Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Infect Dis ; 225(8): 1399-1410, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-32313928

RESUMEN

BACKGROUND: A vaccine (HB-101) consisting of 2 nonreplicating lymphocytic choriomeningitis virus (LCMV) vectors expressing the human cytomegalovirus antigens glycoprotein B (gB) and the 65-kD phosphoprotein (pp65), respectively, is in development to prevent cytomegalovirus infection. METHODS: HB-101 was tested in cytomegalovirus-naive, healthy adults in a randomized, double-blind, placebo-controlled, dose-escalation Phase I trial. Fifty-four subjects received low, medium, or high dose of HB-101 or placebo by intramuscular administration at Month 0, 1, and 3. Safety and immunogenicity were the respective primary and secondary endpoints. Subjects were followed for 12 months after the initial immunization. RESULTS: Vaccination was associated with transient mild to moderate adverse events. HB-101 administration induced dose-dependent gB- and pp65-specific cellular responses, dominated by pp65-specific CD8 T cells, a high fraction of which were polyfunctional. Two administrations were sufficient to elicit dose-dependent gB-binding and cytomegalovirus-neutralizing antibodies (Abs). Cytomegalovirus-specific immune responses were boosted after each administration. Only 1 of 42 vaccine recipients mounted a transient LCMV vector-neutralizing Ab response. CONCLUSIONS: HB-101 was well tolerated and induced cytomegalovirus-specific polyfunctional CD8 T-cell and neutralizing Ab responses in the majority of subjects. Lack of vector-neutralizing Ab responses should facilitate booster vaccinations. These results justify further clinical evaluation of this vaccine candidate.


Asunto(s)
Vacunas contra Citomegalovirus , Vacunas , Adulto , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Citomegalovirus/genética , Humanos , Inmunización Secundaria , Virus de la Coriomeningitis Linfocítica/genética
2.
Proc Natl Acad Sci U S A ; 112(6): 1767-72, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25624487

RESUMEN

Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and the leading viral cause of birth defects after congenital infection. The glycoprotein complexes gH/gL/gO and gH/gL/UL128/UL130/UL131A (Pentamer) are key targets of the human humoral response against HCMV and are required for HCMV entry into fibroblasts and endothelial/epithelial cells, respectively. We expressed and characterized soluble forms of gH/gL, gH/gL/gO, and Pentamer. Mass spectrometry and mutagenesis analysis revealed that gL-Cys144 forms disulfide bonds with gO-Cys351 in gH/gL/gO and with UL128-Cys162 in the Pentamer. Notably, Pentamer harboring the UL128-Cys162Ser/gL-Cys144Ser mutations had impaired syncytia formation and reduced interference of HCMV entry into epithelial cells. Electron microscopy analysis showed that HCMV gH/gL resembles HSV gH/gL and that gO and UL128/UL130/UL131A bind to the same site at the gH/gL N terminus. These data are consistent with gH/gL/gO and Pentamer forming mutually exclusive cell entry complexes and reveal the overall location of gH/gL-, gH/gL/gO-, and Pentamer-specific neutralizing antibody binding sites. Our results provide, to our knowledge, the first structural view of gH/gL/gO and Pentamer supporting the development of vaccines and antibody therapeutics against HCMV.


Asunto(s)
Citomegalovirus/inmunología , Citomegalovirus/fisiología , Glicoproteínas de Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Anticuerpos Neutralizantes/inmunología , Sitios de Unión/genética , Western Blotting , Cromatografía de Afinidad , Secuencia Conservada/genética , Citomegalovirus/metabolismo , Disulfuros/metabolismo , Citometría de Flujo , Humanos , Procesamiento de Imagen Asistido por Computador , Espectrometría de Masas , Glicoproteínas de Membrana/química , Microscopía Electrónica , Complejos Multiproteicos/química , Mutagénesis , Mutagénesis Sitio-Dirigida , Mutación/genética , Unión Proteica , Proteínas del Envoltorio Viral/química
3.
Proc Natl Acad Sci U S A ; 112(19): 6056-61, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25918416

RESUMEN

Varicella-zoster virus (VZV), of the family Alphaherpesvirinae, causes varicella in children and young adults, potentially leading to herpes zoster later in life on reactivation from latency. The conserved herpesvirus glycoprotein gB and the heterodimer gHgL mediate virion envelope fusion with cell membranes during virus entry. Naturally occurring neutralizing antibodies against herpesviruses target these entry proteins. To determine the molecular basis for VZV neutralization, crystal structures of gHgL were determined in complex with fragments of antigen binding (Fabs) from two human monoclonal antibodies, IgG-94 and IgG-RC, isolated from seropositive subjects. These structures reveal that the antibodies target the same site, composed of residues from both gH and gL, distinct from two other neutralizing epitopes identified by negative-stain electron microscopy and mutational analysis. Inhibition of gB/gHgL-mediated membrane fusion and structural comparisons with herpesvirus homologs suggest that the IgG-RC/94 epitope is in proximity to the site on VZV gHgL that activates gB. Immunization studies proved that the anti-gHgL IgG-RC/94 epitope is a critical target for antibodies that neutralize VZV. Thus, the gHgL/Fab structures delineate a site of herpesvirus vulnerability targeted by natural immunity.


Asunto(s)
Anticuerpos Neutralizantes/química , Glicoproteínas/química , Herpesvirus Humano 3/inmunología , Proteínas del Envoltorio Viral/química , Animales , Anticuerpos Monoclonales/química , Anticuerpos Antivirales/inmunología , Cristalografía por Rayos X , Epítopos/química , Humanos , Fragmentos de Inmunoglobulinas/química , Ratones , Modelos Moleculares , Pruebas de Neutralización , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
4.
J Virol ; 90(10): 4926-38, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26937030

RESUMEN

UNLABELLED: Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and is the leading viral cause of birth defects after congenital infection. HCMV infection relies on the recognition of cell-specific receptors by one of the viral envelope glycoprotein complexes. Either the gH/gL/gO or the gH/gL/UL128/UL130/UL131A (Pentamer) complex has been found to fulfill this role, accounting for HCMV entry into almost all cell types. We have studied the UL116 gene product, a putative open reading frame identified by in silico analysis and predicted to code for a secreted protein. Virus infection experiments in mammalian cells demonstrated that UL116 is expressed late in the HCMV replication cycle and is a heavily glycosylated protein that first localizes to the cellular site of virus assembly and then inserts into the virion envelope. Transient-transfection studies revealed that UL116 is efficiently transported to the plasma membrane when coexpressed with gH and that gL competes with UL116 for gH binding. Further evidence for gH/UL116 complex formation was obtained by coimmunoprecipitation experiments on both transfected and infected cells and biochemical characterization of the purified complex. In summary, our results show that the product of the UL116 gene is an HCMV envelope glycoprotein that forms a novel gH-based complex alternative to gH/gL. Remarkably, the gH/UL116 complex is the first herpesvirus gH-based gL-less complex. IMPORTANCE: HCMV infection can cause severe disease in immunocompromised adults and infants infected in utero The dissection of the HCMV entry machinery is important to understand the mechanism of viral infection and to identify new vaccine antigens. The gH/gL/gO and gH/gL/UL128/UL130/UL131 (Pentamer) complexes play a key role in HCMV cell entry and tropism. Both complexes are formed by an invariant gH/gL scaffold on which the other subunits assemble. Here, we show that the UL116 gene product is expressed in infected cells and forms a heterodimer with gH. The gH/UL116 complex is carried on the infectious virions, although in smaller amounts than gH/gL complexes. No gH/UL116/gL ternary complex formed in transfected cells, suggesting that the gH/UL116 complex is independent from gL. This new gH-based gL-free complex represents a potential target for a protective HCMV vaccine and opens new perspectives on the comprehension of the HCMV cell entry mechanism and tropism.


Asunto(s)
Citomegalovirus/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Citomegalovirus/química , Genoma Viral , Humanos , Microscopía Electrónica , Mutación , Multimerización de Proteína , Transfección , Proteínas del Envoltorio Viral/química , Ensamble de Virus , Internalización del Virus
5.
Biotechnol Bioeng ; 112(12): 2505-15, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26058896

RESUMEN

Human cytomegalovirus (HCMV) causes significant disease worldwide. Multiple HCMV vaccines have been tested in man but only partial protection has been achieved. The HCMV gH/gL/UL128/UL130/UL131A complex (Pentamer) is the main target of neutralizing antibodies in HCMV seropositive individuals and raises high titers of neutralizing antibodies in small animals and non-human primates (NHP). Thus, Pentamer is a promising candidate for an effective HCMV vaccine. Development of a Pentamer-based subunit vaccine requires expression of high amounts of a functional and stable complex. We describe here the development of a mammalian expression system for large scale Pentamer production. Several approaches comprising three different CHO-originated cell lines and multiple vector as well as selection strategies were tested. Stable cell pools expressed the HCMV Pentamer at a titer of approximately 60 mg/L at laboratory scale. A FACS-based single cell sorting approach allowed selection of a highly expressing clone producing Pentamer at the level of approximately 400 mg/L in a laboratory scale fed-batch culture. Expression in a 50 L bioreactor led to the production of HCMV Pentamer at comparable titers indicating the feasibility of further scale-up for manufacturing at commercial scale. The CHO-produced HCMV Pentamer bound to a panel of human neutralizing antibodies and raised potently neutralizing immune response in mice. Thus, we have generated an expression system for the large scale production of functional HCMV Pentamer at high titers suitable for future subunit vaccine production.


Asunto(s)
Células CHO , Vacunas contra Citomegalovirus/inmunología , Expresión Génica , Proteínas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Cricetulus , Citomegalovirus/genética , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/genética , Vacunas contra Citomegalovirus/metabolismo , Ratones , Multimerización de Proteína , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
6.
Proc Natl Acad Sci U S A ; 105(50): 19950-5, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19064925

RESUMEN

Rhesus cytomegalovirus infection of rhesus macaques has emerged as a model for human cytomegalovirus pathogenesis. The UL128-UL131 locus of the human virus is a primary determinant for viral entry into epithelial cells, an important cell type during cytomegalovirus infection. Rhesus cytomegalovirus strain 68-1 spreads slowly when grown in cultured rhesus epithelial cells, and it does not code for ORFs corresponding to UL128 and the second exon of UL130. We repaired the UL128-UL131 locus of strain 68-1, using rhesus cytomegalovirus strain 180.92 as template, to generate BRh68-1.1. We also repaired a mutation in the UL36 ORF in BRh68-1.1 to make BRh68-1.2. Both repaired derivatives replicate much more efficiently than parental 68-1 virus in rhesus epithelial cells, suggesting that strain 68-1 may be attenuated. Intriguingly, BRh68-1.1 and BRh68-1.2 replicate efficiently in cultured human epithelial cells and endothelial cells. The extended human cell host range of the repaired viruses raises the possibility that rhesus cytomegalovirus-like viruses will be found in humans.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Macaca mulatta/virología , Animales , Línea Celular , Citomegalovirus/genética , Células Endoteliales/virología , Células Epiteliales/virología , Exones , Humanos , Especificidad de la Especie , Transcripción Genética
7.
J Virol ; 82(5): 2170-81, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18094178

RESUMEN

Rhesus cytomegalovirus (RhCMV) is an emerging model for human cytomegalovirus (HCMV) pathogenesis that facilitates experimental CMV infection of a natural primate host closely related to humans. We have generated a library of RhCMV mutants with lesions in genes whose HCMV orthologues have been characterized as nonessential for replication in human fibroblasts, and we characterized their replication in rhesus fibroblasts and epithelial cells. The RhCMV mutants grew well in fibroblasts, as predicted by earlier studies with HCMV. However, mutations in four genes caused replication defects in rhesus retinal pigment epithelial cells: Rh01 (an HCMV TRL1 orthologue), Rh159 (HCMV UL148), Rh160 (HCMV UL132), and Rh203 (HCMV US22). Growth of the Rh01-deficient mutant was examined in detail. After entry into epithelial cells, the mutant expressed representative viral proteins, accumulated viral DNA, and generated infectious virus, but it failed to spread efficiently. We conclude that Rh01 is a cell tropism determinant that has the potential to dramatically affect virus spread and pathogenesis.


Asunto(s)
Citomegalovirus/genética , Tropismo , Animales , Secuencia de Bases , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/virología , Cartilla de ADN , Genes Virales , Macaca mulatta , Mutagénesis Sitio-Dirigida
8.
Clin Vaccine Immunol ; 24(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27795301

RESUMEN

Subunit vaccines for prevention of congenital cytomegalovirus (CMV) infection based on glycoprotein B (gB) and pp65 are in clinical trials, but it is unclear whether simultaneous vaccination with both antigens enhances protection. We undertook evaluation of a novel bivalent vaccine based on nonreplicating lymphocytic choriomeningitis virus (rLCMV) vectors expressing a cytoplasmic tail-deleted gB [gB(dCt)] and full-length pp65 from human CMV in mice. Immunization with the gB(dCt) vector alone elicited a comparable gB-binding antibody response and a superior neutralizing response to that elicited by adjuvanted subunit gB. Immunization with the pp65 vector alone elicited robust T cell responses. Comparable immunogenicity of the combined gB(dCt) and pp65 vectors with the individual monovalent formulations was demonstrated. To demonstrate proof of principle for a bivalent rLCMV-based HCMV vaccine, the congenital guinea pig cytomegalovirus (GPCMV) infection model was used to compare rLCMV vectors encoding homologs of pp65 (GP83) and gB(dCt), alone and in combination versus Freund's adjuvanted recombinant gB. Both vectors elicited significant immune responses, and no loss of gB immunogenicity was noted with the bivalent formulation. Combined vaccination with rLCMV-vectored GPCMV gB(dCt) and pp65 (GP83) conferred better protection against maternal viremia than subunit or either monovalent rLCMV vaccine. The bivalent vaccine also was significantly more effective in reducing pup mortality than the monovalent vaccines. In summary, bivalent vaccines with rLCMV vectors expressing gB and pp65 elicited potent humoral and cellular responses and conferred protection in the GPCMV model. Further clinical trials of LCMV-vectored HCMV vaccines are warranted.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Portadores de Fármacos , Virus de la Coriomeningitis Linfocítica/genética , Fosfoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Antígenos Virales/inmunología , Infecciones por Citomegalovirus/congénito , Vacunas contra Citomegalovirus/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Cobayas , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Linfocitos T/inmunología , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas de la Matriz Viral/genética
9.
Vaccine ; 35(1): 1-9, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27899229

RESUMEN

An important focus in vaccine research is the design of vaccine vectors with low seroprevalence and high immunogenicity. Replication-incompetent lymphocytic choriomeningitis virus (rLCMV) vectors do not elicit vector-neutralizing antibody responses, and homologous prime-boost regimens with rLCMV vectors induce boostable and protective T cell responses to model antigens in mice. However, cellular and humoral immune responses following homologous rLCMV vaccine regimens have not been rigorously evaluated in non-human primates (NHPs). To test whether rLCMV vectors constitute an effective vaccine platform in NHPs, we developed rLCMV vectors expressing SIVmac239 Env and Gag antigens and assessed their immunogenicity in mice and cynomolgus macaques. Immunization with rLCMV vaccine vectors expressing SIV Env and Gag was effective at generating SIV-specific T cell and antibody responses in both mice and NHPs. Epitope mapping using SIV Env in C57BL/6 mice demonstrated that rLCMV vectors induced sustained poly-functional responses to both dominant and subdominant epitopes. Our results suggest the potential of rLCMV vectors as vaccine candidates. Future SIV challenge experiments in rhesus macaques will be needed to assess immune protection by these vaccine vectors.


Asunto(s)
Antígenos Virales/inmunología , Portadores de Fármacos , Virus de la Coriomeningitis Linfocítica/genética , Vacunas contra el SIDAS/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Macaca fascicularis , Ratones Endogámicos C57BL , Vacunas contra el SIDAS/administración & dosificación , Vacunas contra el SIDAS/genética , Virus de la Inmunodeficiencia de los Simios/genética , Linfocitos T/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
10.
J Mol Biol ; 342(2): 467-78, 2004 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-15327947

RESUMEN

Transcriptional activation by the E.coli NtrC protein can occur via DNA looping between a DNA-bound activator and the target sigma(54) RNA polymerase. NtrC forms an octamer on DNA that is capable of binding two DNA molecules. Its ATPase activity is required for open complex formation. Geometric requirements for activation were assessed using a library of DNA bending sequences created by random ligation of A-tract oligonucleotides, as well as several designed sequences. Thirty random or designed sequences with a variety of DNA lengths and bending geometries were cloned in plasmids, and the library was used to replace the spacer between the NtrC binding sites and the core glnAp2 promoter. The activity of each promoter construct under nitrogen limitation was determined in vivo, in a lambda phage lacZ reporter system integrated as a single-copy lysogen to avoid titrating NtrC or polymerase. A wide variety of bending geometries was found to support a similar level of transcriptional activation ( approximately 3-4-fold). Computer modeling of the DNA trajectories suggests that the most inactive promoters have short spacer DNA and the NtrC sites on the opposite side of the helix as the wild-type sites; otherwise, the loop can form effectively. Flexibility and multivalency of the NtrC-Esigma(54) interaction apparently provides substantial independence from DNA stiffness constraints, and in general activation requires less efficient looping than repression. However, none of the random templates were as active as wild-type promoter. Subsidiary activator binding sites in the wild-type were found to be required for full activity, but, surprisingly, these sites could not be functionally replaced by strong binding sites. This suggests that one or more protomers in the NtrC octamer must form and then release contacts with DNA in order to complete the ATPase cycle and act as an AAA(+) activator of the Esigma(54). This dynamic DNA wrapping around the NtrC octamer is proposed to be necessary for efficient activation, and the wrapping may also reduce adventitious activation of other promoters.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Regiones Promotoras Genéticas , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Elementos de Facilitación Genéticos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli , Modelos Moleculares , Proteínas PII Reguladoras del Nitrógeno
11.
Nat Commun ; 6: 8176, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26365435

RESUMEN

Human cytomegalovirus (HCMV) poses a significant threat to immunocompromised individuals and neonates infected in utero. Glycoprotein B (gB), the herpesvirus fusion protein, is a target for neutralizing antibodies and a vaccine candidate due to its indispensable role in infection. Here we show the crystal structure of the HCMV gB ectodomain bound to the Fab fragment of 1G2, a neutralizing human monoclonal antibody isolated from a seropositive subject. The gB/1G2 interaction is dominated by aromatic residues in the 1G2 heavy chain CDR3 protruding into a hydrophobic cleft in the gB antigenic domain 5 (AD-5). Structural analysis and comparison with HSV gB suggest the location of additional neutralizing antibody binding sites on HCMV gB. Finally, immunoprecipitation experiments reveal that 1G2 can bind to HCMV virion gB suggesting that its epitope is exposed and accessible on the virus surface. Our data will support the development of vaccines and therapeutic antibodies against HCMV infection.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Antígenos Virales/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/ultraestructura , Anticuerpos Antivirales/química , Anticuerpos Antivirales/ultraestructura , Antígenos Virales/química , Antígenos Virales/ultraestructura , Cristalización , Cristalografía por Rayos X , Citomegalovirus/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Inmunoprecipitación , Microscopía Electrónica , Mutagénesis Sitio-Dirigida , Conformación Proteica , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/ultraestructura , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/ultraestructura
12.
Vaccine ; 32(30): 3796-804, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24837507

RESUMEN

Human cytomegalovirus (HCMV) is a member of the ß-herpesvirus family that causes significant disease worldwide. Although evidence exists that neutralizing antibodies and cytotoxic T cell responses to HCMV antigens can prevent HCMV disease and/or infection, there are no approved vaccines to prevent HCMV disease. Over the past 10 years, multiple HCMV vaccines have been tested in man but only partial protection has been achieved in these studies. HCMV contains multiple surface-expressed glycoproteins that are critical to viral entry, including gB, the gM/gN complex, the gH/gL complex, and a pentameric gH/gL/UL128/UL130/UL131A complex. Recently we showed that viral replicon particles (VRPs) expressing the gH/gL complex elicited more potently neutralizing antibodies than VRPs expressing gB in mice. Here we compare the immunogenicity of VRPs encoding the HCMV gH/gL and pentameric complexes, as well as purified gH/gL and pentameric complexes administered in the presence or absence of the MF59 adjuvant. The results of these studies indicate that the pentameric complex elicits significantly higher levels of neutralizing antibodies than the gH/gL complex, and that MF59 significantly increases the potency of each complex. In addition, we show that animals immunized with pentamer encoding VRPs or the pentameric subunit produce antibodies that recognize a broad range of antigenic sites on the complex. Taken together, these studies support the utility of the pentameric complex in HCMV vaccine candidates.


Asunto(s)
Anticuerpos Antivirales/sangre , Vacunas contra Citomegalovirus/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas del Envoltorio Viral/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/sangre , Especificidad de Anticuerpos , Citomegalovirus , Femenino , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Complejos Multiproteicos/inmunología , Polisorbatos/farmacología , Replicón , Escualeno/farmacología , Vacunas de Partículas Similares a Virus/inmunología
13.
Vaccine ; 31(6): 919-26, 2013 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23246547

RESUMEN

Human cytomegalovirus (hCMV) is prevalent worldwide with infection generally being asymptomatic. Nevertheless, hCMV infection can lead to significant morbidity and mortality. Primary infection of seronegative women or reactivation/re-infection of seropositive women during pregnancy can result in transmission to the fetus, leading to severe neurological defects. In addition, hCMV is the most common viral infection in immunosuppressed organ transplant recipients and can produce serious complications. Hence, a safe and effective vaccine to prevent hCMV infection is an unmet medical need. Neutralizing antibodies to several hCMV glycoproteins, and complexes thereof, have been identified in individuals following hCMV infection. Interestingly, a portion of the CMV-specific neutralizing antibody responses are directed to epitopes found on glycoprotein complexes but not the individual proteins. Using an alphavirus replicon particle (VRP) vaccine platform, we showed that bicistronic VRPs encoding hCMV gH and gL glycoproteins produce gH/gL complexes in vitro. Furthermore, mice vaccinated with these gH/gL-expressing VRPs produced broadly cross-reactive complement-independent neutralizing antibodies to hCMV. These neutralizing antibody responses were of higher titer than those elicited in mice vaccinated with monocistronic VRPs encoding gH or gL antigens, and they were substantially more potent than those raised by VRPs encoding gB. These findings underscore the utility of co-delivery of glycoprotein components such as gH and gL for eliciting potent, broadly neutralizing immune responses against hCMV, and indicate that the gH/gL complex represents a potential target for future hCMV vaccine development.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Vacunas contra Citomegalovirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/inmunología , Alphavirus/genética , Animales , Reacciones Cruzadas , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/genética , Femenino , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética
14.
Vaccine ; 30(49): 6980-90, 2012 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-23041121

RESUMEN

Human cytomegalovirus (HCMV) infects the majority of the global population and persists within the infected host for life; infection of healthy adults rarely leads to severe acute clinical symptoms. In contrast, HCMV is a leading infectious cause of congenital disease and a common cause of complications in transplant recipients. A vaccine to prevent HCMV disease in these populations is a widely recognized medical need. We review recent advances in our understanding of the candidate vaccine antigens and published clinical trial data for the four most recent HCMV vaccine candidates: a gB subunit adjuvanted with MF59, a DNA vaccine expressing gB and pp65, alphavirus replicon particles (VRPs) expressing gB and a pp65-IE1 fusion protein, and a pp65 peptide vaccine. The candidates are safe, although some adverse events were reported for an adjuvanted variant of the pp65 peptide vaccine. The gB/MF59 vaccine elicited strong humoral responses with limited durability. The gB/pp65 DNA vaccine elicited cellular immunity, and the pp65 peptide vaccine elicited modest cellular immunity, but only when formulated with an adjuvant. Only the VRP vaccine expressing gB and pp65-IE1 elicited both humoral and cellular immunity. The gB/MF59 vaccine showed a short-term 50% efficacy at preventing infection of seronegative women and significantly reduced viremia and need for antivirals in solid organ transplant recipients, and the gB/pp65 DNA vaccine showed signs of clinical benefit in hematopoietic stem cell transplant recipients. Importantly, the partial efficacy of the subunit and DNA vaccines is new evidence that both humoral and cellular immunity contribute to controlling HCMV-related disease. These data show the clinical feasibility of a recombinant HCMV vaccine. We discuss areas for potential improvements in the next generation of vaccine candidates.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Alphavirus/genética , Anticuerpos Antivirales/sangre , Investigación Biomédica/tendencias , Ensayos Clínicos como Asunto , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/efectos adversos , Vacunas contra Citomegalovirus/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Vectores Genéticos , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Memoria Inmunológica , Linfocitos/inmunología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA