Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Biomed Pharmacother ; 144: 112304, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34634560

RESUMO

Dengue virus (DENV) is a global health threat causing about half of the worldwide population to be at risk of infection, especially the people living in tropical and subtropical area. Although the dengue disease caused by dengue virus (DENV) is asymptomatic and self-limiting in most people with first infection, increased severe dengue symptoms may be observed in people with heterotypic secondary DENV infection. Since there is a lack of specific antiviral medication, the development of dengue vaccines is critical in the prevention and control this disease. Several targets and strategies in the development of dengue vaccine have been demonstrated. Currently, Dengvaxia, a live-attenuated chimeric yellow-fever/tetravalent dengue vaccine (CYD-TDV) developed by Sanofi Pasteur, has been licensed and approved for clinical use in some countries. However, this vaccine has demonstrated low efficacy in children and dengue-naïve individuals and also increases the risk of severe dengue in young vaccinated recipients. Accordingly, many novel strategies for the dengue vaccine are under investigation and development. Here, we conducted a systemic literature review according to PRISMA guidelines to give a concise overview of various aspects of the vaccine development process against DENVs, mainly targeting five potential strategies including live attenuated vaccine, inactivated virus vaccine, recombinant subunit vaccine, viral-vector vaccine, and DNA vaccine. This study offers the comprehensive view of updated information and current progression of immunogen selection as well as strategies of vaccine development against DENVs.


Assuntos
Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Desenvolvimento de Vacinas , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/efeitos adversos , Vacinas contra Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Humanos , Resultado do Tratamento , Eficácia de Vacinas , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/uso terapêutico , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/genética
2.
Commun Biol ; 4(1): 557, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33976375

RESUMO

Dengue virus (DENV) is spread from human to human through the bite of the female Aedes aegypti mosquito and leads to about 100 million clinical infections yearly. Treatment options and vaccine availability for DENV are limited. Defective interfering particles (DIPs) are considered a promising antiviral approach but infectious virus contamination has limited their development. Here, a DENV-derived DIP production cell line was developed that continuously produced DENV-free DIPs. The DIPs contained and could deliver to cells a DENV serotype 2 subgenomic defective-interfering RNA, which was originally discovered in DENV infected patients. The DIPs released into cell culture supernatant were purified and could potently inhibit replication of all DENV serotypes in cells. Antiviral therapeutics are limited for many viral infection. The DIP system described could be re-purposed to make antiviral DIPs for many other RNA viruses such as SARS-CoV-2, yellow fever, West Nile and Zika viruses.


Assuntos
Vírus Defeituosos , Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/crescimento & desenvolvimento , Dengue/prevenção & controle , Replicação Viral , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Vírus Defeituosos/genética , Vírus Defeituosos/metabolismo , Dengue/virologia , Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Genes Reporter , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , RNA Viral/biossíntese , RNA Viral/genética , Células Vero , Carga Viral
3.
Am J Trop Med Hyg ; 103(2): 855-863, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32394880

RESUMO

New dengue vaccines are needed to prevent this globally expanding vector-borne disease. The V180 vaccine candidate consists of four recombinant, soluble, dengue virus envelope glycoproteins and has been previously evaluated in two clinical trials for safety and immunogenicity in Flavivirus-naive participants (NCT01477580 and NCT0093642). Here, we report on a randomized, placebo-controlled, double-blind study of the safety and immunogenicity of the V180 vaccine in subjects who have previously received the live attenuated tetravalent vaccine (LATV) developed by the National Institute of Allergy and Infectious Diseases (protocol #V180-002 [CIR-301]). The study was designed to evaluate whether this recombinant subunit vaccine could boost the neutralizing antibody responses induced by dengue LATV. Twenty participants who had previously received one or two doses of dengue LATV were randomized and received a single dose of V180 nonadjuvanted (N = 8), V180 adjuvanted with Alhydrogel™ (aluminum hydroxide gel, Brenntag Biosector, Frederikssund, Denmark) (N = 8), or placebo (N = 4). Immunogenicity was measured using a plaque reduction neutralization test at days 1, 15, 28, and 180 after vaccination. In addition, vaccine safety (solicited and unsolicited adverse events) was assessed using a vaccination report card for 28 days following vaccination, and serious adverse events were captured from the time of informed consent through the final study visit at 6 months after vaccination. The results of the study demonstrate that the V180 vaccine is generally well tolerated and immunogenic in these dengue-seropositive volunteers.


Assuntos
Vacinas contra Dengue/uso terapêutico , Dengue/prevenção & controle , Imunização Secundária , Adjuvantes Imunológicos/uso terapêutico , Adulto , Hidróxido de Alumínio/uso terapêutico , Anticorpos Neutralizantes/imunologia , Vírus da Dengue/imunologia , Método Duplo-Cego , Feminino , Humanos , Imunogenicidade da Vacina , Reação no Local da Injeção , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Vacinas Atenuadas/uso terapêutico , Vacinas de Subunidades Antigênicas/uso terapêutico , Vacinas Sintéticas/uso terapêutico , Proteínas do Envelope Viral/imunologia , Adulto Jovem
4.
Vaccine ; 37(36): 5137-5146, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31377079

RESUMO

The first licensed dengue vaccine, CYD-TDV (Dengvaxia) is efficacious in seropositive individuals, but increases the risk for severe dengue in seronegative persons about two years after administration of the first dose. For countries considering the introduction of Dengvaxia, WHO recommends a pre-vaccination screening strategy whereby only persons with evidence of a past dengue infection would be vaccinated. Policy-makers need to consider the risk-benefit of vaccination strategies based on such screening tests, the optimal age to introduce the vaccine, communication and implementation strategies. To address these questions, the Global Dengue and Aedes-transmitted diseases Consortium (GDAC) organized a 3-day workshop in January 2019 with country representatives from Asia and Latin America. The meeting discussions highlighted many challenges in introducing Dengvaxia, in terms of screening test characteristics, costs of such tests combined with a 3-dose schedule, logistics, achieving high coverage rates, vaccine confidence and communication; more challenges than for any other vaccine introduction programme. A screening test would require a high specificity to minimize individual risk, and at the same time high sensitivity to maximize individual and population benefit. The underlying seroprevalence dependent positive predictive value is the best indicator for an acceptable safety profile of a pre-vaccination screening strategy. The working groups discussed many possible implementation strategies. Addressing the bottlenecks in school-based vaccine introduction for Dengvaxia will also benefit other vaccines such as HPV and booster doses for tetanus and pertussis. Levels of public trust are highly variable and context specific, and understanding of population perceptions and concerns is essential to tailor interventions, monitor and mitigate risks.


Assuntos
Vacinas contra Dengue/uso terapêutico , Adolescente , Adulto , Anticorpos Antivirais/imunologia , Criança , Dengue/imunologia , Dengue/microbiologia , Dengue/prevenção & controle , Vacinas contra Dengue/imunologia , Vírus da Dengue , Humanos , Programas de Imunização/métodos , Saúde Pública , Estudos Soroepidemiológicos , Vacinas Atenuadas/uso terapêutico , Organização Mundial da Saúde , Adulto Jovem
5.
Rev Neurol ; 69(3): 113-122, 2019 Aug 01.
Artigo em Espanhol | MEDLINE | ID: mdl-31310001

RESUMO

INTRODUCTION: Dengue is an arboviral infection caused by the dengue virus. The neurological complications associated with this infection are reviewed. DEVELOPMENT: The neurotropic nature of dengue virus has been confirmed in epidemiological studies, case series and histopathological studies. The range of neurological complications is 5.6-14.6%, and they are more frequent in serotypes 1 and 3. Encephalopathy is the most common neurological syndrome (0.5-6%) and its prevalence is higher in children and adolescents. The detection of the viral antigen in brain tissue and the presence of pleocytosis or RNA in cerebrospinal fluid are evidence of the neurotropic nature of dengue virus, which manifests itself in the form of encephalitis. Neurological syndromes during convalescence (disseminated acute cerebellitis, opsoclonus-myoclonus syndrome, mononeuritis, poly-radiculoneuritis and plexitis) appear to be immunomediated. Myelitis can occur during acute dengue virus infection and through an immunomediated mechanism in the convalescence phase. Myalgias, myositis, rhabdomyolysis and hypokalemic paralysis are examples of muscular dysfunction associated with the dengue virus. The incidence of stroke is 0.26%, and may be ischaemic or haemorrhagic. Ophthalmological complications include maculopathy, retinal haemorrhage, optic neuropathy and vitritis. CONCLUSIONS: The spectrum of neurological complications from dengue virus is broad. There are no reliable data on its real incidence because most of the studies published to date are isolated series or cases.


TITLE: Complicaciones neurologicas asociadas a la infeccion por el virus del dengue.Introduccion. El dengue es una infeccion arboviral causada por el virus del dengue. Se revisan las complicaciones neurologicas asociadas a dicha infeccion. Desarrollo. El caracter neurotropo del virus del dengue se ha confirmado en estudios epidemiologicos, series de casos y estudios histopatologicos. El rango de complicaciones neurologicas es del 5,6-14,6%, y son mas frecuentes en los serotipos 1 y 3. La encefalopatia es el sindrome neurologico mas comun (0,5-6%); su prevalencia es mayor en los niños y los adolescentes. La deteccion del antigeno viral en el tejido cerebral y la presencia de pleocitosis o ARN en el liquido cefalorraquideo son evidencia del caracter neurotropo del virus del dengue, que se manifiesta en forma de encefalitis. Los sindromes neurologicos durante la fase de convalecencia (encefalomielitis aguda diseminada, cerebelitis, opsoclonia-mioclonia, mononeuritis, polirradiculoneuritis y plexitis) parecen ser inmunomediados. La mielitis puede suceder durante la infeccion aguda por el virus del dengue y por un mecanismo inmunomediado en la fase de convalecencia. Mialgias, miositis, rabdomiolisis y paralisis hipopotasemica son ejemplos de disfuncion muscular asociada al virus del dengue. La incidencia de ictus es del 0,26%, y puede ser isquemico o hemorragico. Las complicaciones oftalmologicas incluyen maculopatia, hemorragia retiniana, neuropatia optica y vitritis. Conclusiones. El espectro de complicaciones neurologicas por el virus del dengue es amplio. No existen datos fiables sobre su incidencia real porque la mayor parte de los estudios publicados son series o casos aislados.


Assuntos
Dengue/complicações , Doenças do Sistema Nervoso/etiologia , Dengue/diagnóstico , Dengue/terapia , Vacinas contra Dengue/uso terapêutico , Diagnóstico Precoce , Encefalite Viral/etiologia , Oftalmopatias/etiologia , Hidratação , Síndrome de Guillain-Barré/etiologia , Humanos , Mielite Transversa/etiologia , Doenças Neuromusculares/etiologia , Hemorragia Retiniana/etiologia , Acidente Vascular Cerebral/etiologia
6.
Nano Lett ; 18(2): 725-733, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29253342

RESUMO

Many viruses often have closely related yet antigenically distinct serotypes. An ideal vaccine against viral infections should induce a multivalent and protective immune response against all serotypes. Inspired by bacterial membrane vesicles (MVs) that carry different protein components, we constructed an agr locus deletion mutant of the Staphylococcus aureus strain (RN4220-Δagr) to reduce potential toxicity. Nanoscale vesicles derived from this strain (ΔagrMVs) carry at least four major components that can deliver heterologous antigens. These components were each fused with a triple FLAG tag, and the tagged proteins could be incorporated into the ΔagrMVs. The presentation levels were (3.43 ± 0.73)%, (5.07 ± 0.82)%, (2.64 ± 0.61)%, and (2.89 ± 0.74)% of the total ΔagrMV proteins for Mntc-FLAG, PdhB-FLAG, PdhA-FLAG, and Eno-FLAG, respectively. With two DENV envelope E domain III proteins (EDIIIconA and EDIIIconB) as models, the DENV EDIIIconA and EDIIIconB delivered by two staphylococcal components were stably embedded in the ΔagrMVs. Administration of such engineered ΔagrMVs in mice induced antibodies against all four DENV serotypes. Sera from immunized mice protected Vero cells and suckling mice from a lethal challenge of DENV-2. This study will open up new insights into the preparation of multivalent nanosized viral vaccines against viral infections.


Assuntos
Proteínas de Bactérias/genética , Micropartículas Derivadas de Células/genética , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Dengue/prevenção & controle , Staphylococcus aureus/genética , Transativadores/genética , Proteínas do Envelope Viral/genética , Animais , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/uso terapêutico , Deleção de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética
7.
Int J Med Sci ; 14(13): 1342-1359, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29200948

RESUMO

Dengue is an important global threat caused by dengue virus (DENV) that records an estimated 390 million infections annually. Despite the availability of CYD-TDV as a commercial vaccine, its long-term efficacy against all four dengue virus serotypes remains unsatisfactory. There is therefore an urgent need for the development of antiviral drugs for the treatment of dengue. Peptide was once a neglected choice of medical treatment but it has lately regained interest from the pharmaceutical industry following pioneering advancements in technology. In this review, the design of peptide drugs, antiviral activities and mechanisms of peptides and peptidomimetics (modified peptides) action against dengue virus are discussed. The development of peptides as inhibitors for viral entry, replication and translation is also described, with a focus on the three main targets, namely, the host cell receptors, viral structural proteins and viral non-structural proteins. The antiviral peptides designed based on these approaches may lead to the discovery of novel anti-DENV therapeutics that can treat dengue patients.


Assuntos
Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Peptídeos/uso terapêutico , Peptidomiméticos/uso terapêutico , Antivirais/uso terapêutico , Dengue/epidemiologia , Dengue/virologia , Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/patogenicidade , Humanos , Internalização do Vírus/efeitos dos fármacos
8.
Lung ; 195(4): 389-395, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28612239

RESUMO

Dengue fever is an arboviral disease transmitted to humans through the bites of infected female Aedes mosquitoes. Dengue virus is a member of the Flaviviridae family, and human infection can be caused by any of the four antigenically distinct serotypes (DENV 1-4). The infection has become recognized as the most important and prevalent arboviral disease in humans, endemic in almost 100 countries worldwide. Nearly 3 billion people live in areas with transmission risk. Autochthonous transmission of the virus in previously disease-free areas, increased incidence in endemic areas, and epidemic resurgence in controlled regions could increase the risk of contracting more severe forms of the disease, such as dengue hemorrhagic fever (DHF)/dengue shock syndrome (DSS). Symptomatic dengue virus infection can present with a wide range of clinical manifestations, from mild fever to life-threatening DSS. Thoracic complications may manifest as pleural effusion, pneumonitis, non-cardiogenic pulmonary edema, and hemorrhage/hemoptysis. No vaccine is currently available and no specific treatment for dengue fever exists, but prevention and prompt management of complications in patients with DHF can help reduce mortality. This review describes the main clinical, pathological, and imaging findings of thoracic involvement in DHF.


Assuntos
Aedes/virologia , Vírus da Dengue/patogenicidade , Hemoptise/virologia , Pulmão/virologia , Dengue Grave/virologia , Animais , Biópsia , Vacinas contra Dengue/uso terapêutico , Diagnóstico Diferencial , Hemoptise/diagnóstico , Hemoptise/mortalidade , Hemoptise/terapia , Humanos , Pulmão/diagnóstico por imagem , Pulmão/patologia , Valor Preditivo dos Testes , Prognóstico , Fatores de Risco , Dengue Grave/diagnóstico , Dengue Grave/mortalidade , Dengue Grave/terapia , Tomografia Computadorizada por Raios X
9.
J Nanobiotechnology ; 15(1): 26, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28376812

RESUMO

BACKGROUND: Dengue is the most prevalent arthropod-borne viral disease in the world. In this article we present results on the development, characterization and immunogenic evaluation of an alternative vaccine candidate against Dengue. METHODS: The MWNT-DENV3E nanoconjugate was developed by covalent functionalization of carboxylated multi-walled carbon nanotubes (MWNT) with recombinant dengue envelope (DENV3E) proteins. The recombinant antigens were bound to the MWNT using a diimide-activated amidation process and the immunogen was characterized by TEM, AFM and Raman Spectroscopy. Furthermore, the immunogenicity of this vaccine candidate was evaluated in a murine model. RESULTS: Immunization with MWNT-DENV3E induced comparable IgG responses in relation to the immunization with non-conjugated proteins; however, the inoculation of the nanoconjugate into mice generated higher titers of neutralizing antibodies. Cell-mediated responses were also evaluated, and higher dengue-specific splenocyte proliferation was observed in cell cultures derived from mice immunized with MWNT-DENV3E when compared to animals immunized with the non-conjugated DENV3E. CONCLUSIONS: Despite the recent licensure of the CYD-TDV dengue vaccine in some countries, results from the vaccine's phase III trial have cast doubts about its overall efficacy and global applicability. While questions about the effectiveness of the CYD-TDV vaccine still lingers, it is wise to keep at hand an array of vaccine candidates, including alternative non-classical approaches like the one presented here.


Assuntos
Formação de Anticorpos , Vacinas contra Dengue/imunologia , Dengue/prevenção & controle , Nanotubos de Carbono/química , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Proliferação de Células , Citocinas/imunologia , Dengue/imunologia , Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/imunologia , Feminino , Imunidade Celular , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Nanoconjugados/química , Nanomedicina , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Análise Espectral Raman , Baço/citologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
10.
Rev. chil. infectol ; 33(4): 452-454, ago. 2016.
Artigo em Espanhol | LILACS | ID: biblio-1042622

RESUMO

Dengue was first diagnosed on Easter Island on year 2002 and thereafter recurrent outbreaks have occurred involving different serotypes of dengue virus. Its vector, Aedes aegypti has not been eliminated despite the small size of the island. Conditions at the local hospital preclude adequate management of severe and hemorrhagic cases due to the absence of a Critical Care Unit as well as no availability of platelets, or plasma units for transfusion. Besides, transfer, of severely affected patients to continental Chile is cumbersome, slow and expensive. In this scenario, it is advisable to implement selective vaccination of Easter Island habitants with an available quadrivalent attenuated dengue vaccine with the aim to reduce hemorrhagic and severe dengue cases. This strategy should not replace permanent efforts to control waste disposal sites, water sources, maintain vector surveillance and increase education of the population.


El dengue surgió el año 2002 en Isla de Pascua y se ha presentado en brotes intercurrentes desde entonces con aparición de diferentes serotipos. El vector Aedes aegypti no ha logrado ser eliminado a pesar del pequeño tamaño de la isla y las condiciones del hospital local no permiten el manejo de casos graves por ausencia de una unidad de cuidados intensivos y disponibilidad de transfusiones de plaquetas o plasma fresco congelado. Además, el traslado de pacientes graves hacia el continente no es inmediato y es muy costoso. En este escenario, es aconsejable vacunar selectivamente a la población residente con la vacuna cuadrivalente atenuada para disminuir la probabilidad de dengue grave. Esta estrategia no debe reemplazar los esfuerzos para el control de basurales, fuentes de agua, vigilancia del vector y educación de la población.


Assuntos
Humanos , Animais , Masculino , Feminino , Criança , Adolescente , Adulto , Pessoa de Meia-Idade , Adulto Jovem , Vacinação , Dengue/prevenção & controle , Vacinas contra Dengue/uso terapêutico , Polinésia/epidemiologia , Comitês Consultivos , Dengue/transmissão , Dengue/epidemiologia , Vírus da Dengue/imunologia , Insetos Vetores/virologia
12.
Medicina (B.Aires) ; 76(2): 98-102, abr. 2016. graf, tab
Artigo em Espanhol | LILACS | ID: biblio-841550

RESUMO

Los brotes de dengue se suceden anualmente en Argentina desde el año 1998. Existen vacunas contra esta enfermedad en distintos grados de desarrollo que han sido probadas en países endémicos. La más avanzada hasta el momento fue autorizada para su comercialización en tres países de América Latina, para niños mayores de 9 años. En este artículo se discuten los beneficios e inconvenientes de las vacunas, así como los desafíos para la implementación de una estrategia de vacunación. Asimismo, se plantea la necesidad de una estratificación de riesgo con nuevos criterios y visión multidisciplinaria como un camino posible para evaluar la pertinencia de un programa de vacunación en las áreas con mayor riesgo de transmisión, y/o en individuos con mayor riesgo de dengue grave. Se sugiere también que la definición del estatus de endemicidad debe tomar en cuenta a las realidades locales. Por último, este artículo propone una discusión amplia de las evidencias, impacto esperado y aspectos instrumentales que estarían involucrados en la incorporación de una vacuna contra el dengue, ya en mercado o en desarrollo, en el programa nacional de inmunizaciones, y especialmente a qué subpoblaciones debería ser dirigida para que la estrategia de inmunización sea costo-efectiva.


Dengue outbreaks have occurred yearly in Argentina since 1998. A number of candidate vaccines have been tested in endemic countries. The most advanced one was licensed in three countries of Latin America for children over 9 years of age. In the present article the benefits and drawbacks of these vaccines as well as the challenges for the implementation of a vaccination strategy in Argentina are discussed. Furthermore, a risk stratification strategy with new criteria and a multidisciplinary vision is suggested as a possible path for the assessment of the pertinence of a vaccination program in areas showing the highest risk of dengue transmission and/or for people at the greatest risk of developing severe dengue. It is also suggested that the definition regarding the status of endemicity should take into account the local realities. Finally, this paper proposes a broad discussion on the evidences, the expected impact and instrumental aspects that would be involved in the incorporation of a dengue vaccine, marketed or in development, into the national immunization program, and especially which subpopulation should be targeted for the immunization strategy to be cost-effective.


Assuntos
Humanos , Criança , Programas de Imunização/métodos , Dengue/prevenção & controle , Vacinas contra Dengue/uso terapêutico , Argentina/epidemiologia , Incidência , Dengue/epidemiologia , Vacinas contra Dengue/economia , Avaliação do Impacto na Saúde/estatística & dados numéricos
13.
Medicina (B.Aires) ; 76(2): 93-97, abr. 2016.
Artigo em Espanhol | LILACS | ID: biblio-841549

RESUMO

Los responsables de la actual pandemia de Chikungunya (alfavirus), dengue y Zika (flavivirus) son virus trasmitidos por artrópodos, arbovirus. Su importancia aumentó en las Américas en los últimos 20 años. Los vectores principales son Aedes aegypti y A. albopictus. La infección por dengue provee inmunidad duradera al serotipo específico y temporaria a otros tres. La posterior infección por otro serotipo determina mayor gravedad. Existe una vacuna contra dengue registrada, Dengvaxia (Sanofi Pasteur). Otras dos (Butantan y Takeda) comienzan la Fase III en 2016. La infección por Zika suele ser asintomática, o presentarse con exantema, conjuntivitis y fiebre no muy elevada. No existen vacunas ni tratamiento específico. Se puede transmitir por vía parental, sexual y por transfusión sanguínea. Se la ha asociado con microcefalia. Chikungunya causa artralgias prolongadas, con respuesta inmune persistente. Hay dos vacunas candidatas en Fase II. El diagnóstico directo del dengue se realiza por cultivo, RT-PCR y ELISA para detección del antígeno NS1; los métodos indirectos son ELISA-IgM (reacción cruzada con otros flavivirus), MAC-ELISA, y neutralización en placas, que diferencia los 4 serotipos DENV y otros flavivirus. Zika se diagnostica por RT-PCR y aislamiento del virus. El diagnóstico serológico presenta reacciones cruzadas con otros flavivirus. Para CHIKV se emplean cultivo y RT-PCR, MAC-ELISA y neutralización en placas. Contra Aedes se emplean larvicidas organofosforados (temefos), insecticidas organofosforados (malation y fenitrotion) y piretroides (permetrina y deltametrina). Puede haber resistencia. Los derivados vegetales son menos costosos y biodegradables, entre ellos el aceite de cetronela, que microencapsulado se preserva de la evaporación.


Arboviruses are transmitted by arthropods, including those responsible for the current pandemic: alphavirus (Chikungunya) and flaviviruses (dengue and Zika). Its importance increased in the Americas over the past 20 years. The main vectors are Aedes aegypti and A. albopictus. Dengue infection provides long lasting immunity against the specific serotype and temporary to the other three. Subsequent infection by another serotype determines more serious disease. There is a registered vaccine for dengue, Dengvaxia (Sanofi Pasteur). Other two (Butantan and Takeda) are in Phase III in 2016. Zika infection is usually asymptomatic or occurs with rash, conjunctivitis and not very high fever. There is no vaccine or specific treatment. It can be transmitted by parental, sexual and via blood transfusion. It has been associated with microcephaly. Chikungunya causes prolonged joint pain and persistent immune response. Two candidate vaccines are in Phase II. Dengue direct diagnosis is performed by virus isolation, RT-PCR and ELISA for NS1 antigen detection; indirect methods are ELISA-IgM (cross-reacting with other flavivirus), MAC-ELISA, and plaque neutralization. Zika is diagnosed by RT-PCR and virus isolation. Serological diagnosis cross-reacts with other flavivirus. For CHIKV culture, RT-PCR, MAC-ELISA and plaque neutralization are used. Against Aedes organophosphate larvicides (temephos), organophosphorus insecticides (malathion and fenitrothion) and pyrethroids (permethrin and deltamethrin) are usually employed. Resistance has been described to all these products. Vegetable derivatives are less expensive and biodegradable, including citronella oil, which microencapsulated can be preserved from evaporation.


