Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Vaccine ; 30(30): 4490-8, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22561143

RESUMEN

A novel vaccine (PEV7) consisting of a truncated, recombinant aspartyl proteinase-2 of Candida albicans incorporated into influenza virosomes was studied. This vaccine candidate generated a potent serum antibody response in mouse and rat following intramuscular immunization. Anti-Sap2 IgG and IgA were also detected in the vaginal fluid of rats following intravaginal or intramuscular plus intravaginal administration. In a rat model of candidal vaginitis, PEV7 induced significant, long-lasting, likely antibody-mediated, protection following intravaginal route of immunization. PEV7 was also found to be safe in a repeated-dose toxicological study in rats. Overall, these data provide a sound basis to envisage the clinical development of this new candidate vaccine against candidal vaginitis.


Asunto(s)
Ácido Aspártico Endopeptidasas/inmunología , Candidiasis Vulvovaginal/prevención & control , Proteínas Fúngicas/inmunología , Vacunas Fúngicas/administración & dosificación , Virosomas/administración & dosificación , Administración Intravaginal , Animales , Anticuerpos Antifúngicos/sangre , Candida albicans/inmunología , Candidiasis Vulvovaginal/inmunología , Reacciones Cruzadas , Femenino , Vacunas Fúngicas/inmunología , Masculino , Ratones , Orthomyxoviridae/inmunología , Ratas , Ratas Wistar , Pruebas de Toxicidad , Virosomas/inmunología
2.
Malar J ; 10: 359, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22166048

RESUMEN

BACKGROUND: In clinical trials, immunopotentiating reconstituted influenza virosomes (IRIVs) have shown great potential as a versatile antigen delivery platform for synthetic peptides derived from Plasmodium falciparum antigens. This study describes the immunogenicity of a virosomally-formulated recombinant fusion protein comprising domains of the two malaria vaccine candidate antigens MSP3 and GLURP. METHODS: The highly purified recombinant protein GMZ2 was coupled to phosphatidylethanolamine and the conjugates incorporated into the membrane of IRIVs. The immunogenicity of this adjuvant-free virosomal formulation was compared to GMZ2 formulated with the adjuvants Montanide ISA 720 and Alum in three mouse strains with different genetic backgrounds. RESULTS: Intramuscular injections of all three candidate vaccine formulations induced GMZ2-specific antibody responses in all mice tested. In general, the humoral immune response in outbred NMRI mice was stronger than that in inbred BALB/c and C57BL/6 mice. ELISA with the recombinant antigens demonstrated immunodominance of the GLURP component over the MSP3 component. However, compared to the Al(OH)(3)-adjuvanted formulation the two other formulations elicited in NMRI mice a larger proportion of anti-MSP3 antibodies. Analyses of the induced GMZ2-specific IgG subclass profiles showed for all three formulations a predominance of the IgG1 isotype. Immune sera against all three formulations exhibited cross-reactivity with in vitro cultivated blood-stage parasites. Immunofluorescence and immunoblot competition experiments showed that both components of the hybrid protein induced IgG cross-reactive with the corresponding native proteins. CONCLUSION: A virosomal formulation of the chimeric protein GMZ2 induced P. falciparum blood stage parasite cross-reactive IgG responses specific for both MSP3 and GLURP. GMZ2 thus represents a candidate component suitable for inclusion into a multi-valent virosomal malaria vaccine and influenza virosomes represent a versatile antigen delivery system suitable for adjuvant-free immunization with recombinant proteins.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/genética , Reacciones Cruzadas , Inmunoglobulina G/sangre , Inyecciones Intramusculares , Vacunas contra la Malaria/administración & dosificación , Manitol/administración & dosificación , Manitol/análogos & derivados , Ratones , Ácidos Oléicos/administración & dosificación , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas de Virosoma/administración & dosificación , Vacunas de Virosoma/inmunología
3.
PLoS One ; 6(7): e22273, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21799810

RESUMEN

BACKGROUND: This trial was conducted to evaluate the safety and immunogenicity of two virosome formulated malaria peptidomimetics derived from Plasmodium falciparum AMA-1 and CSP in malaria semi-immune adults and children. METHODS: The design was a prospective randomized, double-blind, controlled, age-deescalating study with two immunizations. 10 adults and 40 children (aged 5-9 years) living in a malaria endemic area were immunized with PEV3B or virosomal influenza vaccine Inflexal®V on day 0 and 90. RESULTS: No serious or severe adverse events (AEs) related to the vaccines were observed. The only local solicited AE reported was pain at injection site, which affected more children in the Inflexal®V group compared to the PEV3B group (p = 0.014). In the PEV3B group, IgG ELISA endpoint titers specific for the AMA-1 and CSP peptide antigens were significantly higher for most time points compared to the Inflexal®V control group. Across all time points after first immunization the average ratio of endpoint titers to baseline values in PEV3B subjects ranged from 4 to 15 in adults and from 4 to 66 in children. As an exploratory outcome, we found that the incidence rate of clinical malaria episodes in children vaccinees was half the rate of the control children between study days 30 and 365 (0.0035 episodes per day at risk for PEV3B vs. 0.0069 for Inflexal®V; RR  = 0.50 [95%-CI: 0.29-0.88], p = 0.02). CONCLUSION: These findings provide a strong basis for the further development of multivalent virosomal malaria peptide vaccines. TRIAL REGISTRATION: ClinicalTrials.gov NCT00513669.


Asunto(s)
Antígenos de Protozoos/química , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/inmunología , Proteínas de la Membrana/química , Peptidomiméticos/administración & dosificación , Peptidomiméticos/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/química , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Malaria/epidemiología , Malaria/prevención & control , Vacunas contra la Malaria/efectos adversos , Masculino , Persona de Mediana Edad , Peptidomiméticos/efectos adversos , Virosomas , Adulto Joven
4.
Expert Rev Vaccines ; 10(4): 437-46, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21506642

RESUMEN

Influenza virosomes have been used for more than 10 years in commercial vaccines. The technology has been further developed as a carrier and adjuvant system for subunit vaccines, in particular for synthetic peptides. The extensive amount of preclinical and clinical data supports the notion that influenza virosomes represent a platform technology that ensures robust and long-lasting immune responses against subunit antigens with an excellent safety profile. Structurally and functionally, virosomes are enveloped virus-like particles, although they are assembled in vitro. This unique feature ensures a tight control of their composition and at the same time provides the flexibility to adapt the particle to various types of antigens. The mode of action of virosomes is complex and includes carrier as well as immune-stimulatory functions.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Vacunas contra la Influenza/inmunología , Virosomas/administración & dosificación , Humanos , Vacunas contra la Influenza/administración & dosificación , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Virosoma/administración & dosificación , Vacunas de Virosoma/inmunología
5.
Immunity ; 34(2): 269-80, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21315623

RESUMEN

Human immunodeficiency virus (HIV)-1 is mainly transmitted mucosally during sexual intercourse. We therefore evaluated the protective efficacy of a vaccine active at mucosal sites. Macaca mulatta monkeys were immunized via both the intramuscular and intranasal routes with an HIV-1 vaccine made of gp41-subunit antigens grafted on virosomes, a safe delivery carrier approved in humans with self-adjuvant properties. Six months after 13 vaginal challenges with simian-HIV (SHIV)-SF162P3, four out of five vaccinated animals remained virus-negative, and the fifth was only transiently infected. None of the five animals seroconverted to p27gag-SIV. In contrast, all 6 placebo-vaccinated animals became infected and seroconverted. All protected animals showed gp41-specific vaginal IgAs with HIV-1 transcytosis-blocking properties and vaginal IgGs with neutralizing and/or antibody-dependent cellular-cytotoxicity activities. In contrast, plasma IgGs totally lacked virus-neutralizing activity. The protection observed challenges the paradigm whereby circulating antiviral antibodies are required for protection against HIV-1 infection and may serve in designing a human vaccine against HIV-1-AIDS.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Anticuerpos Anti-VIH/biosíntesis , Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Inmunización , Macaca mulatta/inmunología , Fragmentos de Péptidos/inmunología , Vagina/inmunología , Virosomas/inmunología , Vacunas contra el SIDA/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Sitios de Unión , Femenino , Anticuerpos Anti-VIH/inmunología , Proteína gp41 de Envoltorio del VIH/administración & dosificación , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/transmisión , Seropositividad para VIH , Datos de Secuencia Molecular , Fragmentos de Péptidos/administración & dosificación , Transcitosis , Viremia/inmunología , Viremia/prevención & control , Viremia/transmisión , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/análisis
6.
Vaccine ; 27(46): 6415-9, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19559121

RESUMEN

Although respiratory syncytial virus (RSV) causes severe lower respiratory tract infection in infants and adults at risk, no RSV vaccine is currently available. In this report, efforts toward the generation of an RSV subunit vaccine using recombinant RSV fusion protein (rRSV-F) are described. The recombinant protein was produced by transient gene expression (TGE) in suspension-adapted human embryonic kidney cells (HEK-293E) in 4 L orbitally shaken bioreactors. It was then purified and formulated in immunostimulating reconstituted influenza virosomes (IRIVs). The candidate vaccine induced anti-RSV-F neutralizing antibodies in mice, and challenge studies in cotton rats are ongoing. If successful in preclinical and clinical trials, this will be the first recombinant subunit vaccine produced by large-scale TGE in mammalian cells.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Transfección , Proteínas Virales de Fusión/inmunología , Animales , Anticuerpos Antivirales/sangre , Línea Celular , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas de Subunidad/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/aislamiento & purificación , Virosomas/inmunología
7.
Malar J ; 8: 314, 2009 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-20042100

RESUMEN

BACKGROUND: Clinical profiling of two components for a synthetic peptide-based virosomal malaria vaccine has yielded promising results, encouraging the search for additional components for inclusion in a final multi-valent vaccine formulation. This report describes the immunological characterization of linear and cyclized synthetic peptides comprising amino acids 211-237 of Plasmodium falciparum merozoite surface protein (MSP-3). METHODS: These peptides were coupled to phosphatidylethanolamine (PE); the conjugates were intercalated into immunopotentiating reconstituted influenza virosomes (IRIVs) and then used for immunizations in mice to evaluate their capacity to elicit P. falciparum cross-reactive antibodies. RESULTS: While all MSP-3-derived peptides were able to elicit parasite-binding antibodies, stabilization of turn structures by cyclization had no immune-enhancing effect. Therefore, further pre-clinical profiling was focused on FB-12, a PE conjugate of the linear peptide. Consistent with the immunological results obtained in mice, all FB-12 immunized rabbits tested seroconverted and consistently elicited antibodies that interacted with blood stage parasites. It was observed that a dose of 50 microg was superior to a dose of 10 microg and that influenza pre-existing immunity improved the immunogenicity of FB-12 in rabbits. FB-12 production was successfully up-scaled and the immunogenicity of a vaccine formulation, produced according to the rules of Good Manufacturing Practice (GMP), was tested in mice and rabbits. All animals tested developed parasite-binding antibodies. Comparison of ELISA and IFA titers as well as the characterization of a panel of anti-FB-12 monoclonal antibodies indicated that at least the majority of antibodies specific for the virosomally formulated synthetic peptide were parasite cross-reactive. CONCLUSION: These results reconfirm the suitability of IRIVs as a carrier/adjuvant system for the induction of strong humoral immune responses against a wide range of synthetic peptide antigens. The virosomal formulation of the FB-12 peptidomimetic is suitable for use in humans and represents a candidate component for a virosomal multi-valent malaria subunit vaccine.


Asunto(s)
Antígenos de Protozoos/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Técnica del Anticuerpo Fluorescente Indirecta/métodos , Humanos , Ratones , Ratones Endogámicos BALB C , Orthomyxoviridae/química , Péptidos Cíclicos/inmunología , Conejos , Vacunas de Subunidad/inmunología , Vacunas de Virosoma/inmunología , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
8.
Hum Vaccin ; 4(2): 106-14, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18382133

RESUMEN

Presentation of synthetic peptides on immunopotentiating reconstituted influenza virosomes is a promising technology for subunit vaccine development. An optimized virosomally delivered peptide representing 5 NPNA repeats of P. falciparum circumsporozoite protein is highly immunogenic in mice. Antibodies against this peptide (UK-39) inhibit sporozoite invasion of human hepatocytes. A second peptide (AMA49-C1) based on domain III of apical membrane antigen 1, induces antibodies that inhibit blood-stage parasite growth in vitro. Here we show a detailed pre-clinical profiling of these virosomally formulated peptides alone and in combination in mice and rabbits. Two immunizations with virosomally formulated UK-39 or AMA49-C1 were enough to elicit high titers of parasite cross-reactive antibodies in both species. A low dose of 10 microg UK-39 was enough to induce maximal titers in rabbits. Higher doses of peptide did not increase antibody titers. In contrast, AMA49-C1 induced higher antibody titers with 25 and 50 microg peptide. Combination of UK-39 and AMA49- C1 on separate virosomes did not have any negative effect on anti-peptide antibody titers in mice or rabbits. No MHC restriction was observed in the development of humoral responses in outbred rabbits with different immunogenetic backgrounds. All vaccine formulations were safe in toxicity studies in rabbits and rats. Taken together, low amounts of synthetic peptides delivered on virosomes induced high antibody titers in mice and rabbits. Moreover, different peptides could be combined without interfering with individual anti-peptide responses, augmenting the value of this system for the development of a multivalent malaria vaccine.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Proteínas de la Membrana/inmunología , Proteínas Protozoarias/inmunología , Virosomas/inmunología , Animales , Antígenos de Protozoos/química , Reacciones Cruzadas , Relación Dosis-Respuesta Inmunológica , Diseño de Fármacos , Eritrocitos/parasitología , Femenino , Inmunización , Inmunoglobulina G/sangre , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/química , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Masculino , Proteínas de la Membrana/síntesis química , Proteínas de la Membrana/química , Ratones , Ratones Endogámicos BALB C , Péptidos/administración & dosificación , Péptidos/síntesis química , Péptidos/química , Péptidos/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/síntesis química , Proteínas Protozoarias/química , Ratas , Ratas Wistar
9.
Clin Immunol ; 127(2): 188-97, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18337175

RESUMEN

We have demonstrated recently in a phase Ia clinical trial that synthetic malaria peptides delivered by immuno-potentiating reconstituted influenza virosomes (IRIV) induced long-lived peptide-specific antibody responses in all volunteers. In the current ancillary study to this clinical trial we have investigated the cellular immune responses specific for IRIV and the surface bound synthetic malaria peptides tested. After vaccination, in 50% (8/16) of the volunteers at least one positive lymphoproliferative response specific for the 49mer peptide derived from the Plasmodium falciparum apical membrane antigen-1 (AMA-1) was observed with stimulation indices ranging from 2 to 4.5. All volunteers showed pre-existing IRIV specific cellular immunity assessed by ex vivo IFN-gamma ELISpot analysis and lymphoproliferation. The pre-existing influenza specific T cell responses did not interfere negatively with the induction of malaria peptide-specific humoral and cellular immune responses. Our results support the view that IRIV constitute a safe antigen delivery system for induction of peptide-specific immune responses in human populations.


Asunto(s)
Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Orthomyxoviridae/inmunología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Inmunidad Celular/inmunología , Malaria Falciparum/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Datos de Secuencia Molecular , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Virosoma/administración & dosificación , Vacunas de Virosoma/inmunología
10.
Cancer Lett ; 263(2): 291-301, 2008 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-18291576

RESUMEN

BACKGROUND: C-IRIV/PTR-4 is a novel anticancer vaccine construct composed of immune-reconstituted influenza virosomes (IRIV) assembled with the PTH-rP derived peptide (PTR)-4, a synthetic CTL epitope with HLA-A(*)02.01 amino acid binding motifs. This peptide is able to generate a human PTH-rP specific CTL response with anti-tumor activity in vitro and in mice. MATERIALS AND METHODS: We have investigated the immunological and preventive anti-tumor activity of C-IRIV/PTR-4 compared with the soluble PTR-4 peptide, in HHD mice inoculated with autologous PTH-rP+ tumor cells. RESULTS: Peptide vaccination with either a soluble and an IRIV formulation showed similar immunological activity and the ability to purge the tumor tissue of tumor cell clones able to produce the target antigen (PTR-rP). The most efficient protection from tumor growth was however observed in animals vaccinated with C-IRIV/PTR-4 in which an additional IRIV related anti-angiogenetic effect was detected in the tumor tissue. CONCLUSIONS: These results confirm the immunological activity of PTR-4 vaccination and suggest a more efficacious therapeutic potential of C-IRIV/PTR-4 against bone metastases and malignancies like breast, prostate and lung which very often over-express PTH-rP.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Virus de la Influenza A/inmunología , Neovascularización Patológica/tratamiento farmacológico , Proteína Relacionada con la Hormona Paratiroidea/administración & dosificación , Vacunas de Virosoma/farmacología , Animales , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Ratones , Ratones Transgénicos , Vacunas de Subunidad/uso terapéutico , Vacunas de Virosoma/uso terapéutico
11.
PLoS One ; 3(1): e1493, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-18231580

RESUMEN

BACKGROUND: Previous research indicates that a combination vaccine targeting different stages of the malaria life cycle is likely to provide the most effective malaria vaccine. This trial was the first to combine two existing vaccination strategies to produce a vaccine that induces immune responses to both the pre-erythrocytic and blood stages of the P. falciparum life cycle. METHODS: This was a Phase I/IIa study of a new combination malaria vaccine FFM ME-TRAP+PEV3A. PEV3A includes peptides from both the pre-erythrocytic circumsporozoite protein and the blood-stage antigen AMA-1. This study was conducted at the Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Oxford, UK. The participants were healthy, malaria naïve volunteers, from Oxford. The interventions were vaccination with PEV3A alone, or PEV3A+FFM ME-TRAP. The main outcome measure was protection from malaria in a sporozoite challenge model. Other outcomes included measures of parasite specific immune responses induced by either vaccine; and safety, assessed by collection of adverse event data. RESULTS: We observed evidence of blood stage immunity in PEV3A vaccinated volunteers, but no volunteers were completely protected from malaria. PEV3A induced high antibody titres, and antibodies bound parasites in immunofluorescence assays. Moreover, we observed boosting of the vaccine-induced immune response by sporozoite challenge. Immune responses induced by FFM ME-TRAP were unexpectedly low. The vaccines were safe, with comparable side effect profiles to previous trials. Although there was no sterile protection two major observations support an effect of the vaccine-induced response on blood stage parasites: (i) Lower rates of parasite growth were observed in volunteers vaccinated with PEV3A compared to unvaccinated controls (p = 0.012), and this was reflected in the PCR results from PEV3A vaccinated volunteers. These showed early control of parasitaemia by some volunteers in this group. One volunteer, who received PEV3A alone, was diagnosed very late, on day 20 compared to an average of 11.8 days in unvaccinated controls. (ii). Morphologically abnormal parasites were present in the blood of all (n = 24) PEV3A vaccinated volunteers, and in only 2/6 controls (p = 0.001). We describe evidence of vaccine-induced blood stage efficacy for the first time in a sporozoite challenge study.


Asunto(s)
Vacunas contra la Malaria/uso terapéutico , Malaria Falciparum/inmunología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Vacunas contra la Malaria/efectos adversos , Plasmodium falciparum/inmunología
12.
Expert Opin Drug Discov ; 3(4): 415-23, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23489097

RESUMEN

BACKGROUND: The development of a malaria vaccine represents one of the most important scientific public health challenges of our time. One possible approach is based on subunit vaccines that use distinct malarial antigens for which there is some evidence of protective immunity from epidemiological data in the field or animal challenge models. It is generally accepted that an effective malaria subunit vaccine will target antigens of several developmental stages of the parasite. OBJECTIVE: At present, the development of peptide-based vaccines is hampered by their poor immunogenicity and lack of in vivo stability of synthetic peptides and suitable antigen delivery systems driving appropriate immune responses in humans. Most importantly, the synthetic structures delivered have to mimic closely the corresponding native malaria protein to induce effective antibody responses. METHODS: Review of recent publications highlighting the design as well as preclinical and clinical development of conformationally constrained synthetic peptides of two malaria proteins delivered on the surface of influenza virosomes. RESULTS/CONCLUSION: The great potential of influenza virosomes as a flexible, human-compatible antigen delivery platform for the development of a multivalent malaria subunit vaccine is described.

13.
PLoS One ; 2(12): e1278, 2007 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18060072

RESUMEN

OBJECTIVES: Peptides delivered on the surface of influenza virosomes have been shown to induce solid humoral immune responses in experimental animals. High titers of peptide-specific antibodies were also induced in a phase 1a clinical trial in volunteers immunized with virosomal formulations of two peptides derived from the circumsporozoite protein (CSP) and the apical membrane antigen 1 (AMA-1) of Plasmodium falciparum. The main objective of this study was to perform a detailed immunological and functional analysis of the CSP-specific antibodies elicited in this phase 1a trial. METHODOLOGY/PRINCIPAL FINDINGS: 46 healthy malaria-naïve adults were immunized with virosomal formulations of two peptide-phosphatidylethanolamine conjugates, one derived from the NANP repeat region of P. falciparum CSP (designated UK-39) the other from P. falciparum AMA-1 (designated AMA49-C1). The two antigens were delivered in two different concentrations, alone and in combination. One group was immunized with empty virosomes as control. In this report we show a detailed analysis of the antibody response against UK-39. Three vaccinations with a 10 microg dose of UK-39 induced high titers of sporozoite-binding antibodies in all volunteers. This IgG response was affinity maturated and long-lived. Co-administration of UK-39 and AMA49-C1 loaded virosomes did not interfere with the immunogenicity of UK-39. Purified total IgG from UK-39 immunized volunteers inhibited sporozoite migration and invasion of hepatocytes in vitro. Sporozoite inhibition closely correlated with titers measured in immunogenicity assays. CONCLUSIONS: Virosomal delivery of a short, conformationally constrained peptide derived from P. falciparum CSP induced a long-lived parasite-inhibitory antibody response in humans. Combination with a second virosomally-formulated peptide derived from P. falciparum AMA-1 did not interfere with the immunogenicity of either peptide, demonstrating the potential of influenza virosomes as a versatile, human-compatible antigen delivery platform for the development of multivalent subunit vaccines. TRIAL REGISTRATION: ClinicalTrials.gov NCT00400101.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Vacunas contra la Malaria/inmunología , Virosomas/inmunología , Adulto , Animales , Western Blotting , Reacciones Cruzadas , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Plasmodium falciparum/inmunología
14.
PLoS One ; 2(10): e1018, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17925866

RESUMEN

BACKGROUND AND OBJECTIVES: Influenza virosomes represent an innovative human-compatible antigen delivery system that has already proven its suitability for subunit vaccine design. The aim of the study was to proof the concept that virosomes can also be used to elicit high titers of antibodies against synthetic peptides. The specific objective was to demonstrate the safety and immunogenicity of two virosome-formulated P. falciparum protein derived synthetic peptide antigens given in two different doses alone or in combination. METHODOLOGY/PRINCIPAL FINDINGS: The design was a single blind, randomized, placebo controlled, dose-escalating study involving 46 healthy Caucasian volunteers aged 18-45 years. Five groups of 8 subjects received virosomal formulations containing 10 microg or 50 microg of AMA 49-CPE, an apical membrane antigen-1 (AMA-1) derived synthetic phospatidylethanolamine (PE)-peptide conjugate or 10 ug or 50 ug of UK39, a circumsporozoite protein (CSP) derived synthetic PE-peptide conjugate or 50 ug of both antigens each. A control group of 6 subjects received unmodified virosomes. Virosomal formulations of the antigens (designated PEV301 and PEV302 for the AMA-1 and the CSP virosomal vaccine, respectively) or unmodified virosomes were injected i. m. on days 0, 60 and 180. In terms of safety, no serious or severe adverse events (AEs) related to the vaccine were observed. 11/46 study participants reported 16 vaccine related local AEs. Of these 16 events, all being pain, 4 occurred after the 1(st), 7 after the 2(nd) and 5 after the 3(rd) vaccination. 6 systemic AEs probably related to the study vaccine were reported after the 1(st) injection, 10 after the 2(nd) and 6 after the 3(rd). Generally, no difference in the distribution of the systemic AEs between either the doses applied (10 respectively 50 microg) or the synthetic antigen vaccines (PEV301 and PEV302) used for immunization was found. In terms of immunogenicity, both PEV301 and PEV302 elicited already after two injections a synthetic peptide-specific antibody response in all volunteers immunized with the appropriate dose. In the case of PEV301 the 50 microg antigen dose was associated with a higher mean antibody titer and seroconversion rate than the 10 microg dose. In contrast, for PEV302 mean titer and seroconversion rate were higher with the lower dose. Combined delivery of PEV301 and PEV302 did not interfere with the development of an antibody response to either of the two antigens. No relevant antibody responses against the two malaria antigens were observed in the control group receiving unmodified virosomes. CONCLUSIONS: The present study demonstrates that three immunizations with the virosomal malaria vaccine components PEV301 or/and PEV302 (containing 10 microg or 50 microg of antigen) are safe and well tolerated. At appropriate antigen doses seroconversion rates of 100% were achieved. Two injections may be sufficient for eliciting an appropriate immune response, at least in individuals with pre-existing anti-malarial immunity. These results justify further development of a final multi-stage virosomal vaccine formulation incorporating additional malaria antigens. TRIAL REGISTRATION: ClinicalTrials.gov NCT00400101.


Asunto(s)
Vacunas contra la Malaria/química , Malaria/prevención & control , Péptidos/química , Virosomas/química , Adolescente , Adulto , Animales , Antígenos Virales/química , Humanos , Fosfatidiletanolaminas/química , Placebos , Plasmodium falciparum/metabolismo , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento
15.
Expert Rev Vaccines ; 6(5): 711-21, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17931152

RESUMEN

Influenza virosomes are an efficient antigen carrier and adjuvant system that are approved for the use in human vaccines. Structurally, virosomes are spherical vesicles of approximately 150 nm in diameter, composed of a lipid membrane with integrated envelope proteins derived from influenza virus, predominantly hemagglutinin. The particle structure, together with the functions of hemagglutinin--receptor binding, pH-dependent fusion activity and immunostimulation--is responsible for the adjuvant effect of virosomes. In contrast to most other virus-like particles, virosomes are semisynthetic particles reconstituted in vitro from lipids and from virus-derived proteins. The production process has proven to be robust at industrial scale and fully compatible with Good Manufacturing Practice guidelines. At the same time, the formulation procedure is sufficiently flexible to allow modifications of the composition and structure for the intended use, including the positioning of the antigens of interest.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Vacunas contra la Influenza/administración & dosificación , Virosomas/administración & dosificación , Virosomas/inmunología , Animales , Humanos , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control
16.
Peptides ; 28(10): 2051-60, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17875342

RESUMEN

Serine repeat antigen-5 (SERA5) is a candidate antigen for inclusion into a malaria subunit vaccine. During merozoite release and reinvasion the 120 kDa SERA5 precursor protein (P120) is processed, and a complex consisting of an N-terminal 47 kDa (P47) and a C-terminal 18kDa (P18) processing product associates with the surface of merozoites. This complex is thought to be involved in merozoite invasion of and/or egress from host erythrocytes. Here we describe the synthesis and immunogenic properties of virosomally formulated synthetic phosphatidylethanolamine (PE)-peptide conjugates, incorporating amino acid sequence stretches from the N-terminus of Plasmodium falciparum SERA5. Choosing an appropriate sequence was crucial for the development of a peptide that elicited high titers of parasite cross-reactive antibodies in mice. Monoclonal antibodies (mAbs) raised against the optimized peptide FB-23 incorporating amino acids 57-94 of SERA5 bound to both P120 and to P47. Western blotting analysis proved for the first time the presence of SERA5 P47 in sporozoites. In immunofluorescence assays, the mAbs stained SERA5 in all its predicted localizations. The virosomal formulation of peptide FB-23 is suitable for use in humans and represents a candidate component for a multi-valent malaria subunit vaccine targeting both sporozoites and blood stage parasites.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Antígenos de Protozoos/inmunología , Merozoítos/química , Plasmodium falciparum/inmunología , Esporozoítos/química , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/química , Western Blotting , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Plasmodium falciparum/crecimiento & desarrollo
17.
Vaccine ; 25(41): 7065-74, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17766014

RESUMEN

The purpose of a vaccine is the induction of effective cellular and/or humoral immune responses against antigens. Because defined antigens are often poor immunogens when administered alone, an adjuvant is required to potentiate the immune response. Most of these adjuvants are designed to induce humoral immune responses, including immunopotentiating reconstituted influenza virosomes (IRIVs). IRIVs are one of the few adjuvants currently licensed for human use with the advantage of an excellent safety profile. To induce a potent cytotoxic T lymphocyte (CTL) immune response CTL epitopes have to be encapsulated into IRIVs. However, the existing encapsulation methods are inefficient or rather laborious. We have developed and characterised a new generation of influenza virosomes (TIRIVs) that induced both, strong CTL and antibody responses against specific antigens of choice. In addition, these virosomes were stabilised and offer the possibility of lyophilisation while retaining all their structural, functional and immunogenic properties after reconstitution. TIRIVs induce strong cellular and humoral immune responses and are a versatile and efficient carrier system with adjuvant properties for a variety of antigens. TIRIVs are not only stabilised but also allow easy formulation of new and/or labile T cell and B cell antigens. Considering their immunogenic properties, their flexibility and their superior storage characteristics TIRIVs provide a versatile technology platform for any vaccination strategy.


Asunto(s)
Formación de Anticuerpos , Antígenos/inmunología , Citotoxicidad Inmunológica , Orthomyxoviridae/inmunología , Vacunación/métodos , Vacunas de Virosoma/inmunología , Animales , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Orthomyxoviridae/genética , Vacunas de Virosoma/genética
18.
Antiviral Res ; 76(1): 75-85, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17617476

RESUMEN

Cervical cancer results from cervical infection by human papillomaviruses (HPV), especially HPV16. Intramuscular administrations of HPV16 virus-like particle (VLP) vaccines have been shown to induce strong neutralizing antibody responses and protect women against genital HPV16 infection and associated lesions. However, an alternative route of administration that avoids parenteral injection might facilitate vaccine implementation, particularly in developing countries which account for the majority of the worldwide cases of cervical cancer. In addition, inducing mucosal immunity could partially overcome the substantial variation in HPV16 antibodies at the cervix seen in ovulating women. Aerosol vaccination with HPV16 VLPs was previously shown to be immunogenic in mice and in women. Here, we examine whether exposure to other respiratory viral antigens may interfere with the HPV16 VLP-specific humoral response and whether two known mucosal adjuvants, CpG oligodeoxynucleotides and a natural non-toxic Escherichia coli heat-labile enterotoxin (HLT), can enhance the immunogenicity of airway immunization (nasal or aerosol-like) of mice with HPV16 VLPs. Our data show that HLT can significantly improve anti-VLP humoral responses in serum and mucosal secretions, as well as VLP-specific proliferative responses and IFN-gamma production by CD8 T cells, and that recent exposure to influenza surface antigens can diminish mucosal, but not systemic, antibody responses to the VLPs.


Asunto(s)
Adyuvantes Inmunológicos , Enterotoxinas/inmunología , Papillomavirus Humano 16/inmunología , Inmunización , Infecciones por Papillomavirus/inmunología , Infecciones Tumorales por Virus/inmunología , Virión/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración por Inhalación , Aerosoles , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Enterotoxinas/metabolismo , Escherichia coli/metabolismo , Femenino , Calor , Inmunidad Mucosa , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Vacunas contra Papillomavirus
19.
Chem Biol ; 14(5): 577-87, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17524988

RESUMEN

The circumsporozoite protein (CSP) of Plasmodium falciparum is a leading candidate antigen for inclusion in a malaria subunit vaccine. We describe here the design of a conformationally constrained synthetic peptide, designated UK-39, which has structural and antigenic similarity to the NPNA-repeat region of native CSP. NMR studies on the antigen support the presence of helical turn-like structures within consecutive NPNA motifs in aqueous solution. Intramuscular delivery of UK-39 to mice and rabbits on the surface of reconstituted influenza virosomes elicited high titers of sporozoite crossreactive antibodies. Influenza virus proteins were crucially important for the immunostimulatory activity of the virosome-based antigen delivery system, as a liposomal formulation of UK-39 was not immunogenic. IgG antibodies elicited by UK-39 inhibited invasion of hepatocytes by P. falciparum sporozoites, but not by antigenically distinct P. yoelii sporozoites. Our approach to optimized virosome-formulated synthetic peptide vaccines should be generally applicable for other infectious and noninfectious diseases.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria/química , Vacunas contra la Malaria/inmunología , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Virosomas/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Química Farmacéutica , Reacciones Cruzadas , Diseño de Fármacos , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Hibridomas/inmunología , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Fosfatidiletanolaminas/química , Plasmodium falciparum/inmunología , Plasmodium falciparum/patogenicidad , Plasmodium yoelii/inmunología , Plasmodium yoelii/patogenicidad , Conejos , Relación Estructura-Actividad
20.
Vaccine ; 25(19): 3913-21, 2007 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-17336432

RESUMEN

Cytotoxic T lymphocytes (CTL) are crucial in viral clearance and tumor growth control. Thus the induction of CTL activity is an important aim in vaccine development. We investigate an innovative delivery system for peptide transfer to the MHC class I processing pathway of APC with the aim to trigger CTL in the context of an antitumoral response. The strategy relies on a novel antigen delivery system termed "chimeric immunopotentiating reconstituted influenza virosomes" (CIRIV) targeting plasmacytoid dendritic cells (PDC). By using virosomes containing encapsulated Melan-A peptide and a PDC line developed in our laboratory, we evaluated the response of Melan-A specific T cells. Virosomes have the capacity to bind PDC and are endocyted within vesicles in the cytosol. This endocytosis is inhibited by neuraminidase, suggesting that it is mediated by sialic acid present on cell surface. Furthermore, PDC loaded with Melan-A virosomes can induce a Melan-A specific T cell activation. Interestingly, they activate T cells with a better efficiency than PDC loaded with a free peptide and when PDC where previously activated by a TLR ligand. These results indicate that virosomes could be a suitable delivery system for tumor peptide in immunotherapy of cancer.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Melanoma/terapia , Proteínas de Neoplasias/inmunología , Virosomas/administración & dosificación , Antígenos de Neoplasias/inmunología , Humanos , Inmunoterapia Activa , Activación de Linfocitos , Antígeno MART-1 , Orthomyxoviridae/inmunología , Virosomas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA