MXPA01010654A - Vaccines - Google Patents

Vaccines

Info

Publication number
MXPA01010654A
MXPA01010654A MXPA/A/2001/010654A MXPA01010654A MXPA01010654A MX PA01010654 A MXPA01010654 A MX PA01010654A MX PA01010654 A MXPA01010654 A MX PA01010654A MX PA01010654 A MXPA01010654 A MX PA01010654A
Authority
MX
Mexico
Prior art keywords
vaccine
saponin
cpg
antigen
composition according
Prior art date
Application number
MXPA/A/2001/010654A
Other languages
Spanish (es)
Inventor
Nathalie Garcon
Martin Friede
Philippe Hermand
Original Assignee
Smithkline Beecham Biologicals Sa
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Smithkline Beecham Biologicals Sa filed Critical Smithkline Beecham Biologicals Sa
Publication of MXPA01010654A publication Critical patent/MXPA01010654A/en

Links

Abstract

The present invention relates to adjuvant compositions which are suitable to be used in vaccines. In particular, the adjuvant compositions of the present invention comprises a saponin and an immunostimulatory oligonucleotide, optionally with a carrier. Also provided by the present invention are vaccines comprising the adjuvants of the present invention and an antigen. Further provided are methods of manufacture of the adjuvants and vaccines of the present invention and their use as medicaments. Methods of treating an individual susceptible to or suffering from a disease by the administration of the vaccines of the present invention are also provided.

Description

ADJUVANT COMPOSITION COMPRISING SAPONINE AND AN IMMUNOSTIMULATOR OLIGONUCLEOTIDE The present invention relates to novel adjuvant compositions for use in vaccines. In particular, the adjuvant compositions of the present invention comprise a combination of saponin and an immunostimulatory oligonucleotide, said combination optionally further comprising a carrier. Vaccines comprising the adjuvant compositions of the present invention and at least one antigen are also provided by the present invention. Further elaboration methods of the adjuvant compositions and vaccines of the present invention and their use as medicaments are provided. Additionally, the present invention provides methods for treating an individual susceptible to or suffering from a disease by parenteral or mucosal administration of the vaccines of the present invention. Immunostimulatory oligonucleotides containing unmethylated CpG dinucleotides ("CpG") and which are known in the art as adjuvants when administered both systemically and mucosally (WO 96/02555, EP 468520, Davis er al., J. Immunol, 1 998, 1 60 (2): 870-876; McCluskie and Davis, J. Immunol., 1998, 1 61 (9): 4463-6). CpG is an abbreviation for the cytosine-guanosine dinucleotide motifs present in DNA. Historically, it is observed that the BCG DNA fraction could exert an anti-tumor effect. In further studies, synthetic oligonucleotides derived from the BCG gene sequences are shown to be capable of inducing immunostimulatory effects (both in vitro and in vivo). The authors of these studies concluded that certain palindromic sequences, including a central CG motif, carry this activity. The central role of the CG motif in the immunostimulation was then elucidated in a publication by Krieg, Nature 374, p546 1995. Detailed analysis has shown that the CG motif has to be in a certain sequence context, and that such sequences are common in Bacterial DNA but they are rare in vertebrate DNA. The immunostimulatory sequence is often: Purine, Purine, C, G, pyrimidine, pyrimidine; wherein the CG motif of dinucleotide is not methylated, but other unmethylated CpG sequences are known to be immunostimulatory and can be used in the present invention. In certain combinations of the six nucleotides, a palindromic sequence is present. Several of these motifs, either as repeats of a motif or a combination of different motifs, may be present in the same oligonucleotide. The presence of one or more of these immunostimulatory sequences containing oligonucleotides can activate various immune subsets, including natural killer cells (which produce interferon and have cytolytic activity) and macrophages (Wooldrige et al., Vol. 89 (No. 8) 1 977). Although other sequences containing unmethylated CpG do not have their consensus sequence, they have now been shown to be immunomodulatory. CpG when formulated in vaccines, is generally administered in free solution with free antigen (WO 96/02555; McCluskie and Davis, supra) or conjugated covalently with an antigen (PCT Publication No. WO 98/16247), or formula with a vehicle such as aluminum hydroxide (Hepatitis surface antigen) Davis er al., supra; Brazolot-Millan ef al. , Proc. Nati Acad. Sci. USA 1998, 95 (26), 15553-8). Saponins are taught in: Lacaille-Dubois, M and Wagner J. (1996, A review of the biological and pharmacological activities of saponins, Phytomedicine vol 2 pp. 363-386). Saponins are triterpene and steroid glycosides widely distributed in the kingdoms, marine and plant animal. It is observed that saponins form colloidal solutions in water that foam in the agitation, and to precipitate cholesterol. When saponins are found near cell membranes they create pore-like structures in the membranes that cause the membrane to break. Hemolysis of erythrocytes is an example of this phenomenon, which is a property of certain, but not all, saponins. Saponins are known as adjuvants in vaccines for systemic administration. The hemolytic and adjuvant activity of individual saponins has been studied extensively in the art (Lacaille-Dubois and Wagner, supra). For example, Quil A (derived from the bark of the South American tree Quillaja Saponaria Molina), and fractions thereof, are described in US 5,057,540 and "Saponins as vaccine adjuvant", Kensil, C.R. , Crit Rev Ther Drug Carrier Sist, 1996, 12 (1-2): 1 -55; and EP 0 362 279 B1. The particulate structures, called Immune Stimulating Complexes (ISCOMS), which comprise fractions of Quil A are homeolytic and have been used in the manufacture of vaccines (Morein, B., EP 0 109 942 B1). It has been reported that these structures have adjuvant activity (EP 0 109 942 B1, WO 96/1 171 1). The hemolytic saponins QS21 and QS 1 7 (fractions purified by HPLC of Quil A) have been described as potent systemic adjuvants, and the method of their production is described in the U.S. Patent. No. 5,057,540 and EP 0 362 279 B1. The use of QS7 (a non-haemolytic fraction of Quil A) which acts as a potent adjuvant for systemic vaccines is also described in these references. The use of QS21 is further described in Kensil et al., (1991, J. Immunology vol 143, 431-437). The combinations of QS21 and polysorbate or cyclodextrin are also known (WO 99/10008). Particulate adjuvant systems comprising Quil A fractions, such as QS21 and QS7 are described in WO 96/33739 and WO 96/1 171 1. Other saponins that have been used in systemic vaccination studies include those derived from other plant species such as Gisophila and Saponaria (Bomford et al., Vaccine, 10 (9): 572-577, 1992). It is also known that saponins have been used in studies of mucosely applied vaccines, which have found variable success in the induction of immune responses. It has previously been shown that Qui-A saponin has no effect on the induction of an immune response when the antigen is administered intrastrially (Gizurarson er al., 1 994. Vaccine Research 3, 23-29). Meanwhile, other authors have successfully used this adjuvant (Maharaj er al., Can. J. Microbial, 1986, 32 (5): 414-20, Chavali and Campbell, Immunobiology, 174 (3): 347-59). ISCOMs comprising Quil A saponin have been used in intranasal and intragastric vaccine formulations and shown adjuvant activity (Mcl Mowat et al., 1991, Immunology, 72, 317-322; Mcl Mowat and Donachie, Immunology Today, 12, 383- 385). QS21, the non-toxic fraction of Quil A, has also been described as an intranasal or oral adjuvant (Sumino et al., J. Virol., 1 998, 72 (6): 4931-9; WO 98/56415). The use of other saponins in studies of intranasal vaccination has been described. For example, Chenopodium quinoa saponins have been used in both intranasal and intragastric vaccines (Estrada ef al., Comp.Immunol, Microbiol.Infect. Dis., 1998, 21 (3): 225-36). The present invention relates to the surprising discovery that immunostimulatory oligonucleotides (CpG) and combinations of saponin are extremely potent adjuvants. Accordingly, an adjuvant composition comprising a combination of saponin and an immunostimulatory oligonucleotide is provided. Preferably, the adjuvants of the present invention may further comprise a carrier. In a preferred form of the present invention, saponin and oligonucleotides in vaccine and adjuvant compositions act synergistically in the induction of antigen-specific antibody and are potent in the induction of immune responses conventionally associated with the Th1-like immune system. In accordance with the foregoing, adjuvant combinations are not only suitable for the immunoprophylaxis of diseases, but also surprisingly for immunotherapy of diseases such as parasitic, bacterial or persistent viral infections, and also chronic disorders such as cancer. Preferred oligonucleotides for use in adjuvants or vaccines of the present invention preferably contain two or more CpG motifs of oligonucleotides separated by at least three, more preferably at least six or more nucleotides. The oligonucleotides of the present invention are typically deoxynucleotides. In a preferred embodiment, the internucleotide in the oligonucleotide is phosphodiester, or more preferably a phosphorothioate linkage, although phosphodiester linkages and other internucleotides are within the scope of the invention including oligonucleotides with internucleotide linkages. Methods for producing phosphorodithioate or phosphorothioate oligonucleotides are described in EU 5,666, 153, EU 5,278, 302 and WO 95/26204. Examples of preferred oligonucleotides have the following sequences. The sequences preferably contain phosphorothioate modified internucleotide linkages. OLIGO 1 (SEQ ID NO: 1): TCC ATG ACG TTC CTG ACG TT (CpG 1826) OLIGO 2 (SEQ ID NO: 2): TCT CCC AGC GTG CGC CAT (CpG 1758) OLIGO 3 (SEQ ID NO: 3) : ACC GAT GAC GCC GCC GGT GAC GGC ACC ACG OLIGO 4 (SEQ ID NO: 4) TCG TCG TTT TGT CGT TTT GTC GTT (CpG 2006) OLIGO 5 (SEQ ID NO: 5): TCC ATG ACG TTC CTG ATG CT ( CpG 1668). Alternative CpG oligonucleotides can comprise the above preferred sequences in that they have additions or incosequence deletions thereto. The CpG oligonucleotides used in the present invention can be synthesized by any method known in the art (e.g., EP 468520). Conveniently, such oligonucleotides can be synthesized using an automated synthesizer. The oligonucleotides used in the present invention are typically deoxynucleotides. In a preferred embodiment, the internucleotide linkage in the oligonucleotide is phosphorodithioate, or more preferably phosphorothioate linkage, although phosphodiesters are within the scope of the present invention. The oligonucleotide comprising different internucleotide linkages are contemplated, for example, mixed phosphorothioate phosphodiesters. Other internucleotide linkages that stabilize the oligonucleotide can be used. Saponins that can be used in the adjuvant combinations of the present invention include those derived from the bark of Quillaja Saponaria Molina, designated Quil A, and fractions thereof, described in US 5,057,540 and "Saponins as vaccine adjuvant"; Kensil, C.R. , Crit Rev Ther Drug Carrier Sist. , 1 996, 12 (1-2): 1 -55; and EP 0 362 279 B1. Particularly preferred fractions of Quil A are QS21, QS7 and QS1 7. β-Escin is another preferred hemolytic saponin for use in the adjuvant compositions of the present invention. Escina is described in the Merck index (12th edition: entry 3737) as a mixture of saponins that occur in the seed of the horse chestnut tree, Lat: Aesculus hippocastan? M. Its isolation is described by chromatography and purification (Fiedler, Arzneimittel-Forsch: 4, 21 3 (1953)), and by ion exchange resins (Erbring et al., US 3,238, 190). Fractions of escin, a and ß, have been purified and shown to be biologically active (Yoshikawa M, ef al., (Chem Pharm Bull (Tokyo) 1996 Aug: 44 (8): 1454-1464)). β-Escin is also known as Aescin. Another preferred hytic saponin for use in the present invention is Digitonin. Digitonin is described in the Merck index (12th edition: entry 3204) as a saponin, derived from the seeds of Digitalis purpurea and purified according to the procedure described by Gisvold et al., J. Am. Pharm. Assoc. , 1934, 23, 664; and Ruhenstroth-Bauer, Physiol. Chem., 1955, 301, 621. Its use is described as being a clinical reagent for the determination of cholesterol. The adjuvant combinations of the present invention may further comprise a vehicle such as saponin or CpG, or both, may be associated with a particulate carrier entity to increase the adjuvanticity of the combination. Particularly preferred systemic vaccines, for example, comprise a carrier molecule. CpG used in the adjuvant combinations of the present invention can be found in free solution or can be compounded with particulate carriers such as mineral salts (for example, but not limited to, calcium or aluminum salts), liposomes, ISCOMs, emulsions (oil in water , water in oil, water in oil in water), polymers (such as, but not limited to polylactic, polyglycolic, polyphosphazine, polyamino acid, alginate, chitosan) or microparticles. Preferably, said vehicles are cationic. The vaccines of the present invention further comprise an antigen that may be associated with the CpG-vehicle complex, or may not associate with the CpG-carrier complex. In this case, the antigen may be in free suspension or be associated with a separate vehicle. The saponins which form part of the present invention can be separated in the form of micelles, or they can be in the form of ordered long structures such as ISCOMs (EP 0 109 942 B1) or liposomes (WO 96/33739) when formulated with cholesterol and lipid, or in the form of an oil-in-water emulsion (WO 95/1721 0). Saponins can be preferably associated with a metal salt, such as aluminum hydroxide or aluminum phosphate (WO 98/15287). Alternatively, saponin may be associated with a particulate carrier such as chitosan. The saponin can also be found in a dry state such as a powder. The final formulations in the form as administered to the mucosal surface of the vaccines are preferably hytic in nature. The saponin may or may not be physically associated with the antigen either through the direct bond or by co-interaction with the same particulate vehicle molecule (GB 9822712.7, WO 98/16247). CpG and the saponin in the adjuvants or vaccines of the present invention can themselves be separated or associated. For example, CpG and saponin can be in free suspension or associated through a vehicle, more preferably a particulate carrier such as aluminum hydroxide or by a cationic liposome or ISCOM. An adjuvant combination according to the present invention is composed of one or more CpG oligonucleotides containing at least 3, preferably at least 6 nucleotides between two adjacent CG motifs, together with QS21 and a particulate carrier selected from the group comprising an emulsion of oil in water or DQ. More preferably, the adjuvant combination comprises CpG2006 (SEQ ID NO: 4), or CpG 1758 (SEQ ID NO: 2) or CpG 1 826 (SEQ ID NO: 1) mixed with QS21, and a particulate carrier selected from the group comprising an oil in water emulsion or DQ. According to the above, particularly preferred vaccines, for example, comprise such adjuvant combinations and an antigen. The preferred vaccine of the present invention is used to generate systemic immune responses after administration to an individual through the systemic route. The adjuvant combinations of the present invention can be used both as a systemic and mucosal adjuvant. In a particular form of the invention, a systemic vaccine is provided for administration through a systemic or parenteral route such as intramuscular, intradermal, transdermal, subcutaneous, intraperitoneal or intravenous administration. A preferred route of administration is through the transdermal route, for example skin patches. The systemic vaccine preparations of the present invention can be used to protect or treat a mammal susceptible to, or suffering from disease, by administering said vaccine by intramuscular, intraperitoneal, intradermal, transdermal, intravenous or subcutaneous administration. Methods of systemic administration of vaccine preparations may include syringes and needles, or devices designed for ballistic delivery of solid vaccines (WO 99/27961), or needle-free liquid injection device (US 4,596,556).; US 5,993,412), or transdermal patches (WO 97/48440, WO 98/28037). The present invention can also be used to increase the immunogenicity of antigens applied to the skin (transdermal or transcutaneous supply WO 98/20734, WO 98/28037). Therefore, the present invention provides a delivery device for systemic administration, pre-filled with the adjuvant or vaccine compositions of the present invention. In accordance with the foregoing, a method is provided for inducing an immune response in an individual, comprising administering a vaccine comprising an antigen and immunostimulatory oligonucleotide, a saponin, and a vehicle, to the individual, wherein the vaccine is administered. through the systemic or parenteral route. Preferred methods for inducing an immune response comprise administration of a vaccine comprising an oligonucleotide of SEQ ID NO: 1, 2, 3, 4 or 5, with a saponin derived from QuilA, such as QS21, and a carrier, such as an oil in water emulsion, a cholesterol containing liposome or alum. Alternatively, the vaccine preparations of the present invention can be used to protect or treat a mammal susceptible to, or suffering from disease, by administering said vaccine through a mucosal route, such as the oral / alimentary or nasal route. . Alternative mucosal pathways are intravaginal and intra-rectal. The preferred mucosal route of administration is through the nasal route, called intranasal vaccination. Methods of intranasal vaccination are well known in the art, including administration of a dry powder form, spray or drop of vaccines in the nasopharynx of the individual to be immunized. Aerobic or nebulized vaccine formulations also form part of this invention. Enteric formulations such as gastro-resistant granules and capsules for oral administration, suppositories for vaginal or rectal administration also form part of this invention. The adjuvant combinations of the present invention represent a class of mucosal adjuvants suitable for application in humans to replace systemic vaccination by mucosal vaccination. In a preferred form of the present invention, pure saponins such as Quil A, or derivatives thereof, including QS21; Escina; Digitonin; or Gypsohila or Chenopodium quinoa saponins in combination with immunostimulatory oligonucleotides can be used as adjuvants for the mucosal administration of antigens to achieve a systemic immune response. The adjuvant combinations of the present invention are used in the formulation of vaccines, such vaccines can be administered through the systemic or mucosal route. Preferably, when the vaccines are used for mucosal administration, the adjuvant combination comprises a hemolytic saponin. For mucosal administration, preferably the compositions of the invention comprise a hemolytic saponin. The hemolytic saponin, or saponin preparation, within the meaning of this invention is to be determined with reference to the following analysis. 1 . Fresh guinea pig blood is rinsed with phosphate buffered saline (PBS) 3 times in a shelf centrifuge.
After suspension to the original volume, the blood is further diluted 10 times in PBS. 2. 50 μl of this blood suspension is added to 80 μl of PBS containing double dilutions of surfactant and saponin. 3. After 8 hours the hemolysis is assessed visually or by measuring the optical density of the supernatant. The presence of a red supernatant, which absorbs light at 570 nm, indicates the presence of hemolysis. 4. The results are expressed as the concentration of the first dilution of saponin in which hemolysis does not occur anymore. For the purposes of this invention, the adjuvant preparation of saponin is hemolytic if it lyses the erythrocytes in a concentration of less than 0.1%. As a reference medium, the substantially pure samples of QuilA, QS21, QS7, Digitonin, and β-escin are all hemolytic saponins as defined in this analysis, within the inherent experimental variability of such biological analysis, the saponins of the present invention preferably they have a hemolytic activity, of between about 0.5-0.00001%, more preferably between 0.05-0.00001%, even more preferably between 0.005-0.00001%, and more preferably between 0.001-0.004%.
Ideally, said saponins should have similar hemolytic activity (ie, within a tenfold difference) to that of QS21. The vaccine of the present invention can also be administered orally. In such cases the pharmaceutically acceptable excipient may also include alkaline regulators, or enteric capsules or microgranules. The vaccines of the present invention can also be administered vaginally. In such chaos, pharmaceutically acceptable excipients may also include emulsifiers, polymers such as CARBOPOL®, and other known stabilizers of vaginal creams and suppositories. The vaccines of the present invention can also be administered rectally. In such cases, the excipients may also include waxes and polymers known in the art to form rectal suppositories. The preparations of more than one saponin in the adjuvant combinations of the present invention also form part of the present invention. For example, combinations of at least two of the following group comprising QS21, QS7, Quil A, β-escin, or digitonin. Additionally, the compositions of the present invention may comprise a combination of more than one immunostimulatory oligonucleotide. In a similar embodiment of the present invention combinations of CpG / saponin for both systemic and mucosal administration can be further combined with other adjuvants including Monophosphoryl lipid A and its non-toxic derivative of monophosphoryl A 3-de-O-acylated . Alternatively, the saponin formulations can be combined with vaccine carriers composed of chitosan or other polycationic polymers, polylactide and polylactide-co-glycolide particles, polymer matrix based on poly-N-acetyl glucosamine, particles composed of polysaccharides or chemically polysaccharides modified, liposomes and lipid-based particles, particles composed of glycerol monoesters, etc. Saponins can also be formulated in the presence of cholesterol to form particulate structures such as liposomes or ISCOMs. In addition, the saponins can be formulated together with a polyoxyethylene ester or ether, in either a suspension or particulate solution, or in a particulate structure such as a paucilamellar liposome or ISCOM. The saponins can also be formulated with excipients such as Carbopol® to increase viscosity, or they can be formulated into a dry powder form with a powdered carrier such as lactose. The 3-de-O-acylated monophosphoryl lipid A is a well-known adjuvant manufactured by Ribi Immunochem, Montana. It can be prepared by the methods taught in GB 2122204B. A preferred form of 3-de-O-acylated monophosphoryl lipid A is in the form of an emulsion having a small particle size smaller than 0.2 μm in diameter (EP 0 689 454 B1). Particularly preferred adjuvants are combinations of 3D-MPL and QS21 (EP 0 671 948 B1), oil-in-water emulsions comprising 3D-MPL and QS21 (WO 95/17210, WO 98/56414), or 3D-MPL formulated with other vehicles (EP 0 689 454 B1).
Preferably, the formulations. The vaccine of the present invention contains an argenic or antigenic composition capable of producing an immune response against a human pathogen, such an antigenic or antigenic composition of, a of HIV-1, (such as tat, nef, gp1 20 or gp160. ), huma herpes virus or, such as gD or derivative thereof or Immediate Anterior protein, such as ICP27 of HSV1 or HSV2, cytomegalovirus ((human) (such as gB or derivatives thereof), Rotavirus (including live attenuated virus) ), Epstein-Barr virus (such as gp350 or derivative thereof), Viru s Varicella-Zoster (such as gpl, II and IE63), or a hepatitis t virus such as hepatitis B virus (for example surface antigen) of hepatits B, or of other viral pathogens, such as paraxioviruses: Respiratory Sincital virus (such as F and G proteins or derivatives thereof), parainfluenza virus, measles virus, mumps virus, human papilloma virus (e.g. HPV6, 1 1, 1 8, ...), flavivirus (for example, Virus d Yellow Fever, Dengue Virus, Tick-borne Encephalitis Virus, Japanese Encephalitis Virus), or Influenza Virus (inactive or live full virus, influenza virus, grown in eggs or MDCK cells, or complete influenza viruses (such as described by R. Gluck, Vacuna, 1992, 10, 915-920) or recombinant or purified proteins thereof, such as HA, NP, NA or M proteins, or combinations thereof), or derived from bacterial pathogens such as Neisseria spp, including N. gonorrhea and N. meningitidis (for example capsular polysaccharides and conjugates thereof, transferrin binding proteins, lactoferin binding proteins, PilC, adhesins); S. pyrogenes (for example M proteins or fragments thereof, protease C5A, lipoteichoic acid), S. agalactiae, S. mutans; H. ducreyi; Moraxella spp., Including catarrhalis, also known as catarrhalis Branhamella (eg, low molecular weight, high molecular weight adhesins). Berdetella spp, including B. pertussis (for example pertactin, pertussis toxin or derivatives thereof, filamentous haemagglutinin, adenylate cyclase, fimbriae), B. parapertussis and B. bronchiseptica; Mycobaterium spp, including M. tuberculosis (eg, ESAT6, Antigen 85A, -B or -C), M. bovis, M. leprae, M. avium, M. paratuberculosis, M. smegmatis; Legionella spp, including L. pneumophila; Escherichia spp, including £. enterotoxic coli (eg, colonization factors, heat-unstable toxin or derivatives thereof, heat-stable toxin or derivatives thereof), enterohemorrhagic E. coli, enteropathogenic E. coli (eg, toxin similar to Shiga toxin) or derivatives thereof); Vibrio spp, including V. cholera (e.g., cholera toxin or derivative thereof); Shigella spp, including S. sonnei, S. dysenteriae, S. flexnerii; Yersinia spp, including Y. enterocolitica (for example a Yop protein), Y. Perstis, Y. pseudotuberculosis, Campylobacter spp, including, C. jejuni (for example, toxins, adhesins and invasins), and C. coli; Salmonella spp, including S. typhy, S. paratyph, S. choleraesuis, S. enteritidis; Listeria spp, including L. monocytogenes; Helicobacter spp, including H. pylori (for example, urease, catalaza, vacuolation toxin); P. seudomonas spp, Enterococcus spp, including E. faecalis, E. faecium; Clostridium spp; including C. tetan (for example, botulinum toxin and derivative thereof), O difficile (for example clotridium toxins A or B and derivatives thereof); Corynebacterium spp, including C. diphteriae (for example, diphtheria toxin and derivatives thereof), Borrelia spp, including B. burgdorferi (e.g., OspA, OspC, DbpA, DbpB), B. garinii (e.g., OsPA, OspC, DbpA, DbpB), B. afzelii (e.g., OspA, OspC, DbpA, DbpB), B. andersonii (e.g., OsPA, OspC, DbpA, DbpB), 8. hermsii; Ehrlichia spp, including E. equi and the agent of Human Granulotic Erlicosis; Rickettsia spp, including Chlamydia spp, including C. trachomatis (eg MOMP, heparin binding proteins), C. pneuoniae (eg, MOMP, heparin binding proteins), C. psittasci; Leptospira spp, including L. interrogans; Treponema spp, including T. palladum (for example, outer membrane proteins, posterior), T. denticola, T. hydodysenteriae; or it is derived from parasites such as Plasmodium spp; including P. falciparum; Toxoplasma spp, including T. gondii (eg, SAG2, SAG3, Tg34); Entamoeba spp, including E. histolytica, Babesia spp, including 8. microti; Trypanosoma spp, including T. cruzi, Giardia spp, including G. lamblia, Leshmania spp, including L. major, Pneumocystis spp, including P. carinni, Tricomonas spp, including T. vaginalis, Schisostoma spp, including S. mansoni, or yeast derived such as Candida spp, including C. albicans; Cryptococcus spp, including C. peofor / aunt .s. Other specific antigens, preferred for M. tuberculosis are for example Tb Ra1 2, Tb H9, Tb Ra35, Tb38-1, Erd 1 4, DPV, MTI, MSL, mTTC2 and hTCC 1 (WO 99/51 748). The proteins for M. tuberculosis also include fusion proteins and variants thereof wherein at least two, preferably three, M. tuberculosis polypeptides are fused to a larger protein. Preferred mergers include Ra 1 2-TbH9-Ra35, Erd 14-DPV-MTI, DPV-MTI-MSL, Erd 14-DPV-MTI-MSL-mTTC2, Erd14-DPV-MTI-MSL, DPV-MTI-MSL-mTCC2, TbH9- DPV-MTI (WO 99/51748). The most preferred antigens for Chlamydia include for example the High Molecular Weight Protein (HWMP) (WO 99/17741), ORF3 (EP 366 412), and putative membrane proteins (Pmps). Other Chlamydia antigens of the vaccine formulation may be selected from the group described in WO 99/28475. Preferred bacterial vaccines comprise antigens derived from Streptococcus spp, including S. pneumoniae (for example capsular polysaccharides and conjugates thereof, PsaA, PspA, streptolysin, choline binding proteins) and protein antigen Neumolysin (Biochem Biophys Acta, 1 987, 67, 1 007; Rubis et al., Microbial Pathogenesis, 25, 337-342), and detoxified mutant derivatives (WO 90/06951, WO 99/03884). Other preferred bacterial vaccines comprise antigens derived from Haemophilus spp, including H. influenzae type B (eg, PRP and conjugates thereof), H. influenzae without type, eg, OMP26, high molecular weight adhesins, P5, P6, protein D and lipoprotein D, and fimbrine and peptides derived from fimbrine (EU 5,843,464) or multiple copy variants or fusion proteins thereof. The Hepatitis B Surface Antigen derivatives are well known in the art and include, among others, those PreS1, PreS2 antigens established as described in the European Patent Applications EP-A-414 374; EP-A-304 578 and EP 198-474. In a preferred aspect, the vaccine formulation of the invention comprises the HIV-1 antigen, gp120, especially when expressed in CHO cells. In a further embodiment, the vaccine formulation of the invention comprises gD2t as defined hereinbefore. In a preferred embodiment of the present invention vaccines containing the claimed adjuvant comprise antigen derived from the Human Papilloma Virus (HPV) considered responsible for genital warts (HPV 6 or HPV1 1 and others), and HPV viruses responsible for cancer cervical (HPV16, HPV18 and others). Particularly preferred forms of genital wart prophylactic or therapeutic vaccine comprise L1 particles or capsomeres, and the fusion proteins comprise one or more antigens selected from the HPV 6 and HPV 1 1 E6, E7, L1 and L2 proteins. The most preferred forms of fusion protein are: L2E7 as described in WO 96/26277, and protein D (1/3) -E7 described in GB 9717953.5 (PCT / EP98 / 05285). A preferred HPV cervical cancer or infection composition, prophylaxis or therapeutic vaccine, may comprise HPV 16 or 1 8 antigens. For example, L1 or L2 antigen monomers, or L1 or L2 antigens presented together as a virus-like particle. (VLP) or single L1 protein presented alone in a VLP or capsomere structure. Such antigens, virus-like particles and capsomer are known per se. See, for example WO 94/00152, WO 94/20137, WO 94/05792 and. The above, additional proteins can be included alone or as fusion proteins such as E7, E2 or preferably E5 for example; Particularly preferred embodiments of this include a VLP comprising L1 E7 fusion proteins (WO 96/1 1272). Particularly preferred HPV 16 antigens comprise the above proteins E6 or E7 in fusion with a carrier protein D to form the Protein D-E6 or E7 fusions of HPV 16, or combinations thereof; or combinations of E6 or E7 with L2 (WO 96/26277). Alternatively, the above proteins HPV 16 or 18 E6 and E7 can be present in a single molecule, preferably a fusion of Protein D-E6 / E7. Such a vaccine can optionally contain either or both HPV 18 E6 and E7 proteins, preferably in the form of a Protein D-E6 or Protein D-E7 fusion protein or Protein D fusion protein E6 / E7. The vaccine of the present invention may additionally comprise antigens of other strains of HPV, preferably of HPV 31 or 33 strains. The vaccines of the present invention also comprise antigens derived from parasites that cause Malaria. For example, the preferred antigens of Plasmodia falciparum include RTS, S and TRAP. RTS is a hybrid protein that comprises substantially all of the C-terminal portion of the circumsporozoite (CS) protein of P. falciparum linked through four amino acids from the preS2 portion of the Hepatitis B surface antigen to the surface antigen (S) of the hepatitis B virus. Its complete structure is described in International Patent Application No. PCT / EP92 / 02591, published under the number WO 93/101562 which claims the priority of the UK patent application No. 9124390.7. When expressed in yeast RTS it is produced as a lipoprotein particle, and when co-expressed with the HBV S antigen it produces a mixed particle known as RTS.S. TRAP antigens are described in International Patent Application No. PCT / GB89 / 00895, published under WO 90/01496. A preferred embodiment of the present invention is a Malaria vaccine wherein the antigenic preparation comprises a combination of the antigens of TRAP and RTS.S. Other plasmodia antigens that are likely to be components of a multi-stage Malaria vaccine are P. faciparum MSP1, AMA1, MSP3, EBA, GLURP, RAP1, RAP2, Sequestrin, PfEMPI, Pf332, LSA1, LSA3, STARP, SALSA , PfEXPI, Pfs25, Pfs28, PFS27 / 25, Pfs16, Pfs48 / 45, Pfs230 and their analogs in Plasmodium spp. The formulations may also contain an antitumor antigen and be useful for the immunotherapeutic treatment of cancers. For example, the adjuvant formulation finds utility with tumor rejection antigens such as those for cancers of prostate, breast, colorectal, lung, pancreatic, renal or melanoma. Exemplary antigens include MAGE 1 and MAGE 3 or other MAGE antigens (for the treatment of melanoma), PRAME, BAGE or GAGE (Robbins and Kawakami, 1996, Current Opinions in Immunology 8, pps, 628-636; Van den Eynde ef al., International Journal of Clinical & Laboratory Research (presented in 1997); Corréale ef al., (1997), Journal of the National Cancer Institute 89, p293. In fact, these antigens are expressed in a wide range of tumor types such as melanoma, lung carcinoma, sarcoma and bladder carcinoma. Other tumor-specific antigens are suitable for use with the adjuvants of the present invention and include, but are not limited to, tumor-specific gangliosides, prostate-specific antigen (PSA) or Her-2 / neu, KSA (GA733), PAP, mammaglobin, MUC-1, carcinoembryonic antigen (CEA). In accordance with the above, in one aspect of the present invention there is provided a vaccine comprising an adjuvant composition according to the invention and a tumor rejection antigen. It is a particularly preferred aspect of the present invention that the vaccines comprise a tumor antigen; Such vaccines are surprisingly potent in cancer therapy such as prostate, breast, colorectal, lung, pancreatic, renal, ovarian or melanoma cancers. Accordingly, the formulations may contain antigen associated with the tumor, as well as antigens associated with tumor support mechanisms (eg, angiogenesis, tumor invasion). Additionally, antigens particularly relevant to vaccines in cancer therapy also comprise prostate-specific membrane antigen (PSMA), Prostate Stem Cell Antigen (PSCA), tyrosinase, survivin, NY-ESO1, prostase, PS 1 08 (WO 98/50567), RAGE, LAGE, HAGE. Additionally, said antigen can be a self-peptide hormone such as a hormone that releases the full-length Gonadotrophin hormone (GnRH, WO 95/20600), a long peptide of 10 short amino acids, useful in the treatment of many cancers, or in immunocastration. It is noted that the compositions of the present invention will be used to formulate vaccines containing the antigens derived from Borrelia sp. For example, the antigens may include nucleic acid, antigenic or antigenic preparations derived from pathogen, recombinantly prepared peptides or protein, and chimeric fusion proteins. In particular, the antigen is OspA. The OspA can be a complete mature protein in a lipidated form by virtue of the host cell (E.Coli) termed (Lipo-OspA) or a non-lipidated derivative. Such non-lipidated derivatives include the non-lipidated NS 1 -OspA fusion protein having the first 81 amino acids of N-terminus of the non-structural protein (NS1) of the influenza virus, and the complete OspA protein, and others, MDP-OspA is an unlipidated form of OspA that carries 3 amino acids of additional N ends. The vaccines of the present invention can be used for prophylaxis or allergy therapy. Such vaccines would comprise non-specific allergen antigens (e.g., human IgE-derived peptides, including but not limited to Stanworth decapeptide (EP 0 477 231 B1)) and allergen-specific (e.g., Der p1). The vaccines of the present invention can also be used for the prophylaxis or therapy of chronic disorders other than infectious, cancer or allergic diseases. Such chronic disorders are diseases such as atherosclerosis, and Alzheimer's.
The relevant antigens for the prophylaxis and therapy of patients susceptible to or suffering from Alzheimer's neurodegenerative disease are, in particular, the N-terminal 39-43 amino acid fragment (Aβ) of the amyloid precursor protein and smaller fragments. This antigen is described in International Patent Application No. WO 99/27944 - (Athena Neurosciences). The amount of protein in each vaccine dose is selected as an amount that induces an immunoprotective response without adverse side effects, significant in typical vaccines. Such amount will vary depending on what specific immunogen is used and how it is presented. Generally, it is expected that each dose will comprise 1-1000 μg of protein, preferably 1 -500 μg, preferably 1 -100 μg, more preferably 1 to 50 μg. An optimal amount for a particular vaccine can be matched by standard studies that include the observation of appropriate immune responses in vaccinated subjects. After the initial vaccination, subjects may receive one to several adequately separated booster injection immunizations. Such a vaccine formulation can be applied to a mucosal surface of a mammal in any of a booster injection or boot loading vaccination regimen; or alternatively administered systemically, for example through the transdermal, subcutaneous or intramuscular routes. The amount of CpG or immunostimulatory oligonucleotides in the adjuvants or vaccines of the present invention is generally small, but depending on the vaccine formulation it can be in the region of 1 -1000 μg per dose, preferably 1 -500 μg per dose, and more preferably between 1 to 100 μg per dose. The amount of saponin for use in the adjuvants of the present invention may be in the region of 1 -1000 μg per dose, preferably 1 -500 μg per dose, more preferably 1 -250 μg per dose, and more preferably between 1 and 100. μg per dose. The ratio of CpG: saponin (w / w) will therefore be in the range of 1: 1000 to 1000: 1, and will typically be in the range of 1: 100 to 1: 00: 1, and preferably in the range from 1: 1 0 to 1: 1 or 1: 1 to 10: 1, and more preferably 1: 1, 4: 1 or 10: 1. The formulations of the present invention can be used for both prophylactic and therapeutic purposes. Accordingly, the use of a combination of a saponin and a CpG molecule in the manufacture of a vaccine for the prophylaxis and treatment of viral, bacterial disorders, parasitic infections, allergy, cancer, and other chronic disorders is provided. . Accordingly, the present invention provides a method for treating a mammal susceptible to or suffering from an infectious disease or cancer, or allergy, or autoimmune disease. In a further aspect of the present invention there is provided an adjuvant or vaccine combination, comprising a saponin and CpG, as described herein for use as a medicament. The vaccine preparation is generally described in New Trends in Developments in Bacines, edited by Voller, et al., Park Press University, Baltimore, Maryland, U. S.A. 1978 It is noted that the compositions of the present invention will be used to formulate vaccines containing the antigens derived from a wide variety of sources. For example, the antigens may include human, bacterial or viral nucleic acid, and antigenic or antigenic preparations derived from pathogen, antigenic or antigenic preparations derived from tumor, antigens derived from the host, including IgE-derived peptides, such as decapeptide of histamine release. of IgE (known as Stanworth decapeptide), peptides or recombinantly produced protein, and chimeric fusion proteins. A systemic vaccine composition comprising an antigen is provided by the present invention., a saponin and an immunostimulatory immunostimulant. In accordance with the foregoing, a method of treating an individual susceptible to or suffering from a disease is provided by the administration of a composition as substantially described herein through the systemic pathway of said individual. A method is also provided to prevent an individual from contracting a disease selected from the group comprising viral, bacterial and infectious diseases, parasitic diseases, prostate, breast, colorectal, lung, pancreatic, renal, ovarian or melanoma cancers.; Chronic disorders without cancer, allergy, Alzheimer's, atherosclerosis, comprising the administration of a composition as substantially described herein through the systemic pathway of said individual. Alternatively, a mucosal vaccine composition comprising an antigen, and a hemolytic saponin is provided by the present invention. Accordingly, there is provided a method of treating an individual susceptible to or suffering from a disease by the administration of a composition co or substantially described herein to a mucosal surface of said individual. In addition, a method for inducing a systemic antigen-specific immune response in a mammal is described, which comprises administering to a mucosal surface of said mammal a composition comprising an antigen and a hemolytic saponin. In addition, a method of making a vaccine or adjuvant is provided, which comprises taking a saponin and taking a molecule of CpG and mixing them with an antigen. Examples of pharmaceutically suitable excipients for use in the combinations of the present invention include water, phosphate buffered saline, isotonic regulator solutions.
LEGENDS OF THE FIGURES Figure 1: OspA specific IgG concentrations 14 days after the nasal booster injection. Figure 2: Concentrations of LA2 specific for OspA 14 days after the injection of nasal reinforcement. Figure 3: Specific IgG concentrations of the Flu Flu strain 14 days after the nasal booster injection. Figure 4: Concentrations of Hemagglutination Inhibition (HAI) of serum specific to the Flu Flu strain 14 days after the nasal reinforcement injection. Figure 5: Concentrations of LA2 specific for OspA in mice. Figure 6: gp120-specific lymphoproliferation activity of spleen cells of immunized mice. The antigen-specific activity is expressed as SI for different concentrations of antigen for all 4 experimental groups. Figure 7: HBsAg-specific CTL activity of spleen cells from immunized mice. Effector cell activity is assessed by examining the 51 Cr release of P815 cells (open circles) or 5 s-transfected P81 cells (closed circles). Figure 8: HBsAG-specific antibody responses in immunized mice. Specific antibody concentrations (expressed as EU / ml) and isotype profiles were evaluated using ELISA tests. The values of the pooled serum are shown on the tabal, and the isotype distributions are also represented on the graph. Figure 9: gp120-specific lymphoproliferation activity and HBSAg of spleen cells from immunized mice. The antigen-specific activity is expressed as SI for different concentrations of antigen for all 4 experimental groups. Figure 10: CTL activity specific to gp120 and HBsAg of spleen cells from immunized mice. Effector cell activity is assessed by examining 51Cr release of P815 cells (open symbols) or P815 cells displaying a CTL epitope of gp120 or HBsAG (closed symbols). Figure 1 1: Specific antibody responses of HbsAg and Gp1-specific 20 in immunized mice. The concentrations of the specific antibody (expressed in μg / ml) (Figure 1 1 A) and the isotype profiles were evaluated using ELISA tests. The values of grouped sera are shown in the table, and the isotype distributions are also represented in the graph. Figure 1 1 B shows the isotype pattern of gp120-specific antibodies. Figure 1 2: Evolution of average tumor growth by groups of 10 animals over time. The present invention is illustrated by, but not limited to, the following examples. EXAMPLE 1 The use of QS21 and CpG for intranasal booster injection of systemic antibodies to Lipo-OspA In this example, it is investigated whether lytic saponins such as QS21 and immunostimulators such as CpG were able to increase in a synergistic manner the immunological responses Systemic vaccination by intranasal booster injection of mice. Female Balb / c mice (5 animals per group), 8 weeks old, were immunized intramuscularly with lipo-OspA (1 μg) formulated in alum (50 μg). After 3 months, the mice were injected intranasally (under anesthesia) with 10 μl of solution (5 μl through the nostril, supplied as drops per tube) containing 5 μg of lipoprotein. OspA in either A: PBS; B: 20 μg of CpG 1 001 (TCC ATG AGC TTC CTG ACG TT, Krieg 1 826); C: 5 μg of QS21 (obtained from Cambridge Biotech, USA); D: 20 μg of CpG 1001 + 5 μg of QS21; or, E: by intramuscular injection of 1 μg of lipo-OspA absorbed in alum (50 μg). Figures 1 and 2 show the concentrations of OspA specific IgG and LA2 concentrations 14 days after the intranasal booster injection. ELISA methods for measurement of OspA-specific serum IgG in mice: Maxisorp Nunc immunoplates are coated overnight at 4 ° C with 50 μl / 1 μg / ml cavity of OspA diluted in PBS (in columns B to H of the plate), or with 50 μl of 5 μg / ml of purified goat anti-mouse Ig (Boerhinger), in PBS (column A). The free sites on the plates are blocked (1 hour, 37 ° C) using the saturation regulator; PBS containing 1% BSA, 0.1% polyoxyethylene sorbitan monolaurate (TWEEN 20), and 4% Normal Bovine Serum (NBS). Then, serial double dilutions of isotype IgG mixture are diluted in saturation buffer (50 μg per well) and added as a standard curve (mixture of mouse monoclonal antibodies, IgG1, IgG2a and IgG2b from Sigma, starting at 200 ng / ml and placing in column A), and serum samples (starting in 1/1000 dilution and placing it in columns B to H), incubate for 1 hr 30 min at 37 ° C. The plates are then rinsed (x3) with rinse regulator (PBS, 0.1% polyoxyethylene sorbitan monolaurate (TWEEN 20)). Then, biotinylated goat anti-mouse IgG (Amersham) diluted in 1/5000 in saturation buffer is incubated (50 μl / well) for 1 hr 30 min. , at 37 ° C. After 3 rinses, and the subsequent addition of streptavidin-horseradish peroxidase conjugate (Amersham), plates are rinsed 5 times and incubated for 20 minutes at room temperature with 50 μl / revelation buffer cavity (OPDA 0.4 mg / ml (Sigma) and H2O2, 0.03% in 50 mM citrate buffer pH 4.5). The detection is stopped by adding 50 μl / cavity of H2SO42N. Optical densities are read at 492 and 630 nm when using the Biorad 3550 immuno-receptor. Antibody concentrations are calculated by the 4-parameter mathematical method using SoftMaxPro software. Inhibition assay for the measurement of LA2-like antibody concentrations from serum to lipo-OspA The antibody concentrations in the vaccines are studied with respect to their specificity similar to LA2. LA2 is a murine monoclonal antibody that recognizes a conformational OspA epitope on the surface of the bacterium and has been shown to be capable of killing B. burgdorferi in vitro, as well as to protect mice against a change with spirochete grown in the laboratory ( Schaible UE et al., 1990, Proc Nati Acad Sci USA 87: 3768-3772). In addition, mab LA-2 has been shown to correlate with bacterial antibodies, and studies in human serum also showed a good correlation between total anti-OspA IgG concentrations and LA-2 concentrations (as measured by ELISA) . The Maxisorp Nunc immunoplates are coated overnight at 4 ° C with 50 μl / 0.5 μg / ml cavity of lipo-OspA diluted in PBS. Free sites are blocked with saturation buffer for 1 hr at 37 ° C with (100 μl / saturation buffer cavity, PBS / 1% BSA / 0.1% Tween 20/4% NBS). Serial double dilutions of LA2 monoclonal AB (mAb) starting at 4 μg / ml were diluted in a saturation buffer (50 μl per well) to form a standard curve. Dilutions of the serum samples from the vaccines (starting at a 1/0 dilution) were also added and the plates were incubated for 2 hrs at 37 ° C. The plates are rinsed after incubation 3 times with PBS / TWEEN 20 (0.1%). The LA2 peroxidase-mAb conjugate (1/1000) diluted in saturation buffer was added to each well (50 μl / well) and incubated for 1 hr at 37 ° C. After 5 rinsings, the plates are incubated for 20 min at room temperature (in the dark) with 50 μl / revelation buffer cavity (0.4 mg / ml OPDA and 0.03% H2O2 in 50 mM citrate buffer pH 4.5). The reaction and color formation are stopped with 2N H2SO4. Optical densities are read at 492 and 630 nm using the Biorad 3550 immuno-receptor. LA2-like Ab concentrations are calculated by the 4-parameter mathematical method using SoftMaxPro software. LA2-like antibody concentrations were determined by comparison with the standard curve. CpG results as well as QS21 significantly improve intranasal booster injection of systemic antibodies to Lipo-OspA. Furthermore, when both adjuvants are combined, a synergistic effect in those responses is clearly demonstrated, especially in terms of LA2 antibodies. The humoral responses produced in the presence of QS21 and CpG are significantly higher than those induced by the injection of parenteral reinforcement. Taken together, these results clearly show the potential of intranasal formulations by combining a lytic saponin and an immunostimulator. EXAMPLE 2: Synergistic combination of QS21 and CpG to increase intranasal booster injection of systemic antibodies to influenza virus. In this example, it is investigated whether hemolytic sapins such as QS21 (see example) and immunostimulators such as CpG are capable of synergistically increasing intranasal booster injection of systemic antibodies in mice loaded intranasally with inactivated complete influenza virus. Female Balb / c mice (10 animals per group), 8 weeks old, were loaded intranasally with trivalent complete influenza virus, inactive with β-propiolactone (A / Beijing / 262/95; A / Johannesburg / 33/94; B / Panama / 45/90; 5 μg DE 5A / strain) to mimic the load of natural onset occurring in humans. After 28 days, the mice were injected with intranasal reinforcement (under anesthesia) with 20 μl of solution (10 μl through the nostril, supplied as drops per test piece) containing 1.5 μg of HA / trivalent complete influenza virus strain, inactive with β-propiolactone either A: PBS; B: 50 μg of CpG (TCC TQG TTT TGT CGT TTT GTC GTT, Krieg 2006); C: 4.5 μg of QS21 (obtained from Cambridge Biotech, USA); D: 50 μg of CpG + 4.5 μg of QS21; or, E: by intramuscular injection of 1.5 μg HA / divided influenza virus strain (same strains as in the initial loading immunization). The Flu antigens were supplied by the manufacturer SSD G BH (Dresden, Germany).
Figures 3 and 4 show the specific IgG concentrations of the Flu Flu strain and the Hemagglutination Inhibition (HAI) concentrations 14 days after the nasal booster injection. Elisa methods for measurement of anti-influenza IgG concentrations in mice: Maxisorp Nunc immunoplates are coated overnight at 4 ° C with 50 μl / 1 μg / ml influenza virus antigen in PBS (in the columns B to H of the plate), or with 50 μl of 5 μg / ml of purified goat anti-mouse Ig (Boerhinger), in PBS (column A). The free sites on the plates are blocked (1 hour, 37 ° C) using the saturation regulator; PBS containing 1% BSA, 0.1% polyoxyethylene sorbitan monolaurate (TWEEN 20), and 4% Normal Bovine Serum (NBS). Then, serial double dilutions of isotype IgG mixture are diluted in saturation buffer (50 μg per well) and added as a standard curve (mixture of mouse monoclonal antibodies, IgG1, IgG2a and IgG2b from Sigma, starting at 200 ng / ml and placing in column A), and serum samples (starting in 1/1000 dilution and placing it in columns B to H), are incubated for 1 hr 30 min at 37 ° C. The plates are then rinsed (x3) with rinse regulator (PBS, 0.1% polyoxyethylene sorbitan monolaurate (TWEEN 20)). Then, biotinylated goat anti-mouse IgG (Amersham) diluted in 1/5000 in saturation buffer is incubated (50 μl / well) for 1 hr 30 min. , at 37 ° C. After 3 rinses, and the subsequent addition of streptavidin-horseradish peroxidase conjugate (Amersham), plates are rinsed 5 times and incubated for 20 minutes at room temperature with 50 μl / revelation buffer cavity (OPDA 0.4 mg / ml (Sigma) and H2O2, 0.03% in 50 mM citrate buffer pH 4.5). The detection is stopped by adding 50 μl / cavity of H2SO42N. The optical densities are read at 492 and 630 nm when using the Biorad 3550 immuno-receptor. Antibody concentrations are calculated by the 4-parameter mathematical method using the SoftMaxPro software. The complete influenza virus for coating (strain A / Beijing / 262/95), inactive with β-propiolactone (BPL), is supplied by the manufacturer SSD GmBH (Dresden, Germany). HemAglutination Inhibition Activity (HAI) of Flu-specific serum Abs in mice Sera (25 μl) is first treated for 20 minutes at room temperature (RT) with 100 μl of 0.5M borate buffer (pH 9) and 125 μl of kaolin purchased from Dade Behring. After centrifugation (30 minutes, 3000 RPM or 860 g), 1 00 μl of supernatant (corresponding to a dilution of 1/10 of the serum) are taken and incubated for 1 hour at 4 ° C with 0.5% blood cells. red lines of chicken. The supernatant is collected after centrifugation for 10 minutes at 3200 RPM (970 g). Both operations are done to eliminate the haemagglutination factors contained in the sera. Then, 25 μl of treated sera are diluted in 25 μl of PBS (serial double dilutions starting at 1/20) in Greiner 96-well plates. The inactive complete BPL virus is added (25 μl / well) at a concentration of 4 units of haemagglutination (ie, at a dilution that is 4 times lower than the last one that causes agglutination of red blood cells) for 30 minutes at RT under agitation. Red chicken blood cells are then added (25 μl / well) for 1 hour at RT. The plates are finally maintained overnight at 4 ° C before being read. The concentration of HAI corresponds to the last dilution of serum that inhibits the hemagglutination induced by virus. CpG results as well as QS21 do not improve the injection of intranasal reinforcement of IgG or antibodies of HAI to Flu strains. However, when both adjuvants are combined, a synergistic effect in those responses is clearly demonstrated. The HAI responses produced in the presence of QS21 and CpG are still similar to those induced by the injection of parenteral reinforcement. These results confirm the potential of intranasal formulations that combine hemolytic saponin in this context (Krieg 2006 in the present and Krieg 1826 in examples 3 and 5). EXAMPLE 3: Synergistic combination of β-Escin and CpG to increase intranasal booster injection of systemic antibodies to Lipo-OspA We appreciate in this example the possibility that a synergy similar to that observed between QS21 and CpG could be obtained with other hemolytic saponins (see example) such as ß-Escin. The non-hemolytic saponin, glycyrrhizic acid, is also tested. The female Balb / c mice (6 animals per group), 8 weeks old, were loaded intramuscularly with lipo-OspA (1 μg) formulated in alum (50 μg). After 3 months, the mice were injected intranasally (under anesthesia) with 10 μl of solution (5 μl through the nostril, supplied as drops per tube) containing 5 μg of lipoprotein. OspA in either A: PBS; B: 50 μg of CpG 1001 (TCC ATG AGC TTC CTG ACG TT, Krieg 1826); C: 5 μg of β-Escin (purchased from Sigma); D: 50 μg of CpG 1001 + 5 μg of β-Escin; or, E: 5 μg glycyrrhizic acid (purchased from Sigma); F: 50 μg CpG 1001 + 5 μg glycyrrhizic acid or, G: by intramuscular injection of 1 μg of lipo-OspA absorbed in alum (50 μg). Figure 5 shows the LA2-specific concentrations of OspA 14 days after the nasal booster injection. Methods The methods are the same as those detailed in Example 1. Results ß-Escin and CpG act synergistically to increase intranasal booster injection of systemic Abs LA2. This combination produces more high Ab responses than the injection of parenteral reinforcement. On the other hand, such synergy is not obtained by combining CpG with glycyrrhizic acid. These results and the previous ones in this patent taken together show the ability of CpG and different hemolytic saponins to adjuvant immune responses in a synergistic manner. EXAMPLE 4: Immunogenicity studies using P. falciparum RTS.S and HIV-1 gp 120 formulated with CpG and / or DQS21 1. Profile of the experiment Two mouse immunogenicity studies are conducted to evaluate the potential additive or synergistic effects of oligonucleotides of CpG (CpG) and QS21. Groups of mice were immunized with RTS.S and gp120 formulated with CpG and QS21 alone or in combination. These adjuvant combinations are also tested in the presence of the vehicle AI (OH) 3 or an oil in water emulsion (o / p). The immunogenicity of the formulations is examined after two parenteral immunizations. The sera were analyzed for the presence of antigen-specific antibodies, and for the distribution of antibody isotypes. The spleen cells were used to evaluate the immune responses mediated by cell. Those cells were tested for the presence of cytotoxic T lymphocytes (CTL) and lymphoproliferative cells (lymphoproliferation). Table 1: Groups of mice in experiment 1 Table 2: Groups of mice in experiment 2 2. Formulation 2.1 Experiment 1 The formulations were prepared three days before each injection. When needed, RTS, S (10 μg) and gp 120 (10 μg) were absorbed in 100 μg of AL (OH) 3. When needed, MPL (5 μg) was added and incubated 30 minutes before addition of the regulator as a mixture of PBS 10 times concentrated pH 7.4 and H2O expected by the group without DQ for which the regulator was PO4l NaCl 10/150 pH 6.8. After 30 min, if needed, QS21 (5 μg) mixed with liposomes in a weight ratio QS21 / cholesterol of 1/5 (referred to as DQ) is added to the formulation. Thirty minutes later, for the oligo formulations, 100 μg of CpG was added 30 min before the addition of 50 μg / ml of thiomersal as a preservative. AI (OH) 2 + RTSS + gp120 + 1h-MPL-30 min-premix-30-min-DQ-30 min-CpG-30 min-Thio All combinations are carried out at room temperature with stirring. 2.2 Experiment 2 Formulation process: The formulations are performed simultaneously for both injections. The injection volume for a mouse is 100 μl. Fifty μg / ml of thiomersal are added as preservatives. Group 1: RTS.S (10 μg) and gp120 (10 μg) are diluted with H2O and PBS pH 6.8 for isotonicity. After 5 min, the formulation is absorbed in CpG 1856 (100 μg). Group 2: RTS.S (10 μg) and gp120 (10 μg) are diluted with H2O and PBS pH 7.4 for isotonicity. After 30 min, RTS.S and gp120 are absorbed in DQ (5 μg). After 30 min. absorption, the formulation is absorbed in CpG 1856 (100 μg). Group 3: RTS.S (10 μg) and gp120 (10 μg) are diluted with H2O and PBS pH 6.8 for isotonicity. After 5 min, the formulation is absorbed in an o / p emulsion. After 5 min of absorption, the formulation is absorbed in QS21 (5 μg) before the addition of CpG (1 00 μg) -3. Immunological methods Nine mice (Balb / C x C57B1 / 6) F1 per group received in the plants of the occult feet 2 x 50 μl vaccines twice at an interval of two weeks. Two weeks later the sera were obtained to assess the antibody responses, and the spleen cells were collected to determine the immune responses mediated by cell. For analysis of lymphoproliferation, the cells were seeded in quadriplicates in microconcentration plates of round bottom of 96 cavities in a concentration of 2x106 per ml. The cells were cultured for 72 or 96 hrs in RPMI-1640 supplemented with antibiotics, glutamine and 1% (v / v) of normal mouse serum in the presence of different concentrations of RTS.S or gp120 antigen. Control cells were cultured without antigen. The cells were then propelled overnight with 1 μCi / [3 H] -thymidia cavity, harvested and the incorporated radioactivity was determined in a beta counter. The results are expressed as average accounts per minute (cpm). For CTL analysis the cells were cultured for 7 days in 6-well plates in the presence of 10 μg per ml of synthetic peptide pCM1003 (IPQSLDSWWTSL) which corresponds to a CTL epitope HbsAfg (Schirmbeck et al., 1995) or peptide pCM1007 (GIHIGPGRAFYAARK) representing an epitope of CTL gp120 (Casement et al., 1995). At the end of the culture period, the effector cells are titrated in duplicate for the specific cytolytic activity of HBsAg in standard [51 Cr] release analysis using P815 S-transfected and control cells. The specific cytotoxicity of gp120 is determined using P81 target cells 5 that were either left untreated or boosted for 1 hr with peptide pCM1007. The minimum and maximum release was determined with target cells without effector cells and by the addition of 3% (v / v) Triton X-100, respectively. The results are expressed as% [51 Cr] release (cpm culture experiment-cpm Spontaneous Release / cp, maximum release - cpm spontaneous release). The concentration and isotyping of pooled sera was performed in a standard enzyme-linked immunosorbent assay (ELISA) format using plates coated with HbsAg. The sera were diluted in PBS / BSA starting at 1: 400. The specific specific biotinylated antibodies for Ig or the IgG1 isotypes, IgG2 and IgG2 followed by a conjugate of horseradish peroxidase-streptavidin were used for the detection of bound antibodies. ELISA concentrations are calculated from a reference by SoftmaxPro and expressed in ELISA units (EU / ml). The concentrations of gp120-specific antibody is determined in a standard ELISA using plates coated with gp120 protein. The sera were diluted in PBS / Tween20 / BSA starting at 1: 100. Biotinylated secondary antibodies specific for Ig or the isotypes lgG1, IgG2 and IgG2 followed by a conjugate of horseradish peroxidase-streptavidin were used for the detection of bound antibodies . The concentrations were calculated in relation to a standard mouse Ig and expressed as μg / ml. 4. Results Experiment 1 In analysis of lymphoproliferation responses did not show any significant difference in reactivity to RTS, S between groups. In contrast, groups 1 and 3 containing both CpG and DQS21 showed better gp120-specific lymphoproliferation responses than groups containing CpG or DQS21 alone (Figure 6). In this experiment only specific CTL of HBsAg were used. There was no pronounced difference in CTL induction between groups 1 and 2 that had received CpG and QS21 in combination and groups 2 and 4 immunized with only one of the two adjuvant components, while the presence of AI (OH) 3 decreased the CTL activity observed for the combination of CpG and DQS21 in group (Figure 7). However, there is a trend that CpG and DQS21 was better than DQS21 alone, and the combination induced more CTL in the presence of AI (OH) 3 than CpG alone (Figure 7). The humoral immune response of the mice was examined only by the presence of antibodies specific for HBsAg. The concentrations were similar in all groups except group 3, which showed an approximately triple increase, demonstrating that, in the presence of AI (OH) 3, the combination of DQS21 and CpG is more immunogenic than CpG alone (Figure 8). The isotype distribution was similar to groups 3 and 4 containing AI (OH) 3, whereas in the absence of AI (OH) 3 the combination of CpG and DQS21 induced a isotype pattern similar to T, which DQS21 alone ( Figure 8). Experiment 2 The specific lymphoproliferation responses for RTS.S, and gp120 were very similar in this experiment. The data indicate that the addition of DQS21 (either alone or with an o / p emulsion) increases the lymphoproliferation responses to both antigens (Figure 9). The CTL responses were evaluated using both a CTL epitope peptide gp1 20 and HBsAg. In both cases, CTL could be detected after immunization of group 1 with CpG alone (Figure 10). However, the addition of DQS21 resulted in a considerable increase in CTL for both antigens (Figure 10). The presence of an o / p emulsion either neutralized the positive effect of DQS21 (gp120) or increased the background of the in vitro analysis (HBsAg). Antibody responses to HBsAg and gp120 were increased by the addition of DQS21 to the CpG adjuvant (Figure 11A). A further increase is observed when a w / p emulsion is included in the formulation (Figure 1 1 A). The addition of DQS21 to CpG changed the gp120 isotype profiles towards a more pronounced TH deviation (Figure 1 1 B), whereas the impact of the HBsAg isotype profiles was less pronounced in this experiment. 5. Conclusions Immunization with RTS.S and gp120 was formulated with the combination of CpG and DQS21 results in strong antigen-specific immune responses. The combination of adjuvant components CpG and DQS21 - increases lymphoproliferation responses increases CTL activity - increases antibody concentrations and isotype patterns of TH .. compared to single components. EXAMPLE 5: Therapeutic Potential of CpG and / or DQS21 Formulations in TC1 Tumor Model 1. Experimental Design Four groups of 10 C57b1 / 6 mice received 10e6 (200 μl) TC1 cells (tumor cells expressing E7) subcutaneously on day 0 in free space. Mice were vaccinated twice on day 14 and 21 after tumor change, with 5 μg of PD1 / 3E7 H PV1 6 formulated injected into the skin plant. The growth of the tumor is measured individually twice a week. Groups of mice: 1. unvaccinated 2. PDI / 3E7 + CPG (10 μg ODN 2006) 3. PDI / 3E7 + DQS21 (0.5 μg) 4. PDI / 3E7 + CPG + DQS21 Tumor growth is monitored when measuring individual tumors, twice per week. 2. Formulations The formulations were made on the days of injection. The injection volume for a mouse was 100 μl. When needed, PD1 / E37 (5 μg) were diluted with H2O and PBS pH 7.4 for isotonicity. After 5 min, if needed QS21 (0.5 μg) mixed with liposomes in a weight ratio of QS21 / cholesterol of 1/5 (referred to as DQ) is added to the formulation, 30 min later, for the formulation with the oligo , 1 μg of CpG (ODN 2006) were added 30 min before the addition of 1 μg / ml thio ersal as conservative. H2O + PBS pH 7.4 + PD1 / 3E7 - 5 m, "+ DQ - 30 m, n + CpG - 3o m - Thio 3. Results The evolution of the average tumor growth by groups of 1 0 animals with time are shown in Figure 1 2. 1 00% of the animals that received a tumor change from 10e6 TC1 cells progressively developed growing tumor. 70-80% of the unvaccinated animals or animals vaccinated with the E7 protein in DQS21 died by day 35.
Two vaccinations with the E7 protein formulated in DQS21 had almost no effect on tumor growth. In contrast, 2 vaccinations, IFP (day 14, 21) with 5 μg of ProtD 1/3 E7 HVP16 in adjuvant CPG induced regression of these pre-established tumors and protect the mice from dying: 70-80% of the mice were still alive on day 35. The combination of the 2 immunostimulants CPG and DQS21 showed a beneficial libero effect on the CpG used alone.
LIST OF SEQUENCES < 110 > Friede, Martin Gar with. Nathalie Hermand Pt-ilippe < 120 > ADJUVANT COMPOSITION COMPRISING SAPONINE AND AN IMMUNOSTIMULATOR OLIGONUCLEOTIDE < 130 > B45181 < 160 > 5 < 170 > FastSEQ for Windows Version 3.0 * < 210 > 1 < 211 > 20 < 212 > DNA < 213 > Human < 400 > 1 tccatgacgt tcetgacgtt 20 < 210 > 2 < 211 > 18 < 212 > DNA < 213 > Human < 400 > 2 tc cccagcg tgcgccat 18 < 210 > 3 < 211 > 30 < 212 > DNA < 213 > Human < 400 > 3 accgatgacg tcgccggtga c? Gcaccacg 30 < 2X0 > 4 < 211 > 24 < 212 > DNA < 213 > Human < 400 > 4 tcgtcgtttt g cg tttgt cgtt 24 < 210 > 5 < 211 > 20 < 212 > DNA < 213 > Human < 400 > 5 tccatgacgt tcctgatgct 20

Claims (34)

  1. CLAIMS 1. An adjuvant composition comprising a saponin and an immunostimulatory oligonucleotide, wherein said immunostimulatory oligonucleotide comprises a Purine, Purine, C, G, pyrimidine, pyrimidine sequence.
  2. 2. An adjuvant composition comprising a saponin and an immunostimulatory oligonucleotide, wherein said immunostimulatory oligonucleotide is selected from the group comprising: TCC ATG ACG TTC CTG ACG TT (SEQ ID NO: 1); TCT CCC AGC GTG CGC CAT (SEQ ID NO: 2), ACC GAT GAC GTC GCC GGT GAC GGC ACC ACG (SEQ ID NO: 3); TCG TCG TTT TGT CGT TTT GTC GTT (SEQ ID NO: 4), TCC ATG ACG TTC CTG ATG CT (SEQ ID NO: 5).
  3. 3. An adjuvant composition comprising a saponin and an immunostimulatory oligonucleotide, wherein the immunostimulatory oligonucleotide contains at least two unmethylated GC repeats that are separated by at least 3 nucleotides.
  4. 4. An adjuvant composition according to claim 3, characterized in that the immunostimulatory oligonucleotide contains at least two unmethylated GC repeats that are separated by 6 nucleotides.
  5. 5. An adjuvant composition according to any of claims 1 to 4, characterized in that the saponin is derived from quilA.
  6. 6. An adjuvant composition according to claim 5, characterized in that the quilA derivative is QS21.
  7. 7. An adjuvant composition according to claims 5 or 6, characterized in that it additionally comprises a vehicle.
  8. An adjuvant composition according to claim 7, characterized in that the carrier is a particulate carrier selected from the group comprising an oil in water emulsion, a cholesterol containing liposome or alum.
  9. 9. An adjuvant composition comprising a saponin and an immunostimulatory oligonucleotide containing unmethylated CpG dinucleotide, wherein the saponin is in the form of a liposome.
  10. 10. An adjuvant composition comprising a saponin and an immunostimulatory oligonucleotide containing unmethylated CpG dinucleotide, wherein the saponin is in the form of an oil-in-water emulsion. eleven .
  11. An adjuvant composition according to claims 9 and 11, characterized in that it also comprises a metal salt particle.
  12. 12. An adjuvant composition according to claim 1, characterized in that the metal salt particle is aluminum hydroxide or aluminum phosphate.
  13. 1 3. A vaccine composition comprising an adjuvant composition as claimed in any of claims 1 to 12, further comprising an antigen.
  14. 14. A vaccine composition according to claim 13, characterized in that said antigen is derived from an organism selected from the group comprising: Immunodeficiency Virus Humana, Varicella-Zoster Virus, Simple Herpes Virus type 1, virus Simple herpes type 2, human cytomegalovirus, dengue virus, Hepatitis A, B, C or E, respiratory virus Sincital, human papilloma virus, influenza virus, Hib, meningitis virus, Salmonella, Neisseria, Borrelia, Chlamydia, Bordetella, Streptococcus, Mycoplasma, Mycobacteria, Haemophilus, Plasmodium or Toxoplasma, Depeptide of Stanworth; antigens associated with Tumor (TAA), MAGE, BAGE, GAGE, MUC-1, Her-2 neu, CEA, PSA, KSA, or PRAME; or a self-peptide hormone, GnRH. 5.
  15. A vaccine composition according to claim 13, characterized in that said antigen is derived from the group comprising (a) antigens associated with tumor PSMA, PSCA, tyrosinase, survivin, NY-ESO1, prostase, PS108, RAGE, LAGE, HAGE.; (b) or the N-terminal 39-43 amino acid fragment (Abeta) of the amyloid precursor protein; (c) or antigens associated with atherosclerosis.
  16. 16. A vaccine composition according to claims 13 to 15, characterized in that the vaccine is administered systemically.
  17. 17. A vaccine composition according to claims 13 to 15, characterized in that the vaccine is administered mucosally.
  18. 1 8. A vaccine composition according to claim 1 7, characterized in that the saponin of the adjuvant composition is haemolytic.
  19. 19. A delivery device pre-filled with the vaccine of claims 13 to 15, said device being designed to administer the vaccine systemically.
  20. 20. A method for inducing an immune response in an individual, comprising the systemic administration of a safe and effective amount of the vaccine composition according to claims 1 to 15.
  21. A method of treating an individual suffering from a disease by administering to an individual an effective amount of the vaccine as claimed in any of claims 13 to 18.
  22. 22. A method for preventing an individual from contracting a disease that comprises administering to said individual an effective amount of the vaccine as claimed in any of claims 13 to 18.
  23. 23. A method according to claims 21 to 22, characterized in that the disease is selected from the group comprising prostate, breast, colorectal, lung, pancreatic, renal, ovarian or melanoma, chronic disorders without cancer, allergy, Alzheimer's, atherosclerosis.
  24. 24. A method of treatment according to claims 21 to 23, characterized in that the administration of the vaccine is through a systemic route.
  25. 25. A method of treatment according to claims 23 and 24, characterized in that the vaccine is administered through a systemic route.
  26. 26. A vaccine according to claim 1 3 or 1 5 for use as a medicament.
  27. 27. The use of a combination of a saponin and a CpG molecule according to claims 1 to 6 in the manufacture of a vaccine for the prophylaxis and treatment of viral, bacterial, parasitic, allergy, cancer or other chronic disorders.
  28. 28. The use of a combination of a saponin, an immunostimulatory oligonucleotide and a vehicle according to claims 7, 8, 11 and 12 in the manufacture of a vaccine for the prophylaxis and treatment of viral, bacterial, parasitic, allergic, cancer or other chronic disorders.
  29. 29. A method for inducing a systemic antigen-specific immune response in a mammal, comprising administering to a mucosal surface of said mammal a composition comprising an antigen and a hemolytic saponin and a CpG molecule. C
  30. 30. A method for making an adjuvant composition according to claims 1 to 6 comprising mixing a saponin with an immunostimulatory oligonucleotide.
  31. 31 A method for making an adjuvant composition according to claims 7, 8, 11 and 12 comprising mixing a saponin, a non-stimulatory oligonucleotide and a vehicle.
  32. 32. A method for making a vaccine comprising mixing the following (a) a saponin, (b) an immunostimulatory oligonucleotide, and (c) an antigen, wherein the saponin is in the form of a liposome and wherein the oligonucleotide contains CpG dinucleotides without methylation.
  33. 33. A method for making a vaccine comprising mixing the following (a) a saponin, (b) an immunostimulatory oligonucleotide, and (c) an antigen, wherein the saponin is in the form of an oil-in-water emulsion and wherein the oligonucleotide contains unmethylated CpG dinucleotides.
  34. 34. The method for making a vaccine according to claim 28, further comprises mixing a vehicle.
MXPA/A/2001/010654A 1999-04-19 2001-10-19 Vaccines MXPA01010654A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9908885.8 1999-04-19
US09301829 1999-04-29

Publications (1)

Publication Number Publication Date
MXPA01010654A true MXPA01010654A (en) 2002-06-05

Family

ID=

Similar Documents

Publication Publication Date Title
CA2370697C (en) Adjuvant composition comprising saponin and an immunostimulatory oligonucleotide
US6558670B1 (en) Vaccine adjuvants
EP1432442B1 (en) The use of liposomes containing saponins and sterols in the manufacture of intradermal vaccines
KR20030044016A (en) Adjuvant composition comprising an immunostimulatory oligonucleotide and a tocol
JP2007508272A (en) Vaccine composition comprising interleukin 18 and saponin adjuvant system
JP5307859B2 (en) vaccine
MXPA01010654A (en) Vaccines
MXPA00009887A (en) Adjuvant compositions