Assuntos
Humanos , Animais , Dengue/diagnóstico , Dengue/prevenção & controle , Dengue/transmissão , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/prevenção & controle , Febre de Chikungunya/transmissão , América/epidemiologia , Ensaio de Imunoadsorção Enzimática , Vacinas Virais/uso terapêutico , Vírus Chikungunya/imunologia , Aedes/virologia , Vírus da Dengue/imunologia , Vacinas contra Dengue/uso terapêutico , Zika virus/imunologia , Infecção por Zika virus/transmissão , Insetos Vetores/fisiologia , Inseticidas
14.
Salud pública Méx ; 58(1): 71-83, ene.-feb. 2016. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-773571

RESUMO

El dengue es un importante problema de salud pública global, que afecta a América Latina y México. Las medidas de prevención y control centradas en vigilancia epidemiológica y control de vectores han resultado parcialmente efectivas y costosas, por lo que el desarrollo de una vacuna contra el dengue ha creado grandes expectativas entre las autoridades sanitarias y las comunidades científicas en el mundo. Sólo la vacuna CYD-TDV, producida por Sanofi-Pasteur, ha sido evaluada en ensayos clínicos controlados fase 3. No obstante a pesar de la importante contribución que esto significa para el desarrollo de una vacuna contra el dengue, los tres estudios clínicos fase 3 de CYD-TDV y el metaanálisis de seguimiento a largo plazo derivado de los mismos proporcionan evidencia de que esta vacuna tiene una eficacia parcial para proteger contra dengue virológicamente confirmado. Al respecto, surgen cuatro consideraciones: a) eficacia adecuada contra infecciones por virus de dengue (DENV) 3 y 4, menor eficacia contra infecciones por DENV 1 y prácticamente nula protección contra infecciones por DENV 2; b) disminución de la eficacia en individuos seronegativos a dengue al inicio de la vacunación; c) 83 y 90% de protección contra hospitalizaciones y formas de dengue grave, respectivamente, a 25 meses de seguimiento, y d) incremento de hospitalización por dengue, en el grupo de vacunados, en niños menores de nueve años de edad al momento de la vacunación, detectado a partir del tercer año de seguimiento. El beneficio de la vacuna CYD-TDV se puede resumir en la protección contra infecciones por DENV 3 y 4, así como en la protección de hospitalizaciones y casos graves en individuos mayores de nueve años y en quienes han tenido infección previa por dengue, pues funciona principalmente como una vacuna de refuerzo. En esta revisión se identificaron elementos sobre eficacia y seguridad de esta vacuna que deben ser tomados en cuenta ante el potencial registro e inclusión en el programa de vacunación en la población mexicana. La evidencia científica disponible sobre la vacuna CYD-TDV demuestra méritos, pero también da lugar a preguntas relevantes que deberían ser contestadas para evaluar apropiadamente el perfil de seguridad del producto, así como las poblaciones blanco de potencial beneficio. Al respecto, consideramos que sería informativo completar el seguimiento indicado de seis años después de iniciar la vacunación, de acuerdo con el protocolo propuesto en los propios estudios del fabricante como una recomendación de la Organización Mundial de la Salud. Al igual que con cualquier nueva vacuna, el potencial registro e implementación de uso de CYD-TDV en el programa nacional de vacunación de México requiere una definición clara de cuál es el balance entre los beneficios y riesgos esperados. En particular, ante una vacuna con eficacia variable y algunas señales de riesgo, en caso de aprobar el registro, se deben desarrollar protocolos de manejo de riesgos detallados que permitan identificar de manera oportuna cualquier evento de salud asociado con la vacunación.


Dengue is a major global public health problem affecting Latin America and Mexico Prevention and control measures, focusing on epidemiological surveillance and vector control, have been partially effective and costly, thus, the development of a vaccine against dengue has created great expectations among health authorities and scientific communities worldwide. The CYD-TDV dengue vaccine produced by Sanofi-Pasteur is the only dengue vaccine evaluated in phase 3 controlled clinical trials. Notwithstanding the significant contribution to the development of a vaccine against dengue, the three phase 3 clinical studies of CYD-TDV and the meta-analysis of the long-term follow up of those studies, have provided evidence that this vaccine exhibited partial vaccine efficacy to protect against virologically confirmed dengue and lead to four considerations: a) adequate vaccine efficacy against dengue virus (DENV) infections 3 and 4, less vaccine efficacy against DENV 1 and no protection against infection by DENV 2; b) decreased vaccine efficacy in dengue seronegative individuals at the beginning of the vaccination; c) 83% and 90% protection against hospitalizations and severe forms of dengue, respectively, at 25 months follow-up; and d) increased hospitalization for dengue in the vaccinated group, in children under nine years of age at the time of vaccination, detected since the third year of follow-up. The benefit of the CYD-TDV vaccine can be summarized in the protection against infection by DENV 3 and 4, as well as protection for hospitalizations and severe cases in people over nine years, who have had previous dengue infection, working mainly as a booster. In this review we identified elements on efficacy and safety of this vaccine that must be taken into account in the licensing process and potential inclusion in the national vaccination program of Mexico. The available scientific evidence on the CYD-TDV vaccine shows merits, but also leads to relevant questions that should be answered to properly assess the safety profile of the product and the target populations of potential benefit. In this regard we consider it would be informative to complete the 6-year follow-up after starting vaccination, according to the company's own study protocol recommended by the World Health Organization. As with any new vaccine, the potential licensing and implementation of the CYD-TDV as part of Mexico's vaccination program, requires a clear definition of the balance between the expected benefits and risks. Particularly with a vaccine with variable efficacy and some signs of risk, in the probable case of licensing, the post-licensed period must involve the development of detailed protocols to immediately identify risks or any health event associated with vaccination.


Assuntos
Humanos , Aprovação de Drogas/legislação & jurisprudência , Programas de Imunização/legislação & jurisprudência , Dengue/prevenção & controle , Vacinas contra Dengue/uso terapêutico , Vacinas Atenuadas/uso terapêutico , Saúde Pública , Resultado do Tratamento , Hospitalização , México
15.
Indian J Med Res ; 144(4): 587-591, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28256468

RESUMO

BACKGROUND & OBJECTIVES: Epitope-based vaccines (EVs) are specific, safe and easy to produce. However, vaccine failure has been frequently reported due to variation within epitopic regions. Therefore, development of vaccines based on conserved epitopes may prevent such vaccine failure. This study was undertaken to identify highly conserved antigenic regions in the four dengue serotypes to produce an epitope-based dengue vaccine. METHODS: Polyprotein sequences of all four dengue serotypes were collected and aligned using MAFFT multiple sequence alignment plugin with Geneious Pro v6.1. Consensus sequences of the polyproteins for all four dengue serotypes were designed and screened against experimentally proven epitopes to predict potential antigenic regions that are conserved among all four dengue serotypes. RESULTS: The antigenic region VDRGWGNGCGLFGKG was 100 per cent conserved in the consensus polyprotein sequences of all four dengue serotypes. Fifteen experimentally proven epitopes were identical to the immunodominant antigenic region. INTERPRETATION & CONCLUSIONS: Computationally predicted antigenic regions may be considered for use in the development of EVs for protection against dengue virus. Such vaccines would be expected to provide protection against dengue infections caused by all dengue serotypes because these would contain antigenic regions highly conserved across those serotypes. Therefore, the immunodominant antigenic region (VDRGWGNGCGLFGKG) and 15 potential epitopes may be considered for use in dengue vaccines.


Assuntos
Antígenos Virais/genética , Vacinas contra Dengue/genética , Dengue/tratamento farmacológico , Epitopos Imunodominantes/genética , Sequência de Aminoácidos/genética , Antígenos Virais/imunologia , Biologia Computacional , Dengue/genética , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/imunologia , Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Humanos , Epitopos Imunodominantes/imunologia , Epitopos Imunodominantes/uso terapêutico , Alinhamento de Sequência , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
17.
J Virol ; 88(13): 7276-85, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24741106

RESUMO

UNLABELLED: Dengue virus (DENV) infects an estimated 400 million people every year, causing prolonged morbidity and sometimes mortality. Development of an effective vaccine has been hampered by the lack of appropriate small animal models; mice are naturally not susceptible to DENV and only become infected if highly immunocompromised. Mouse models lacking both type I and type II interferon (IFN) receptors (AG129 mice) or the type I IFN receptor (IFNAR(-/-) mice) are susceptible to infection with mouse-adapted DENV strains but are severely impaired in mounting functional immune responses to the virus and thus are of limited use for study. Here we used conditional deletion of the type I IFN receptor (IFNAR) on individual immune cell subtypes to generate a minimally manipulated mouse model that is susceptible to DENV while retaining global immune competence. Mice lacking IFNAR expression on CD11c(+) dendritic cells and LysM(+) macrophages succumbed completely to DENV infection, while mice deficient in the receptor on either CD11c(+) or LysM(+) cells were susceptible to infection but often resolved viremia and recovered fully from infection. Conditional IFNAR mice responded with a swift and strong CD8(+) T-cell response to viral infection, compared to a weak response in IFNAR(-/-) mice. Furthermore, mice lacking IFNAR on either CD11c(+) or LysM(+) cells were also sufficiently immunocompetent to raise a protective immune response to a candidate subunit vaccine against DENV-2. These data demonstrate that mice with conditional deficiencies in expression of the IFNAR represent improved models for the study of DENV immunology and screening of vaccine candidates. IMPORTANCE: Dengue virus infects 400 million people every year worldwide, causing 100 million clinically apparent infections, which can be fatal if untreated. Despite many years of research, there are no effective vaccine and no antiviral treatment available for dengue. Development of vaccines has been hampered in particular by the lack of a suitable small animal model. Mouse models used to test dengue vaccine are deficient in interferon (IFN) type I signaling and severely immunocompromised and therefore likely not ideal for the testing of vaccines. In this study, we explored alternative models lacking the IFN receptor only on certain cell types. We show that mice lacking the IFN receptor on either CD11c- or LysM-expressing cells (conditional IFNAR mice) are susceptible to dengue virus infection. Importantly, we demonstrate that conditional IFN receptor knockout mice generate a better immune response to live virus and a candidate dengue vaccine compared to IFNAR mice and are resistant to subsequent challenge.


Assuntos
Células Dendríticas/imunologia , Vacinas contra Dengue/uso terapêutico , Dengue/imunologia , Modelos Animais de Doenças , Interferon Tipo I/fisiologia , Interferon gama/fisiologia , Macrófagos/imunologia , Animais , Citocinas/metabolismo , Células Dendríticas/virologia , Dengue/prevenção & controle , Dengue/virologia , Vírus da Dengue/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Imunização , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Viral/genética , Replicação Viral
18.
PLoS One ; 8(3): e58357, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23472186

RESUMO

The dengue envelope glycoprotein (E) is the major component of virion surface and its ectodomain is composed of domains I, II and III. This protein is the main target for the development of a dengue vaccine with induction of neutralizing antibodies. In the present work, we tested two different vaccination strategies, with combined immunizations in a prime/booster regimen or simultaneous inoculation with a DNA vaccine (pE1D2) and a chimeric yellow fever/dengue 2 virus (YF17D-D2). The pE1D2 DNA vaccine encodes the ectodomain of the envelope DENV2 protein fused to t-PA signal peptide, while the YF17D-D2 was constructed by replacing the prM and E genes from the 17D yellow fever vaccine virus by those from DENV2. Balb/c mice were inoculated with these two vaccines by different prime/booster or simultaneous immunization protocols and most of them induced a synergistic effect on the elicited immune response, mainly in neutralizing antibody production. Furthermore, combined immunization remarkably increased protection against a lethal dose of DENV2, when compared to each vaccine administered alone. Results also revealed that immunization with the DNA vaccine, regardless of the combination with the chimeric virus, induced a robust cell immune response, with production of IFN-γ by CD8+ T lymphocytes.


Assuntos
Vacinas contra Dengue/uso terapêutico , Dengue/prevenção & controle , Vacinas de DNA/uso terapêutico , Proteínas do Envelope Viral/imunologia , Vírus da Febre Amarela/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Vírus da Dengue/genética , Interferon gama/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Terciária de Proteína , Vacinação/métodos
19.
Am J Trop Med Hyg ; 87(4): 743-753, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22890035

RESUMO

The immunogenicity and safety of three novel host-range vaccines containing deletions in the transmembrane domain of dengue virus serotype 2 (DV2) E glycoprotein were evaluated in African green monkeys. The shorter transmembrane domains are capable of functionally spanning an insect but not a mammalian cell membrane, resulting in production of viral mutants that have reduced infectivity in mammalian hosts but efficient growth in insect cells. Groups of four monkeys received one dose each of test vaccine candidate with no booster immunization. After immunization, levels of viremia produced by each vaccine were determined by infectious center assay. Vaccine recipient immune response to wild-type DV2 challenge was measured on Day 57 by enzyme-linked immunosorbent assay and plaque reduction neutralization test. Two vaccines, DV2ΔGVII and DV2G460P, generated neutralizing antibody in the range of 700-900 50% plaque reduction neutralization test units. All three vaccine strains decreased the length of viremia by at least two days. No safety concerns were identified.


Assuntos
Vacinas contra Dengue , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Cultivadas , Chlorocebus aethiops , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/efeitos adversos , Vacinas contra Dengue/genética , Vacinas contra Dengue/imunologia , Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/genética , Imunização , Testes de Neutralização , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Ensaio de Placa Viral
20.
Arch Virol ; 153(12): 2215-23, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19002647

RESUMO

In an effort to develop a suitable DNA vaccine candidate for dengue, using dengue-3 virus (DENV-3) as a prototype, the genes coding for premembrane (prM) and envelope proteins (E) were inserted into an expression plasmid. After selecting recombinant clones containing prM/E genes, protein expression in the cell monolayer was detected by indirect immunofluorescence and immunoprecipitation assays. After selecting three vaccine candidates (pVAC1DEN3, pVAC2DEN3 and pVAC3DEN3), they were analyzed in vivo to determine their ability to induce a DENV-3-specific immune response. After three immunizations, the spleens of the immunized animals were isolated, and the cells were cultivated to measure cytokine levels by ELISA and used for lymphoproliferation assays. All of the animals inoculated with the recombinant clones induced neutralizing antibodies against DENV-3 and produced a T cell proliferation response after specific stimuli. Immunized and control mice were challenged with a lethal dose of DENV-3 and observed in order to assess their survival capability. The groups that presented the best survival rate after the challenge were the animals vaccinated with the pVAC3DEN3 clones, with an 80% survival rate. Thus, these data show that we have manufactured a vaccine candidate for DENV-3 that is able to induce a specific immune response and protects mice against a lethal challenge.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Linhagem Celular Tumoral , Proliferação de Células , Chlorocebus aethiops , Citocinas/imunologia , Citocinas/metabolismo , Dengue/imunologia , Dengue/mortalidade , Vacinas contra Dengue/genética , Vacinas contra Dengue/uso terapêutico , Vírus da Dengue/genética , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Vacinas de DNA/genética , Vacinas de DNA/uso terapêutico , Células Vero , Proteínas do Envelope Viral/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA