Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Wien Klin Wochenschr ; 118(19-20 Suppl 3): 50-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17131241

RESUMEN

Malaria remains a serious cause of morbidity and mortality in millions of individuals each year. The development of widespread resistance of the parasite to drugs as well as resistance of the transmitting mosquito-vector to insecticides in combination with the poor economic situation of many malaria-endemic countries make the development of an effective and inexpensive treatment and prevention a main focus of research. Vaccines remain to be one of the most cost effective and feasible means of disease control and have remarkable success in the control of many infectious disease: eradication of small pox, virtual eradication of polio and the reduction of measles and diphtheria. Next generation vaccines should focus on specific antigens rather than whole inactivated or attenuated pathogens, since the requirements by regulatory authorities concerning safety are becoming more stringent over time. But sub-unit and in particular peptide-based vaccines are poorly immunogenic themselves, and alum represents only a sub-optimal adjuvant for recombinant proteins and synthetic peptides. This emphasizes the need for suitable carrier- and adjuvant systems promoting protective immune responses by delivering protein and peptide antigens in an appropriate conformation. Here, we review the development of a new approach combining peptide-based malaria vaccine candidate antigens with an immune stimulatory carrier-system based on influenza virosomes focusing on the induction of protective antibodies.


Asunto(s)
Antígenos de Protozoos/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium/inmunología , Vacunas de Virosoma/inmunología , Virosomas/inmunología , Adyuvantes Inmunológicos , Animales , Formación de Anticuerpos/inmunología , Liposomas , Activación de Linfocitos/inmunología , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
7.
J Mol Neurosci ; 27(2): 157-66, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16186626

RESUMEN

Active vaccination with amyloid peptides shows promise for the treatment and prevention of Alzheimer's disease (AD). Several studies in transgenic mouse models of AD have revealed the potency of vaccination to prevent or even clear amyloid plaques from mouse brain. However, the idea that soluble oligomeric species of beta-amyloid (Abeta), rather than plaques, trigger the disease has gained momentum, and current active vaccination strategies affect the levels of total or soluble brain Abeta little or not at all. We describe an active vaccination method based on Abeta1-16 presented on the surface of virosomes, which triggered a dramatic decrease in both soluble Abeta40 (75% reduction; p=0.01) and soluble Abeta42 (62% reduction; p=0.03) in a double transgenic mouse model of AD. Whereas Abeta40 and Abeta42 levels in the insoluble fraction tended to be reduced (by 30% and 27%, respectively), the number of thioflavine-S-positive amyloid plaques was not affected. The high specific antibody responses, obtained without eliciting T-cell reactivity, demonstrate that immunostimulating reconstituted influenza virosomes are a promising antigen carrier system against the neuropathology of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/inmunología , Placa Amiloide/inmunología , Vacunas de Virosoma/inmunología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Animales , Biomarcadores , Química Encefálica , Modelos Animales de Enfermedad , Inmunización , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Placa Amiloide/patología , Distribución Aleatoria
8.
Epigenetics ; 8(11): 1226-35, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24071829

RESUMEN

The adaptive immune system is involved in tumor establishment and aggressiveness. Tumors of the ovaries, an immune-privileged organ, spread via transceolomic routes and rarely to distant organs. This is contrary to tumors of non-immune privileged organs, which often disseminate hematogenously to distant organs. Epigenetics-based immune cell quantification allows direct comparison of the immune status in benign and malignant tissues and in blood. Here, we introduce the "cellular ratio of immune tolerance" (immunoCRIT) as defined by the ratio of regulatory T cells to total T lymphocytes. The immunoCRIT was analyzed on 273 benign tissue samples of colorectal, bronchial, renal and ovarian origin as well as in 808 samples from primary colorectal, bronchial, mammary and ovarian cancers. ImmunoCRIT is strongly increased in all cancerous tissues and gradually augmented strictly dependent on tumor aggressiveness. In peripheral blood of ovarian cancer patients, immunoCRIT incrementally increases from primary diagnosis to disease recurrence, at which distant metastases frequently occur. We postulate that non-pathological immunoCRIT values observed in peripheral blood of immune privileged ovarian tumor patients are sufficient to prevent hematogenous spread at primary diagnosis. Contrarily, non-immune privileged tumors establish high immunoCRIT in an immunological environment equivalent to the bloodstream and thus spread hematogenously to distant organs. In summary, our data suggest that the immunoCRIT is a powerful marker for tumor aggressiveness and disease dissemination.


Asunto(s)
Biomarcadores de Tumor/inmunología , Tolerancia Inmunológica , Neoplasias/inmunología , Neoplasias/patología , Adulto , Anciano , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Epigénesis Genética , Femenino , Humanos , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Linfocitos T/inmunología , Linfocitos T/patología , Adulto Joven
9.
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
10.
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
11.
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.

12.
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
13.
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
14.
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
15.
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
16.
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
17.
J Pept Sci ; 11(11): 707-12, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16059967

RESUMEN

Vaccines have been well accepted and used effectively for more than 100 years. Traditional vaccines are generally composed of whole inactivated or attenuated microorganisms that have lost their disease-causing properties. These classical prophylactic live vaccines evoke protective immune responses, but have often been associated with an unfavorable safety profile, as observed, for example, for smallpox and polio myelitis vaccines [1,2]. First improvements were subunit vaccines that do not focus on attenuation of whole organisms but concentrate on particular proteins. These vaccines are able to generate protective immune responses (e.g. diphtheria, tetanus, pertussis)3. However, next generation vaccines should focus on specific antigens (e.g. proteins, peptides), since the requirements by regulatory authorities to crude biological material are becoming more stringent over time. An increasing number of such antigens capable of inducing protective humoral or cellular immune responses have been identified in the last few years. But most of these are weak immunogens. This reemphasizes the need for adjuvants to promote a potent immune response and also for delivery antigens to the immune system in an appropriate way (carrier capability). Here we review a new approach for prophylactic and therapeutic vaccines, which focuses on the induction of highly specific immune responses directed against antigen-derived peptides using a suitable carrier system.


Asunto(s)
Orthomyxoviridae/inmunología , Péptidos/administración & dosificación , Vacunas de Virosoma/administración & dosificación , Animales , Anticuerpos Antivirales/biosíntesis , Humanos , Inmunidad Celular , Péptidos/inmunología , Vacunas de Virosoma/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA