Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Vaccine ; 41(45): 6762-6773, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37739888

RESUMO

Novavax, a global vaccine company, began evaluating NVX-CoV2373 in human studies in May 2020 and the pivotal placebo-controlled phase 3 studies started in November 2020; five clinical studies provided adult and adolescent clinical data for over 31,000 participants who were administered NVX-CoV2373. This extensive data has demonstrated a well-tolerated response to NVX-CoV2373 and high vaccine efficacy against mild, moderate, or severe COVID-19 using a two-dose series (Dunkle et al., 2022) [1], (Heath et al., 2021) [2], (Keech et al., 2020) [3], (Mallory et al., 2022) [4]. The most common adverse events seen after administration with NVX-CoV2373 were injection site tenderness, injection site pain, fatigue, myalgia, headache, malaise, arthralgia, nausea, or vomiting. In addition, immunogenicity against variants of interest (VOI) and variants of concern (VOC) was established with high titers of ACE2 receptor-inhibiting and neutralizing antibodies in these studies (EMA, 2022) [5], (FDA, 2023) [6]. Further studies on correlates of protection determined that titers of anti-Spike IgG and neutralizing antibodies correlated with efficacy against symptomatic COVID-19 established in clinical trials (p < 0.001 for recombinant protein vaccine and p = 0.005 for mRNA vaccines for IgG levels) (Fong et al., 2022) [7]. Administration of a booster dose of the recombinant protein vaccine approximately 6 months following the primary two-dose series resulted in substantial increases in humoral antibodies against both the prototype strain and all evaluated variants, similar to or higher than the antibody levels observed in phase 3 studies that were associated with high vaccine efficacy (Dunkle et al., 2022) [1], (Mallory et al., 2022) [4]. These findings, together with the well tolerated safety profile, support use of the recombinant protein vaccine as primary series and booster regimens.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Adolescente , Adulto , Humanos , Vacinas contra COVID-19/efeitos adversos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/genética , COVID-19/prevenção & controle , Adjuvantes Imunológicos , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Anticorpos Neutralizantes , Medição de Risco , Imunoglobulina G , Anticorpos Antivirais , Imunogenicidade da Vacina
2.
Hum Vaccin Immunother ; 19(1): 2167907, 2023 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-36880669

RESUMO

Individuals who are immunocompromised (IC) due to therapy or underlying disease are at increased risk of herpes zoster (HZ). This study evaluates the public health impact of recombinant zoster vaccine (RZV) relative to no HZ vaccination for the prevention of HZ among adults aged ≥18 years diagnosed with selected cancers in the United States (US). A static Markov model was used to simulate three cohorts of individuals who are IC with cancer (time horizon of 30 years; one-year cycle length): hematopoietic stem cell transplant (HSCT) recipients, patients with breast cancer (BC; a solid tumor example), and patients with Hodgkin's lymphoma (HL; a hematological malignancy example). Cohort sizes reflect the estimated annual incidence of each condition in the US population (19,671 HSCT recipients, 279,100 patients with BC, and 8,480 patients with HL). Vaccination with RZV resulted in 2,297; 38,068; and 848 fewer HZ cases for HSCT recipients, patients with BC, and patients with HL, respectively (each versus no vaccine). Vaccination with RZV also resulted in 422; 3,184; and 93 fewer postherpetic neuralgia cases for HSCT, BC, and HL, respectively. Analyses estimated the quality-adjusted life years gained to be 109, 506, and 17 for HSCT, BC, and HL, respectively. To prevent one HZ case, the number needed to vaccinate was 9, 8, and 10, for HSCT, BC, and HL, respectively. These results suggest RZV vaccination may be an effective option to significantly reduce HZ disease burden among patients diagnosed with selected cancers in the US.


Shingles cases can be prevented by recombinant zoster vaccine (RZV). People who have a weakened immune system (immunocompromised) due to disease or therapy are more likely to develop shingles. For example, shingles occurs in nearly a quarter of patients receiving immunosuppressive treatment for blood cancers. To estimate the public health impact of vaccination against shingles in people who are immunocompromised due to cancer in the United States (US), we used a model to simulate groups with selected types of cancer. The results indicate vaccination with RZV can significantly reduce shingles cases and related complications among these groups in the US.


Assuntos
Neoplasias da Mama , Vacina contra Herpes Zoster , Herpes Zoster , Neuralgia Pós-Herpética , Humanos , Adulto , Estados Unidos , Adolescente , Feminino , Vacina contra Herpes Zoster/efeitos adversos , Saúde Pública , Análise Custo-Benefício , Herpes Zoster/epidemiologia , Herpes Zoster/prevenção & controle , Herpesvirus Humano 3 , Neuralgia Pós-Herpética/epidemiologia , Vacinas Sintéticas/efeitos adversos
3.
PLoS Pathog ; 18(6): e1010658, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35759511

RESUMO

Nipah virus (NiV) disease is a bat-borne zoonosis responsible for outbreaks with high lethality and is a priority for vaccine development. With funding from the Coalition of Epidemic Preparedness Innovations (CEPI), we are developing a chimeric vaccine (PHV02) composed of recombinant vesicular stomatitis virus (VSV) expressing the envelope glycoproteins of both Ebola virus (EBOV) and NiV. The EBOV glycoprotein (GP) mediates fusion and viral entry and the NiV attachment glycoprotein (G) is a ligand for cell receptors, and stimulates neutralizing antibody, the putative mediator of protection against NiV. PHV02 is identical in construction to the registered Ebola vaccine (Ervebo) with the addition of the NiV G gene. NiV ephrin B2 and B3 receptors are expressed on neural cells and the wild-type NiV is neurotropic and causes encephalitis in affected patients. It was therefore important to assess whether the NiV G alters tropism of the rVSV vector and serves as a virulence factor. PHV02 was fully attenuated in adult hamsters inoculated by the intramuscular (IM) route, whereas parental wild-type VSV was 100% lethal. Two rodent models (mice, hamsters) were infected by the intracerebral (IC) route with graded doses of PHV02. Comparator active controls in various experiments included rVSV-EBOV (representative of Ebola vaccine) and yellow fever (YF) 17DD commercial vaccine. These studies showed PHV02 to be more neurovirulent than both rVSV-EBOV and YF 17DD in infant animals. PHV02 was lethal for adult hamsters inoculated IC but not for adult mice. In contrast YF 17DD retained virulence for adult mice inoculated IC but was not virulent for adult hamsters. Because of the inconsistency of neurovirulence patterns in the rodent models, a monkey neurovirulence test (MNVT) was performed, using YF 17DD as the active comparator because it has a well-established profile of quantifiable microscopic changes in brain centers and a known reporting rate of neurotropic adverse events in humans. In the MNVT PHV02 was significantly less neurovirulent than the YF 17DD vaccine reference control, indicating that the vaccine will have an acceptable safety profile for humans. The findings are important because they illustrate the complexities of phenotypic assessment of novel viral vectors with tissue tropisms determined by transgenic proteins, and because it is unprecedented to use a heterologous comparator virus (YF vaccine) in a regulatory-enabling study. This approach may have value in future studies of other novel viral vectors.


Assuntos
Infecções por Henipavirus , Estomatite Vesicular , Vacinas Virais , Animais , Modelos Animais de Doenças , Vacinas contra Ebola , Glicoproteínas/genética , Doença pelo Vírus Ebola/prevenção & controle , Infecções por Henipavirus/prevenção & controle , Humanos , Camundongos , Vírus Nipah/genética , Vacinas Atenuadas/efeitos adversos , Vacinas Sintéticas/efeitos adversos , Estomatite Vesicular/prevenção & controle , Vacinas Virais/efeitos adversos
4.
Arthritis Rheumatol ; 74(11): 1833-1841, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35666070

RESUMO

OBJECTIVE: Persons with immune-mediated inflammatory diseases (IMIDs) are at an increased risk of herpes zoster (HZ). In 2018, the Centers for Disease Control and Prevention recommended a highly efficacious vaccine, recombinant zoster vaccine (RZV), for prevention of HZ in immunocompetent patients ≥50 years of age. This study was undertaken to estimate RZV vaccination among adults ages ≥50 years with IMIDs during 2018-2019 and to examine possible vaccine-related flares following RZV. METHODS: We identified a cohort of IMID patients using medical claims data from the IBM MarketScan (ages 50-64 years) and Centers for Medicare and Medicaid Services Medicare (ages ≥65 years) databases. Presumed flares were defined as hospitalization/emergency department visit for their respective IMIDs, or steroid treatment with a short-acting oral glucocorticoid or parenteral glucocorticoid injection. We conducted a self-controlled case series (SCCS) analysis to examine a temporal association between RZV and flares. RESULTS: Among enrollees with IMIDs, 14.8% of 55,654 MarketScan enrollees and 43.2% of 160,545 Medicare enrollees received ≥1 dose of RZV in 2018-2019. Two-dose series completion rates were 76.6% in MarketScan enrollees and 85.4% in Medicare enrollees. In the SCCS analysis, 10% and 13% developed flares in the control window, compared to 9% and 11-12% in the risk window following 1 or 2 doses of RZV among MarketScan and Medicare enrollees, respectively. We found no statistically significant increase in flares following RZV administration for any IMID in either age group following RZV dose 1 or dose 2. CONCLUSION: We did not find an increase in presumed flares following RZV vaccination. Among adults ages ≥50 years with IMIDs, a substantial proportion received RZV compared to general zoster coverage estimates, and series completion rates were high.


Assuntos
Vacina contra Herpes Zoster , Herpes Zoster , Estados Unidos , Humanos , Idoso , Pessoa de Meia-Idade , Vacina contra Herpes Zoster/uso terapêutico , Glucocorticoides/uso terapêutico , Medicare , Herpes Zoster/epidemiologia , Herpes Zoster/prevenção & controle , Herpes Zoster/tratamento farmacológico , Herpesvirus Humano 3 , Vacinas Sintéticas/efeitos adversos
5.
Am J Epidemiol ; 191(5): 957-964, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35152283

RESUMO

The recombinant herpes zoster vaccine (RZV), approved as a 2-dose series in the United States in October 2017, has proven highly effective and generally safe. However, a small risk of Guillain-Barré syndrome after vaccination was identified after approval, and questions remain about other possible adverse events. This data-mining study assessed RZV safety in the United States using the self-controlled tree-temporal scan statistic, scanning data on thousands of diagnoses recorded during follow-up to detect any statistically unusual temporal clustering of cases within a large hierarchy of diagnoses. IBM MarketScan data on commercially insured persons at least 50 years of age receiving RZV between January 1, 2018, and May 5, 2020, were used, including 56 days of follow-up; 1,014,329 doses were included. Statistically significant clustering was found within a few days of vaccination for unspecified adverse effects, complications, or reactions to immunization or other medical substances/care; fever; unspecified allergy; syncope/collapse; cellulitis; myalgia; and dizziness/giddiness. These findings are consistent with the known safety profile of this and other injected vaccines. No cluster of Guillain-Barré syndrome was detected, possibly due to insufficient sample size. This signal-detection method has now been applied to 5 vaccines, with consistently plausible results, and seems a promising addition to vaccine-safety evaluation methods.


Assuntos
Síndrome de Guillain-Barré , Vacina contra Herpes Zoster , Herpes Zoster , Síndrome de Guillain-Barré/epidemiologia , Síndrome de Guillain-Barré/etiologia , Herpes Zoster/etiologia , Herpes Zoster/prevenção & controle , Vacina contra Herpes Zoster/efeitos adversos , Humanos , Estados Unidos/epidemiologia , Vacinação , Vacinas Sintéticas/efeitos adversos
6.
Ann Intern Med ; 175(3): JC35, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35226533

RESUMO

SOURCE CITATION: Goud R, Lufkin B, Duffy J, et al. Risk of Guillain-Barré syndrome following recombinant zoster vaccine in Medicare beneficiaries. JAMA Intern Med. 2021;181:1623-30. 34724025.


Assuntos
Síndrome de Guillain-Barré , Vacina contra Herpes Zoster , Herpes Zoster , Vacinas contra Influenza , Idoso , Síndrome de Guillain-Barré/etiologia , Herpes Zoster/complicações , Herpes Zoster/prevenção & controle , Vacina contra Herpes Zoster/efeitos adversos , Humanos , Medicare , Estados Unidos , Vacinas Sintéticas/efeitos adversos
7.
JAMA Intern Med ; 181(12): 1623-1630, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34724025

RESUMO

Importance: Guillain-Barré syndrome can be reported after vaccination. This study assesses the risk of Guillain-Barré syndrome after administration of recombinant zoster vaccine (RZV or Shingrix), which is administered in 2 doses 2 to 6 months apart. Objective: Use Medicare claims data to evaluate risk of developing Guillain-Barré syndrome following vaccination with zoster vaccine. Design, Setting, and Participants: This case series cohort study included 849 397 RZV-vaccinated and 1 817 099 zoster vaccine live (ZVL or Zostavax)-vaccinated beneficiaries aged 65 years or older. Self-controlled analyses included events identified from 2 113 758 eligible RZV-vaccinated beneficiaries 65 years or older. We compared the relative risk of Guillain-Barré syndrome after RZV vs ZVL, followed by claims-based and medical record-based self-controlled case series analyses to assess risk of Guillain-Barré syndrome during a postvaccination risk window (days 1-42) compared with a control window (days 43-183). In self-controlled analyses, RZV vaccinees were observed from October 1, 2017, to February 29, 2020. Patients were identified in the inpatient, outpatient procedural (including emergency department), and office settings using Medicare administrative data. Exposures: Vaccination with RZV or ZVL vaccines. Main Outcomes and Measures: Guillain-Barré syndrome was identified in Medicare administrative claims data, and cases were assessed through medical record review using the Brighton Collaboration case definition. Results: Amongst those who received RZV vaccinees, the mean age was 74.8 years at first dose, and 58% were women, whereas among those who received the ZVL vaccine, the mean age was 74.3 years, and 60% were women. In the cohort analysis we detected an increase in risk of Guillain-Barré syndrome among RZV vaccinees compared with ZVL vaccinees (rate ratio [RR], 2.34; 95% CI, 1.01-5.41; P = .047). In the self-controlled analyses, we observed 24 and 20 cases during the risk and control period, respectively. Our claims-based analysis identified an increased risk in the risk window compared with the control window (RR, 2.84; 95% CI, 1.53-5.27; P = .001), with an attributable risk of 3 per million RZV doses (95% CI, 0.62-5.64). Our medical record-based analysis confirmed this increased risk (RR, 4.96; 95% CI, 1.43-17.27; P = .01). Conclusions and Relevance: Findings of this case series cohort study indicate a slightly increased risk of Guillain-Barré syndrome during the 42 days following RZV vaccination in the Medicare population, with approximately 3 excess Guillain-Barré syndrome cases per million vaccinations. Clinicians and patients should be aware of this risk, while considering the benefit of decreasing the risk of herpes zoster and its complications through an efficacious vaccine, as risk-benefit balance remains in favor of vaccination.


Assuntos
Síndrome de Guillain-Barré/induzido quimicamente , Vacina contra Herpes Zoster/efeitos adversos , Herpes Zoster/prevenção & controle , Medicare/economia , Vacinação/efeitos adversos , Vacinas Sintéticas/efeitos adversos , Idoso , Análise Custo-Benefício , Feminino , Síndrome de Guillain-Barré/epidemiologia , Humanos , Incidência , Masculino , Estudos Retrospectivos , Estados Unidos/epidemiologia , Vacinação/economia
11.
ACS Nano ; 14(10): 12522-12537, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33034449

RESUMO

Humanity is experiencing a catastrophic pandemic. SARS-CoV-2 has spread globally to cause significant morbidity and mortality, and there still remain unknowns about the biology and pathology of the virus. Even with testing, tracing, and social distancing, many countries are struggling to contain SARS-CoV-2. COVID-19 will only be suppressible when herd immunity develops, either because of an effective vaccine or if the population has been infected and is resistant to reinfection. There is virtually no chance of a return to pre-COVID-19 societal behavior until there is an effective vaccine. Concerted efforts by physicians, academic laboratories, and companies around the world have improved detection and treatment and made promising early steps, developing many vaccine candidates at a pace that has been unmatched for prior diseases. As of August 11, 2020, 28 of these companies have advanced into clinical trials with Moderna, CanSino, the University of Oxford, BioNTech, Sinovac, Sinopharm, Anhui Zhifei Longcom, Inovio, Novavax, Vaxine, Zydus Cadila, Institute of Medical Biology, and the Gamaleya Research Institute having moved beyond their initial safety and immunogenicity studies. This review analyzes these frontrunners in the vaccine development space and delves into their posted results while highlighting the role of the nanotechnologies applied by all the vaccine developers.


Assuntos
Ensaios Clínicos como Assunto , Indústria Farmacêutica/métodos , Nanotecnologia/métodos , Vacinas Virais/imunologia , Vacinas contra COVID-19 , Infecções por Coronavirus/economia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Humanos , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia , Vacinas Virais/efeitos adversos , Vacinas Virais/economia
12.
Vaccine ; 38(35): 5734-5739, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32653276

RESUMO

Several protein vaccine candidates are among the COVID-19 vaccines in development. The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) has prepared a standardized template to describe the key considerations for the benefit-risk assessment of protein vaccines. This will help key stakeholders to assess potential safety issues and understand the benefit-risk of such a vaccine platform. The structured and standardized assessment provided by the template would also help contribute to improved public acceptance and communication of licensed protein vaccines.


Assuntos
Vacinas Virais/efeitos adversos , Vacinas Virais/imunologia , Antígenos Virais/administração & dosagem , Antígenos Virais/efeitos adversos , Antígenos Virais/imunologia , Vacinas contra COVID-19 , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Humanos , Segurança do Paciente , Medição de Risco , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia , Proteínas Virais/administração & dosagem , Proteínas Virais/efeitos adversos , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem
14.
Ann Intern Med ; 170(6): 380-388, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30776797

RESUMO

Background: The U.S. Advisory Committee on Immunization Practices recently developed recommendations for use of a new recombinant zoster vaccine (RZV). Objective: To evaluate the cost-effectiveness of vaccination with RZV compared with zoster vaccine live (ZVL) and no vaccination, the cost-effectiveness of vaccination with RZV for persons who have previously received ZVL, and the cost-effectiveness of preferential vaccination with RZV over ZVL. Design: Simulation (state-transition) model using U.S. epidemiologic, clinical, and cost data. Data Sources: Published data. Target Population: Hypothetical cohort of immunocompetent U.S. adults aged 50 years or older. Time Horizon: Lifetime. Perspective: Societal and health care sector. Intervention: Vaccination with RZV (recommended 2-dose regimen), vaccination with ZVL, and no vaccination. Outcome Measures: The primary outcome measure was the incremental cost-effectiveness ratio (ICER). Results of Base-Case Analysis: For vaccination with RZV compared with no vaccination, ICERs ranged by age from $10 000 to $47 000 per quality-adjusted life-year (QALY), using a societal perspective and assuming 100% completion of the 2-dose RZV regimen. For persons aged 60 years or older, ICERs were less than $60 000 per QALY. Vaccination with ZVL was dominated by vaccination with RZV for all age groups 60 years or older. Results of Sensitivity Analysis: Results were most sensitive to changes in vaccine effectiveness, duration of protection, herpes zoster incidence, and probability of postherpetic neuralgia. Vaccination with RZV after previous administration of ZVL yielded an ICER of less than $60 000 per QALY for persons aged 60 years or older. In probabilistic sensitivity analyses, RZV remained the preferred strategy in at least 95% of simulations, including those with 50% completion of the second dose. Limitation: Few data were available on risk for serious adverse events, adherence to the recommended 2-dose regimen, and probability of recurrent zoster. Conclusion: Vaccination with RZV yields cost-effectiveness ratios lower than those for many recommended adult vaccines, including ZVL. Results are robust over a wide range of plausible values. Primary Funding Source: Centers for Disease Control and Prevention.


Assuntos
Vacina contra Herpes Zoster/economia , Herpes Zoster/prevenção & controle , Neuralgia Pós-Herpética/prevenção & controle , Vacinação/economia , Idoso , Idoso de 80 Anos ou mais , Análise Custo-Benefício , Projetos de Pesquisa Epidemiológica , Política de Saúde , Vacina contra Herpes Zoster/efeitos adversos , Humanos , Adesão à Medicação , Pessoa de Meia-Idade , Guias de Prática Clínica como Assunto , Anos de Vida Ajustados por Qualidade de Vida , Prevenção Secundária , Sensibilidade e Especificidade , Vacinação/efeitos adversos , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/economia
15.
Hum Vaccin Immunother ; 13(11): 2612-2625, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-28933657

RESUMO

Dengue virus (DV) is the etiologic agent of dengue fever, the most significant mosquito-borne viral disease in humans. Most DV vaccine approaches are focused on generating antibody mediated responses; one such DV vaccine is approved for use in humans but its efficacy is limited. While it is clear that T cell responses play important role in DV infection and subsequent disease manifestations, fewer studies are aimed at developing vaccines that induce robust T cells responses. Potent T cell based vaccines require 2 critical components: the identification of specific T cell stimulating MHC associated peptides, and an optimized vaccine delivery vehicle capable of simultaneously delivering the antigens and any required adjuvants. We have previously identified and characterized DV specific HLA-A2 and -A24 binding DV serotypes conserved epitopes, and the feasibility of an epitope based vaccine for DV infection. In this study, we build on those previous studies and describe an investigational DV vaccine using T cell epitopes incorporated into a calcium phosphate nanoparticle (CaPNP) delivery system. This study presents a comprehensive analysis of functional immunogenicity of DV CaPNP/multipeptide formulations in vitro and in vivo and demonstrates the CaPNP/multipeptide vaccine is capable of inducing T cell responses against all 4 serotypes of DV. This synthetic vaccine is also cost effective, straightforward to manufacture, and stable at room temperature in a lyophilized form. This formulation may serve as an effective candidate DV vaccine that protects against all 4 serotypes as either a prophylactic or therapeutic vaccine.


Assuntos
Fosfatos de Cálcio/química , Vacinas contra Dengue/imunologia , Epitopos de Linfócito T/química , Imunização/métodos , Nanopartículas/administração & dosagem , Nanopartículas/química , Animais , Animais Geneticamente Modificados , Antígenos Virais/química , Antígenos Virais/imunologia , Fosfatos de Cálcio/administração & dosagem , Dengue/imunologia , Dengue/prevenção & controle , Dengue/terapia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/efeitos adversos , Vacinas contra Dengue/economia , Vírus da Dengue/química , Vírus da Dengue/imunologia , Sistemas de Liberação de Medicamentos , Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Humanos , Imunogenicidade da Vacina , Camundongos , Linfócitos T/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/efeitos adversos , Vacinas de Subunidades Antigênicas/economia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/economia , Vacinas Sintéticas/imunologia
16.
PLoS Med ; 13(11): e1002181, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27898668

RESUMO

BACKGROUND: Large Phase III trials across Asia and Latin America have recently demonstrated the efficacy of a recombinant, live-attenuated dengue vaccine (Dengvaxia) over the first 25 mo following vaccination. Subsequent data collected in the longer-term follow-up phase, however, have raised concerns about a potential increase in hospitalization risk of subsequent dengue infections, in particular among young, dengue-naïve vaccinees. We here report predictions from eight independent modelling groups on the long-term safety, public health impact, and cost-effectiveness of routine vaccination with Dengvaxia in a range of transmission settings, as characterised by seroprevalence levels among 9-y-olds (SP9). These predictions were conducted for the World Health Organization to inform their recommendations on optimal use of this vaccine. METHODS AND FINDINGS: The models adopted, with small variations, a parsimonious vaccine mode of action that was able to reproduce quantitative features of the observed trial data. The adopted mode of action assumed that vaccination, similarly to natural infection, induces transient, heterologous protection and, further, establishes a long-lasting immunogenic memory, which determines disease severity of subsequent infections. The default vaccination policy considered was routine vaccination of 9-y-old children in a three-dose schedule at 80% coverage. The outcomes examined were the impact of vaccination on infections, symptomatic dengue, hospitalised dengue, deaths, and cost-effectiveness over a 30-y postvaccination period. Case definitions were chosen in accordance with the Phase III trials. All models predicted that in settings with moderate to high dengue endemicity (SP9 ≥ 50%), the default vaccination policy would reduce the burden of dengue disease for the population by 6%-25% (all simulations: -3%-34%) and in high-transmission settings (SP9 ≥ 70%) by 13%-25% (all simulations: 10%- 34%). These endemicity levels are representative of the participating sites in both Phase III trials. In contrast, in settings with low transmission intensity (SP9 ≤ 30%), the models predicted that vaccination could lead to a substantial increase in hospitalisation because of dengue. Modelling reduced vaccine coverage or the addition of catch-up campaigns showed that the impact of vaccination scaled approximately linearly with the number of people vaccinated. In assessing the optimal age of vaccination, we found that targeting older children could increase the net benefit of vaccination in settings with moderate transmission intensity (SP9 = 50%). Overall, vaccination was predicted to be potentially cost-effective in most endemic settings if priced competitively. The results are based on the assumption that the vaccine acts similarly to natural infection. This assumption is consistent with the available trial results but cannot be directly validated in the absence of additional data. Furthermore, uncertainties remain regarding the level of protection provided against disease versus infection and the rate at which vaccine-induced protection declines. CONCLUSIONS: Dengvaxia has the potential to reduce the burden of dengue disease in areas of moderate to high dengue endemicity. However, the potential risks of vaccination in areas with limited exposure to dengue as well as the local costs and benefits of routine vaccination are important considerations for the inclusion of Dengvaxia into existing immunisation programmes. These results were important inputs into WHO global policy for use of this licensed dengue vaccine.


Assuntos
Vacinas contra Dengue/economia , Vacinas contra Dengue/normas , Modelos Teóricos , Saúde Pública , Segurança , Vacinação/métodos , Criança , Análise Custo-Benefício , Vacinas contra Dengue/efeitos adversos , Humanos , Estudos Soroepidemiológicos , Vacinação/efeitos adversos , Vacinação/economia , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/economia , Vacinas Atenuadas/normas , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/economia , Vacinas Sintéticas/normas
17.
Vaccine ; 34(44): 5376-5383, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27591952

RESUMO

Shigellosis is an acute bacillary diarrheal disease caused by the gram negative bacillus Shigella. The existence of multiple Shigella serotypes and their growing resistance to antibiotics stress the urgent need for the development of vaccine that is protective across all serotypes. Shigella's IpaB antigen is involved in translocon pore formation, promotes bacterial invasion and induces apoptosis in macrophages. S. Typhi GroEL (Hsp 60) is the immunodominant antigen inducing both arms of immunity and has been explored as adjuvant in this study. The present study evaluates the immunogenicity and protective efficacy of recombinant IpaB domain-GroEL fusion protein in mice against lethal Shigella infection. The IpaB domain and GroEL genes were fused using overlap extension PCR and cloned in pRSETA expression vector. Fused gene was expressed in Escherichia coli BL-21 cells and the resulting 90 KDa fusion protein was purified by affinity chromatography. Intranasal (i.n.) immunization of mice with fusion protein increased the IgG and IgA antibody titers as compared to the group immunized with IpaB and GroEL and control PBS immunized group. Also IgG1 and IgG2a antibodies induced in fusion protein immunized mice were higher than co-immunized group. Significant increase in lymphocyte proliferation and cytokine levels (IFN-γ, IL-4 and IL-10), indicates induction of both Th1 and Th2 immune responses in both immunized groups. Immunization with fusion protein protected 90-95% of mice whereas 80-85% survivability was observed in co-immunized group against lethal challenge with S. flexneri, S. boydii and S. sonnei. Passive immunization conferred 60-70% protection in mice against all these Shigella species. Organ burden and histopathology studies also revealed significant decrease in lung infection as compared to the co-immunized group. Since IpaB is the conserved dominant molecule in all Shigella species, this study will lead to an ideal platform for the development of safe, efficacious and cost-effective recombinant vaccine against Shigella serotypes.


Assuntos
Anticorpos Antibacterianos/sangue , Disenteria Bacilar/prevenção & controle , Proteínas Recombinantes de Fusão/imunologia , Vacinas contra Shigella , Shigella/imunologia , Adjuvantes Imunológicos , Animais , Proteínas de Bactérias/genética , Chaperonina 60/genética , Citocinas/biossíntese , Escherichia coli/genética , Imunização Passiva , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Interleucina-10/biossíntese , Interleucina-4/biossíntese , Pulmão/microbiologia , Pulmão/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Salmonella typhi/química , Shigella/isolamento & purificação , Vacinas contra Shigella/efeitos adversos , Vacinas contra Shigella/economia , Vacinas contra Shigella/genética , Vacinas contra Shigella/imunologia , Células Th1/imunologia , Células Th2/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
18.
Vaccine ; 34(51): 6597-6609, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27395563

RESUMO

The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety of live, recombinant viral vaccines incorporating genes from heterologous viral and other microbial pathogens in their genome (so-called "chimeric virus vaccines"). Many such viral vector vaccines are now at various stages of clinical evaluation. Here, we introduce an attenuated form of recombinant vesicular stomatitis virus (rVSV) as a potential chimeric virus vaccine for HIV-1, with implications for use as a vaccine vector for other pathogens. The rVSV/HIV-1 vaccine vector was attenuated by combining two major genome modifications. These modifications acted synergistically to greatly enhance vector attenuation and the resulting rVSV vector demonstrated safety in sensitive mouse and non-human primate neurovirulence models. This vector expressing HIV-1 gag protein has completed evaluation in two Phase I clinical trials. In one trial the rVSV/HIV-1 vector was administered in a homologous two-dose regimen, and in a second trial with pDNA in a heterologous prime boost regimen. No serious adverse events were reported nor was vector detected in blood, urine or saliva post vaccination in either trial. Gag specific immune responses were induced in both trials with highest frequency T cell responses detected in the prime boost regimen. The rVSV/HIV-1 vector also demonstrated safety in an ongoing Phase I trial in HIV-1 positive participants. Additionally, clinical trial material has been produced with the rVSV vector expressing HIV-1 env, and Phase I clinical evaluation will initiate in the beginning of 2016. In this paper, we use a standardized template describing key characteristics of the novel rVSV vaccine vectors, in comparison to wild type VSV. The template facilitates scientific discourse among key stakeholders by increasing transparency and comparability of information. The Brighton Collaboration V3SWG template may also be useful as a guide to the evaluation of other recombinant viral vector vaccines.


Assuntos
Vacinas contra a AIDS/efeitos adversos , Vacinas contra a AIDS/imunologia , Portadores de Fármacos , Vesiculovirus/genética , Vacinas contra a AIDS/genética , Animais , Ensaios Clínicos Fase I como Assunto , Avaliação Pré-Clínica de Medicamentos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Vetores Genéticos , Humanos , Primatas , Medição de Risco , Linfócitos T/imunologia , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA