MXPA01002532A - Vaccine against sexually transmitted diseases - Google Patents

Vaccine against sexually transmitted diseases

Info

Publication number
MXPA01002532A
MXPA01002532A MXPA/A/2001/002532A MXPA01002532A MXPA01002532A MX PA01002532 A MXPA01002532 A MX PA01002532A MX PA01002532 A MXPA01002532 A MX PA01002532A MX PA01002532 A MXPA01002532 A MX PA01002532A
Authority
MX
Mexico
Prior art keywords
vaccine
use according
adjuvant
disease
antigens
Prior art date
Application number
MXPA/A/2001/002532A
Other languages
Spanish (es)
Inventor
Moncef Mohamed Slaoui
Pierre G Vandepapeliere
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 MXPA01002532A publication Critical patent/MXPA01002532A/en

Links

Abstract

A method of administering a vaccine to females to prevent or treat infections associated with pathogens which cause sexually transmitted diseases is described. The vaccine comprises one or more antigens for the prevention or treatment of sexually transmitted diseases, for example an HSV glycoprotein D or an immunological fragment thereof, and an adjuvant, especially a TH-1 inducing adjuvant. The use of the vaccine components for the formulation of a vaccine composition for the prevention or treatment of sexually transmitted diseases in female subjects is also described.

Description

VACCINE AGAINST SEXUALLY TRANSMITTED DISEASES The present invention relates to one or more antigens for the prevention or treatment of sexually transmitted diseases and the use thereof in the formulation of a vaccine, for administration to human subjects, for the prevention or treatment of infections associated with pathogens. that cause sexually transmitted diseases. The invention also relates to a method of administering the vaccine to women to prevent or treat infections associated with pathogens that cause sexually transmitted diseases. Pathogens that cause sexually transmitted diseases (STDs) are known and there is an urgent need for effective vaccines to treat or prevent such conditions. Sometimes sexually transmitted diseases are caused by one or more pathogens. Therefore, combination vaccines capable of preventing and / or treating one or more STDs are also required. It has been found that certain vaccine formulations are surprisingly effective in preventing or treating STDs in human subjects women, who are susceptible to or suffer from such STDs. The present invention provides a method for treating a female human subject suffering from or susceptible to one or more sexually transmitted diseases (STDs), such method comprising administering to a subject woman in need thereof an effective amount of a vaccine formulation. comprising one or more antigens J & Z m ... > < m * & *, ... ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^ Preferably the adjuvant is an adjuvant that induces TH-1. In a related aspect, the invention provides the use of one or more antigens derived from or associated with a pathogen causing STD and an adjuvant, especially an adjuvant that induces TH-1, in the preparation of a vaccine for administration to a subject human woman for the prevention and / or treatment of one or more STDs. Examples of antigens derived from or associated with a pathogen causing STD include those derived from or associated with herpes viruses (HSV-1 and HSV-2), human papillomarviruses (all types of HPV), Chlamydia trachomatis, Neiserria gonorrhea, Treponema pallidum (syphilis) and Haemophilus ducreyi (chancre). Other sources of antigens including recombinant bacteria, recombinant viruses, fusion proteins, peptides and mimotopes can also be used. The above list is not exhaustive and other pathogens are well known to medical practitioners and other experts in the field and are listed in standard textbooks. Suitable adjuvants in the invention include those well known in the field of vaccine formulation. By "adjuvant inducing TH-1" is meant an adjuvant which is a preferential stimulator of cellular response TH1. A recoed signal that a TH1 response has been :, ¿T¿asfe * ¿t- .- * t * i * j-i ui ~ ^ Ád¿i & stimulated is the increased production of TH1 type cytokines, for example, IFN-? and IL-2. The secretion of IFN-? It is associated with protective responses against intracellular pathogens, including parasites, bacteria and viruses. The activation of leukocytes by IFN-? increases the death of intracellular pathogens and increases the expression of Fe receptors. Direct cytotoxicity can also occur, especially in synergism with lymphotoxin (another product of TH1 cells). IFN-? it is also both an inducer and a product of NK cells, which are major innate effectors of protection. The answers of type TH1, already through IFN-? or other mechanisms, provide preferential support for murine IgG2a immunoglobulin isotypes. In contrast, TH-2 type responses are associated with humoral mechanisms and the secretion of IL-4, IL-5, IL-6, IL-10 and beta-tumor necrosis factor. Adjuvants that are capable of preferential stimulation of the TH1 cellular response are described in International Patent Applications Nos. WO 94/00153 and WO 95/17209. Lipid A 3 Acylated De-O-monophosphoryl (3D-PL) is an adjuvant. It is known from GB 222021 1 (Ribi). Chemically it is a mixture of lipid A 3 De-O monophosphoryl acylated with 4, 5 or 6 acylated chains and manufactured by Ribi Immunochem Montana. A preferred "small particle" form of acylated 3 De-O-monophosphoryl lipid is described in EP 0 689 454BI (SmithKine Beecham Biologicals SA). In such a 'small particle' 3-DMPL, the 3D-MPL particles are small enough to filter sterile through a 0.22 micron membrane (as described in the European Patent 0 Another preferred adjuvant that can be used in the present invention comprises QS21, a non-toxic purified fraction of Hplc derived from the bark of Quillaja Saponaria Molina. Optionally it can be mixed with 3 De-O acylated monophosphoryl lipid (3D-DMPL), optionally together with a carrier. The production method of QS21 is described (as QS21) in the U.S. Patent. No. 5,057,540 and is available from Auilla 0 Pharmaceuticals. Non-reactogenic adjuvant formulations containing QS21 have been previously described (WO 96/33739). It has been shown that such formulations comprising QS21 and cholesterol are adjuvants that stimulate successful TH1 when formulated together with an antigen. In this manner, the vaccine compositions forming part of the present invention can include a combination of QS21 and cholesterol. Preferential adjuvants that are preferential stimulators of TH 1 cellular response include oligonucleotides or immunomodulators, for example, unmethylated CpG sequences as described in WO 96/02555. The combinations of different adjuvants that stimulate TH1, such as those mentioned hereinbefore, are also contemplated as providing an adjuvant which is a preferential cell response stimulator TH1. For example, QS21 ÉmS? St3! AÁÍ A-BSife m. - ^^ g¡m ^ g ^^^^^^^^ ^^^^ can be formulated together with 3D-MPL. The ratio of QS21: 3D-MPL will typically be in the order of 1: 1 0 to 10: 1, preferably 1: 5 to 5: 1 and often substantially 1: 1. The preferred range for optimal synergy is 2.5: 1 to 1: 1 of 3D MPL: QS21. Preferably, also a vehicle is present in the vaccine composition according to the invention. The vehicle can be an oil in water emulsion, or an aluminum salt. Other mineral salts can also be used as a vehicle such as calcium, iron or zinc salts. Other vehicles include polyphosphazene, liposomes and ISCOMS. The non-toxic oil in water emulsions preferably contains a non-toxic oil, for example, squalene or squalane, an emulsifier, for example, Tween 80, in an aqueous vehicle. The aqueous vehicle can be, for example, phosphate buffered saline. An oil Preferred in water emulsion comprises a metabolisable oil, such as squalene, alpha tocopherol and Tween 80. Additionally the water emulsion oil may contain space 85 and / or lecithin. Typically for human administration, QS21 and 3D MPL will be present in a vaccine in the range of 1 μg - 500 μg, such as -100 μg, preferably 10 μg-50 μg per dose. Typically, the oil in water will comprise 2 to 10% squalene, 2 to 10% alpha tocopherol and 0.3 to 3% Tween 80. Preferably, the squalene: alpha tocopherol ratio is equal to or less than 1 since provides a more stable emulsion. Space 85 can also be present in a level of 1%. In some cases it may be advantageous for the vaccines of present invention further contain a stabilizer. A particularly potent adjuvant formulation including QS21, 3D-MPL and tocopherol in a water emulsion oil is described in WO 95/1721 0. In a preferred aspect aluminum (alum) or aluminum phosphate hydroxide will be included in the composition of vaccine that is used or manufactured according to the invention. In a particularly preferred aspect, the antigens in the vaccine composition used or manufactured according to the invention are combined with 3D-MPL and alum. The vaccines employed in the present invention can, if desired, comprise adjuvant molecules of the general formula (I): HO (CH2CH2O) nAR where, n is 1 -50, A is a bond or -C (O) -, R is C? -50 alkyl or C? -50 phenyl alkyl. One embodiment of the present invention consists of a vaccine formulation comprising a polyoxyethylene ether of the general formula (I), wherein n is between 1 and 50, preferably 4-24, more preferably 9; the R component is C? -50 alkyl, preferably C2o and more preferably C12 alkyl, and A is a bond. The concentration of the polyoxyethylene ethers should be in the range of 0.1-20%, preferably 0.1-10%, and more preferably in the 0.1-1% range. Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether, polyoxyethylene-9-stearyl ether, polyoxyethylene-8-stearyl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether and polyoxyethylene-23-lauryl ether. Polyoxyethylene ethers such as polyoxyethylene lauryl ether are described in the Merck index (ed 12 {, Entry 7717). HSV-2 is the primary etiologic agent of genital herpes. HSV-1 is the causative agent of genital herpes. Together, these viruses are characterized by their ability to induce both acute diseases and to establish a latent infection, mainly in cells of neuronal ganglia. WO 92/16231 provides additional background information about genital herpes and describes a vaccine that can be used to treat people susceptible to HSV infections comprising HSV glycoprotein D or an immunological fragment thereof in conjunction with lipid A 3 Dehydrogenase. Acylated monophosphoryl and a suitable vehicle. The specification of WO 92/16231 provides details of the glycoprotein D, immunological fragments thereof, and 3-DMPL and methods for obtaining it. The specification describes some promising tests of a candidate vaccine in animal models but no data are given in humans. In a preferred aspect the method or use according to the invention relates to the prevention or treatment of infections associated with genital herpes, in particular HSV-2 infections. The vaccine that can be used in the present invention comprises glycoprotein D or an immunological fragment thereof which is Typically HSV-2 Glycoprotein D is located in the viral membrane, and is also found in the cytoplasm of infected cells (Eisenberg R.J. et al., J. of Virol 1 980 35 428-435). It comprises 393 amino acids that include a signal peptide and has a molecular weight of approximately 60 kD. Of all the proteins that develop HSV, this is probably the best characterized (Cohen et al., J. Virology 60 157-166). In vivo, it is known to play a central role in viral binding to cell membranes. In addition, it has been shown that glycoprotein D is capable of producing neutralizing antibodies in vivo (Eing ef al., J. Med. Virology 127: 59-65). However, the HSV-2 virus can still be reactivated and induce the recurrence of the disease despite the presence of high neutralizing antibody concentrations in the patient area. As described in WO 92/16231, a preferred embodiment thereof is a HSV-2 glycoprotein of 308 amino acids comprising amino acids 1 to 306 of the glycoprotein that occurs naturally with the addition of aparagin and glutamine at the end. C terminal of the free truncated protein of its membrane support region. This form of the protein includes the signal peptide that divides to produce a mature 283 amino acid protein. The production of such protein in Chinese Hamster Ovary cells has been described in EP-B-139 417. The mature truncate preferably used in the vaccine formulation within the scope of the invention can be designated recombinant gD2t (rgD2t) or simply (as in the present below) gD2t. The HSV antigen can be conjugated chemically or otherwise to a particulate carrier as described in WO 92/16231. In a preferred aspect the vaccine for use in the invention comprises gD2t, 3-DMPL (especially 3-DMPL of small particle) and aluminum hydroxide (alum). Papilomaviruses are small DNA tumor viruses, which are highly specific species. Still, more than 70 individual human papillomavirus (HPV) genotypes have been described. HPVs are usually specific for either the skin (for example HPV-1 and -2) or mucosal surfaces (for example, HPV-6 and -1 1) and usually cause benign tumors (warts) that persist for several months or years. Such benign tumors can afflict troubled individuals but tend not to be life threatening, with a few exceptions. 15 Some HPVs are also associated with cancers. The strongest positive association between an HPV and human cancer is that it exists between HPV-16 and HPV-17 and cervical carcinoma. Cervical cancer is the most common malignancy in developing countries, with approximately 500,000 new cases occurring in the world each year. Is now It is technically feasible to actively combat primary HPV-16 infections, and even established HPV-16-containing cancers, using vaccines. For a review of the prospects for therapeutic and prophylactic vaccination against HPV-16 see Cason J., Clin. Immunother. 1994: 1 (4) 293-306 and Hagenesee M.E. Infections in Medicine 1997 14 (7) 555-556, 559-564. Preferably, a vaccine composition of according to the invention comprises the major capsid protein, the L1 protein. Currently, different types of HPVs have been isolated and characterized with the help of cloning systems in bacteria and more recently by PCR amplification. The molecular organization of HPV genomes has been defined on a comparative basis with that of type 1 well characterized bovine papillomavirus (BPV1). Although minor variations occur, all the genomes of HPVs described have at least seven previous genes, E1 to E7 and two subsequent genes L1 and L2. In addition, an upstream regulatory region harbors the regulatory sequences that appear to control most of the transcriptional events of the HPV genome. The E1 and E2 genes include viral replication and transcriptional control, respectively and have to be interrupted by viral integration. E6 and E7, and recent evidence that also involves E5, are included in the viral transformation. In HPVs included in cervical carcinoma such as HPV 16 and 18, the oncogenic process starts after the integration of viral DNA. The integration results in the inactivation of genes encoding capsid proteins L1 and L2 and the continuous installation of the overexpression of the two previous proteins E6 and E7 that will lead to the gradual loss of normal cell differentiation and the development of the carcinoma. Carcinoma of the cervix is common in women and develops through an intermediate pre-cancerous stage to invasive carcinoma that frequently leads to death. The intermediate stages of the disease are known as cervical intraepithelial neoplasia and are classified I to III in terms of increasing severity. Clinically, HPV infection of the anogenital tract of women manifests as cervical flat condylomata, the hallmark of which is the coilocytosis that predominantly affects the intermediate and superficial cells of the cervical squamous epithelium. The coilocytes that are the consequence of a cytopathic effect of the virus appear as multinucleated cells with a transparent perinuclear haloe. The epithelium thickens with abnormal keratinisation responsible for the warty appearance of the lesion. Such flat condylomata when positive for serotypes HPV 16 or 18, are high risk factors for the evolution towards cervical intraepithelial neoplasia (CIN) and carcinoma in situ (CIS) that are considered by themselves as precursor lesions of invasive cervical carcinoma. . International Patent Application No. WO 96/19496 describes variants of E6 and E7 proteins of human papilloma virus, particularly E6 / E7 fusion proteins with a deletion in both E6 and E7 proteins. These elimination fusion proteins are said to be immunogenic. Vaccines based on HPV L1 are described in WO 94/00152, WO 94/20137, WO 93/02184 and WO 94/05792. Such a vaccine may comprise an L1 antigen such as a monomer, a capsomer or a particle as a virus. Such particles may additionally comprise L2 proteins. Other HPV vaccines are based on the above proteins, such as E7 or fusion proteins such as L2-E7. In the vaccine of the invention it is preferred to use compositions comprising either an E6 or E7 protein linked to an immunological fusion pattern having T cell epitopes. In a preferred form of the invention, the immunological fusion pattern is derived from the protein D of Heamophiluos influenza B.
Preferably, the protein D derivative comprises approximately the first 1/3 of the protein, in particular approximately the first 100-1 10 amino acids of N-terminal. Accordingly, the present invention can employ fusion proteins comprising Protein D -E6 of HPV 16, Protein D-E7 of HPV 16, Protein D-E7 of HPV 18 and Protein D-E6 of HPV 18. The protein D part preferably comprises the first 1/3 of the D protein. The obligate intracellular bacterium Chlamydia trachomatis infects the mucosal epithelial cells of the conjunctiva and the urogenital tract, causing a broad spectrum of human diseases such as genital infections and trachoma that can result in long-term sequelae. Trachoma, which is endemic in several developing countries, is the world's leading cause of preventable blindness; Genital infections, which represent around 3 million cases per year in the US, annually convert 200,000 infertile women following Chlamydia salpingitis (1). Therefore, this pathogen is a significant public health problem and efforts are being made to establish a «^^^^^^^^^^^ vaccine against human Chlamydia infections. Vaccination tests performed on men and nonhuman primates using the whole organism as an immunogen give specific protection for serovariation but some of the vaccines developed more severe reactions after reinfection (2). Several studies have shown that the pathology associated with Chlamydia infection is immunologically mediated (3); Besides; it was shown that a 57 kDa purified chlamydia (Hsp60) produces a pathology similar to reinfection in previously infected animals (4,5). This observation leads to the conclusion that protection against Chlamydia trachomatis could only be achieved using a subunit vaccine. The Chlamydia trachomatis species is stereotyped in 15 serovaries that are placed in 3 serogroups: the B complex (B, Ba, D, E, L1 and L2 serovariations), the intermediate complex (F, G, K, L3 serovariations) and the C complex (serovariations A, C, H, I and J) (6). Sexually transmitted diseases (STD) are caused by serovariations D to K that cover all 3 serogroups. In this way, a chlamydia subunit vaccine against STD should protect against multiple serovaries that are more or less antigenically related. For the design of a subunit vaccine, much attention has been focused on the serotype antigen consisting of the 40 kDa main outer membrane protein (MOMP). This protein, which was shown to function in vivo as a porin (7), is present during the complete life cycle of the bacterium (8); This major surface protein is highly immunogenic in humans and animals. The MOMP displays 4 variable domains (VD) surrounded by five constant regions that are highly conserved between serovariations 5 (9, 10). In vitro and in vivo the neutralizing B-cell epitopes have been mapped in VDs (11, 12, 13, 14, 15), while the T cell epitopes have been identified in both variable and constant domains (16, 17 ). Recombinant MOMP has been expressed in E. coli by different authors (18, 19, 20); Nevertheless; Manning et al. , show that their The recombinant protein failed to react with a monoclonal antibody that recognizes an epitope of conformational MOMP (18). Immunizations with purified or recombinant MOMP followed by the heterotypic or homotypic Chlamydia test have been carried out in different animal models with variable effects in the parameters of the infection (21, 22, 23). An elegant experimental model of salpingitis has been developed in mice in which intrauterine inoculation of a human strain of Chlamydia trachomatis leads to long-term infertility (24,25). In a heterotypic test experiment, Tuffrey et al., Has shown that mucosal immunization and parenteral with rMOMP absorbed in alhydrogel reduced the severity of salpingitis and the duration of colonization of the lower genital tract, respectively. However, the preparation did not confer protection against infertility resulting from infection (23). Both cell-mediated and humoral immunity seem play a protective role in the genital diseases caused by Chlamydia trachomatis. However, Rank's group suggests that in T-cell-mediated immunity in mice it is the main immune mechanism to control chlamydial genital disease (26, 27, 28) and it has been shown that CD4 and CD8 positive T cells contribute to anti-chlamydial immunity in vivo (29,30). In one embodiment of the invention, the MOMP antigen is Serovariation 2 and is produced in E. coli by means of recombinant DNA techniques. In such circumstances the protein is produced without its signal sequence. Antigens derived from or associated with N. gonorrhea include the transfer binding protein (Tbp). Two proteins are included to make the Tbp-TbpA and TbpB complex. The DNA / protein sequence of gonococcal TbpA is described in WO 92/03467 (University of North Carolina). A recent paper that refers especially to TbpA and TbpB of gonococcus and as required for infection is Mol. Microbiol., 1998 Feb; 27 (3): 61 1 -616. Other antigens include the Por B protein, see Proc Nati Acad Sci U S A 1987 Nov; 84 (22): 8135-8139 and Mol Biol Evol 1995 May; 12 (3): 363-370. Still an additional antigen is a lipopolysaccharide (type R) described in Can J Microbiol 1978 Feb; 24 (2): 1 17-123. See also J Immunol 1996 Jul 1; 151 (1): 234-243. the FrpB protein is also a candidate antigen; see J Bacteriol 1995 Apr; 177 (8): 2041 -2049 and WO 96/31618. A Pilus vaccine is described in J Clin Invest 1981 Oct; 68 (4): 881-888. Antigens derived from or associated with the pathogen for syphilis include Treponema outer membrane proteins; see Emerg - ^ á ^ ^ ^^^^^^^., ^ .. ^ - ^ - ^ rtri ^ M ^ iÉM ^ - ^^^^^^^ - ^^^^^ - ^ l ~ ^ a¡ ^ s * Infect Dis 1997 Jan: 3 (1): 1 1 -20. A unique physical characteristic of Treponema pallidum, the venereally transmitted agent of human syphilis, is that its outer membrane contains 100 times less protein that rotates the membrane than the outer membranes of negative bacteria per typical gram; a property that has been linked to the chronicity of syphilitic infection. These foreign membrane proteins of membrane spinning T. pallidum, TROMPs, represent potential virulence determinants of potential surface and host immunity targets. The outer membrane of T. pallidum has been isolated and its constituent proteins identified. Five proteins of molecular mass 17-, 28-, 31-, 45- and 65-kDa were associated by the outer membrane. Tubes 1, 2 and 3 were antigenic when tested with infection serum and human and immune syphilitic rabbits. An additional candidate is the outer layer protein P6; see J. Exp Med 1986 Oct 1; 164 (4): 1 160-1 170. See also Microbiol Rev 1993 Sep; 57 (3): 750-779. The chancre is a sexually transmitted disease caused by Haemophilus ducreyi. Antigens derived from or associated with Haemophilus ducreyi include an outer membrane protein of 18,000 MW described in Infecí Immun 1996 Jun; 64 (6): 1950-209. A novel lipoprotein expressed by Haemophilus ducreyi is described in Infecí Immun 1996 Dec; 64 (12): 5047-5052. An outer membrane protein binding hemoglobin is included in virulence expression pro Haemophilus ducreyi in an animal model. See Infect Immun 1996 May; 64 (5): 1724-1735. The characterization of the hgbA locus which encodes a hemoglobin receptor of Haemophilus ducreyi is describes in Infect Immun 1 995 Jun; 63 (6): 2194-2200. See also J Med Microbiol 1992 Dec; 37 (6): 413-419 for the identification of species-specific and highly conserved polypeptides of Haemophilus ducreyi. Combination vaccines administered or prepared according to the present invention will contain an immunoprotective amount of the antigens and can be prepared and administered by conventional techniques. Vaccine preparation is generally described in New Trends and Developments in Vaccines, edited by Voller ef al. , 10 University of Park Press, Baltimore, Maryland, U.S. A. 1978. Encapsulation within liposomes is described, for example, by Fullerton, US Pat. 4,235,877. The conjugation of proteins to macromolecules is described, for example, by Likhite, US Pat. 4,372,945 and by Armor ef al., Patent of E.U. 4,474,757. The amount of antigen in each vaccine dose is selected as an amount that induces a therapeutic or immunoprotective response without adverse side effects, significant in typical vaccines. Such amount will vary depending on which specific immunogen is used. Generally, it is expected that each dose will comprise 1-1000 μg of protein, preferably 2-100 μg, more preferably 4-40 μg. An optimal amount for a particular vaccine can be ascertained by standard studies that include the observation of antibody concentrations and other responses in subjects. Following an initial vaccination, subjects can receive a boost in about 4 weeks. * rA '? 4 r? ^ j The amount of antigen in each vaccine dose is an amount that induces a therapeutically effective or immunoprotective response without adverse side effects in typical women's vaccines. Generally, each dose is expected to comprise 1-1000 μg of antigen, preferably 2-100 μg, more preferably 4-40 μg. The adjuvant that induces TH-1, for example, 3-DMPL, will normally be present in a range of 10-200-200 μg, preferably 25-75 μg, especially about 50 μg per dose. The amount of vehicle may vary and may be selected according to the knowledge of a person skilled in the art. If aluminum hydroxide (alum) or aluminum phosphate is used, the amount employed will generally be in the range of 100-1000 μg, for example 250-750 μg, preferably about 500 μg per dose of vaccine.
Typical amounts of each component in the vaccine are antigen (20 μg), alum (500 μg) and an adjuvant, especially an adjuvant that induces TH-1 such as 3-DMPL (50 μg). In a preferred aspect, the vaccine for use in the invention comprises gD2t, 3-DMPL (especially particle 3-DMPL) small) and aluminum hydroxide (alum). In a preferred regimen, the vaccine can be given in intervals of 0, 1 and 6 months. Other dose regimens, including stimulant doses, may also be used. The vaccine can be administered intramuscularly. The manufacture of a vaccine according to the invention it can also be carried out by conventional techniques, such as those described in WO 92/16231. The method typically includes mixing one or more antigens derived from or associated with an STD with an adjuvant, especially an adjuvant that induces ^ TH-1, and optionally a vehicle as described herein above. The resulting vaccine composition can be used for administration to female subjects according to the method of the invention, especially sexually active women suffering from or at risk of contracting an STD. Generally, women will be in an age range of 12-70 years, more usually adolescents and women of 60 or less, for example 14-60, typically 18-45 as in the study described below. In one aspect, an adequate group of women includes those who suffer from or are at risk of contracting genital herpes infection. The method or use of the invention can, for example, be applied in healthy seronegative consorts of subjects with genital herpes disease. The invention is illustrated, without limitation, by the following examples, which show results when a vaccine against herpes was administered to female subjects. Similar results can be obtained with vaccines against other STDs such as chlamydia and HPV antigens and with combination of polyvalent vaccines against more than one STD, especially combination vaccines comprising an antigen associated with Herpes Simplex, more especially HSV-2 gD or fragments immunogens thereof such as gD2t as described herein previously . EXAMPLE 1 - STUDY DESIGN Vaccine under study Herpes Simplex candidate vaccine from SmithKIine Beecham Biologicals (gD2t-20 μg) with Alum (500 μg) and 3-DMPL (50 μg). Concentration A randomized, double-blind, placebo-controlled study to evaluate the efficacy of the candidate Herpes Simplex vaccine from SmithKine Beecham Biologicals (gD2t) with 3-DMPL to prevent genital herpes disease in healthy consorts of subjects with genital herpes disease . Indication / study population Healthy adult volunteers, women and men, aged 18 to 45 years with negative serological markers of Herpes Simplex infection (HSV-1 and -2) and whose consorts have herpes disease ^ - ^ ia ^. ^ ". MSiiA ^^ f ^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^^ second vaccination, the protective efficacy of the gD-Alum-3-DMPL vaccine to prevent clinical disease of genital herpes. Second To compare with placebo, starting one month after the second vaccination, the protective efficacy of the gD-Alum-3-DMPL vaccine to prevent clinical disease of genital herpes. Compare with placebo, after the full vaccination period, the protective efficacy of the gD-Alum-3-DMPL vaccine to prevent genital herpes infection. 15 Compare with placebo, after the full vaccination period, the protective efficacy of the gD-Alum-3-DMPL vaccine to prevent genital herpes infection during a period of extended clinical follow-up. Compare with placebo, after the full vaccination period, the protective efficacy of the gD-Aium-3-DMPL vaccine to prevent genital herpes disease. Compare with placebo, after the full vaccination period, the protective efficacy of the gD-Alum-3-DMPL vaccine to prevent clinical disease of genital herpes during a period of extended clinical follow-up.
Evaluate, starting one month after the second vaccination, the time of occurrence of the disease in each group. Evaluate, starting a month after the second vaccination, the time of infection in each group. 5 Evaluate, in each group, the number of typical and atypical cases of genital herpes disease. Evaluate the severity of the primary disease in both groups. To evaluate the cellular and humoral immune response (excluding 10 subjects from the study centers initiated after July 1, 1995) of the vaccine. Determine immunological or serological correlations for protective efficacy (excluding subjects from study centers after July 1, 1995). 15 In case of infection or primary disease, evaluate the number of subsequent recurrences in the two groups. To assess the safety and reactogenicity of the candidate vaccine for Herpes Simples from SmithKIine Beecham Biologicals (with 3-DMPL) in healthy seronegative HSV subjects. 20 Evaluate the number of cases of orolabial (or non-genital) herpes disease. Compare with place, beginning a month after the second vaccination, the protective efficacy of gD-Alum-3-DMPL to prevent the symptoms and signs of suspected genital herpes associated with either Seroconversion of Western Blot in non-vaccine antigens or with the Detection of HSV DNA in a genital swab by PCR. To assess the incidence of genital herpes disease and HSV infection in vaccine recipients during the period of extended clinical follow-up. 5 Study design Controlled placebo study, aleotirazdo, double blind. Vaccination program: 0-1 -6 months Initial follow-up period - 17 months for each subject 10 starting 1 month after the second vaccination. Extended follow-up period - 24 months for each subject (from the month 19 view to the 43 month visit). Phase A (recipients of placebo, vaccine and double anonymity) - ends when the last subject recruited completes the initial follow-up period (approximately the time the study is not hidden for analysis). Phase B (only vaccine recipients, open) - begins when the last subject recruited completes the initial follow-up period (month 19 visit) and ends when the last subject 20 recruited completes the month 43 visit. Because there may be a period of several months between the date on which the last recruited subject completes the initial follow-up period and the date on which the study is fully discovered for analysis (due to the time required to code and clear all the study data collected during the period of , ^^^ AÍ ^ ^ .y ^ f, ^^^ ^ ^, ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^ ^. After initial follow-up and phase A of the extended follow-up period, the initial part of phase B of the extended follow-up period may include both vaccine and placebo recipients. 2 groups: I gD2t-Alum-3-DMP-Alum-3-DMPL as a placebo. Scheme of the study design HSV-007 - Study periods: Vaccination phase (V); Initial follow-up * (f / u starts); Phase A of extended monitoring; Phase B of extended monitoring. * Note: The "initial follow-up period" now includes 2-19 months. 10 Number of subjects 800 couples will be recruited in the study to allow at least 640 evaluable subjects. Primary Efficacy end point During the 17 month period, starting one month after the second 15 vaccination (2-19 months), the primary efficacy endpoint will be as follows: Disease prevention: A comparison between the two groups of the number of subject 20 with at least one compatible symptom of genital herpes disease and either a concurrent positive culture OR appearance of antibodies to non-vaccinating antigens by Western Blot within six months and positive local detection of herpes simplex DNA by Chain Reaction of Polymerase (PCR). 25 NA: Not Applicable Secondary efficacy end points 1) Infection prevention: A comparison will be made (between the placebo and vaccine groups), of the number of subjects that develop the antibodies for non-vaccine antigens (seroconversion) and of subjects that develop the disease (proven culture). This final point will be evaluated for the following periods. Initial follow-up period (2-19 months) 7-19 Months Phase A of extended follow-up 20 Initial follow-up period (2-19 months) and phase A of the combined extended follow-up 7-19 months and phase A of the combined extended follow-up. The data analysis of phase A of the extended follow-up period will include all events that occur after each visit 25 of month 19 of the subject and the end of phase A (when the last subject recruited completes the month's visit 19) Case definition NA: Not applicable 2) Prevention of disease between 7-19 months A comparison with placebo after the full vaccination period (7-19 months) of the subject number with at least one compatible symptom of genital herpes disease AND either an appearance of OR of concurrent positive culture of antibodies for non-vaccinating antigens by Western Blot and positive local detection of herpes simplex DNA by Polymerase Chain Reaction (PCR). 3) Prevention of disease during phase A of the extended follow-up period 20 During phase A of the extended follow-up period, a comparison will be made between the two groups of the number of subjects with at least one compatible symptom of genital herpes disease. And either an appearance of OR of positive concurrent culture of antibodies for non-vaccinal antigens by Western Blot and detection local positive herpes simplex DNA by Chain Reaction Polymerase (PCR). In addition, this final point will also be evaluated for the new initial follow-up period (2-19 months) and phase A of the combined extended follow-up period and also for 7-19 months and phase A of the combined extended follow-up. 4) Evaluate during the period of 17 months starting one month after the second vaccination, in each group, the time of occurrence of the genital herpes disease. 5) Evaluate during the period of 17 months beginning one month after the second vaccination, in each group, the time of occurrence of genital herpes infection. 6) Evaluate in each group the number of clinical disease cases of typical genital herpes and atypical genital herpes disease. The case definitions are described in the primary endpoint. 7) evaluate in each group the symptoms and general and local subjective signs of the patient of genital HSV disease and its duration. 8) Evaluate the humoral response (anti-gD2 antibodies by ELIS and anti-HSV neutralizing antibodies) and cellular response (lymphoproliferation, interferon gamma secretion) to the vaccine (excluding subjects from study centers initiated after July 1, 1995) . 9) If clinical efficacy is demonstrated, immunological and serological markers will be evaluated extensively using the sera and Peripheral Blood Lymphocytes stored when program, in an attempt to determine the correlations between protective efficacy and laboratory parameters (excluding subjects from study centers initiated after July 1, 1995). 5 10) In case of infection or primary disease, evaluate the number of subsequent recurrences of genital herpes in each group. 1 1) General and local reactogenicity and safety will be assessed after each vaccination by recording symptoms and general and local signs after each dose and experiences adverse events during the study period. The biochemical and haematological parameters will be verified in baseline and after the last vaccination. During the extended follow-up period, all serious adverse experiences reported by vaccine recipients will be registered. 12) Evaluate the number of clinical cases of non-genital herpes disease, including gold-labial herpes disease, in each group. 13) Compare with placebo, during the follow-up period of 17 20 months, starting a month after the second vaccination, the number of vaccine recipients that develop symptoms and signs of genital herpes associated with either seroconversion to non-vaccine antigens by Western Blot (within a period of six months from the onset of symptoms and signs of genital herpes) or with the detection of HSV DNA in a swab genital by PCR. 14) During phase B of the extended follow-up period, the number of vaccine recipients that develop antibodies to non-vaccine antigens (seroconversion) and of subjects who develop the disease (which has been tested), will be analyzed in relation to the interval from the administration of the last vaccine. These data will be used to calculate infection speed and genital herpes disease at attack speed. EXAMPLE 2 10 The analysis of the primary endpoint is based on comparison with attack rates between the placebo and vaccine groups as described in RAP. The analysis of secondary endpoints is based on either the comparison of attack speeds or comparison of the end points of the time of occurrence of the disease or infection as described below. The statistical tests are bilateral and are carried out using SAS software and an a-level of 0.05. It should be noted that many statistical analyzes are reported, but for the secondary endpoints the speed of error (a) is not under control. Since no adjustment of a is made for secondary endpoints, the p-values should be interpreted with care and only as descriptive. Populations analyzed for Vaccine Efficacy Efficacy analyzes are carried out in populations of two subjects: the population with intention to treat (ITT) and the population according to the procedure (ATP). The ATP group is also referred to in the '".. .. ^ ^ ^ ^ _ ^ ¿i k k k k k k......... ¿¿¿¿¿¿¿¿¿¿¿¿K k................... The population according to the procedure is the primary analysis.The definition of the ATP population (or PP) is defined by the study period under consideration: 5 1) For the period between months 2-19, the ATP population consists of subjects: who meet all the eligibility criteria of the procedure who have received three doses of vaccine / placebo 10 - or who have received two doses of vaccine / placebo and for whom the event considered (illness or infection) has occurred prior to the visit of the month 6 for whom the event considered (illness or infection) has not occurred before the start of the 2-15-19 month period 2) For the period between months 2-19, the ATP population consists of subjects: who meet all the eligibility criteria of the procedure 20 - who have received three doses of vaccine / placebo or who have received two doses of vaccine / placebo and for whom the event considered (illness or infection) has occurred prior to the month 6 visit for whom the event considered (illness or infection) has not occurred before the start of the 2- period • £ se 1 9 months 2) For the period between months 7-1 9, the ATP population consists of subjects - who meet all the eligibility criteria of the procedure that have received three doses of vaccine / placebo or for whom the event considered (illness or infection) has not occurred before the start of the 7-19 month period. 0 The analysis of the ITT population is considered as the secondary analysis. This analysis includes all subjects who receive at least one dose of study vaccine and have at least one evaluation in the vaccine. The purpose of the two analyzes is to ensure that the 5 violations of the procedure, falls of the subject and disadvantages are not related to the treatment and do not lead to any selection deviation in the efficacy results. The populations to be included in the analyzes of safety and immunogenicity they will be fully described in the final study report. Evaluation periods The evaluation periods, in which the analyzes are carried out, include: 2-1 9 months (ATP population) 5 - 7-19 months (ATP population) 0-1 9 months (ITT population) The selected results are shown below, together with a summary of the total conclusions of the study. Table 1 a. Adjusting effect of vafe one on the occurrence of genital herpes disease by the gender population ITT Table 1 b. Adjustment of the effect of the vaccine on the occurrence of genital herpes infection by the ITT gender population twenty Distribution of the Cases of Infection efe HSV v Genital Herpes Disease by the Treatment Group Table 2a. Cases of Genital Herpes Disease by the Treatment Group - Women and Men * For each interval, the number of evaluable subjects per-procedure is shown in the (). Table 2b Cases of Genital Herpes Disease by the Treatment Group - Men only * For each interval, the number of evaluable subjects per-procedure is shown in the ().
**** A m ^ m ^ á ^, ^^^ ^^^ Table 2c Cases of Genital Herpes Disease by the Treatment Group - Only Women * For each interval, the number of evaluable subjects per-procedure is shown in ei (). Table 3a. Cases of Genital Herpes Infection by the Treatment Group - Women and Men * For each interval, the number of evaluable subjects per-procedure is shown in the (). Table 3b Cases of Genital Herpes Infection by the Treatment Group - Men only For each interval, the number of subjects evaluable per procedure ** "*** - • ^ * Ut- ± ** ^ a & z is shown in the ().
Table 3c Cases of Genital Herpes Infection by the Treatment Group - Only Women * For each interval, the number of subject evaluable per-procedure is shown in the (). Preliminary Efficacy Analysis Primary efficacy end point During the 17 month period, starting one month after the second vaccination months 2-19), the primary efficacy endpoint will be as follows: Disease prevention: A comparison between the two groups of the number of subjects with at least one compatible symptom of genital herpes disease AND either an OR appearance of concurrent positive culture of antibodies to non-vaccinating antigens by Western Blot within six months and positive local detection of herpes simplex DNA by Reaction in Chain of Polymerase (PCR).
Table 4a. Prevention of Genital Herpes Disease - Men and Women Table 4b. Prevention of Genital Herpes Disease - Men only Table 4c. Prevention of Genital Herpes Disease - Only women 15 Secondary efficacy endpoints 20 1) Infection prevention: A comparison will be made (between placebo and vaccine groups), the number of subjects that develop antibodies to non-vaccine antigens (seroconversion) and of subjects who develop the disease (tested culture) for the initial follow-up period (months 2-1 9).
•? ^ ^^ - ,,, ^? ^ Ustí. ? i.í ^ ^ ittÉ. ^ M ^ I.
Table 5a. Prevention of Genital Herpes Infection - Men and Women Table 5b. Prevention of Genital Herpes Infection - Men only Table 5c. Prevention of Genital Herpes Infection - Only women 15 2) Infection prevention: 20 A comparison will be made (between placebo and vaccine groups), the number of subjects that develop antibodies to non-vaccinal antigens (seroconversion) and of subjects who develop the disease (proven culture) for months 7-19 .
Table 6a. Prevention of Genital Herpes Infection - Men and Women Table 6b. Prevention of Genital Herpes Infection - Men only Table 6c. Prevention of genital herpes infection - Only women 1) Prevention of infection: A comparison will be made (between placebo and vaccine groups), the number of subjects that develop antibodies to non-vaccine antigens (seroconversion) and of subjects who develop the disease (tested culture) for the initial follow-up period (months 2-19). t? »£ & l & ^. «- - ^^ - ^ S1 !! ^ Table 5a. Prevention of Genital Herpes Infection - Men and Women Table 5b. Prevention of Genital Herpes Infection - Men only Table 5c. Prevention of Genital Herpes Infection - Only women 15 3) Prevention of disease between months 7-1 9: 20 A comparison will be made with placebo after the complete vaccination period (months 7-19) of the number of subjects with at least one compatible symptom of genital herpes disease AND either an OR appearance of concurrent positive culture of antibodies to non-vaccinal antigens by Western Blot within six months and local detection positive herpes simplex DNA by Polymerase Chain Reaction -? - mtA ?? ai * .., ^ atJ ^ j ^ J ^ s ^ b ^ ^^ || L £ &(CRP).
Table 7a. Prevention of Genital Herpes Disease - Men and 5 Women Table 7b. Prevention of Genital Herpes Disease - Only men Table 7c. Prevention of Genital Herpes Disease - Only women Evaluate during the 17 month period starting one month after the second vaccination, in each group, the time of occurrence of genital herpes disease. The time of occurrence of genital herpes disease for the ITT population is shown in figures 1 a (men and women), 1 b (only men), and 1 c (only women). Efficiency analyzes of time for the occurrence of genital herpes disease for the ITT population is shown in | §¿ -tables 8a (women and man), 8b (only men) and 8c (solamerpe women) The time of occurrence was not analyzed for the per-procedure populations (months 2-19) since a previous difference without disease was observed among recipients before month 2. Note: the time of the analysis of occurrence excludes cases of genital herpes disease that occur after month 19.
Table 8a. Prevention of Genital Herpes Disease due to Occurrence Time - Men and Women Table 8b. Prevention of Genital Herpes Disease due to Occurrence Time - Only men Table 8c. Prevention of Genital Herpes Disease due to Occurrence Time - Only Women Summary and conclusions after detailed analysis of the results of the test Demographic characteristics and evaluation of risk factors Total, of the 847 (425 vaccine and 422 placebo) subjects 5 recruited, 697 (344 vaccine and 353 placebo) subjects completed the study until month 1 9. One hundred and fifty subjects (150) withdrew from the study; none of the withdrawals resulted from a serious adverse event. Three hundred and seventy subjects (370) in the vaccine group and 369 in the placebo group were evaluated in the ATP population for months 2-19. 10 The treatment groups were balanced for all demographic characteristics and the selection of process consenters for the ATP population analyzes did not result in the introduction of deviation by treatment groups. Risk factors that may impact the rate of acquisition of genital herpes disease or infection were assessed and included the duration of the relationship before studying the entry, the average time to separation of the source pattern, the frequency of sexual intercourse ( baseline and during the effectiveness monitoring period) and frequency of condom use (baseline and during the efficacy monitoring period). These results indicate that the placebo and vaccine groups were balanced in baseline for all risk factors and the balance was maintained during the study. The similarity of the ITT population profile to that of the ATP population in terms of risk factors also confirms that the elimination of the non-consenting for the The procedure is not diverted by the treatment groups. The sub-analyzes by gender indicate that within each gender group, the risk factors that can impact the acquisition of infection or genital herpes disease are balanced by the group treatment. End-point analysis of primary efficacy The end-point analysis of primary efficacy does not demonstrate efficacy of the genital herpes vaccine in a combined population of seronegative healthy consorts, men and women, of subjects with genital herpes disease. The results of the analysis of the endpoint of primary efficacy are summarized as follows: 1) The effectiveness of the relative vaccine in the total population (month 2-19 ATP) IS 25.4% (95% ci: -55.5, 64.2, p = 0.449 ). The effectiveness of the relative vaccine for the ITT population is 37.9% (95% CI: -16.6, 67.0, p = 0. 143). 2) A statistically significant gender by group interaction in the analysis of efficacy of the ITT population (p = 0.03). 3) A separate analysis by gender shows an efficacy of 20% of the population of 54.2% in the population of women ATP in the months 2-19 (95% Cl, -47.7, 85.8, p = 0.238) and a statistically significant efficacy of the vaccine of 72.7% (95% Cl, 19.1, 90.8, p = 0.014) in the ITT population of women. In the male population, there is no evidence of efficacy of the vaccine. In the population of men ATP in the months 2-19, the efficacy of the vaccine is 3.6% (95% CI.-171, 60.5) and -1.1 .1% (95% Cl: -157.6, 52.1) **. *** ...., », * ^. ^^, ... ^ - ^ .... ^ ^ ¿_, _ ..., .. o. ^^^ fe-rf ^ .a, ^^^^ á ^^^^ = ¿^ ¿-Í in the ITT male population. Several baseline covariates were investigated to determine if they can influence efficacy outcomes; gender, age, frequency of condom use in baseline, frequency of sexual intercourse and 5 duration of relationships before studying entry. The trend towards efficacy of the vaccine was associated with gender, age over 30 years, use of condoms not frequent, sexual intercourse less than the average frequency and shorter duration of relationships. These observations were applied to both ATP and ITT populations. 10 Secondary efficacy endpoint analysis Prevention of genital herpes disease (month 7-19). After 3 doses of vaccine (between study months 7-19), vaccine efficacy of 81.1% (95% CI: -58.9, 97.8) was observed for the prevention of genital herpes disease in women (p = 0.1 1 1).
No trend towards efficacy was observed in men during the observation period of 7-19 months (-2.9% efficacy of the vaccine, 95% CI: -624.7, 85.4, p = 0.99). This trend towards efficacy of the vaccine in the ATP population of women aged 7-19 months is consistent with the observation of the efficacy of the vaccine in women in the ITT analysis. of the primary end point. Prevention of HSV infection A comparison was made between the placebo and vaccine groups for the efficacy of the vaccine to prevent HSV infection. Overall, there was no vaccine efficacy against HSV infection. Nevertheless, consistent with disease end-point analyzes, a trend towards efficacy of vaccine against HSV infection was suggested in women in the ITT population (vaccine efficacy 46.0%, 95% Cl: -2.1, 71.4, p = 0.072) and in the ATP population of 7-1 9 months (vaccine efficacy of 52.8%, 95% Cl: -33.4, 83.3, p = 0.184). , 5 Time of occurrence of genital herpes The time of occurrence of the genital herpes disease is calculated from the entrance of the study until the occurrence of the disease. The main analysis has been carried out by the Gradolog test; Kaplan-Meier curves were drawn for each group. In women (ATP population) months 2-19), the separation of the curves that denote the occurrence of the disease cases is apparent for approximately nine months with cases of disease that continue to occur in the placebo group. The vaccine efficacy is estimated at 53.6% (95% CI, -54.2, 86.0) in women. In the ITT women population where the separation of placebo and vaccine curves are apparent from month 0, statistically the efficacy of the significant vaccine is estimated at 73.2% (95% Cl, 18.7, 91.2, p = 0.013). No efficacy of the vaccine was observed for the male population. Again, the results of the "time of occurrence" analysis is consistent with the primary endpoint analysis. 20 Severity of genital herpes disease Parameters including duration of injuries, duration of symptoms per episode, number of symptoms per episode and intensity of symptoms per episode were used to assess the severity of the disease in both treatment groups. In the ATP population of 2-25 19 months combined, the duration of symptoms per episode is significantly longer in the small number of cases that occur in e! vaccine group (P = 0 \ 031). The gender specific severity data also reveal that in women, there is a statistically significant higher number of genital herpes disease 5 lesions per episode (p = 0.010) in the vaccine group. These observations follow that although vaccination can prevent mild to moderate disease in the vaccine group, the disease with more severe manifestations is not prevented by vaccination. Total Conclusions 10 The analysis demonstrates that although there may be a trend towards efficacy of the vaccine against genital herpes, the primary endpoint analysis does not demonstrate efficacy of the vaccine in a combined population of healthy seronegative consorts of men and women, of subjects with genital herpes disease. However, a sub separate analysis by the gender group, based on the observed gender interaction, surprisingly shows a trend towards the effectiveness of the vaccine in women that is statistically significant in the ITT population. There is no evidence of the effectiveness of the vaccine in the male population. twenty - 4 / - ADDITIONAL REFERENCES 1. Washington AE, Johnson RE and Sanders LL. Chlamydia trachomatis infections in the United States: what are they costing us. JAMA 1987, 257, 2070-2072. 2. Grayston JT and Wang SP. New nowledge of Chlamydiae and the diseases they cause. The Journal of Infectious Diseases 1975, 132: 87-105. 3. Grayston JT, Wang SP, Yeh LJ, and Kuo CC. Importance of reinfection in the pathogenesis of trachoma. Reviews of Infectious Diseases 1985, 7, 717-725 4. Mopison RP, RJ Belland, K Lyng and HD Caldwell. Chal ydial disease pathogepesis. The 57-kD Chlamydial hypersepsitivity anugen is a stress response protein. J. Exp. Med. 1989, 170, 1271-1283 5. Blander SJ and Amortegui AJ. Mice immunized with a chlamydial extract have not increased in ear and protective immunity despite ipcreased inflammation following genital infection by the mouse pneumonitis agent of Chlamydia trachomatis. Infect Immun. 1994, 62, 3617-3624. μg 6. Wang SP, Kuo CC, Barnes RC, Stephens RS and Grayston JT. Immunotyping of Chlamydia trachomatis with monoclonal antibodies. The Journal of Infectious Diseases 1985, 152, 791-800. 7. Bavoil P, Ohlin A, and Schachter J. Role of disulfide bonding in outer membrane structure and permeability in Chlamydia trachomatis. Infect. Im a. 1984, 44, 479-485. 8. Hatch TP, Miceli M, Sublett JE. Synthesis of disulfide-bonded outer membrane proteins during development cycle of Chlamydia psittaci and Chlamydia trachomatis. J. Bacteriol. 1986, 165, 379-385. 9. Stephens RS, R Sánchez-Pescador, EA Wagar, C Inouye and MS Urdea. Diversity of Chlamydia trachomatis Major Outer Membrane Protein genes. J. Bacteriol. 1987, 169, 3879-3885. 10. Yuan Y, 22? Ang YX, Watkins NG, and Caldwell HD. Nucleotide and deduced amino acid sequences for the four variable domains of the major outer mebrane proteins of the 15 Chlamydia trachomatis serovars. Infect. Immun. 1989, 57, 1040-1049. 4cJ 11. Baehr W, Zhang YX, Joseph T, Su H, Nano FE, Everett EK and Calwell HD. Mapping antigenic domaips expressed by Chlamydia trachomaxis major outer membrane protein genes. Proc. Nati Acad. Soi. USA 1988. 85, 4000-4004 12. Lucero ME and Kuo CC. Neutralization of Chlamydia trachomatis cell culture infection by serovar-specific mdhoclonal antibodies. Infect. Immun. 1985, 50, 595-597 13. Zhang YX, Stewart S, Joseph T, Taylor HR and HD Caldwell. Protective monoclonal antibodies recognize epitopes located on the major outer membrane protein of Chlamydia trachomatis. J. Imunoi. 1987, 138, 575-581. 14. Peterson E, Zhong G, Carlson E and de la Maza LM. Protective role of magnesium in the neutralization by monoclonal antibodies of Chlamydia trachomatis ipfectivity. Infected Immun. 1988, 56, 885-891. 15. Zhang YX, Stewart SJ and Caldwell HD. Protective monoclonal antibodies to Chlamydia trachomatis serovar- and serogroup- specific major outer membrane protein determinants. Infect. Immun. 1989, 57, 636-638 16. Alien JE, RM Loksley and RS Stephens. A single peptide from the major outer membrane protein of Chlamydia trachomatis eiicits T cell help for the production of antibodies to protective determinants. J. Immunol. 1991, 147, 674-679 17. His H, RP Morrison, NG Watkins and HD Caldwell. Identification and characterization of T heiper cell epitopes of the major outer membrane protein of Chlamydia trachomatis. J. Exp. Med. 1990, 172, 203-212 18. Manning DS and SJ Stewart. Expression of the major outer membrane protein of Chalmydia trachomatis in Escherichia coli. Infect. Immun. 1993, 61, 4093 ^ 098. 19. Koehier JE, Birkelund S and Stephens RS. Overexpression and surface localization of the Chlamydia trachomatis major outer membrane protein in Escherichia coli. Molecular Microbiology 1992, 6, 1087-1094 20. Pickett MA, ME Ward and IN Clarke. High level expression and epitope locaiization of the major outer membrane protein of Chlamydia trachomatis serovar Ll. Molecular Microbiology 1988, 2, 681-685.
- A M - 21. Taylor HR, J Whittum-Hudson, J Schachter, HD Caldwell and RA Prendergast. Oral immunization with chlamydial major outer membrane protein (MOMP). Investigative Ophthalmology and visual Science 1988, 29, 1847-1853 22. Batteiger BE, RG Rank, PM Bavoil and LSF Soderberg. Partial protection against genital reinfection by immunization of guinea-pig with isolated membrane proteins of the chlamydial agent of guinea-pig inclusion conjunctivitis. Journal of General Microbiology 1993, 139, 2965-2972. 23. Tuffrey M, F Alexander, W Conian, C Woods and M Ward. Heterotypic protection of mice against chlamydial salpingitis and colonization of the lower genital tract with a human serovar F isolate of Chlamydia trachomatis by prior immunization with recombinant Ll major outer-membrane protein. Journal of General Microbiology 1992, 138, 1707-1715. 24. Tuffrey M, P Falder, J Gale and D Taylor-Robinson. Salpingitis in mice induced by human strains of Chlamydia trachomatis. Br. J. exp. Path. 1986, 67, 605-616. 25. Tuffrey M, P Falder, J Gale, R Quinn and D Taylor -Robinson.Infertility in mice infected genitally with a human strain of Chlamydia trachomatis. Br. J. exp. Path. 1986, 78, 251-260 26. Ramsey KH, LSF Soderberg and RG Rank. Resoiution of Chlamydial genital infection in B-cell deficient mice and im unity to reinfection. Infect. Im a. 1988, 56, 1320-1325. 27. Rank RG, LSF Soderberg and AL Barron. Chronic chlamydial genital infection in congenitally athymic pude ice. Infect. Immun. 1985, 48, 847-849 28. Igietseme JU and RG Rank. Susceptibility to reinfection after a primary chlamydial genital infection is associated with a decrease in antigen-specific T cell in the genital tract. Infect. Immun. 1991, 59, 1346-1351 29. Igietseme JU, KH Ramsey, DM Magee, DM Williams TJ Kincy and RG Rank. Resolution of murine chlamydial genital infection by the adoptive transfer of a biovar-specific, THl Lymphocyte clone. Regional Immunology 1993, 5, 317-324. 30. Igietseme JU, Dfvt Magee, DM Williams and RG Rank. Role for CD8 + T cell in antichlamydial immunity defined by chlamydial-specific T-lymphocyte clones. Infect. Immun. 1994, 62, 5195-5197.

Claims (20)

  1. CLAIMS 1. A method for treating a female human subject suffering from or susceptible to one or more sexually transmitted diseases (STDs), such method comprises administering to a female subject in need thereof an effective amount of a vaccine formulation comprising one or more antigens derived from or associated with a pathogen that causes STD and an adjuvant.
  2. 2. The use of one or more antigens derived from or associated with a pathogen causing STD and an adjuvant in the preparation of a vaccine for administration to a human female subject for the prevention and / or treatment of one or more STDs.
  3. 3. Method or use according to claim 1 or claim 2, characterized in that said adjuvant is an adjuvant that induces TH-1. Method or use according to any of claims 1 to 3, characterized in that said one or more antigens include HSV glycoprotein D or an immunological fragment thereof. Method or use according to claim 4, characterized in that the glycoprotein D of HSV-2 is a truncated glycoprotein. Method or use according to claim 5, characterized in that the truncated glycoprotein is HSV gD2 and is free from the C-terminal support region (gD2t). Method or use according to any of the preceding claims, characterized in that said one or more antigens include a . ^. ^^^^^^^^^^ i ^^^^^. t. ^ t. ^ JMfe ^^ -. a ^^ antigen derived from or associated with HPV. Method and use according to any of the preceding claims, characterized in that said one or more antigens include an antigen derived from or associated with Chlamydia. 9. Method or use according to any of the preceding claims, characterized in that said one or more antigens include an antigen derived from or associated with Neiserria gonorrhea. Method or use according to any of the preceding claims, characterized in that said one or more antigens include an antigen derived from or associated with Treponema pallidum (syphilis) or Haemophilus ducreyi (chancre). eleven . Method or use according to any of the preceding claims, characterized in that said antigen or combination of antigens is formulated with a suitable vehicle. Method or use according to claim 10, characterized in that the vehicle is aluminum hydroxide (alum), aluminum phosphate or an oil in water emulsion. 13. Method or use according to any of the preceding claims, characterized in that the adjuvant is the adjuvant 3-DMPL that induces TH-1. Method or use according to claim 13, characterized in that the 3D-MPL particles are sufficiently small to filter sterile through a 0.22 micron membrane. 15. Method or use according to any of claims 4 to 14, characterized in that the vaccine is used to immunize or treat A to ... A ^^? ^^ ¿fe¿ ^ ^ subjects women at risk of getting herpes simplex infections. 16. Method or use according to claim 15, characterized in that the vaccine is used to treat or prevent genital herpes infections. 5. Method or use according to claim 15 or 16, characterized in that the vaccine formulation comprises gD2t (1-1000 μg), 3-DMPL (10-200 μg) and an aluminum salt (100-1000 μg). Method or use according to claim 17, characterized in that the vaccine formulation comprises gD2t (20 μg), 3-DMPL (50 μg) 10 and alum (500 μg). 19. Method or use according to any of the preceding claims, characterized in that the vaccine formulation is administered to, or manufactured for administration to, female subjects at intervals of 0, 1 and 6 months. 20. Method or use according to any of the preceding claims, characterized in that the vaccine formulation is administered intramuscularly.
MXPA/A/2001/002532A 1998-09-11 2001-03-09 Vaccine against sexually transmitted diseases MXPA01002532A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB9819898.9 1998-09-11

Publications (1)

Publication Number Publication Date
MXPA01002532A true MXPA01002532A (en) 2001-11-21

Family

ID=

Similar Documents

Publication Publication Date Title
US6692752B1 (en) Methods of treating human females susceptible to HSV infection
JP4896715B2 (en) Immunogenic composition against Chlamydiatrachomatis
AU744989B2 (en) Vaccine against sexually transmitted diseases
JP4510283B2 (en) Combined vaccine composition
RU2585961C9 (en) Vaccine against herpes simplex virus type 2: compositions and methods for triggering immune response
JP2008544745A (en) Immunogenic composition for Chlamydiatrachomatis
Odegard et al. A novel HSV-2 subunit vaccine induces GLA-dependent CD4 and CD8 T cell responses and protective immunity in mice and guinea pigs
JP2012180367A (en) Vaccine against chlamydial infection
Roth et al. HSV-2 vaccine: current state and insights into development of a vaccine that targets genital mucosal protection
RU2723046C2 (en) Chlamydia sp vaccines
JP2010505795A (en) Vaccines against Chlamydia infection
Wang et al. Evaluation of a tandem Chlamydia psittaci Pgp3 multiepitope peptide vaccine against a pulmonary chlamydial challenge in mice
Adimora et al. Vaccines for classic sexually transmitted diseases
CA2275896A1 (en) Chlamydia vaccines
MXPA01002532A (en) Vaccine against sexually transmitted diseases
US8541007B2 (en) Vaccines against chlamydial infection
AU2932502A (en) Vaccine against sexually transmitted diseases
EP4032545A1 (en) Immunogenic composition and vaccine containing chlamydia ssp. surface antigens and its use
Farris Mechanisms of major outer membrane protein (MOMP)-vaccine induced protective immunity against Chlamydia muridarum genital infection