Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Eur J Epidemiol ; 34(8): 793-799, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30993509

RESUMO

Chronological age alone is not a sufficient measure of the true physiological state of the body. The aims of the present study were to: (1) quantify biological age based on a physiological biomarker composite model; (2) and evaluate its association with death and age-related disease onset in the setting of an elderly population. Using structural equation modeling we computed biological age for 1699 individuals recruited from the first and second waves of the Rotterdam study. The algorithm included nine physiological parameters (c-reactive protein, creatinine, albumin, total cholesterol, cytomegalovirus optical density, urea nitrogen, alkaline phosphatase, forced expiratory volume and systolic blood pressure). We assessed the association between biological age, all-cause mortality, all-cause morbidity and specific age-related diseases over a median follow-up of 11 years. Biological age, compared to chronological age or the traditional biomarkers of age-related diseases, showed a stronger association with all-cause mortality (HR 1.15 vs. 1.13 and 1.10), all-cause morbidity (HR 1.06 vs. 1.05 and 1.03), stroke (HR 1.17 vs. 1.08 and 1.04), cancer (HR 1.07 vs. 1.04 and 1.02) and diabetes mellitus (HR 1.12 vs. 1.01 and 0.98). Individuals who were biologically younger exhibited a healthier life-style as reflected in their lower BMI (P < 0.001) and lower incidence of stroke (P < 0.001), cancer (P < 0.01) and diabetes mellitus (P = 0.02). Collectively, our findings suggest that biological age based on the biomarker composite model of nine physiological parameters is a useful construct to assess individuals 65 years and older at increased risk for specific age-related diseases.


Assuntos
Envelhecimento/fisiologia , Biomarcadores/sangue , Adulto , Idade de Início , Idoso , Pressão Sanguínea , Índice de Massa Corporal , Doenças Cardiovasculares/mortalidade , Causas de Morte , Creatinina , Diabetes Mellitus/mortalidade , Feminino , Humanos , Longevidade , Masculino , Pessoa de Meia-Idade , Modelos Teóricos , Morbidade , Mortalidade , Estudos Prospectivos
3.
PLoS One ; 9(7): e103550, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25075622

RESUMO

Current influenza vaccines are believed to confer protection against a narrow range of virus strains. The identification of broadly influenza neutralizing antibodies (bnAbs) has triggered efforts to develop vaccines providing 'universal' protection against influenza. Several bnAbs were isolated from humans recently vaccinated with conventional influenza vaccines, suggesting that such vaccines could, in principle, be broadly protective. Assessing the breadth-of-protection conferred to humans by influenza vaccines is hampered by the lack of in vitro correlates for broad protection. We designed and employed a novel human-to-mouse serum transfer and challenge model to analyze protective responses in serum samples from clinical trial subjects. One dose of seasonal vaccine induces humoral protection not only against vaccine-homologous H1N1 challenge, but also against H5N1 challenge. This heterosubtypic protection is neither detected, nor accurately predicted by in vitro immunogenicity assays. Moreover, heterosubtypic protection is transient and not boosted by repeated inoculations. Strategies to increase the breadth and duration of the protective response against influenza are required to obtain 'universal' protection against influenza by vaccination. In the absence of known correlates of protection for broadly protective vaccines, the human-to-mouse serum transfer and challenge model described here may aid the development of such vaccines.


Assuntos
Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Área Sob a Curva , Linhagem Celular Tumoral , Proteção Cruzada , Cães , Feminino , Testes de Inibição da Hemaglutinação , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/prevenção & controle , Curva ROC , Estações do Ano , Vacinação
4.
J Infect Dis ; 207(2): 248-56, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23125443

RESUMO

BACKGROUND: Adenovirus serotype 26 (Ad26) has been developed as a novel candidate vaccine vector for human immunodeficiency virus type 1 (HIV-1) and other pathogens. The primary safety and immunogenicity data from the Integrated Preclinical/Clinical AIDS Vaccine Development Program (IPCAVD) 001 trial, the first-in-human evaluation of a prototype Ad26 vector-based vaccine expressing clade A HIV-1 Env (Ad26.ENVA.01), are reported concurrently with this article. Here, we characterize in greater detail the humoral and cellular immune responses elicited by Ad26.ENVA.01 in humans. METHODS: Samples from the IPCAVD 001 trial were used for humoral and cellular immunogenicity assays. RESULTS: We observed a dose-dependent expansion of the magnitude, breadth, and epitopic diversity of Env-specific binding antibody responses elicited by this vaccine. Antibody-dependent cell-mediated phagocytosis, virus inhibition, and degranulation functional activity were also observed. Env-specific cellular immune responses induced by the vaccine included multiple CD8(+) and CD4(+) T-lymphocyte memory subpopulations and cytokine secretion phenotypes, although cellular immune breadth was limited. Baseline vector-specific T-lymphocyte responses were common but did not impair Env-specific immune responses in this study. CONCLUSION: Ad26.ENVA.01 elicited a broad diversity of humoral and cellular immune responses in humans. These data support the further clinical development of Ad26 as a candidate vaccine vector. CLINICAL TRIALS REGISTRATION: NCT00618605.


Assuntos
Vacinas contra a AIDS/imunologia , Adenovírus Humanos/genética , Produtos do Gene env/imunologia , Anticorpos Anti-HIV/sangue , HIV-1/imunologia , Imunidade Celular/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/efeitos adversos , Adenovírus Humanos/classificação , Método Duplo-Cego , Produtos do Gene env/genética , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Humanos , Leucócitos Mononucleares/imunologia , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
5.
J Infect Dis ; 207(2): 240-7, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23125444

RESUMO

BACKGROUND: We report the first-in-human safety and immunogenicity assessment of a prototype Ad26 vector-based human immunodeficiency virus (HIV) vaccine in humans. METHODS: Sixty Ad26-seronegative, healthy, HIV-uninfected subjects were enrolled in a randomized, double-blinded, placebo-controlled, dose-escalation phase 1 study. Five groups of 12 subjects received 10(9)-10(11) vp of the Ad26-EnvA vaccine (N = 10/group) or placebo (N = 2/group) at weeks 0 and 24 or weeks 0, 4, and 24. Safety and immunogenicity were assessed. RESULTS: Self-limited reactogenicity was observed after the initial immunization at the highest (10(11) vp) dose. No product-related SAEs were observed. All subjects who received the Ad26-EnvA vaccine developed Ad26 NAb titers, EnvA-specific enzyme-linked immunosorbent assays (ELISA) titers, and EnvA-specific enzyme-linked immunospot assays (ELISPOT) responses. These responses persisted at week 52. At week 28 in the 10(9), 10(10), 10(11) vp 3-dose and the 10(10) and 5 × 10(10) vp 2-dose groups, geometric mean EnvA ELISA titers were 6113, 12 470, 8545, 3470, and 9655 and mean EnvA ELISPOT responses were 397, 178, 736, 196, and 1311 SFC/10(6) peripheral blood mononuclear cells, respectively. CONCLUSION: This Ad26 vectored vaccine was generally safe and immunogenic at all doses tested. Reactogenicity was minimal with doses of 5 × 10(10) vp or less. Ad26 is a promising new vaccine vector for HIV-1. CLINICAL TRIALS REGISTRATION: NCT00618605.


Assuntos
Vacinas contra a AIDS/efeitos adversos , Vacinas contra a AIDS/imunologia , Adenovírus Humanos/genética , Produtos do Gene env/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Vacinas contra a AIDS/administração & dosagem , Adenovírus Humanos/classificação , Método Duplo-Cego , Feminino , Produtos do Gene env/genética , Anticorpos Anti-HIV/sangue , Infecções por HIV/imunologia , Humanos , Leucócitos Mononucleares/imunologia , Masculino , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
6.
PLoS One ; 7(12): e44115, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23236343

RESUMO

Filoviruses cause sporadic but highly lethal outbreaks of hemorrhagic fever in Africa in the human population. Currently, no drug or vaccine is available for treatment or prevention. A previous study with a vaccine candidate based on the low seroprevalent adenoviruses 26 and 35 (Ad26 and Ad35) was shown to provide protection against homologous Ebola Zaire challenge in non human primates (NHP) if applied in a prime-boost regimen. Here we have aimed to expand this principle to construct and evaluate Ad26 and Ad35 vectors for development of a vaccine to provide universal filovirus protection against all highly lethal strains that have caused major outbreaks in the past. We have therefore performed a phylogenetic analysis of filovirus glycoproteins to select the glycoproteins from two Ebola species (Ebola Zaire and Ebola Sudan/Gulu,), two Marburg strains (Marburg Angola and Marburg Ravn) and added the more distant non-lethal Ebola Ivory Coast species for broadest coverage. Ad26 and Ad35 vectors expressing these five filovirus glycoproteins were evaluated to induce a potent cellular and humoral immune response in mice. All adenoviral vectors induced a humoral immune response after single vaccination in a dose dependent manner that was cross-reactive within the Ebola and Marburg lineages. In addition, both strain-specific as well as cross-reactive T cell responses could be detected. A heterologous Ad26-Ad35 prime-boost regime enhanced mainly the humoral and to a lower extend the cellular immune response against the transgene. Combination of the five selected filovirus glycoproteins in one multivalent vaccine potentially elicits protective immunity in man against all major filovirus strains that have caused lethal outbreaks in the last 20 years.


Assuntos
Reações Cruzadas , Vacinas contra Ebola/imunologia , Filoviridae/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Imunidade Humoral/imunologia , Linfócitos T/imunologia , Vacinas Virais/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Vacinas contra Ebola/genética , Feminino , Vetores Genéticos , Doença pelo Vírus Ebola/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Virais/genética
7.
Mol Ther ; 20(12): 2268-81, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22929662

RESUMO

The development of adenoviral vectors for intravascular (i.v.) delivery will require improvements to their in vivo safety and efficacy. The hypervariable regions (HVRs) of the Ad5 hexon are a target for neutralizing antibodies, but also interact with factor X (FX), facilitating hepatocyte transduction. Ad48, a species D adenovirus, does not bind FX and has low seroprevalence. Therefore, it has been suggested that Ad5HVR48(1-7), a hexon-chimeric vector featuring the seven HVRs from Ad48, should display advantageous properties for gene therapy, by evading pre-existing Ad5 immunity and blocking FX interactions. We investigated the in vivo biodistribution of Ad5, Ad5HVR48(1-7), and Ad48 following i.v. delivery. Ad5HVR48(1-7) displayed reduced hepatocyte transduction and accumulation in Kupffer cells (KCs), but triggered a robust proinflammatory response, even at relatively low doses of vector. We detected elevated serum transaminases (48 hours) and increased numbers of periportal CD11b(+)/Gr-1(+) cells in the livers of Ad5HVR48(1-7)-treated animals following i.v., but not intramuscular (i.m.), delivery. In contrast, Ad48 did not elevate transaminases or result in the accumulation of CD11b(+)/Gr-1(+) cells. Collectively, these findings suggest that substantial hexon modifications can lead to unexpected properties which cannot be predicted from parental viruses. Therefore, refined mutations may be preferential for the successful development of targeted vector systems which require i.v. administration.


Assuntos
Adenoviridae/imunologia , Administração Intravenosa , Vetores Genéticos/efeitos adversos , Vetores Genéticos/imunologia , Animais , Vetores Genéticos/administração & dosagem , Células Hep G2 , Humanos , Masculino , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transaminases/genética , Transaminases/metabolismo
8.
Vaccine ; 30(41): 5991-8, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22732429

RESUMO

Licensed live attenuated virus vaccines capable of expressing transgenes from other pathogens have the potential to reduce the number of childhood immunizations by eliciting robust immunity to multiple pathogens simultaneously. Recombinant attenuated measles virus (rMV) derived from the Edmonston Zagreb vaccine strain was engineered to express simian immunodeficiency virus (SIV) Gag protein for the purpose of evaluating the immunogenicity of rMV as a vaccine vector in rhesus macaques. rMV-Gag immunization alone elicited robust measles-specific humoral and cellular responses, but failed to elicit transgene (Gag)-specific immune responses, following aerosol or intratracheal/intramuscular delivery. However, when administered as a priming vaccine to a heterologous boost with recombinant adenovirus serotype 5 expressing the same transgene, rMV-Gag significantly enhanced Gag-specific T lymphocyte responses following rAd5 immunization. Gag-specific humoral responses were not enhanced, however, which may be due to either the transgene or the vector. Cellular response priming by rMV against the transgene was highly effective even when using a suboptimal dose of rAd5 for the boost. These data demonstrate feasibility of using rMV as a priming component of heterologous prime-boost vaccine regimens for pathogens requiring strong cellular responses.


Assuntos
Produtos do Gene gag/imunologia , Imunização Secundária/métodos , Vacina contra Sarampo/imunologia , Linfócitos T/imunologia , Adenoviridae , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Produtos do Gene gag/genética , Vetores Genéticos , Imunidade Celular , Macaca mulatta , Sarampo/imunologia , Sarampo/prevenção & controle , Vacina contra Sarampo/administração & dosagem , Vacina contra Sarampo/genética , Testes de Neutralização , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Transgenes , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
9.
J Infect Dis ; 206(2): 158-66, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22551811

RESUMO

BACKGROUND: Vaccination is the best measure to protect the population against a potential influenza H5N1 pandemic, but 2 doses of vaccine are needed to elicit protective immune responses. An immunological marker for H5N1 vaccine effectiveness is needed for early identification of the best vaccine candidate. METHODS: We conducted a phase I clinical trial of a virosomal H5N1 vaccine adjuvanted with Matrix M. Sixty adult volunteers were vaccinated intramuscularly with 2 doses of either 30 µg hemagglutinin (HA) alone or with 1.5, 7.5, or 30 µg HA and Matrix M adjuvant (50 µg). The humoral response was measured by the hemagglutination inhibition (HI), microneutralization (MN), and single radial hemolysis (SRH) assays, and the CD4(+) T-helper 1 (Th1)-cell response was measured by intracellular staining for the cytokines interleukin 2, interferon γ, and tumor necrosis factor α. RESULTS: The adjuvanted vaccine effectively induced CD4(+) Th1-cell responses, and the frequency of influenza-specific Th1 cells after the first vaccine dose predicted subsequent HI, MN, and SRH seroprotective responses after the second vaccination. CONCLUSIONS: These results support early identification of Th1-cell responses as a predictive biomarker for an efficient vaccine response, which could have great implications for early identification of persons with low or no response to vaccine when evaluating future pandemic influenza vaccines.


Assuntos
Anticorpos Antivirais/sangue , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Linfócitos T Auxiliares-Indutores/fisiologia , Adjuvantes Imunológicos/fisiologia , Adulto , Citocinas/sangue , Relação Dose-Resposta Imunológica , Humanos , ISCOMs/imunologia , Influenza Humana/imunologia , Influenza Humana/virologia , Pessoa de Meia-Idade , Vacinação , Vacinas Virossomais/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Adulto Jovem
10.
Vaccine ; 30(12): 2098-108, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22296955

RESUMO

BACKGROUND: Despite the availability of Bacille Calmette Guérin (BCG) vaccines, Mycobacterium tuberculosis currently infects billions of people and millions die annually from tuberculosis (TB) disease. New TB vaccines are urgently needed. METHODS: We studied the ability of AERAS-402, a recombinant, replication-deficient adenovirus type 35 expressing the protective M. tuberculosis antigens Ag85A, Ag85B, and TB10.4, to boost BCG immunity in an area of low TB endemicity. RESULTS: In volunteers primed with BCG 3 or 6 months prior to AERAS-402 boosting, significant CD4(+) and CD8(+) T cell responses were induced. Ag85-specific responses were more strongly boosted than TB10.4-specific responses. Frequencies of TB-specific CD8(+) T cells reached>50 fold higher than pre-AERAS boosting levels, remarkably higher than reported in any previous human TB vaccine trial. Multiparameter flow cytometric assays demonstrated that AERAS-402-boosted CD4(+) and CD8(+) T cells were multifunctional, producing multiple cytokines and other immune effector molecules. Furthermore, boosted T cells displayed lymphoproliferative capacity, and tetramer analyses confirmed that antigen-specific CD8(+) T cells were induced. BCG and AERAS-402 vaccinations given 3 and 6 months apart appeared equivalent. CONCLUSIONS: Our results indicate that AERAS-402 is a promising TB vaccine candidate that can significantly enhance both CD4(+) and CD8(+) TB-specific T cell responses after BCG priming. ClinicalTrials.gov Identifier: NCT01378312.


Assuntos
Aciltransferases/imunologia , Adenovírus Humanos/genética , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Vetores Genéticos , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Aciltransferases/genética , Adulto , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Citocinas/biossíntese , Citometria de Fluxo , Experimentação Humana , Humanos , Mycobacterium tuberculosis/genética , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/genética
11.
Nature ; 482(7383): 89-93, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22217938

RESUMO

Preclinical studies of human immunodeficiency virus type 1 (HIV-1) vaccine candidates have typically shown post-infection virological control, but protection against acquisition of infection has previously only been reported against neutralization-sensitive virus challenges. Here we demonstrate vaccine protection against acquisition of fully heterologous, neutralization-resistant simian immunodeficiency virus (SIV) challenges in rhesus monkeys. Adenovirus/poxvirus and adenovirus/adenovirus-vector-based vaccines expressing SIV(SME543) Gag, Pol and Env antigens resulted in an 80% or greater reduction in the per-exposure probability of infection against repetitive, intrarectal SIV(MAC251) challenges in rhesus monkeys. Protection against acquisition of infection showed distinct immunological correlates compared with post-infection virological control and required the inclusion of Env in the vaccine regimen. These data demonstrate the proof-of-concept that optimized HIV-1 vaccine candidates can block acquisition of stringent, heterologous, neutralization-resistant virus challenges in rhesus monkeys.


Assuntos
Anticorpos Neutralizantes/imunologia , Macaca mulatta/imunologia , Vacinas contra a SAIDS/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas contra a AIDS/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , HIV-1/imunologia , Masculino , Testes de Neutralização , Vacinas Virais/imunologia
12.
Vaccine ; 30(37): 5578-84, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21983157

RESUMO

A protective malaria vaccine may induce both high levels of neutralising antibodies and strong T-cell responses. The Plasmodium falciparum circumsporozoite protein (CSp) is a leading pre-erythrocytic vaccine candidate. CSp is a week immunogen per se, but Mycobacterium bovis Bacille Calmette-Guérin (BCG) has excellent adjuvant activity and has been utilized as a vector to deliver heterologous vaccine candidate antigens. It is safe in immunocompetent individuals and inexpensive to produce. We assessed in vitro and in vivo a recombinant BCG-expressing CSp (BCG-CS) as malaria vaccine candidate. Immunisation of BALB/c mice with BCG-CS augmented numbers of dendritic cells (DCs) in draining lymph nodes and in the spleen. The activation markers MHC-class-II, CD40, CD80 and CD86 on DCs were significantly upregulated by BCG-CS as compared to wild-type BCG (wt-BCG). In vitro stimulation of bone marrow-derived DCs and macrophages with BCG-CS induced IL-12 and TNF-α production. BCG-CS induced higher phagocytic activity in macrophages as compared to wt-BCG. Immunogenicity studies show that BCG-CS induced CS-specific antibodies and IFN-γ-producing memory cells. In conclusion, BCG-CS is highly efficient in activating antigen-presenting cells (APCs) for priming of adaptive immunity. Implications for the rational design of novel vaccines against malaria and TB, the two major devastating poverty-related diseases, are discussed.


Assuntos
Células Dendríticas/imunologia , Memória Imunológica , Vacinas Antimaláricas/imunologia , Mycobacterium bovis/genética , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinas Sintéticas/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Antígeno B7-1/imunologia , Antígeno B7-2/imunologia , Biomarcadores/metabolismo , Antígenos CD40/imunologia , Feminino , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-12/metabolismo , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/imunologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Baço/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Vacinas Sintéticas/genética
13.
Vaccine ; 29(32): 5203-9, 2011 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-21619905

RESUMO

Recombinant adenovirus serotype 5 (rAd5) vaccine vectors for HIV-1 and other pathogens have been shown to be limited by high titers of Ad5 neutralizing antibodies (NAbs) in the developing world. Alternative serotype rAd vectors have therefore been constructed. Here we report Ad5, Ad26, Ad35, and Ad48 NAb titers in 4381 individuals from North America, South America, sub-Saharan Africa, and Southeast Asia. As expected, Ad5 NAb titers were both frequent and high magnitude in sub-Saharan Africa and Southeast Asia. In contrast, Ad35 NAb titers proved infrequent and low in all regions studied, and Ad48 NAbs were rare in all regions except East Africa. Ad26 NAbs were moderately common in adults in sub-Saharan Africa and Southeast Asia, but Ad26 NAb titers proved markedly lower than Ad5 NAb titers in all regions, and these relatively low Ad26 NAb titers did not detectably suppress the immunogenicity of 4×10(10)vp of a rAd26-Gag/Pol/Env/Nef vaccine in rhesus monkeys. These data inform the clinical development of alternative serotype rAd vaccine vectors in the developing world.


Assuntos
Infecções por Adenoviridae/epidemiologia , Infecções por Adenoviridae/imunologia , Adenoviridae/imunologia , Anticorpos Antivirais/imunologia , Vacinas Virais/imunologia , Vacinas contra a AIDS/imunologia , Adenoviridae/isolamento & purificação , Infecções por Adenoviridae/sangue , Infecções por Adenoviridae/virologia , Adolescente , Adulto , Animais , Anticorpos Antivirais/sangue , Criança , HIV-1/imunologia , Humanos , Lactente , Macaca mulatta/imunologia , Vacinas Sintéticas/imunologia , Adulto Jovem
14.
Clin Vaccine Immunol ; 18(5): 776-82, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21411600

RESUMO

Various pre-erythrocyte malaria vaccines are currently in clinical development, and among these is the adenovirus serotype 35-based circumsporozoite (CS) vaccine produced on PER.C6 cells. Although the immunological correlate of protection against malaria remains to be established, the CS antibody titer is a good marker for evaluation of candidate vaccines. Here we describe the validation of an anti-Plasmodium falciparum circumsporozoite antibody enzyme-linked immunosorbent assay (ELISA) based on the binding of antibodies to a peptide antigen mimicking the CS repeat region. The interassay variability was determined to be below a coefficient of variation (CV) of 15%, and sensitivity was sufficient to detect low antibody titers in subjects from endemic regions. Antibody titers were in agreement with total antibody responses to the whole CS protein. Due to its simplicity and high performance, the ELISA is an easy and rapid method for assessment of pre-erythrocyte malaria vaccines based on CS.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários , Técnicas de Laboratório Clínico/métodos , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adulto , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Malária Falciparum/prevenção & controle , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
15.
J Virol ; 85(9): 4222-33, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21325402

RESUMO

The use of adenoviruses (Ad) as vaccine vectors against a variety of pathogens has demonstrated their capacity to elicit strong antibody and cell-mediated immune responses. Adenovirus serotype C vectors, such as Ad serotype 5 (Ad5), expressing Ebolavirus (EBOV) glycoprotein (GP), protect completely after a single inoculation at a dose of 10(10) viral particles. However, the clinical application of a vaccine based on Ad5 vectors may be hampered, since impairment of Ad5 vaccine efficacy has been demonstrated for humans and nonhuman primates with high levels of preexisting immunity to the vector. Ad26 and Ad35 segregate genetically from Ad5 and exhibit lower seroprevalence in humans, making them attractive vaccine vector alternatives. In the series of studies presented, we show that Ad26 and Ad35 vectors generate robust antigen-specific cell-mediated and humoral immune responses against EBOV GP and that Ad5 immune status does not affect the generation of GP-specific immune responses by these vaccines. We demonstrate partial protection against EBOV by a single-shot Ad26 vaccine and complete protection when this vaccine is boosted by Ad35 1 month later. Increases in efficacy are paralleled by substantial increases in T- and B-cell responses to EBOV GP. These results suggest that Ad26 and Ad35 vectors warrant further development as candidate vaccines for EBOV.


Assuntos
Adenovírus Humanos/imunologia , Portadores de Fármacos , Vacinas contra Ebola/imunologia , Vetores Genéticos/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Proteínas do Envelope Viral/imunologia , Adenovírus Humanos/genética , Animais , Anticorpos Antivirais/sangue , Vacinas contra Ebola/administração & dosagem , Doença pelo Vírus Ebola/imunologia , Linfócitos/imunologia , Macaca fascicularis , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética
16.
Proc Natl Acad Sci U S A ; 107(51): 22213-8, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21135247

RESUMO

Successful vaccination against respiratory infections requires elicitation of high levels of potent and durable humoral and cellular responses in the lower airways. To accomplish this goal, we used a fine aerosol that targets the entire lung surface through normal respiration to deliver replication-incompetent recombinant adenoviral vectors expressing gene products from several infectious pathogens. We show that this regimen induced remarkably high and stable lung T-cell responses in nonhuman primates and that it also generated systemic and respiratory tract humoral responses of both IgA and IgG isotypes. Moreover, strong immunogenicity was achieved even in animals with preexisting antiadenoviral immunity, overcoming a critical hurdle to the use of these vectors in humans, who commonly are immune to adenoviruses. The immunogenicity profile elicited with this regimen, which is distinct from either intramuscular or intranasal delivery, has highly desirable properties for protection against respiratory pathogens. We show that it can be used repeatedly to generate mucosal humoral, CD4, and CD8 T-cell responses and as such may be applicable to other mucosally transmitted pathogens such as HIV. Indeed, in a lethal challenge model, we show that aerosolized recombinant adenoviral immunization completely protects ferrets against H5N1 highly pathogenic avian influenza virus. Thus, genetic immunization in the lung offers a powerful platform approach to generating protective immune responses against respiratory pathogens.


Assuntos
Adenoviridae , Linfócitos T CD8-Positivos/imunologia , Vetores Genéticos/farmacologia , Imunização/métodos , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Administração Intranasal , Aerossóis , Animais , Furões , Imunidade Celular/imunologia , Virus da Influenza A Subtipo H5N1/genética , Pulmão , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia
17.
Proc Natl Acad Sci U S A ; 107(44): 18950-5, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-20956322

RESUMO

Many flaviviruses are significant human pathogens, with the humoral immune response playing an essential role in restricting infection and disease. CR4354, a human monoclonal antibody isolated from a patient, neutralizes West Nile virus (WNV) infection at a postattachment stage in the viral life-cycle. Here, we determined the structure of WNV complexed with Fab fragments of CR4354 using cryoelectron microscopy. The outer glycoprotein shell of a mature WNV particle is formed by 30 rafts of three homodimers of the viral surface protein E. CR4354 binds to a discontinuous epitope formed by protein segments from two neighboring E molecules, but does not cause any detectable structural disturbance on the viral surface. The epitope occurs at two independent positions within an icosahedral asymmetric unit, resulting in 120 binding sites on the viral surface. The cross-linking of the six E monomers within one raft by four CR4354 Fab fragments suggests that the antibody neutralizes WNV by blocking the pH-induced rearrangement of the E protein required for virus fusion with the endosomal membrane.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Fragmentos Fab das Imunoglobulinas/química , Proteínas Estruturais Virais/química , Vírus do Nilo Ocidental/química , Sequência de Aminoácidos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/farmacologia , Sítios de Ligação , Endossomos/imunologia , Endossomos/virologia , Epitopos/química , Epitopos/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Dados de Sequência Molecular , Proteínas Estruturais Virais/imunologia , Internalização do Vírus/efeitos dos fármacos , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/ultraestrutura
18.
Clin Vaccine Immunol ; 17(11): 1687-94, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20826614

RESUMO

The most advanced malaria vaccine, RTS,S, is comprised of a portion of the Plasmodium falciparum circumsporozoite (CS) protein, fused to and admixed with the hepatitis B virus surface antigen, and an adjuvant [corrected].This vaccine confers short-term protection against malaria infection, with an efficacy of about 50%, and induces particularly B-cell and CD4(+) T-cell responses.In the present study, we tested the hypothesis that the Th1 immune response to CS protein,in particular the CD8(+) T-cell response, which is needed for strong and lasting malaria immunity, is boosted to sustainable levels by adenovirus vectors 35 and 26 with a homologous insert (Ad35.CS/Ad26.CS) [corrected]. In this study, we evaluated immune responses induced with vaccination regimens based on an adjuvant-containing, yeast-produced complete CS protein followed by two recombinant low-seroprevalence adenoviruses expressing P. falciparum CS antigen, Ad35.CS (subgroup B) and Ad26.CS (subgroup D). Our results show that (i) the yeast (Hansenula polymorpha)produced, adjuvanted full-length CS protein is highly potent in inducing high CS-specific humoral responses in mice but produces poor T-cell responses, (ii) the Ad35.CS vector boosts the gamma interferon-positive (IFN-γ(+)) CD8(+) T-cell response induced by the CS protein immunization and shifts the immune response toward the Th1 type, and (iii) a three-component heterologous vaccination comprised of a CS protein prime followed by boosts with Ad35.CS and Ad26.CS elicits an even more robust and sustainable IFN-γ(+) CD8(+) T-cell response than one- or two-component regimens. The Ad35.CS/Ad26.CS combination boosted particularly the IFN-γ(+) and tumor necrosis factor alpha-positive (TNF-α(+)) T cells, confirming the shift of the immune response from the Th2 type to the Th1 type. These results support the notion of first immunizations of infants with an adjuvanted CS protein vaccine, followed by a booster Ad35.CS/Ad26.CS vaccine at a later age, to induce lasting protection against malaria for which the Th1 response and immune memory is required.


Assuntos
Adenoviridae/genética , Vetores Genéticos/genética , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Células Th1/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Linfócitos T CD8-Positivos/imunologia , Feminino , Humanos , Imunização Secundária/métodos , Interferon gama/metabolismo , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Pichia/genética , Proteínas de Protozoários/genética , Vacinação/métodos , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
19.
J Virol ; 84(19): 10386-94, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660192

RESUMO

The immunogenicity and durability of genetic vaccines are influenced by the composition of gene inserts and choice of delivery vector. DNA vectors are a promising vaccine approach showing efficacy when combined in prime-boost regimens with recombinant protein or viral vectors, but they have shown limited comparative efficacy as a stand-alone platform in primates, due possibly to suboptimal gene expression or cell targeting. Here, regimens using DNA plasmids modified for optimal antigen expression and recombinant adenovirus (rAd) vectors, all encoding the glycoprotein (GP) gene from Angola Marburg virus (MARV), were compared for their ability to provide immune protection against lethal MARV Angola infection. Heterologous DNA-GP/rAd5-GP prime-boost and single-modality rAd5-GP, as well as the DNA-GP-only vaccine, prevented death in all vaccinated subjects after challenge with a lethal dose of MARV Angola. The DNA/DNA vaccine induced humoral responses comparable to those induced by a single inoculation with rAd5-GP, as well as CD4(+) and CD8(+) cellular immune responses, with skewing toward CD4(+) T-cell activity against MARV GP. Vaccine regimens containing rAd-GP, alone or as a boost, exhibited cellular responses with CD8(+) T-cell dominance. Across vaccine groups, CD8(+) T-cell subset dominance comprising cells exhibiting a tumor necrosis factor alpha (TNF-alpha) and gamma interferon (IFN-gamma) double-positive functional phenotype was associated with an absence or low frequency of clinical symptoms, suggesting that both the magnitude and functional phenotype of CD8(+) T cells may determine vaccine efficacy against infection by MARV Angola.


Assuntos
Vetores Genéticos , Doença do Vírus de Marburg/imunologia , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/genética , Marburgvirus/imunologia , Vacinas Virais/farmacologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Genes Virais , Imunidade Celular , Imunização Secundária , Macaca fascicularis , Modelos Animais , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
20.
Vaccine ; 28(27): 4369-75, 2010 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-20444437

RESUMO

A powder vaccine intended for aerosol delivery was formulated by spray drying the Ad35-vectored tuberculosis (TB) AERAS-402 vaccine with mannitol-based stabilizers. Thermodynamic properties, water absorption, particle size distribution and morphology of the powders were evaluated. Virus survival during spray drying and storage was determined by medium Tissue Culture Infectious Dose (TCID(50)). A mannitol-based powder (mannitol-cyclodextrin-trehalose-dextran, MCTD) had a narrow size distribution with a median volume diameter around 3.2-3.5microm (suitable for pulmonary vaccination of humans) and good aerosolization characteristics. The spray dry powders generated from mannitol-based formulations were hydrophobic, which benefits virus survival during both production and storage at 4 degrees C, 25 degrees C and 37 degrees C as compared to the hygroscopic formulations (trehalose, sucrose, dextran, PVP, leucine). In conclusion, this study demonstrates that it is possible to produce in a one-step spray drying process a stable dry powder formulation of a TB vaccine suitable for mass vaccination.


Assuntos
Adenoviridae/genética , Química Farmacêutica/métodos , Vetores Genéticos/genética , Vacinas contra a Tuberculose/genética , Vacinas contra a Tuberculose/imunologia , Linhagem Celular , Humanos , Vacinas contra a Tuberculose/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA