said ingredients. 20 The pharmaceutical composition may also comprise genetic molecules such as a vector capable of transfecting target cells where the vector carries a nucleic acid molecule capable of expressing, for example, a functional equivalent to a GPI inositolglycan domain or derivative thereof. The vector may, for example, be a viral vector and it may be administered by any suitable method including, for example transfection 25 directly into the cells of the mammal being treated or transfection into a host cell, such as a bacterium, yeast or attenuated parasite, which is then introduced into the mammal. Administration of the immunogenic GPI inositolglycan domain of the present invention induces antibody production and in particular IgG production. Said antibodies are 30 involved in inhibiting, halting or delaying the onset or progression of symptoms associated with micro-organism infection such as, for example, pathalogical responses WO 00/15254 PCT/AU99/00770 - 52 to a parasitic infection. Said antibodies function, for example, by neutralising parasite induced TNF induction or by direct antiperasitic effect such as killing the parasite by binding to its surface and inhibiting its growth or development or otherwise inhibiting its onward progression. Antibodies directed to the GPI inositolglycan domain or 5 derivatives thereof may therefore also be utilised in treating parasitic infections therapeutically or prophylactically. Accordingly, another aspect of the present invention is directed to antibodies to GPI inositolglycan domains but substantially incapable of interacting with the lipidic domain 10 of a GPI. Such antibodies may be monoclonal or polyclonal, may be of any isotyope and may be selected from naturally occuring antibodies to endogenous or exogenous GPI inositolglycan domains or may be specifically raised to GPI inositolglycan domains. 15 Antibodies may also have been raised against antigens other than the GPI inositolglycan domain but are cross-reactive with one or more epitopes of the GPI inositolglycan domain. In the case of antibodies raised to the GPI inositolglycan domain, a GPI inositolglycan may first need to be associated with a carrier molecule as hereinbefore described. 20 The antibodies and/or GPI inositolglycan domains of the present invention are particularly useful as therapeutic or diagnostic agents. For example, a GPI inositolglycan domain can be used to screen for naturally occuring antibodies to GPI inositolglycan domain. These may occur, for example in some infectious and 25 autoimmune diseases. Alternatively, specific antibodies can be used to screen for GPI inositolglycan domains. Techniques for such assays are well known in the art and include, for example, sandwich assays, ELISA, Western blot and flow cytometry. Knowledge of GPI inositolglycan domain levels may be important for diagnosis of certain diseases, such as parasitic infections, autoimmune diseases (e.g. Type 1 30 diabetes), degenerative diseases (e.g. Type 2 diabetes) and somatically acquired genetic defects (e.g. Paroxysmal Nocturnal Haemoglobinurea) or for monitoring certain WO 00/15254 PCT/AU99/00770 -53 therapeutic protocols. Said antibodies would be useful as research tools or reagents for the detection of GPI inositolglycan domains. Said antibodies would also be important for example as a means for screening for levels of GPI inositolglycan domains in cell extract or other biological fluid or purifying a GPI made by recombinant means from 5 culture supernatant fluids. Techniques for the assays contemplated herein and known in the art and include, for example, sandwich assays and ELISA, Western blot and affinity chromatography. Antibodies to GPI inositolglycan domain of the present invention may be monoclonal or 10 polyclonal. Alternatively, fragments of antibodies may be used such as Fab fragments. Furthermore, the present invention extends to recombinant and synthetic antibody, to antibody hybrid and to humanised antibody. A "synthetic antibody" is considered herein to include fragments and hybrids of antibodies. The antibodies of this aspect of the present invention are particularly useful for immunotherapy and immunoprophylaxis 15 and may also be used as a diagnostic tool for assessing, for example, parasitic infection or for monitoring the program of therapeutic regimen. It is within the scope of this invention to include any second antibodies (monoclonal, polyclonal or fragments of antibodies or synthetic antibodies) directed to the first 20 mentioned antibodies discussed above. Both the first and second antibodies may be used in detection assays or a first antibody may be used with a commercially available anti-immunoglobulin antibody. An antibody as contemplated herein includes any antibody specific to any region of the GPI inositolglycan domain. 25 Both polyclonal and monoclonal antibodies are obtainable by immunization with the GPI inositolglycan domain and are utilizable for immunoassays. The methods of obtaining both types of sera are well known in the art. Polyclonal sera are less preferred but are relatively easily prepared by injection of a suitable laboratory animal with an effective amount of a GPI inositolglycan domain, or antigenic parts thereof, 30 collecting serum from the animal, and isolating specific sera by any of the known immunoadsorbent techniques. Although antibodies produced by this method are WO 00/15254 PCT/AU99/00770 - 54 utilizable in virtually any type of immunoassay, they are generally less favoured because of the potential heterogeneity of the product. The use of monoclonal antibodies in an immunoassay is particularly preferred because 5 of the ability to produce them in large quantities and the homogeneity of the product. The preparation of hybridoma cell lines for monoclonal antibody production derived by fusing an immortal cell line and lymphocytes sensitized against the immunogenic preparation can be done by techniques which are well known to those who are skilled in the art. 10 Yet another aspect of the present invention relates to a pharmaceutical composition comprising an antibody directed to a GPI inositolglycan domain together with one or more pharmaceutically acceptable carriers or diluents as hereinbefore described. 15 A further aspect of the present invention relates to the use of the antibodies of the present invention in relation to disease conditions. For example, the present invention is particularly useful but in no way limited to use in treating parasitic infections, their symptoms and pathologies. 20 Accordingly, another aspect of the present invention relates to a method of inhibiting, halting or delaying the onset of progression of a mammalian disease condition characterised by a micro-organism infection said method comprising administering to said mammal an effective amount of an antibody has hereinbefore described. 25 Preferably said disease condition is a parasite infection and most preferably malaria. In yet another aspect the present invention relates to the use of an antibody in the manufacture of a medicament for inhibiting, halting or delaying the onset or progression of a disease condition characterised by the infection of a mammal by a 30 micro-organism.
WO 00/15254 PCT/AU99/00770 - 55 Preferably said disease condition is a parasite infection and most preferably malaria. The present invention is further described by the following non-limiting Examples.
WO 00/15254 PCT/AU99/00770' - 56 EXAMPLE 1 REAGENTS, ANIMALS AND PREPARATION OF PARASITES Pronase was obtained from Boehringer Mannheim. Phosphatidylinositol-specific 5 phospholipase C was from Calbiochem. Octyl-Sepharose, Protein-G Sepharose, n-octylthioglucopyranoside (n-otg), phenylmethylsulfonylfluoride (PMSF), p-tosyl-L-lysine-chloromethylketone (TLCK), N-tosyl-L-phenylalaninechloromethylketone (TPCK), p-chloro-mercuriphenylsulphonic acid (p-CMPS), aprotinin, leupeptin, pepstatin, iodoacetamide, n-ethyl-maleimide 10 (NEM), and Concanavalin-A were obtained from Sigma Chemical Co. Sephadex was from Pharmacia. Biogel P4 was from Biorad. Analytical or HPLC grade, acetic acid, butanol, chloroform, diethyl ether, ethanol, methanol and water were obtained from BDH and Waters. Silica G60 TLC plates were from Merck Darmstadt. Tritiated mannose, glucosamine, myristic and palmitic acids were from Amersham. 15 Adult female C57BL/6 and C3H/HeJ mice were bred and maintained in the Walter and Eliza Hall Institute specific pathogen free animal facility. The FCB-1 line of Plasmodium falciparum were grown in vitro by standard methods, 20 and confirmed free of Mycoplasma contamination. For the biosynthetic labelling of parasite proteins, 3H-palmitic acid conjugated to defatted bovine serum albumin in molar ratio 1:1, 3H-glucosamine or 3H-mannose were added at a final specific activity of 10pCurie/ml, to RPMI 1640 cultures of 2x101 0 parasites at the late trophozoite/early schizont stage for 2 hours (for labelling of GPI precursors) or 8 hours (for labelling of 25 protein-bound GPI). Parasites were harvested by 0.05% Saponin lysis and centrifugation in the cold at 15,000g for 20 minutes, followed by two washes in PBS and storage at -70 0
C.
WO 00/15254 PCT/AU99/00770 - 57 EXAMPLE 2 PURIFICATION OF THE 195KD MSP-1 AND 56KD MSP-2 ANTIGENS The GPI-anchored MSP-1 and MSP-2 merozoite surface proteins were purified to 5 homogeneity as described previously (Schofield and Hackett, 1993). Biosynthetically labelled malaria parasites at the late schizont stage were lysed in 0.05% Saponin and centrifuged at 15,000g for 20 minutes, and washed as above. The pellet was extracted in 25mM n-octyl-thioglucopyranoside (n-otg), 1% BSA, 1mM EDTA, 0. 1mM EGTA, 1mM PMSF, 1mM TPCK, 0. lmM TLCK, 5mM pCMPS, 1pg/ml pepstatin, lpg/ml 10 leupeptin, 1mM NEM, 5mM iodoacetamide, 150mM NaC1, 25mM Tris/HCl pH 7.4 by sonication on ice. The extact was clarified by centrifugation at 20,000g for 30 minutes in the cold, and the supernatant decanted and loaded onto two immunoaffinity columns arranged in sequence, containing approximately 10mg monoclonal antibody 111.4 or monoclonal antibody 113.1, each cross-linked to Protein G-Sepharose by 15 gluteraldehyde (all procedures on ice). The protein extract was passed through the column at a rate of 0.3ml/min. The columns were washed first with with 100ml 10mM n-otg, 1% BSA, 300mM NaC1, followed by 100ml 10mM n-otg, 300mM NaC1. Antigen was eluted from each column with four column volumes of 10mM n-otg, 200mM glycine pH 2.8. The pH of the eluate was neutralized with 2M Tris. Aliquots 20 of protein were analysed for purity by SDS-PAGE followed by staining with Coomassie brilliant blue. The remaining purified proteins were dialysed exhaustively against 100mM NH 4
HCO
3 using dialysis membrane previously boiled exhaustively in 10mM EDTA followed by boiling in 10 changes of double distilled water. Protein concentration was determined by standard methods. 25 The remaining detergent soluble extract was made up to 1mM CaCl 2 , 1mM MgCl 2 , and 1mM MnCl 2 , and passed over a Con-A sepharose column, followed by washing with 10 column volumes of extraction buffer. The column was first eluted with detergent buffer containing 0.5M a-methyl-mannopyranoside and 0.5M a-glucopyranoside, followed by 30 25mM n-otg in 8M urea. Aliquots were subject to SDS-PAGE and fluorography or staining with Coomassie blue.
WO 00/15254 PCT/AU99/00770~ - 58 EXAMPLE 3 PURIFICATION OF THE C-TERMINAL GPI ANCHORS OF DEFINED PARASITE ANTIGENS 5 To purify the intact C-terminal GPIs free of detergent, non-covalently bound lipids, glycolipids, phospholipids and protein or peptide fragments, affinity purified MSP-1 and MSP-2 were first scrubbed with organic solvents. 10mg/ml GPI-anchored proteins were placed in 150iA aliquots in clean glass tubes. 600A MeOH was added and vortexed, followed by 150A1 1 Chloroform and 4 50tl water and further vortexing. The 10 samples were centrifuged at 14000rpm for 3min, the supernatant discarded, and the interphase and lower phase mixed with 450[d MeOH and re- centrifuged at 14000rpm for 3min. The protein pellet was extracted 5 times with C/M/W 10:10:3, and finally extracted with acetone over night at -20oC. The acetone was removed completely and the proteins taken up with sonication in 6M Urea, 1mM DTT, 1mM iodoacetic acid. 15 After 15 minutes at room temperature, the sample was diluted 6 fold and made to 5mM CaCl 2 . 2.5% pre-digested Pronase B was added for 72h at 37oC with 2 additions of 0.3 % pronase. The digested sample was phase separated between water and water-saturated butanol, and the organic phase back extracted with water. The butanol phase was spotted onto TLC plates (Si-60) and run in the solvent system C/M/HAc/W 20 25:15:4:2. The pronase-digested GPI fragment free of contaminants remains close to the origin, and was detected by Berthold Digital Autoradiograph. The appropriate region was scraped and the material eluted twice with C/M/W 10:10:3 followed by 40% 1-propanol in water. The material was dried under nitrogen gas, and once more separated between water and water-saturated butanol. 25 WO 00/15254 PCT/AU99/00770 - 59 EXAMPLE 4 PURIFICATION OF GIPLs AND GPI BIOSYNTHETIC PRECURSORS BY TLC GPI biosynthetic intermediates and non-protein bound mature GPI species were purified 5 by TLC. 2x10 1 0 P. falciparum schizonts were labelled with lmCi 3 [H]-mannose or 3 [H]-palmitic acid in 250ml glucose deficient RPMI 1640 supplemented with 40mM fructose and 0.5% Albumax (GIBCO) for 2 hours. Parasites were harvested by saponin lysis and washed twice in PBS. They were extracted three times in chloroform/ methanol (2:1) and three times in chloroform/methanol/water (1:1:0.3). The 10 chloroform/methanol extracts were subject to repeated Folch washing and the chloroform phase dried in a Speedvac. The chloroform/methanol/water extracts were dried down and partitioned between water and water-saturated butanol. The butanol phase was washed with water and dried in a Speedvac. Both residues were then separated by TLC and the plates scanned by Bertold Digital Autoradiograph TLC 15 scanner. The radiolabelled peaks were identified and removed by scraping and re-extraction followed by drying. Areas lying between and outside the identifiable peaks were treated in the same way, as were sham plates. In some experiments, the GPIs were further purified over Octyl-Sepharose. Samples were taken up in 5% 1-propanol in 100mM Ammonium acetate and loaded at a flow rate of 0. lml/min onto 20 an Octyl-Sepharose column, and the column washed with 100mM Ammonium acetate, 5% 1-propanol. The column was eluted in a gradient running from 100mM Ammonium acetate, 5 % 1-propanol to 60% 1-propanol in water. GPI containing fractions were lyophilised and flash evaporated in methanol. 25 EXAMPLE 5 GENERATION OF CHEMICAL AND ENZYMATIC HYDROLYSIS FRAGMENTS OF GPIs Purified, glucosamine-labelled Pfalciparum GPIs, in which all dpms were detected in 30 the organic phase following butanol/water partitioning, were subject to base hydrolysis by suspension in methanol/ammonia 1:1 for 6 hours at 50oC, followed by partioning WO 00/15254 PCT/AU99/00770 - 60 between water and water saturated butanol. Essentially 100% of label was then recovered from the aqueous phase. The aqueous phase was twice extracted with water-saturated butanol, lyophilized, and flash evaporated with methanol. 5 EXAMPLE 6 DEAE ANION EXCHANGE CHROMATOGRAPHY GPIs were loaded onto a A DEAE column in 99% methanol, 1% water and washed with ten column volumes of solvent. They were subsequently eluted in 100mM 10 Ammonium Acetate in 99% methanol, 1% water and dried under Nitrogen. EXAMPLE 7 BIOGEL P4 SIZE-EXCLUSION CHROMATOGRAPHY 15 Base-hydrolysed GPI glycans were spiked with phenol red and blue dextran in 100mM Ammonium Acetate and further size-fractionated by passage through a 1cm x 1.2 metre Biogel P4 column equilibrated in 100mM Ammonium acetate in water. The column had previously been exhaustively calibrated by repeated analytical runs with GPI mixed with acid hydrolysed dextran markers to yield the relative elution position of glucose 20 units detected by staining with orcinol in concentrated sulfuric acid. The column runs proved to be highly reproducible. For preparative purposes the dextran markers were omitted. The GPI peak was detected by scintillation counting of aliquots. EXAMPLE 8 25 COMPOSITIONAL ANALYSIS BY GC/MS Glycan concentration and compositional purity was determined by GC-MS, following acid methanolysis and trimethylsilyl (TMS) derivatization. myo-Inositol content was measured following acid hydrolysis (6N HC1, 1 10oC, 16 h) and TMS derivatization, 30 with selected ion monitoring for m/z 305 and 318. scyllo-Inositol was used as internal standard throughout.
WO 00/15254 PCT/AU99/00770' -61 EXAMPLE 9 COUPLING OF GPI GLYCAN TO MALEIMIDE-ACTIVATED KLH The GPI glycan was exposed to 1mM Traut's reagent (2-iminothiolane) in 60mM 5 triethanolamine, 7mM potassium phosphate, 100mM NaC1, 1mM EDTA, pH 8.0 in the cold for 90 minutes under nitrogen. The sample was then desalted by gel filtration at 4oC through a small Biogel P4 column equilibrated in 7mM potassium phosphate, 100mM NaC1, 1mM EDTA, pH 7.2 and added to maleimide-activated KeyHole Limpet Haemocyanin (KLH) or Ovalbumin (OVA) in coupling buffer (7mM potassium 10 phosphate, 100mM NaC1, 1mM EDTA, pH 7.2) overnight. The degree of conjugation was estimated by comparison of cpms before and after dialysis of the sample against PBS, or by use of Ellman's reagent for the quantitation of sulfhydryl groups. Excess reactive sites were blocked with cysteine. 15 EXAMPLE 10 EPITOPE MAPPING OF ANTI-GPI ANTIBODIES Coupling of the purified GPI glycan to proteins was undertaken as above. To measure anti-lipid reactivities, we utilized commercially available phosphatidylinositol from 20 Sigma with identical composition to the malarial GPI, namely dipalmitoyl-PI. 2mg PI was coupled to defatted BSA according to published protocols (Bate et al, 1993). EXAMPLE 11 ELISA ASSAY 25 Antigen (GPI-OVA, Glycan-OVA, BSA-PI, OVA or BSA alone) at 20/g/ml in phosphate binding buffer was incubated overnight in 50pl volumes in flat-bottomed Immunlon 96-well plates, followed by extensive washing with buffer. The plates were blocked with 1% BSA, 1% OVA in PBS for several hours. From a 1/32 dilution, sera 30 were titrated two-fold in 1% BSA, 1% OVA in PBS, and 501l aliquots incubated in triplicate for 2 hours at room temperature, followed by extensive washing with 1% WO 00/15254 PCT/AU99/00770 - 62 BSA, 1% OVA 0.05% Tween-20 in PBS. An aliquot of affinity purified, biotin-labelled isotype specific goat anti-mouse second antibody was incubated as above, followed by further washing and the addition of streptavidin-alkaline phosphatase. After 30 minutes the plates were washed again and colourimetric 5 development initiated by the addition of p-Nitrophenylphosphate in diethanolamine buffer. Background binding to BSA/OVA-coated plates was determined in parallel. The end-titres derived are the last point giving values statistically different by two-way analysis of variance from non-specific binding by the same serum to the BSA/OVA-coated plates. 10 EXAMPLE 12 COMPETITION ELISA From a 1/32 dilution, sera or mAbs were titrated two-fold in 1% BSA in PBS, 0.05% 15 Tween-20, and pre-incubated for 4 hours at room temperature with a molar excess of competitior (20pg/ml PI, or phosphatidylserine (PS), or diluent alone). Antigen (BSA-PI or BSA alone) at 20[tg/ml in phosphate binding buffer was incubated overnight in 501l volumes in flat-bottomed Immunlon 96-well plates, followed by extensive washing with buffer. The plates were blocked with 1% BSA in PBS for several hours. 20 50tl aliquots of titrated antibody with or without competitor were incubated in triplicate for 2 hours at room temperature, followed by extensive washing with 1% BSA 0.05% Tween-20 in PBS. An aliquot of affinity purified, biotin-labelled isotype specific goat anti-mouse second antibody was incubated as above, followed by further washing and the addition of streptavidin-alkaline phosphatase. After 30 minutes the plates were 25 washed again and colourimetric development initiated by the addition of p-Nitrophenylphosphate in diethanolamine buffer. Background binding to BSA-coated plates was determined in parallel. The end-titres derived are the last point giving values statistically different by two-way analysis of variance from non-specific binding by the same serum to the BSA-coated plates.
WO 00/15254 PCT/AU99/00770 - 63 EXAMPLE 13 PRODUCTION OF MONOCLONAL ANTIBODIES Monoclonal antibodies to the lipid domain of the GPI were produced as previously 5 described (Tachado et al, 1996). Monoclonal antibodies to the glycan were generated by immunization of OVA-TCR transgenic mice on a Balb/c background with OVA-glycan, followed by fusion and screening of hybridoma culture supernatants against BSA vs. BSA-glycan. 10 EXAMPLE 14 MACROPHAGE CULTURE AND TNF OUTPUT LPS-nonresponsive C3H/HeJ macrophages were obtained as previously described (Schofield and Hackett, 1993 and Tachado et al, 1996). 2x10 5 adherent cells/well were 15 given medium alone or test agents. 3 hrs after incubation TNF-a levels in the supernatant and standard curve were determined by capture ELISA (Pharmingen). Tyrosine phosphorylation. Rapid onset tyrosylphosphorylation was determined as previously described (Tachado et 20 al, 1997). EXAMPLE 15 PI-PLC TREATMENT AND FACS ANALYSIS 25 2x10 5 cells were exposed to 1U/ml PI-PLC at 37oC for 2 hours, followed by washing. They were then incubated in ice cold murine tonicity RPMI 1640 with 0.05% Sodium azide and 1% BSA with monoclonal antibodies or murine sera followed by washing and a further incubation with isotype-specific FITC-conjugated antibody to mouse immunoglobulins. After washing in the same medium the cells were counter-stained 30 with 0.5Lg/ml propidium iodide and analysed by FACSscan.
WO 00/15254 PCT/AU99/00770' - 64 EXAMPLE 16 IMMUNIZATION OF MICE WITH FREE GPI Mice were immunized by three successive boosts of free intact malarial GPI emulsified 5 in Incomplete Freund's Adjuvant spaced two weeks apart. Control mice received an equal amount of IFA alone. After immunization, sera were bled and the titres of anti-GPI antibodies determined by ELISA. All animals immunized with GPI developed broadly similar levels of anti-GPI antibodies (range 1/1024 - 1/4096) among individual animals. The anti-GPI response was predominantly IgM, and epitope mapping studies 10 by competition ELISA revealed that the antibody response was directed predominantly towards the lipidic (phosphatidylinositol, PI) domain of the molecule, with some cross-reactivity to other phospholipid determinants (Fig. 1). Two weeks after the final boost mice were challenged with P. berghei ANKA. Parasitaemia, the development of neurological complications, and mortality were recorded daily. No difference in 15 parasitaemia was observed. In the control group, 100% of animal manifested between day 5 and 9 an aggressive cerebral syndrome with neurological signs proceeding to rapid death with 12 hours. In animals immunized with intact free GPI, however, deaths occurred at a noticable faster rate (Fig.2). 20 The increased death rate in animals immunized with free GPI and subsequently challenged with malaria may result from unanticipated autoreactivity of anti-GPI antibodies. A panel of IgM monoclonal antibodies was derived from mice immunized with free GPIs. mAbs selected at random from this panel were shown by PI-specific ELISA to be reactive with PI domain of the molecule (Fig. 3), as was expected given 25 the established serological specificity of the polyclonal sera of the donor immunized mice (Fig. 1). In addition, these mAbs and the polyclonal antisera of GPI-immunized mice were shown by FACS analysis to react with host GPI molecules expressed at the cell surface. Although suprising, the recognition of GPI-associated lipidic determinants at the cells surface is not without precedence (Xia et al, 1993). Pre-treatment of host 30 cells with phosphatidylinositol-specifc phospholipase C resulted in loss of binding of these mAbs, demonstrating formally that a lipidic moiety of GPI molecules is exposed WO 00/15254 PCT/AU99/007701 - 65 at the cell surface and is accessible for binding by autoreactive antibodies generated in response to exposure to free malarial GPI (Fig. 4). The binding was also shown to cause massive rapid onset intracellular tyrosine phosphorylation (Fig. 5), a well-known and predictable consequence of cross-linking host GPIs at the cell surface (Shenoy 5 Scaria et al, 1992 and Stefanova et al, 1993). Following binding of these antibodies to macrophages, the cells responded more vigorously to stimulation with GPI, phorbol esters or malaria parasite extracts (Fig. 6). Upon passive transfer into mice, these mAbs were sufficient to cause an increased rate of death as compared with control IgM mAbs (Fig. 7). 10 Thus to summarize: (i) immunization of mice with the free P. falciparum GPI generates IgM reacting predominantly with the PI domain of the GPI; (ii) this immunization appears to exacerbate the P. berghei cerebral malaria syndrome; (iii) exarcerbated pathogenicity as detected by increased death rate was also observed upon passive 15 transfer of IgM monoclonals with the same reactivity; (iv) the mAbs were shown to cross-react with host GPIs by FACS analysis, thereby causing massive intracellular tyrosylphosphorylation and sensitization of macrophages resulting in increased TNF output in response to addition agonists. Therefore it is proposed that a novel mechanism exists by which the acquisition of certain auto-reactive immunological 20 specificities results in increased physiological sensitization to malarial toxins. EXAMPLE 17 IMMUNIZATION OF MICE WITH THE GPI GLYCAN CONJUGATED TO KLH 25 Previous publications dealing with the prospect of anti-disease vaccines against malaria have proposed immunizing against a phospholipid domain within the putative toxin (Bate et al, 1992c, Bate et al, 1992a, Bate et al, 1992b, Bate et al, 1993, Jakobsen et al, 1993b, Bate and Kwiatkowski, 1994 and Playfair, 1994). The present data indicate 30 strongly that this may be deleterious and should be avoided. It was sought to develop a novel approach, namely to detoxify and deacylate the GPI and to determine whether WO 00/15254 PCT/AU99/00770 - 66 immunization against the glycan domain of the molecule would exarcerbate disease or be sufficient to protect mice against malarial pathology. Mice (n= 7) were immunized by three successive boosts of 501kg KLH-glycan emulsified in Incomplete Freund's Adjuvant spaced two weeks apart. Two separate control groups (n=8 each) comprised 5 animals receiving an equal amount of sham conjugated KLH in IFA, or those left untreated. After immunization, sera were bled and the titres of anti-GPI antibodies determined by ELISA. All animals immunized with KLH-glycan developed detectable anti-GPI glycan IgG antibodies, although there were differences in end-titre (range 1/128 - 1/4096) among individual animals. The sera from vaccine recipients (but not 10 sham-KLH controls) were able to inhibit TNF output from macrophages stimulated with crude P. falciparum extracts, providing convincing proof-of-principle for the neutralization of pathogenicity (Fig. 8). In contrast to the host-reactive antibodies to the GPI lipid domain, pre-exposure of macrophages to these sera did not result in increased TNF output in response to additional agonists. With these sera it was not 15 possible to detect significant cross-reactivity with host GPIs at the cell surace as judged by FACS analysis of antibody binding to host cells. The P. berghei ANKA murine cerebral malaria model has many features in common with the human cerebral malaria syndrome. It is a TNF-a and interferon-y(IFN-y) 20 dependent encephalitis associated with upregulation of ICAM-1 on the cerebral microvascular endothelium, an increase in both parasite and macrophage/neutrophil adherence to these target cells, and attendant neurological complications. Unlike human cerebral malaria, there is a breakdown of the blood-brain barrier in the terminal stages of the murine syndrome. However, in the proximal stages the murine disease reflects more 25 accurately the inflammatory cascade leading to cerebral involvement in humans. To determine whether anti-GPI immunization prevents cerebral pathogenesis in vivo, mice were immunized with P. falciparum IPG conjugated to KLH. Two weeks after the final boost mice were challenged with P. berghei ANKA. Parasitaemia, the development of neurological complications, and mortality were recorded daily. No difference in 30 parasitaemia was observed among groups. In both control groups, 87.5% of animal manifested between day 7 and 12 an aggressive cerebral syndrome with neurological WO 00/15254 PCT/AU99/00770 - 67 signs proceeding to rapid death with 12 hours, and 12.5% did not develop the syndrome. As there were no significant differences between sham-immunized and untreated groups, the data from these two control groups are pooled (Fig.9). In recipents of KLH-glycan, one animal (14.2%) died with similar kinetics, two animals 5 (28.5%) developed the cerebral syndrome with sustantially delayed kinetics (on days 10 and 11, and showing prolonged course of syndrome before succumbing), and four animals (57.2%) were completely protected, failing to develop the cerebral syndrome at any stage (Fig.9). Thus immunization of mice with the P. falciparum GPI glycan covalently linked to a carrier protein affords substantial protection against the P. 10 berghei cerebral malaria syndrome. A panel of monoclonal antibodies was made from mice immunized with purifed GPI glycan conjugated to OVA (OVA-glycan). The hybridoma fusion products were initially screened for binding to BSA-glycan as compared to BSA alone. Over 80 15 glycan-reactive IgG monoclonal antibodies were detected. Of these, many were reactive with parasites but not host erythrocytes as judged by the Indirect Fluorescent Antibody Test. Purified monoclonal antibodies 1G7 and 3G6 were sufficient to block the induction of TNF by 100% when added at low concentration to total crude parasite extracts (Fig. 10). To determine whether anti-GPI antibodies alone are sufficient to 20 prevent severe malarial pathology, mice were infected with 106 P. berghei ANKA i.p. On day 4 they were divided at random into 10 controls receiving an irrelevant IgG and groups of 5 receiving mAbs 1D12, 2C4, 3G5 and 4C3 raised against the P. falciparum GPI inositolphosphoglycan. All mice received 100pg antibody/day i.p. for 7 days. Mice were monitored for parasitaemia daily and clinical signs every 6 hours. 100% of 25 controls died of the cerebral malaria syndrome between days 6 and 8 post-infection. Throughout this period, no animals receiving either of the 4 anti-GPI monoclonal antibodies showed signs of illness, despite being equally parasitized as controls. On day 10 one of the 5 animals receiving monoclonal 3G5 died. Other than this individual, no others showed cerebral signs and none died (Fig. 11). Thus 19/20 (95%) of the 20 30 animals receiving anti-GPI mAbs survived, vs. zero survival in controls (n= 30 total). Parasitaemias were identical in test and control groups throughout the experiment. For WO 00/15254 PCT/AU99/00770 ~ - 68 visual clarity, the figure shows the 4 treatment groups in aggregate. In addition, 5 mice received antibodies alone without parasite challenge. There were no detectable acute or toxic reactions in these mice receiving antibodies alone. 5 In addition a standard murine model of TNF-driven lethality was used to determine whether GPI mediates parasite-induced acute toxic shock. This model manifests disseminated intravascular coagulation, peripheral vascular failure and shock, and thus has clinical features in common with the human "algid malaria" syndrome. LPS-non responsive C3H/HeJ and C57B16 mice were primed with 20 mg D-galactosamine 10 followed after 1 hour by purified GPI, PE or PBS alone. Mice receiving D galactosamine followed by vehicle alone showed 100% survival. Both PE and purified GPI induced lethal shock in 100% of D-galactosamine-primed C3H/HeJ and C57B16 recipients. mAbs to the GPI glycan substantially prevented TNF-driven lethality in vivo (Fig. 12). 15 Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred 20 to or indicated in this specification, individually or collectively, and any and all combinations of any two or more said steps or features.
WO 00/15254 PCT/AU99/00770 - 69 BIBLIOGRAPHY: Bate, C. A. W., and Kwiatkowski, D. (1994) Infection and Immunity 62:5261-5266. Bate, C. A. W., Taverne, J., and Playfair, J. H. L. (1992c) Infection and Immunity 60:1894-1901. Bate, C.A., Taverne, J., and Playfair, J.H. (1989) Immunology 66:600. Bate, C.A., Taverne, J., and Playfair, J.H. (1988) Immunology 64:227. Bate, C. A., Taverne, J., Romin, E., Moreno, C., and Playfair, J. H. L. (1992a) Immunology 75:129-135. Bate, C. A. W., Taverne, J., Kwiatkowski, D., and Playfair, J. H. L. (1993) Immunology 79:138-145. Bate, C. A. W., Taverne, J., Bootsma, H. J., Mason, R. C. S. H., Skalko, N., Gregoriadis, G., and Playfair, J. H. L. (1992b) Immunology 76:35-41. Berendt, A.R., Turner, G.D.H. and Newbold, C.I. (1994) Parasitol Today 10:412. Clark, I.A. (1978) Lancet 2:75. Clark, I.A., Virelizier, J.-L., Carswell, E.A., and Wood, P.R. (1981) Infect. Immun. 32:1058. Gerold, P., Schofield, L., Brackman M., Holder, A.A., Schwarz, R.T. (1996) Mol. Biochem. Parasitol 75:131.
WO 00/15254 PCT/AU99/00770 - 70 Gerold, P., Dieckman-Schuppert A. and Schwarz, R.T. (1992) Bio. Soc. Trans. 29:297. Golgi, C. (1886) Arch. Sci. med. (Torino) 10:109. Jakobsen, P. H., Morris-Jones, S. D., Hviid, L., Theander, T. G., Hoier-Madsen, M., Bayoumi, R., and Greenwood, B. M. (1993b) Immunology 79:653-657. McConville, M.J. and Ferguson M.A. (1993), Biochem. J. 294:305. Playfair, J. H. L. (1994) Immunology Letters 43:83-86. Schofield, L., Novakovic, S., Gerold, P., Schwarz, R.T., McConville, M.J. and Tachado, S.D. (1996) J. Immunol. 156:1886-1896. Schofield, L., and Hackett, F. (1993) Journal of Experimental Medicine 177:145-153. Shenoy-Scaria, A. M., Kwong, J., Fujita, T., Olszowy, M. W., Shaw, A. S., and Lublin, D. M. (1992) Journal of Immunology 149:3535-3541. Stefanova, I., Corcoran, M. L., Horak, E. M., Wahl, L. M., Bolen, J. B., and Horak, I. D. (1993) Journal of Biological Chemistry 268:20725-20728. Stevens, V.L. (1995) Biochem. J. 310:361. Tachado, S. D., Gerold, P., McConville, M. J., Baldwin, T., Quilici, D., Schwarz, R. T., and Schofield, L. (1996) Journal of Immunology 156:1897-1907. Tachado, S. D., Gerold, P., Schwarz, R., Novakovic, S., McConville, M., and Schofield, L. (1997) Proceedings of the National Academy of Sciences USA 94:4022-4027.
WO 00/15254 PCT/AU99/00770 -71 Xia, M.-Q., Hale, G., Lifely, M. R., Ferguson, M. A. J., Campbell, D., Packman, L., and Waldmann, H. (1993) Biochemical Journal 293:633-640.

Claims (53)

1. A method of eliciting or inducing, in a mammal, an immune response directed to a micro-organism said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositol glycan domain of a GPI but which molecule is substantially incapable of inducing an immune response directed to a lipidic domain of GPI.
2. A method according to claim 1 wherein said molecule is a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain.
3. A method according to claim 2 wherein said modified GPI molecule is the inositolglycan domain portion of GPI or derivative or equivalent thereof.
4. A method according to claim 2 or 3 wherein said modified GPI molecule is a modified parasite GPI molecule or derivative or equivalent thereof.
5. A method according to claim 4 wherein said parasite is Plasmodium.
6. A method according to claim 5 wherein said Plasmodium is Plasmodium falciparum.
7. A method according to claim 6 wherein said modified Plasmodiumfalciparum GPI molecule is a Plasmodiumfalciparum GPI inositolglycan domain. WO 00/15254 PCT/AU99/00770 - 73
8. A method according to claim 7 wherein said GPI inositol glycan domain comprises the structure ethanolamine-phosphate-(Manal 1,2)-Manal 1,2Mana 1,6Man 1,4GlcN myo-inositol phosphoglycerol or derivative or equivalent thereof.
9. A method according to claim 7 wherein said GPI inositol glycan domain comprises the structure XI - X2 - X 3 -X 4 - ethanolamine-phosphate-(Mana 1,2) Manal,2Manal,6Manal,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain.
10. A method according to claim 7 wherein said GPI inositolglycan domain comprises a structure selected from: EtN-P-[Mca2]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Mca2][G]Ma2 Mca6 Ma4Ga6Ino EtN-P-[Ma2][X]Mca2 Ma6 Ma4Gca6Ino EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4Gc6Ino EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Ma4G EtN-P-Ma2 Ma6 M EtN-P-[Mca2][G]Ma2 Ma6 Ma4G EtN-P-[Ma2][X]Ma2 Mca6 Ma4G EtN-P-[Ma2][EtN-P]Mca2 Ma6 Ma4G Mu2 [Ma2][G]Ma2 Ma6 Ma4G WO 00/15254 PCT/AU99/00770 - 74 Ma2 [Ma2][X]Ma2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Ma6 Ma4G Ma6 Ma4Ga6Ino Ma2 Ma6 Ma4Ga6Ino Ma2 [Ma2]Ma6 Ma4Ga6Ino Ma2 [Ma2][G]M(6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 M EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M Ma2 [Ma2][G]Ma2 Ma6 M Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a-linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required.
11. A method of therapeutically or prophylactically treating a mammal for a micro organism infection said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI, but substantially incapable of inducing an WO 00/15254 PCT/AU99/00770' - 75 immune response directed to the lipid domain of a GPI, for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with infection of said mammal by said micro-organism.
12. A method according to claim 11 wherein said molecule is a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipid domain to induce or elicit an immune response directed to a GPI lipidic domain.
13. A method according to claim 12 wherein said micro-organism infection is a parasite infection.
14. A method according to claim 13 wherein said parasite is Plasmodium.
15. A method according to claim 14 wherein said Plasmodium is Plasmodium falciparum.
16. A method according to any one of claims 13-15 wherein said modified GPI molecule is a modified parasite GPI molecule or derivative or equivalent thereof.
17. A method according to claim 16 wherein said parasite is Plasmodium.
18. A method according to claim 17 wherein a said Plasmodium is Plasmodium falciparum.
19. A method according to claim 18 wherein said modified Plasmodiumfalciparum GPI molecule is a Plasmodiumfalciparum GPI inositolglycan domain. WO 00/15254 PCT/AU99/00770 ~ - 76
20. A method according to claim 19 wherein said GPI inositolglycan domain comprises the structure ethanolamine-phosphate-(Manal 1,2)-Manal 1,2Man 1,6Man 1,4GlcN myo-inositol phosphoglycerol or derivative or equivalent thereof.
21. A method according to claim 19 wherein said GPI inositolglycan domain comprises the structure XI - X 2 - X 3 -X 4 - ethanolamine-phosphate-(Manal,2) Mancal 1,2Mancal ,6Manal 1,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain.
22. A method according to claim 19 wherein said GPI inositolglycan domain comprises a structure selected from: EtN-P-[Ma2]Ma2 Ma6 Mca4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 Ma4Gcu6Ino EtN-P-[Ma2][X]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][EtN-P]Ma2 Ma6 Mca4Ga6Ino EtN-P-Ma2 Mca6 M4G Ma2 Ma6 Mca4G EtN-P-Ma2 Mca6 M EtN-P-[Ma2][G]Ma2 Mca6 Ma4G EtN-P-[Ma2][X]M2 Ma6 Ma4G EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4G Ma2 [Mca2][G]Ma2 Ma6 Ma4G WO 00/15254 PCT/AU99/00770 - 77 Ma2 [Ma2][X]Ma2 Ma6 Ma4G Mca2 [Ma2][EtN-P]Ma6 Ma4G Ma6 Ma4Gac6Ino Mca2 Ma6 Ma4Gca6Ino Ma2 [Ma2]M(6 Ma4Ga6Ino Ma2 [Ma2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 M EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M Ma2 [Ma2][G]Mo2 Ma6 M Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a-linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required.
23. A method for the treatment and/or prophylaxis of a mammalian disease condition characterised by a micro-organism infection, said method comprising administering to said mammal an effective amount of an immunogenic composition which composition comprises a molecule capable of inducing an immune response directed to the inositolglycan domain of a GPI, but WO 00/15254 PCT/AU99/00770 - 78 substantially incapable of inducing an immune response directed to the lipid domain of a GPI, for a time and under conditions sufficient for said immune response to reduce, inhibit or otherwise alleviate any one or more symptoms associated with said micro-organism infections.
24. A method according to claim 23 wherein said molecule is a modified GPI molecule or derivative or equivalent thereof and which modified GPI molecule comprises insufficient lipid domain to induce or elicit an immune response directed to a GPI lipidic domain.
25. A method according to claim 24 wherein said modified GPI molecule is the inositolglycan domain portion of GPI or derivative or equivalent thereof.
26. A method according to claim 24 or 25 wherein said modified GPI molecule is a modified parasite GPI molecule or derivative or equivalent thereof.
27. A method according to claim 26 wherein said parasite is Plasmodium.
28. A method according to claim 27 wherein said Plasmodium is Plasmodium falciparum.
29. A method according to claim 28 wherein said modified Plasmodiumfalciparum GPI molecule is a Plasmodiumfalciparum GPI inositolglycan domain.
30. A method according to claim 29 wherein said GPI inositolglycan domain comprises the structure ethanolamine-phosphate-(Manal ,2)-Manal 1,2Mana 1l,6Mancal 1,4GlcN myo-inositol phosphoglycerol or derivative or equivalent thereof. WO 00/15254 PCT/AU99/00770 - 79
31. A method according to claim 29 wherein said GPI inositolglycan domain comprises the structure XI - X 2 - X 3 -X 4 - ethanolamine-phosphate-(Manal,2) Man al,2Manal ,6Manal ,4GlcN-rnyo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain.
32. A method according to claim 29 wherein said GPI inositolglycan domain comprises the structure: EtN-P-[Ma2]Mo2 Mcx6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 Ma4Gc6Ino EtN-P-[Ma2][X]Ma2 Mc6 Ma4Ga6Ino EtN-P-[Mx2][EtN-P]Ma2 Ma6 Ma4Ga6Ino EtN-P-Ma2 Ma6 Ma4G Ma2 Ma6 Mca4G EtN-P-Ma2 Ma6 M EtN-P-[Ma2][G]Ma2 Ma6 Ma4G EtN-P-[Ma2][X]Ma2 Ma6 Mc4G EtN-P-[Mca2][EtN-P]Ma2 Mcx6 Ma4G Ma2 [Ma2][G]Ma2 Ma6 Ma4G Ma2 [Ma2][X]Ma2 Ma6 Ma4G Ma2 [Ma2][EtN-P]Mu6 Ma4G Mc6 Ma4Ga6Ino Ma2 Ma6 Ma4Ga6Ino Ma2 [Mu2]Mca6 Ma4Gac6Ino Ma2 [Mo2][G]Ma6 Ma4Ga6Ino Ma2 [Ma2][X]Ma6 Ma4Gca6Ino EtN-P-[Ma2][G]Ma2 Ma6 M WO 00/15254 PCT/AU99/00770 - 80 EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M Ma2 [Ma2][G]Ma2 Ma6 M Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Mca2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a-linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required.
33. A method according to any one of claims 24-31 wherein said disease condition is malaria.
34. Use of a composition comprising a molecule capable of inducing an immune response directed to a micro-organism GPI inositolglycan domain but substantially incapable of inducing an immune response directed to a lipidic domain of GPI in the manufacture of a medicament for the therapeutic and/or prophylactic treatment of a mammalian disease condition characterised by infection with said micro-organism.
35. A method according to claim 34 wherein said composition comprises a Plasmodium GPI inositolglycan domain or derivative or equivalent thereof which inositolglycan domain comprises insufficient lipidic domain of a WO 00/15254 PCT/AU99/00770 -81 Plasmodium GPI to elicit or induce an immune response directed to a GPI lipidic domain.
36. A composition capable of inducing an immune response directed to a micro organism said composition comprising a molecule capable of inducing an immune response against a micro-organism GPI inositolglycan domain but substantially incapable of inducing an immune response to a lipidic domain of a GPI.
37. A composition according to claim 36 wherein said molecule comprises a modified GPI molecule or derivative or equivalent thereof which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain.
38. A vaccine composition comprising as the active component a molecule capable of inducing an immune response directed to a micro-organism GPI inositolglycan domain but substantially incapable of inducing an immune response directed to a lipidic domain of a GPI together with one or more pharmaceutically acceptable carriers and/or diluents.
39. A vaccine composition according to claim 38 wherein said molecule comprises a modified GPI molecule or derivative or equivalent thereof which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain.
40. A pharmaceutical composition comprising a molecule capable of inducing an immune response directed to a micro-organism GPI inositolglycan domain but substantially incapable of inducing an immune response directed to a lipidic domain of a GPI together with one or more pharmaceutically acceptable carriers and/or diluents. WO 00/15254 PCT/AU99/00770 - 82
41. A pharmaceutical composition according to claim 40 wherein said molecule comprises a modified GPI molecule or derivative or equivalent thereof which modified GPI molecule comprises insufficient lipidic domain to induce or elicit an immune response directed to a GPI lipidic domain.
42. A composition according to any one of claims 37, 39 or 41 wherein said modified GPI molecule is the inositolglycan domain portion of GPI or a derivative or equivalent thereof.
43. A composition according to any one or claims 37, 39, 41 or 42 wherein said modified GPI molecule is a modified parasite GPI molecule or derivative or equivalent thereof.
44. A composition according to claim 43 wherein said parasite is Plasmodium.
45. A composition according to claim 41 wherein said Plasmodium is P.falciparum.
46. A composition according to claim 42 wherein said modified Plasmodium falciparum GPI molecule is a Plasmodiumfalciparum GPI inositolglycan domain.
47. A composition according to claim 46 wherein said GPI inositolglycan domain comprises the structure ethanolamine-phosphate-(Manal,2)-Manal,2Manal,6Manal 1,4GlcN myo-inositol phosphoglycerol or derivative or equivalent thereof.
48. A composition according to claim 46 wherein said GPI inositolglycan domain comprises the structure WO 00/15254 PCT/AU99/00770 - 83 X, - X2 - X 3 -X 4 - ethanolamine-phosphate-(Manal,2) Manal 1,2Manal ,6Mana 1l,4GlcN-myo-inositol phosphoglycerol wherein X 1 , X 2 , X 3 and X 4 are any 4 amino acids, or derivative or equivalent of said GPI inositolglycan domain.
49. A composition according to claim 46 wherein said GPI inositolglycan domain comprises the structure: EtN-P-[Mo2]Ma2 Ma6 Ma4Ga6Ino EtN-P-[Ma2][G]Ma2 Ma6 M4Ga6Ino EtN-P-[Ma2][X]Mca2 Ma6 Ma4Gca6Ino EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4Gcu6Ino EtN-P-Mo2 Ma6 Ma4G Ma2 Mca6 Ma4G EtN-P-Ma2 Mc6 M EtN-P-[Ma2][G]Mca2 Ma6 Mc4G EtN-P-[Moc2][X]Ma2 Ma6 Ma4G EtN-P-[Ma2][EtN-P]Ma2 Ma6 Ma4G Ma2 [Mca2][G]Ma2 Ma6 Mc4G Ma2 [Ma2][X]Ma2 Mca6 Ma4G Ma2 [Ma2][EtN-P]Ma6 Ma4G Ma6 Ma4Ga6Ino Mc2 Mu6 Ma4Gca6Ino Ma2 [Ma2]Ma6 M4Ga6Ino Ma2 [Ma2][G]Ma6 M4Ga6Ino Ma2 [Ma2][X]M(6 Ma4Ga6Ino EtN-P-[Ma2][G]Mo2 Ma6 M EtN-P-[Ma2][X]Ma2 Ma6 M EtN-P-[Ma2][EtN-P]Ma2 Ma6 M Ma2 [Ma2][G]Ma2 Ma6 M WO 00/15254 PCT/AU99/00770 - 84 Ma2 [Ma2][X]Ma2 Ma6 M Ma2 [Ma2][EtN-P]Ma6 M Ma2 Ma6 M Ma6 Ma4G EtN-P-[Ma2][G]Ma2 M EtN-P-[Ma2][X]Ma2 M EtN-P-[Ma2][EtN-P]Ma2 M or derivative or equivalent thereof wherein EtN is ethanolamine, P is phosphate, M is mannose, G is non-N-acetylated glucosamine, [G] is any non-N-acetylated hexosamine, Ino is inositol or inositol-phosphoglycerol, [X] is any other substituent, a represents a-linkages which may be substituted with P-linkages wherever required, and numeric values represent positional linkages which may be substituted with any other positional linkages as required.
50. An antibody directed to a GPI inositolglycan domain but which antibody is substantially incapable of interacting with the lipidic domain of a GPI.
51. A pharmaceutical composition comprising an antibody directed to a GPI inositolglycan domain, but which antibody is substantially incapable of interacting with a GPI lipidic domain, together with one or more pharmaceutically acceptable carriers and/or diluents.
52. A method of inhibiting, halting or delaying the onset or progression of a mammalian disease condition characterised by a micro-organism infection said method comprising administering to said mammal an effective amount of an antibody as claimed in claim 50 or 51. WO 00/15254 PCT/AU99/00770 - 85
53. Use of an antibody according to claim 50 or 51 in the manufacture of a medicament for inhibiting, halting or delaying the onset or progression of a disease condition characterised by the infection of a mammal by a micro organism.
AU58420/99A 1998-09-14 1999-09-14 Immunogenic compositions and uses thereof Expired AU766837C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU58420/99A AU766837C (en) 1998-09-14 1999-09-14 Immunogenic compositions and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AUPP5893A AUPP589398A0 (en) 1998-09-14 1998-09-14 Immunogenic compositions and uses thereof
AUPP5893 1998-09-14
AU58420/99A AU766837C (en) 1998-09-14 1999-09-14 Immunogenic compositions and uses thereof
PCT/AU1999/000770 WO2000015254A1 (en) 1998-09-14 1999-09-14 Immunogenic compositions and uses thereof

Publications (3)

Publication Number Publication Date
AU5842099A true AU5842099A (en) 2000-04-03
AU766837B2 AU766837B2 (en) 2003-10-23
AU766837C AU766837C (en) 2006-01-12

Family

ID=25632095

Family Applications (1)

Application Number Title Priority Date Filing Date
AU58420/99A Expired AU766837C (en) 1998-09-14 1999-09-14 Immunogenic compositions and uses thereof

Country Status (1)

Country Link
AU (1) AU766837C (en)

Also Published As

Publication number Publication date
AU766837B2 (en) 2003-10-23
AU766837C (en) 2006-01-12

Similar Documents

Publication Publication Date Title
US8058401B2 (en) Immunogenic compositions and uses thereof
US8501183B2 (en) Immunogenic compositions and diagnostic and therapeutic uses thereof
US20110008383A1 (en) Compositions of toll-like receptor agonists and malaria antigens and methods of use
WO2007107488A2 (en) Vaccine against trypanosoma cruzi infection
US5932225A (en) Vaccine comprising eimeria spp. gametocyte antigen
US20030161838A1 (en) Isolation and purification of P. falciparum merozoite protein-142 vaccine
US6015882A (en) Vaccines, antibodies, proteins, glycoproteins, DNAs and RNAs for prophylaxis and treatment of Cryptosporidium parvum infections
JP2617946B2 (en) Coccidiosis vaccine
AU766837B2 (en) Immunogenic compositions and uses thereof
WO2000024406A1 (en) A method of activating t cells and agents useful for same
AU714841B2 (en) Toxoplasma gondii glycoconjugates
EP1808443A1 (en) Purified polypeptide comprising or consisting of a C-terminus MSP1 antigen from Plasmodium falciparum carrying a Glycosyl-phosphatidyl-inositol group (GPI)
AU775222B2 (en) A method of activating T cells and agents useful for same
US20030161839A1 (en) Recombinant P. falciparum merozoite protein-142 vaccine
SCHOFIELD Patent 2493782 Summary
US8188213B2 (en) Purified polypeptide comprising or consisting of a C-terminus MSP1 antigen from Plasmodium falciparum carrying a Glycosyl-phosphatidyl-inositol group (GPI)
Rao et al. Developing peptide based immunogen against human malaria delivered in liposomes containing non-toxic adjuvants
Jansen Identification of non-procyclin molecules expressed by Trypanosoma brucei brucei procyclic culture forms
Chai The humoral response to a chemically defined synthetic vaccine comprised of epitopes derived from the circumsporozoite protein of Plasmodium berghei

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired