Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 30(12): 3639-3657, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-35949171

RESUMEN

Adenovirus vector vaccines have been widely and successfully deployed in response to coronavirus disease 2019 (COVID-19). However, despite inducing potent T cell immunity, improvement of vaccine-specific antibody responses upon homologous boosting is modest compared with other technologies. Here, we describe a system enabling modular decoration of adenovirus capsid surfaces with antigens and demonstrate potent induction of humoral immunity against these displayed antigens. Ligand attachment via a covalent bond was achieved using a protein superglue, DogTag/DogCatcher (similar to SpyTag/SpyCatcher), in a rapid and spontaneous reaction requiring only co-incubation of ligand and vector components. DogTag was inserted into surface-exposed loops in the adenovirus hexon protein to allow attachment of DogCatcher-fused ligands on virus particles. Efficient coverage of the capsid surface was achieved using various ligands, with vector infectivity retained in each case. Capsid decoration shielded particles from vector neutralizing antibodies. In prime-boost regimens, adenovirus vectors decorated with the receptor-binding domain of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike induced >10-fold higher SARS-CoV-2 neutralization titers compared with an undecorated vector encoding spike. Importantly, decorated vectors achieved equivalent or superior T cell immunogenicity against encoded antigens compared with undecorated vectors. We propose capsid decoration using protein superglues as a novel strategy to improve efficacy and boostability of adenovirus-based vaccines and therapeutics.


Asunto(s)
Vacunas contra el Adenovirus , COVID-19 , Humanos , SARS-CoV-2 , Inmunidad Humoral , Ligandos , COVID-19/prevención & control
3.
Nat Microbiol ; 6(8): 1031-1042, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34282309

RESUMEN

The antiviral cytokine interferon activates expression of interferon-stimulated genes to establish an antiviral state. Myxovirus resistance 2 (MX2, also known as MxB) is an interferon-stimulated gene that inhibits the nuclear import of HIV-1 and interacts with the viral capsid and cellular nuclear transport machinery. Here, we identified the myosin light chain phosphatase (MLCP) subunits myosin phosphatase target subunit 1 (MYPT1) and protein phosphatase 1 catalytic subunit-ß (PPP1CB) as positively-acting regulators of MX2, interacting with its amino-terminal domain. We demonstrated that serine phosphorylation of the N-terminal domain at positions 14, 17 and 18 suppresses MX2 antiviral function, prevents interactions with the HIV-1 capsid and nuclear transport factors, and is reversed by MLCP. Notably, serine phosphorylation of the N-terminal domain also impedes MX2-mediated inhibition of nuclear import of cellular karyophilic cargo. We also found that interferon treatment reduces levels of phosphorylation at these serine residues and outline a homeostatic regulatory mechanism in which repression of MX2 by phosphorylation, together with MLCP-mediated dephosphorylation, balances the deleterious effects of MX2 on normal cell function with innate immunity against HIV-1.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/inmunología , Inmunidad Innata , Proteínas de Resistencia a Mixovirus/química , Proteínas de Resistencia a Mixovirus/inmunología , Secuencias de Aminoácidos , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Células HeLa , Humanos , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/inmunología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Fosforilación , Dominios Proteicos , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/inmunología , Serina/metabolismo
4.
Cell Rep ; 29(7): 1923-1933.e3, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31722207

RESUMEN

Myxovirus resistance 2 (MX2/MXB) is an interferon (IFN)-induced HIV-1 restriction factor that inhibits viral nuclear DNA accumulation. The amino-terminal domain of MX2 binds the viral capsid and is essential for inhibition. Using in vitro assembled Capsid-Nucleocapsid (CANC) complexes as a surrogate for the HIV-1 capsid lattice, we reveal that the GTPase (G) domain of MX2 contains a second, independent capsid-binding site. The importance of this interaction was addressed in competition assays using the naturally occurring non-antiviral short isoform of MX2 that lacks the amino-terminal 25 amino acids. Specifically, these experiments show that the G domain enhances MX2 function, and the foreshortened isoform acts as a functional suppressor of the full-length protein in a G-domain-dependent manner. The interaction of MX2 with its HIV-1 capsid substrate is therefore multi-faceted: there are dual points of contact that, together with protein oligomerization, contribute to the complexity of MX2 regulation.


Asunto(s)
Cápside/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Proteínas de Resistencia a Mixovirus/metabolismo , Infecciones por VIH/genética , VIH-1/genética , Células HeLa , Humanos , Proteínas de Resistencia a Mixovirus/genética , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
PLoS Pathog ; 14(11): e1007408, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30496303

RESUMEN

Human myxovirus resistance 2 (MX2/MXB) is an interferon-induced post-entry inhibitor of human immunodeficiency virus type-1 (HIV-1) infection. While the precise mechanism of viral inhibition remains unclear, MX2 is localized to the nuclear envelope, and blocks the nuclear import of viral cDNAs. The amino-terminus of MX2 (N-MX2) is essential for anti-viral function, and mutation of a triple arginine motif at residues 11 to 13 abrogates anti-HIV-1 activity. In this study, we sought to investigate the role of N-MX2 in anti-viral activity by identifying functionally relevant host-encoded interaction partners through yeast-two-hybrid screening. Remarkably, five out of seven primary candidate interactors were nucleoporins or nucleoporin-like proteins, though none of these candidates were identified when screening with a mutant RRR11-13A N-MX2 fragment. Interactions were confirmed by co-immunoprecipitation, and RNA silencing experiments in cell lines and primary CD4+ T cells demonstrated that multiple components of the nuclear pore complex and nuclear import machinery can impact MX2 anti-viral activity. In particular, the phenylalanine-glycine (FG) repeat containing cytoplasmic filament nucleoporin NUP214, and transport receptor transportin-1 (TNPO1) were consistently required for full MX2, and interferon-mediated, anti-viral function. Both proteins were shown to interact with the triple arginine motif, and confocal fluorescence microscopy revealed that their simultaneous depletion resulted in diminished MX2 accumulation at the nuclear envelope. We therefore propose a model whereby multiple components of the nuclear import machinery and nuclear pore complex help position MX2 at the nuclear envelope to promote MX2-mediated restriction of HIV-1.


Asunto(s)
Infecciones por VIH/metabolismo , VIH-1/fisiología , Proteínas de Resistencia a Mixovirus/metabolismo , Transporte Activo de Núcleo Celular , Antivirales/metabolismo , Células HEK293 , Infecciones por VIH/virología , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Interferones/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Membrana Nuclear/metabolismo , Poro Nuclear/metabolismo , Poro Nuclear/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Replicación Viral , beta Carioferinas/metabolismo
6.
Sci Rep ; 6: 18848, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26743316

RESUMEN

Transmission-blocking vaccines (TBV) target the sexual-stages of the malaria parasite in the mosquito midgut and are widely considered to be an essential tool for malaria elimination. High-titer functional antibodies are required against target antigens to achieve effective transmission-blocking activity. We have fused Pfs25, the leading malaria TBV candidate antigen to IMX313, a molecular adjuvant and expressed it both in ChAd63 and MVA viral vectors and as a secreted protein-nanoparticle. Pfs25-IMX313 expressed from viral vectors or as a protein-nanoparticle is significantly more immunogenic and gives significantly better transmission-reducing activity than monomeric Pfs25. In addition, we demonstrate that the Pfs25-IMX313 protein-nanoparticle leads to a qualitatively improved antibody response in comparison to soluble Pfs25, as well as to significantly higher germinal centre (GC) responses. These results demonstrate that antigen multimerization using IMX313 is a very promising strategy to enhance antibody responses against Pfs25, and that Pfs25-IMX313 is a highly promising TBV candidate vaccine.


Asunto(s)
Adyuvantes Inmunológicos/genética , Anticuerpos Antiprotozoarios/biosíntesis , Inmunogenicidad Vacunal , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Culicidae/efectos de los fármacos , Culicidae/parasitología , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/inmunología , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Humanos , Insectos Vectores/efectos de los fármacos , Insectos Vectores/parasitología , Estadios del Ciclo de Vida/efectos de los fármacos , Estadios del Ciclo de Vida/inmunología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos BALB C , Pichia/genética , Pichia/metabolismo , Plásmidos/química , Plásmidos/inmunología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Proteínas Protozoarias/administración & dosificación , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunación , Vacunas Sintéticas
7.
J Virol ; 90(1): 22-32, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26446602

RESUMEN

UNLABELLED: Human myxovirus resistance 2 (MX2/MXB) is an interferon-stimulated gene (ISG) and was recently identified as a late postentry suppressor of human immunodeficiency virus type 1 (HIV-1) infection, inhibiting the nuclear accumulation of viral cDNAs. Although the HIV-1 capsid (CA) protein is believed to be the viral determinant of MX2-mediated inhibition, the precise mechanism of antiviral action remains unclear. The MX family of dynamin-like GTPases also includes MX1/MXA, a well-studied inhibitor of a range of RNA and DNA viruses, including influenza A virus (FLUAV) and hepatitis B virus but not retroviruses. MX1 and MX2 are closely related and share similar domain architectures and structures. However, MX2 possesses an extended N terminus that is essential for antiviral function and confers anti-HIV-1 activity on MX1 [MX1(NMX2)]. Higher-order oligomerization is required for the antiviral activity of MX1 against FLUAV, with current models proposing that MX1 forms ring structures that constrict around viral nucleoprotein complexes. Here, we performed structure-function studies to investigate the requirements for oligomerization of both MX2 and chimeric MX1(NMX2) for the inhibition of HIV-1 infection. The oligomerization state of mutated proteins with amino acid substitutions at multiple putative oligomerization interfaces was assessed using a combination of covalent cross-linking and coimmunoprecipitation. We show that while monomeric MX2 and MX1(NMX2) mutants are not antiviral, higher-order oligomerization does not appear to be required for full antiviral activity of either protein. We propose that lower-order oligomerization of MX2 is sufficient for the effective inhibition of HIV-1. IMPORTANCE: Interferon plays an important role in the control of virus replication during acute infection in vivo. Recently, cultured cell experiments identified human MX2 as a key effector in the interferon-mediated postentry block to HIV-1 infection. MX2 is a member of a family of large dynamin-like GTPases that includes MX1/MXA, a closely related interferon-inducible inhibitor of several viruses, including FLUAV, but not HIV-1. MX GTPases form higher-order oligomeric structures, and the oligomerization of MX1 is required for inhibitory activity against many of its viral targets. Through structure-function studies, we report that monomeric mutants of MX2 do not inhibit HIV-1. However, in contrast to MX1, oligomerization beyond dimer assembly does not seem to be required for the antiviral activity of MX2, implying that fundamental differences exist between the antiviral mechanisms employed by these closely related proteins.


Asunto(s)
VIH-1/inmunología , VIH-1/fisiología , Interacciones Huésped-Patógeno , Inmunidad Innata , Proteínas de Resistencia a Mixovirus/metabolismo , Multimerización de Proteína , Replicación Viral , Sustitución de Aminoácidos , Línea Celular , Análisis Mutacional de ADN , Humanos , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas de Resistencia a Mixovirus/genética , Conformación Proteica
8.
Sci Rep ; 5: 16756, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26576856

RESUMEN

Replication defective adenoviruses are promising vectors for the delivery of vaccine antigens. However, the potential of a vector to elicit transgene-specific adaptive immune responses is largely dependent on the viral serotype used. HAdV-5 (Human adenovirus C) vectors are more immunogenic than chimpanzee adenovirus vectors from species Human adenovirus E (ChAdOx1 and AdC68) in mice, though the mechanisms responsible for these differences in immunogenicity remain poorly understood. In this study, superior immunogenicity was associated with markedly higher levels of transgene expression in vivo, particularly within draining lymph nodes. To investigate the viral factors contributing to these phenotypes, we generated recombinant ChAdOx1 vectors by exchanging components of the viral capsid reported to be principally involved in cell entry with the corresponding sequences from HAdV-5. Remarkably, pseudotyping with the HAdV-5 fiber and/or penton RGD loop had little to no effect on in vivo transgene expression or transgene-specific adaptive immune responses despite considerable species-specific sequence heterogeneity in these components. Our results suggest that mechanisms governing vector transduction after intramuscular administration in mice may be different from those described in vitro.


Asunto(s)
Adenoviridae/inmunología , Adenovirus Humanos/inmunología , Proteínas de la Cápside/inmunología , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Receptores de Orexina/genética , Adenoviridae/genética , Adenovirus Humanos/genética , Animales , Anticuerpos Antivirales/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proteínas de la Cápside/química , Expresión Génica , Vectores Genéticos/administración & dosificación , Humanos , Inmunidad , Inmunización , Inyecciones Intramusculares , Ratones , Pan troglodytes , Transducción Genética , Transgenes , Vacunas/genética , Vacunas/inmunología
9.
Vaccine ; 33(9): 1121-8, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25629523

RESUMEN

Adenovirus vaccine vectors generated from new viral serotypes are routinely screened in pre-clinical laboratory animal models to identify the most immunogenic and efficacious candidates for further evaluation in clinical human and veterinary settings. Here, we show that studies in a laboratory species do not necessarily predict the hierarchy of vector performance in other mammals. In mice, after intramuscular immunization, HAdV-5 (Human adenovirus C) based vectors elicited cellular and humoral adaptive responses of higher magnitudes compared to the chimpanzee adenovirus vectors ChAdOx1 and AdC68 from species Human adenovirus E. After HAdV-5 vaccination, transgene specific IFN-γ(+) CD8(+) T cell responses reached peak magnitude later than after ChAdOx1 and AdC68 vaccination, and exhibited a slower contraction to a memory phenotype. In cattle, cellular and humoral immune responses were at least equivalent, if not higher, in magnitude after ChAdOx1 vaccination compared to HAdV-5. Though we have not tested protective efficacy in a disease model, these findings have important implications for the selection of candidate vectors for further evaluation. We propose that vaccines based on ChAdOx1 or other Human adenovirus E serotypes could be at least as immunogenic as current licensed bovine vaccines based on HAdV-5.


Asunto(s)
Adenoviridae/genética , Portadores de Fármacos , Vectores Genéticos , Proteínas Recombinantes/inmunología , Transgenes , Vacunas Virales/inmunología , Animales , Animales de Laboratorio , Linfocitos T CD8-positivos/inmunología , Bovinos , Interferón gamma/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
10.
Virol J ; 10: 349, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24304565

RESUMEN

BACKGROUND: Rift Valley Fever (RVF) is a viral zoonosis that historically affects livestock production and human health in sub-Saharan Africa, though epizootics have also occurred in the Arabian Peninsula. Whilst an effective live-attenuated vaccine is available for livestock, there is currently no licensed human RVF vaccine. Replication-deficient chimpanzee adenovirus (ChAd) vectors are an ideal platform for development of a human RVF vaccine, given the low prevalence of neutralizing antibodies against them in the human population, and their excellent safety and immunogenicity profile in human clinical trials of vaccines against a wide range of pathogens. METHODS: Here, in BALB/c mice, we evaluated the immunogenicity and efficacy of a replication-deficient chimpanzee adenovirus vector, ChAdOx1, encoding the RVF virus envelope glycoproteins, Gn and Gc, which are targets of virus neutralizing antibodies. The ChAdOx1-GnGc vaccine was assessed in comparison to a replication-deficient human adenovirus type 5 vector encoding Gn and Gc (HAdV5-GnGc), a strategy previously shown to confer protective immunity against RVF in mice. RESULTS: A single immunization with either of the vaccines conferred protection against RVF virus challenge eight weeks post-immunization. Both vaccines elicited RVF virus neutralizing antibody and a robust CD8+ T cell response. CONCLUSIONS: Together the results support further development of RVF vaccines based on replication-deficient adenovirus vectors, with ChAdOx1-GnGc being a potential candidate for use in future human clinical trials.


Asunto(s)
Adenoviridae/genética , Portadores de Fármacos , Vectores Genéticos , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/genética , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
11.
Int J Parasitol ; 43(11): 869-74, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23872520

RESUMEN

The mosquito innate immune response is able to clear the majority of Plasmodium parasites. This immune clearance is controlled by a number of regulatory molecules including serine protease inhibitors (serpins). To determine whether such molecules could represent a novel target for a malaria transmission-blocking vaccine, we vaccinated mice with Anopheles gambiae serpin-2. Antibodies against Anopheles gambiae serpin-2 significantly reduced the infection of a heterologous Anopheles species (Anopheles stephensi) by Plasmodium berghei, however this effect was not observed with Plasmodium falciparum. Therefore, this approach of targeting regulatory molecules of the mosquito immune system may represent a novel approach to transmission-blocking malaria vaccines.


Asunto(s)
Anopheles/parasitología , Proteínas de Insectos/inmunología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/inmunología , Serpinas/inmunología , Animales , Inmunidad Innata , Proteínas de Insectos/antagonistas & inhibidores , Ratones , Serpinas/metabolismo
12.
Sci Rep ; 3: 1706, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23609325

RESUMEN

Rodent malaria species Plasmodium yoelii and P. chabaudi have been widely used to validate vaccine approaches targeting blood-stage merozoite antigens. However, increasing data suggest the P. berghei rodent malaria may be able to circumvent vaccine-induced anti-merozoite responses. Here we confirm a failure to protect against P. berghei, despite successful antibody induction against leading merozoite antigens using protein-in-adjuvant or viral vectored vaccine delivery. No subunit vaccine approach showed efficacy in mice following immunization and challenge with the wild-type P. berghei strains ANKA or NK65, or against a chimeric parasite line encoding a merozoite antigen from P. falciparum. Protection was not improved in knockout mice lacking the inhibitory Fc receptor CD32b, nor against a Δsmac P. berghei parasite line with a non-sequestering phenotype. An improved understanding of the mechanisms responsible for protection, or failure of protection, against P. berghei merozoites could guide the development of an efficacious vaccine against P. falciparum.


Asunto(s)
Formación de Anticuerpos/inmunología , Antimaláricos/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Merozoítos/inmunología , Plasmodium berghei/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Línea Celular , Femenino , Células HEK293 , Humanos , Inmunización/métodos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Conejos , Receptores de IgG/inmunología , Roedores/inmunología
13.
PLoS One ; 7(7): e40385, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22808149

RESUMEN

Recombinant adenoviruses are among the most promising tools for vaccine antigen delivery. Recently, the development of new vectors has focused on serotypes to which the human population is less exposed in order to circumvent pre-existing anti vector immunity. This study describes the derivation of a new vaccine vector based on a chimpanzee adenovirus, Y25, together with a comparative assessment of its potential to elicit transgene product specific immune responses in mice. The vector was constructed in a bacterial artificial chromosome to facilitate genetic manipulation of genomic clones. In order to conduct a fair head-to-head immunological comparison of multiple adenoviral vectors, we optimised a method for accurate determination of infectious titre, since this parameter exhibits profound natural variability and can confound immunogenicity studies when doses are based on viral particle estimation. Cellular immunogenicity of recombinant E1 E3-deleted vector ChAdY25 was comparable to that of other species E derived chimpanzee adenovirus vectors including ChAd63, the first simian adenovirus vector to enter clinical trials in humans. Furthermore, the prevalence of virus neutralizing antibodies (titre >1:200) against ChAdY25 in serum samples collected from two human populations in the UK and Gambia was particularly low compared to published data for other chimpanzee adenoviruses. These findings support the continued development of new chimpanzee adenovirus vectors, including ChAdY25, for clinical use.


Asunto(s)
Adenovirus de los Simios/genética , Adenovirus de los Simios/inmunología , Vectores Genéticos/genética , Pan troglodytes/inmunología , Pan troglodytes/virología , Vacunas contra el Adenovirus/inmunología , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Adenovirus de los Simios/patogenicidad , Animales , Anticuerpos Neutralizantes/inmunología , Secuencia de Bases , Femenino , Gambia/epidemiología , Genes Virales/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Filogenia , Estudios Seroepidemiológicos , Volumetría , Reino Unido/epidemiología , Virión/genética
14.
PLoS One ; 7(2): e31208, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363582

RESUMEN

BACKGROUND: Traditionally, vaccine development against the blood-stage of Plasmodium falciparum infection has focused on recombinant protein-adjuvant formulations in order to induce high-titer growth-inhibitory antibody responses. However, to date no such vaccine encoding a blood-stage antigen(s) alone has induced significant protective efficacy against erythrocytic-stage infection in a pre-specified primary endpoint of a Phase IIa/b clinical trial designed to assess vaccine efficacy. Cell-mediated responses, acting in conjunction with functional antibodies, may be necessary for immunity against blood-stage P. falciparum. The development of a vaccine that could induce both cell-mediated and humoral immune responses would enable important proof-of-concept efficacy studies to be undertaken to address this question. METHODOLOGY: We conducted a Phase Ia, non-randomized clinical trial in 16 healthy, malaria-naïve adults of the chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient viral vectored vaccines encoding two alleles (3D7 and FVO) of the P. falciparum blood-stage malaria antigen; apical membrane antigen 1 (AMA1). ChAd63-MVA AMA1 administered in a heterologous prime-boost regime was shown to be safe and immunogenic, inducing high-level T cell responses to both alleles 3D7 (median 2036 SFU/million PBMC) and FVO (median 1539 SFU/million PBMC), with a mixed CD4(+)/CD8(+) phenotype, as well as substantial AMA1-specific serum IgG responses (medians of 49 µg/mL and 41 µg/mL for 3D7 and FVO AMA1 respectively) that demonstrated growth inhibitory activity in vitro. CONCLUSIONS: ChAd63-MVA is a safe and highly immunogenic delivery platform for both alleles of the AMA1 antigen in humans which warrants further efficacy testing. ChAd63-MVA is a promising heterologous prime-boost vaccine strategy that could be applied to numerous other diseases where strong cellular and humoral immune responses are required for protection. TRIAL REGISTRATION: ClinicalTrials.gov NCT01095055.


Asunto(s)
Adenovirus de los Simios/genética , Antígenos de Protozoos/inmunología , Vectores Genéticos/genética , Vacunas contra la Malaria/efectos adversos , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Virus Vaccinia/genética , Adolescente , Adulto , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antiprotozoarios/inmunología , Ensayo de Immunospot Ligado a Enzimas , Femenino , Humanos , Inmunización , Interferón gamma/inmunología , Estadios del Ciclo de Vida , Malaria Falciparum/inmunología , Masculino , Persona de Mediana Edad , Plasmodium falciparum/crecimiento & desarrollo , Linfocitos T/inmunología , Adulto Joven
15.
Mol Ther ; 19(12): 2269-76, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21862998

RESUMEN

Efficacy trials of antibody-inducing protein-in-adjuvant vaccines targeting the blood-stage Plasmodium falciparum malaria parasite have so far shown disappointing results. The induction of cell-mediated responses in conjunction with antibody responses is thought to be one alternative strategy that could achieve protective efficacy in humans. Here, we prepared chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient vectors encoding the well-studied P. falciparum blood-stage malaria antigen merozoite surface protein 1 (MSP1). A phase Ia clinical trial was conducted in healthy adults of a ChAd63-MVA MSP1 heterologous prime-boost immunization regime. The vaccine was safe and generally well tolerated. Fewer systemic adverse events (AEs) were observed following ChAd63 MSP1 than MVA MSP1 administration. Exceptionally strong T-cell responses were induced, and these displayed a mixed of CD4(+) and CD8(+) phenotype. Substantial MSP1-specific serum immunoglobulin G (IgG) antibody responses were also induced, which were capable of recognizing native parasite antigen, but these did not reach titers sufficient to neutralize P. falciparum parasites in vitro. This viral vectored vaccine regime is thus a leading approach for the induction of strong cellular and humoral immunogenicity against difficult disease targets in humans. Further studies are required to assess whether this strategy can achieve protective efficacy against blood-stage malaria infection.


Asunto(s)
Adenoviridae/genética , Linfocitos T CD4-Positivos/inmunología , Vectores Genéticos/uso terapéutico , Malaria Falciparum/inmunología , Malaria Falciparum/terapia , Proteína 1 de Superficie de Merozoito/inmunología , Virus Vaccinia/genética , Adyuvantes Inmunológicos , Adulto , Animales , Anticuerpos Antiprotozoarios/inmunología , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunidad Celular , Inmunoglobulina G/inmunología , Memoria Inmunológica , Macaca mulatta , Malaria Falciparum/sangre , Masculino , Proteína 1 de Superficie de Merozoito/sangre , Proteína 1 de Superficie de Merozoito/genética , Ratones , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Vacunación , Adulto Joven
16.
PLoS One ; 6(6): e20977, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698193

RESUMEN

BACKGROUND: Apical membrane antigen 1 (AMA1) is a leading candidate vaccine antigen against blood-stage malaria, although to date numerous clinical trials using mainly protein-in-adjuvant vaccines have shown limited success. Here we describe the pre-clinical development and optimization of recombinant human and simian adenoviral (AdHu5 and ChAd63) and orthopoxviral (MVA) vectors encoding transgene inserts for Plasmodium falciparum AMA1 (PfAMA1). METHODOLOGY/PRINCIPAL FINDINGS: AdHu5-MVA prime-boost vaccination in mice and rabbits using these vectors encoding the 3D7 allele of PfAMA1 induced cellular immune responses as well as high-titer antibodies that showed growth inhibitory activity (GIA) against the homologous but not heterologous parasite strains. In an effort to overcome the issues of PfAMA1 antigenic polymorphism and pre-existing immunity to AdHu5, a simian adenoviral (ChAd63) vector and MVA encoding two alleles of PfAMA1 were developed. This antigen, composed of the 3D7 and FVO alleles of PfAMA1 fused in tandem and with expression driven by a single promoter, was optimized for antigen secretion and transmembrane expression. These bi-allelic PfAMA1 vaccines, when administered to mice and rabbits, demonstrated comparable immunogenicity to the mono-allelic vaccines and purified serum IgG now showed GIA against the two divergent strains of P. falciparum encoded in the vaccine. CD8(+) and CD4(+) T cell responses against epitopes that were both common and unique to the two alleles of PfAMA1 were also measured in mice. CONCLUSIONS/SIGNIFICANCE: Optimized transgene inserts encoding two divergent alleles of the same antigen can be successfully inserted into adeno- and pox-viral vaccine vectors. Adenovirus-MVA immunization leads to the induction of T cell responses common to both alleles, as well as functional antibody responses that are effective against both of the encoded strains of P. falciparum in vitro. These data support the further clinical development of these vaccine candidates in Phase I/IIa clinical trials.


Asunto(s)
Adenoviridae/genética , Alelos , Vectores Genéticos , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Orthopoxvirus/genética , Plasmodium falciparum/genética , Transgenes , Animales , Vacunas contra la Malaria/genética , Ratones , Plasmodium falciparum/inmunología , Conejos
17.
J Immunol ; 185(12): 7583-95, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21098232

RESUMEN

Protein-in-adjuvant formulations and viral-vectored vaccines encoding blood-stage malaria Ags have shown efficacy in rodent malaria models and in vitro assays against Plasmodium falciparum. Abs and CD4(+) T cell responses are associated with protective efficacy against blood-stage malaria, whereas CD8(+) T cells against some classical blood-stage Ags can also have a protective effect against liver-stage parasites. No subunit vaccine strategy alone has generated demonstrable high-level efficacy against blood-stage infection in clinical trials. The induction of high-level Ab responses, as well as potent T and B cell effector and memory populations, is likely to be essential to achieve immediate and sustained protective efficacy in humans. This study describes in detail the immunogenicity of vaccines against P. falciparum apical membrane Ag 1 in rhesus macaques (Macaca mulatta), including the chimpanzee adenovirus 63 (AdCh63), the poxvirus modified vaccinia virus Ankara (MVA), and protein vaccines formulated in Alhydrogel or CoVaccine HT adjuvants. AdCh63-MVA heterologous prime-boost immunization induces strong and long-lasting multifunctional CD8(+) and CD4(+) T cell responses that exhibit a central memory-like phenotype. Three-shot (AdCh63-MVA-protein) or two-shot (AdCh63-protein) regimens induce memory B cells and high-titer functional IgG responses that inhibit the growth of two divergent strains of P. falciparum in vitro. Prior immunization with adenoviral vectors of alternative human or simian serotype does not affect the immunogenicity of the AdCh63 apical membrane Ag 1 vaccine. These data encourage the further clinical development and coadministration of protein and viral vector vaccine platforms in an attempt to induce broad cellular and humoral immune responses against blood-stage malaria Ags in humans.


Asunto(s)
Adenoviridae , Adyuvantes Inmunológicos , Antígenos de Protozoos/inmunología , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Poxviridae , Proteínas Protozoarias/inmunología , Virus Vaccinia , Animales , Anticuerpos Antiprotozoarios/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos , Inmunoglobulina G/inmunología , Memoria Inmunológica/inmunología , Macaca mulatta , Malaria Falciparum/inmunología , Ratones , Vacunas de Subunidad/inmunología
18.
Vaccine ; 28(44): 7167-78, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20937436

RESUMEN

Subunit vaccination modalities tend to induce particular immune effector responses. Viral vectors are well known for their ability to induce strong T cell responses, while protein-adjuvant vaccines have been used primarily for induction of antibody responses. Here, we demonstrate in mice using a Plasmodium falciparum merozoite surface protein 1 (PfMSP1) antigen that novel regimes combining adenovirus and poxvirus vectored vaccines with protein antigen in Montanide ISA720 adjuvant can achieve simultaneous antibody and T cell responses which equal, or in some cases surpass, the best immune responses achieved by either the viral vectors or the protein vaccine alone. Such broad responses can be achieved either using three-stage vaccination protocols, or with an equally effective two-stage protocol in which viral vectors are admixed with protein and adjuvant, and were apparent despite the use of a protein antigen that represented only a portion of the viral vector antigen. We describe further possible advantages of viral vectors in achieving consistent antibody priming, enhanced antibody avidity, and cytophilic isotype skew. These data strengthen the evidence that tailored combinations of vaccine platforms can achieve desired combinations of immune responses, and further encourage the co-administration of antibody-inducing recombinant protein vaccines with T cell- and antibody-inducing recombinant viral vectors as one strategy that may achieve protective blood-stage malaria immunity in humans.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Linfocitos T CD8-positivos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Proteína 1 de Superficie de Merozoito/inmunología , Adenoviridae/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antiprotozoarios/inmunología , Afinidad de Anticuerpos , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Malaria Falciparum/inmunología , Manitol/análogos & derivados , Manitol/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ácidos Oléicos/farmacología , Plasmodium falciparum/inmunología , Poxviridae/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...