Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Immunol ; 10(1): 14-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19088735

RESUMEN

A systems biology approach provides correlates of successful vaccination, which allows a new method for measuring early vaccine efficiency and suggests hypotheses for the mechanisms that underlie immunogenicity.


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica/métodos , Biología de Sistemas/métodos , Vacunación , Vacuna contra la Fiebre Amarilla/inmunología , Animales , Ensayos Clínicos como Asunto , Humanos , Ratones , Proteoma , Resultado del Tratamiento , Fiebre Amarilla/prevención & control , Vacuna contra la Fiebre Amarilla/uso terapéutico
2.
Nat Immunol ; 10(1): 116-125, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19029902

RESUMEN

A major challenge in vaccinology is to prospectively determine vaccine efficacy. Here we have used a systems biology approach to identify early gene 'signatures' that predicted immune responses in humans vaccinated with yellow fever vaccine YF-17D. Vaccination induced genes that regulate virus innate sensing and type I interferon production. Computational analyses identified a gene signature, including complement protein C1qB and eukaryotic translation initiation factor 2 alpha kinase 4-an orchestrator of the integrated stress response-that correlated with and predicted YF-17D CD8(+) T cell responses with up to 90% accuracy in an independent, blinded trial. A distinct signature, including B cell growth factor TNFRS17, predicted the neutralizing antibody response with up to 100% accuracy. These data highlight the utility of systems biology approaches in predicting vaccine efficacy.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Inmunidad Innata/genética , Biología de Sistemas/métodos , Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control , Virus de la Fiebre Amarilla/inmunología , Adolescente , Adulto , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Proteínas Portadoras/genética , Células Cultivadas , Ensayos Clínicos Controlados como Asunto , Humanos , Inmunidad Activa/genética , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Análisis Multivariante , Pruebas de Neutralización , Proteínas Serina-Treonina Quinasas/genética , Factor de Necrosis Tumoral alfa/genética , Vacunación , Vacuna contra la Fiebre Amarilla/uso terapéutico , Adulto Joven
3.
BMC Immunol ; 19(1): 15, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29801432

RESUMEN

BACKGROUND: Although a safe and effective yellow fever vaccine was developed more than 80 years ago, several issues regarding its use remain unclear. For example, what is the minimum dose that can provide immunity against the disease? A useful tool that can help researchers answer this and other related questions is a computational simulator that implements a mathematical model describing the human immune response to vaccination against yellow fever. METHODS: This work uses a system of ten ordinary differential equations to represent a few important populations in the response process generated by the body after vaccination. The main populations include viruses, APCs, CD8+ T cells, short-lived and long-lived plasma cells, B cells and antibodies. RESULTS: In order to qualitatively validate our model, four experiments were carried out, and their computational results were compared to experimental data obtained from the literature. The four experiments were: a) simulation of a scenario in which an individual was vaccinated against yellow fever for the first time; b) simulation of a booster dose ten years after the first dose; c) simulation of the immune response to the yellow fever vaccine in individuals with different levels of naïve CD8+ T cells; and d) simulation of the immune response to distinct doses of the yellow fever vaccine. CONCLUSIONS: This work shows that the simulator was able to qualitatively reproduce some of the experimental results reported in the literature, such as the amount of antibodies and viremia throughout time, as well as to reproduce other behaviors of the immune response reported in the literature, such as those that occur after a booster dose of the vaccine.


Asunto(s)
Algoritmos , Modelos Teóricos , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Virus de la Fiebre Amarilla/efectos de los fármacos , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Humanos , Vacunación/métodos , Viremia/inmunología , Viremia/prevención & control , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/fisiología
4.
BMC Infect Dis ; 18(1): 185, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29665797

RESUMEN

BACKGROUND: A yellow fever epidemic occurred in Angola in 2016 with 884 laboratory confirmed cases and 373 deaths. Eleven unvaccinated Chinese nationals working in Angola were also infected and imported the disease to China, thereby presenting the first importation of yellow fever into Asia. In Angola, there are about 259,000 Chinese foreign workers. The fact that 11 unvaccinated Chinese workers acquired yellow fever suggests that many more Chinese workers in Angola were not vaccinated. METHODS: We applied a previously developed model to back-calculate the number of unvaccinated Chinese workers in Angola in order to determine the extent of lack of vaccine coverage. RESULTS: Our models suggest that none of the 259,000 Chinese had been vaccinated, although yellow fever vaccination is mandated by the International Health Regulations. CONCLUSION: Governments around the world including China need to ensure that their citizens obtain YF vaccination when traveling to countries where such vaccines are required in order to prevent the international spread of yellow fever.


Asunto(s)
Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/epidemiología , Angola/epidemiología , Pueblo Asiatico , China/epidemiología , Brotes de Enfermedades/prevención & control , Epidemias , Humanos , Migrantes/estadística & datos numéricos , Viaje , Vacunación , Fiebre Amarilla/prevención & control
5.
Biochem Biophys Res Commun ; 492(4): 548-557, 2017 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-28109882

RESUMEN

Despite the availability of a safe and efficacious yellow fever vaccine since 1937, yellow fever remains a public health threat as a re-emerging disease in Africa and South America. We reviewed the trend of reported yellow fever outbreaks in eastern African countries, identified the risk epidemiological factors associated with the outbreaks and assessed the current situation of Yellow Fever vaccination in Africa. Surveillance and case finding for yellow fever in Africa are insufficient primarily due to lack of appropriate diagnostic capabilities, poor health infrastructure resulting in under-recognition, underreporting and underestimation of the disease. Despite these challenges, Ethiopia reported 302,614 cases (30,505 deaths) in 1943-2015, Kenya had 207 cases (38 deaths) in 1992-2016, Sudan experienced 31,750 suspected cases (1855 deaths) from 1940 to 2012 and Uganda had 452 cases (65 deaths) in 1941-2016. Major risk factors associated with past yellow fever outbreaks include climate, human practices and virus genetics. Comparisons between isolates from different outbreaks after 45 years have revealed the genetic stability of the structural proteins of YFV which are the primary targets of the host immune cells. This probably explains why yellow fever 17D vaccine is considered as outstandingly efficacious and safe after being used for 75 years. However, the 14 amino-acid changes among these isolates may have a greater impact on the changing disease epidemiology, virulence and transmission rate. Low population immunity against YF influences outbreak frequency especially in countries where the incorporation of YF vaccination is not combined with mass vaccination campaigns or vaccination is limited to international travellers. Understanding Yellow fever virus epidemiology as determined by its evolution underscores appropriate disease mitigation strategies and immunization policies. Mobilizing scarce resources to enhance population immunity through sufficient vaccination, promoting environmental sanitation/hygienic practices, driving behavioral change and community-based vector control are significant to preventing future epidemics.


Asunto(s)
Brotes de Enfermedades/economía , Brotes de Enfermedades/estadística & datos numéricos , Accesibilidad a los Servicios de Salud/estadística & datos numéricos , Vacunación Masiva/estadística & datos numéricos , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/mortalidad , Fiebre Amarilla/prevención & control , África Oriental/epidemiología , Países en Desarrollo/economía , Países en Desarrollo/estadística & datos numéricos , Brotes de Enfermedades/prevención & control , Enfermedades Endémicas/economía , Enfermedades Endémicas/prevención & control , Enfermedades Endémicas/estadística & datos numéricos , Costos de la Atención en Salud/estadística & datos numéricos , Accesibilidad a los Servicios de Salud/economía , Humanos , Vacunación Masiva/economía , Prevalencia , Factores de Riesgo , Tasa de Supervivencia , Revisión de Utilización de Recursos , Fiebre Amarilla/economía , Vacuna contra la Fiebre Amarilla/economía
6.
Euro Surveill ; 22(28)2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28749337

RESUMEN

States in south-eastern Brazil were recently affected by the largest Yellow Fever (YF) outbreak seen in a decade in Latin America. Here we provide a quantitative assessment of the risk of travel-related international spread of YF indicating that the United States, Argentina, Uruguay, Spain, Italy and Germany may have received at least one travel-related YF case capable of seeding local transmission. Mitigating the risk of imported YF cases seeding local transmission requires heightened surveillance globally.


Asunto(s)
Brotes de Enfermedades/prevención & control , Modelos Teóricos , Riesgo , Viaje , Fiebre Amarilla/prevención & control , Fiebre Amarilla/transmisión , Virus de la Fiebre Amarilla/aislamiento & purificación , Virus de la Fiebre Amarilla/patogenicidad , Animales , Argentina , Brasil/epidemiología , Alemania , Salud Global , Humanos , Insectos Vectores , Italia , Factores de Riesgo , España , Estados Unidos , Uruguay , Fiebre Amarilla/epidemiología , Vacuna contra la Fiebre Amarilla/uso terapéutico
7.
Rev Med Suisse ; 10(429): 1014-9, 2014 May 07.
Artículo en Francés | MEDLINE | ID: mdl-24908746

RESUMEN

Indication for yellow fever vaccination is not always easy to assess. The decision to immunize is not only based on the actual risk of the disease in a specific location, but also on public health considerations in the visited country (in order to respectively avoid epidemics in endemic countries or the introduction of the virus in zones where the vectors mosquitoes are present) and on travelers' risk factors for severe or even fatal vaccine adverse events. WHO has recently published new recommendations regarding vaccination against yellow fever after concluding that one dose of vaccine generates a life-long protection. This article tends to clarify the strategy to adopt in 2013 using cases frequently encountered in the practice of travel medicine.


Asunto(s)
Guías de Práctica Clínica como Asunto , Viaje , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Adulto , África , Femenino , Humanos , Masculino , Vacunación Masiva/normas , Persona de Mediana Edad , Organización Mundial de la Salud
8.
BMC Cancer ; 13: 4, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23282240

RESUMEN

BACKGROUND: Cancer is a significant and growing problem worldwide. While this increase may, in part, be attributed to increasing longevity, improved case notifications and risk-enhancing lifestyle (such as smoking, diet and obesity), hygiene-related factors resulting in immuno-regulatory failure may also play a major role and call for a revision of vaccination strategies to protect against a range of cancers in addition to infections. DISCUSSION: Human endogenous retroviruses (HERVs) are a significant component of a wider family of retroelements that constitutes part of the human genome. They were originated by the integration of exogenous retroviruses into the human genome millions of years ago. HERVs are estimated to comprise about 8% of human DNA and are ubiquitous in somatic and germinal tissues.Physiologic and pathologic processes are influenced by some biologically active HERV families. HERV antigens are only expressed at low levels by the host, but in circumstances of inappropriate control their genes may initiate or maintain pathological processes. Although the precise mechanism leading to abnormal HERVs gene expression has yet to be clearly elucidated, environmental factors seem to be involved by influencing the human immune system.HERV-K expression has been detected in different types of tumors.Among the various human endogenous retroviral families, the K series was the latest acquired by the human species. Probably because of its relatively recent origin, the HERV-K is the most complete and biologically active family.The abnormal expression of HERV-K seemingly triggers pathological processes leading to melanoma onset, but also contributes to the morphological and functional cellular modifications implicated in melanoma maintenance and progression.The HERV-K-MEL antigen is encoded by a pseudo-gene incorporated in the HERV-K env-gene. HERV-K-MEL is significantly expressed in the majority of dysplastic and normal naevi, as well as other tumors like sarcoma, lymphoma, bladder and breast cancer. An amino acid sequence similar to HERV-K-MEL, recognized to cause a significant protective effect against melanoma, is shared by the antigenic determinants expressed by some vaccines such as BCG, vaccinia virus and the yellow fever virus.HERV-K are also reactivated in the majority of human breast cancers. Monoclonal and single-chain antibodies against the HERV-K Env protein recently proved capable of blocking the proliferation of human breast cancer cells in vitro, inhibiting tumor growth in mice bearing xenograft tumors. SUMMARY: A recent epidemiological study provided provisional evidence of how melanoma risk could possibly be reduced if the yellow fever virus vaccine (YFV) were received at least 10 years before, possibly preventing tumor initiation rather than culling melanoma cells already compromised. Further research is recommended to confirm the temporal pattern of this protection and eliminate/attenuate the potential role of relevant confounders as socio-economic status and other vaccinations.It appears also appropriate to examine the potential protective effect of YFV against other malignancies expressing high levels of HERV-K antigens, namely breast cancer, sarcoma, lymphoma and bladder cancer.Tumor immune-therapy, as described for the monoclonal antibodies against breast cancer, is indeed considered more complex and less advantageous than immune-prevention. Cellular immunity possibly triggered by vaccines as for YFV might also be involved in anti-cancer response, in addition to humoral immunity.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Neoplasias/prevención & control , Infecciones por Retroviridae/prevención & control , Vacunas Virales/uso terapéutico , Animales , Antígenos Virales/inmunología , Neoplasias de la Mama/prevención & control , Neoplasias de la Mama/virología , Retrovirus Endógenos/clasificación , Retrovirus Endógenos/genética , Retrovirus Endógenos/inmunología , Femenino , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Masculino , Melanoma/prevención & control , Melanoma/virología , Neoplasias/epidemiología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/virología , Neoplasias Ováricas/prevención & control , Neoplasias Ováricas/virología , Infecciones por Retroviridae/epidemiología , Infecciones por Retroviridae/virología , Factores de Riesgo , Replicación Viral , Fiebre Amarilla/prevención & control , Fiebre Amarilla/virología , Vacuna contra la Fiebre Amarilla/uso terapéutico
9.
Antiviral Res ; 203: 105331, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35533777

RESUMEN

Yellow fever virus (YFV) continues to cause periodic outbreaks of severe disease throughout tropical regions of South America and Africa despite the availability of an effective vaccine. Despite efforts to control this virus for the last century, no antivirals have been approved for the treatment of YFV. The purpose of this study was to evaluate the broadly active antiviral compound remdesivir (RDV) in a hamster model of disease. Yellow fever (YF) disease in hamsters was prevented when treatment with RDV was initiated just prior to virus challenge, which was confirmed in a second study. Disease parameters including viremia, serum ALT and weight loss were significantly improved with RDV treatment in a dose-dependent manner. RDV was also effective when treatment was initiated as late as 4 days post-virus infection (dpi). These results demonstrate therapeutic efficacy of RDV in the treatment of YF in a relevant animal model of disease.


Asunto(s)
Vacuna contra la Fiebre Amarilla , Fiebre Amarilla , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Animales , Antivirales/uso terapéutico , Cricetinae , Viremia/tratamiento farmacológico , Fiebre Amarilla/prevención & control , Vacuna contra la Fiebre Amarilla/uso terapéutico , Virus de la Fiebre Amarilla
10.
Pediatr Clin North Am ; 69(1): 171-184, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34794673

RESUMEN

The pretravel management of the international pediatric traveler is based on provision of preventive education, chemoprophylaxis against malaria and traveler's diarrhea, as well as travel vaccinations. Immunization requirements are determined based on the traveler's pretravel immunization status, age, medical history, and destination. Immunization needs also vary depending on the exposures during the trip. Potential exposure to water, insects, or animals as well as duration of travel will help tailor risk avoidance education and travel immunizations. This review provides clinicians an overview of vaccines recommended for children traveling internationally.


Asunto(s)
Inmunización/métodos , Viaje , Enfermedades Prevenibles por Vacunación/prevención & control , Vacunas/uso terapéutico , Adolescente , Antimaláricos/uso terapéutico , Niño , Preescolar , Diarrea/prevención & control , Humanos , Esquemas de Inmunización , Lactante , Malaria/prevención & control , Infecciones Meningocócicas/prevención & control , Vacunas Meningococicas/uso terapéutico , Factores de Riesgo , Vacunación/métodos , Fiebre Amarilla/prevención & control , Vacuna contra la Fiebre Amarilla/uso terapéutico
11.
Elife ; 102021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33722340

RESUMEN

Yellow fever (YF) is a viral, vector-borne, haemorrhagic fever endemic in tropical regions of Africa and South America. The vaccine for YF is considered safe and effective, but intervention strategies need to be optimised; one of the tools for this is mathematical modelling. We refine and expand an existing modelling framework for Africa to account for transmission in South America. We fit to YF occurrence and serology data. We then estimate the subnational forces of infection for the entire endemic region. Finally, using demographic and vaccination data, we examine the impact of vaccination activities. We estimate that there were 109,000 (95% credible interval [CrI] [67,000-173,000]) severe infections and 51,000 (95% CrI [31,000-82,000]) deaths due to YF in Africa and South America in 2018. We find that mass vaccination activities in Africa reduced deaths by 47% (95% CrI [10%-77%]). This methodology allows us to evaluate the effectiveness of vaccination and illustrates the need for continued vigilance and surveillance of YF.


Asunto(s)
Carga Global de Enfermedades , Fiebre Amarilla/epidemiología , África/epidemiología , Brotes de Enfermedades , Salud Global , Humanos , Vacunación Masiva/estadística & datos numéricos , Modelos Teóricos , Estudios Seroepidemiológicos , América del Sur/epidemiología , Encuestas y Cuestionarios , Vacunación/métodos , Fiebre Amarilla/prevención & control , Fiebre Amarilla/transmisión , Vacuna contra la Fiebre Amarilla/uso terapéutico
12.
Elife ; 102021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34165077

RESUMEN

Background: Childhood immunisation services have been disrupted by the COVID-19 pandemic. WHO recommends considering outbreak risk using epidemiological criteria when deciding whether to conduct preventive vaccination campaigns during the pandemic. Methods: We used two to three models per infection to estimate the health impact of 50% reduced routine vaccination coverage in 2020 and delay of campaign vaccination from 2020 to 2021 for measles vaccination in Bangladesh, Chad, Ethiopia, Kenya, Nigeria, and South Sudan, for meningococcal A vaccination in Burkina Faso, Chad, Niger, and Nigeria, and for yellow fever vaccination in the Democratic Republic of Congo, Ghana, and Nigeria. Our counterfactual comparative scenario was sustaining immunisation services at coverage projections made prior to COVID-19 (i.e. without any disruption). Results: Reduced routine vaccination coverage in 2020 without catch-up vaccination may lead to an increase in measles and yellow fever disease burden in the modelled countries. Delaying planned campaigns in Ethiopia and Nigeria by a year may significantly increase the risk of measles outbreaks (both countries did complete their supplementary immunisation activities (SIAs) planned for 2020). For yellow fever vaccination, delay in campaigns leads to a potential disease burden rise of >1 death per 100,000 people per year until the campaigns are implemented. For meningococcal A vaccination, short-term disruptions in 2020 are unlikely to have a significant impact due to the persistence of direct and indirect benefits from past introductory campaigns of the 1- to 29-year-old population, bolstered by inclusion of the vaccine into the routine immunisation schedule accompanied by further catch-up campaigns. Conclusions: The impact of COVID-19-related disruption to vaccination programs varies between infections and countries. Planning and implementation of campaigns should consider country and infection-specific epidemiological factors and local immunity gaps worsened by the COVID-19 pandemic when prioritising vaccines and strategies for catch-up vaccination. Funding: Bill and Melinda Gates Foundation and Gavi, the Vaccine Alliance.


Asunto(s)
COVID-19/epidemiología , Programas de Inmunización/estadística & datos numéricos , Sarampión/prevención & control , Infecciones Meningocócicas/prevención & control , Fiebre Amarilla/prevención & control , Adolescente , Adulto , África/epidemiología , Bangladesh/epidemiología , Niño , Preescolar , Brotes de Enfermedades , Humanos , Programas de Inmunización/métodos , Lactante , Sarampión/epidemiología , Vacuna Antisarampión/uso terapéutico , Infecciones Meningocócicas/epidemiología , Vacunas Meningococicas/uso terapéutico , Pandemias , Medición de Riesgo , SARS-CoV-2 , Vacunación/estadística & datos numéricos , Fiebre Amarilla/epidemiología , Vacuna contra la Fiebre Amarilla/uso terapéutico , Adulto Joven
13.
Voen Med Zh ; 331(6): 32-6, 2010 Jun.
Artículo en Ruso | MEDLINE | ID: mdl-20731094

RESUMEN

At the beginning of 21 century endemic areas of yellow fever are in tropical forests of Africa and South America, covering 43 countries of the around the world. In a period of 11 years 2005 citizens from 14 countries of Africa and 871 citizens from 8 countries of South America were taken ill with of a yellow fever. On the both continentals were observed distinct, limited (no more than 40 people) ictuses of urban of a yellow fever related to mosquito Aedes aegypti. Still, the total mortality is high--up to 35%. The systems of epidemiological surveillance and informational connection are created; the live vaccine in sufficient amount for general is delivered to the hot spots of disease. Civil wars and ethnic conflicts hinder the introduction of compulsory vaccination in national calendars of the inoculation of children in endemic countries. Because of it the vaccination is realized slowly: nowadays the vaccination is introduce in 19 among 25 countries of Africa with high risk of disease.


Asunto(s)
Enfermedades Endémicas , Personal Militar , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla , Enfermedades Endémicas/prevención & control , Humanos , América Latina/epidemiología , Federación de Rusia/epidemiología , Sudáfrica/epidemiología , Fiebre Amarilla/epidemiología , Fiebre Amarilla/prevención & control , Vacuna contra la Fiebre Amarilla/administración & dosificación
14.
EMBO Mol Med ; 12(1): e10375, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31746149

RESUMEN

Live 17D is widely used as a prophylactic vaccine strain for yellow fever virus that induces potent neutralizing humoral and cellular immunity against the wild-type pathogen. 17D replicates and kills mouse and human tumor cell lines but not non-transformed human cells. Intratumoral injections with viable 17D markedly delay transplanted tumor progression in a CD8 T-cell-dependent manner. In mice bearing bilateral tumors in which only one is intratumorally injected, contralateral therapeutic effects are observed consistent with more prominent CD8 T-cell infiltrates and a treatment-related reduction of Tregs. Additive efficacy effects were observed upon co-treatment with intratumoral 17D and systemic anti-CD137 and anti-PD-1 immunostimulatory monoclonal antibodies. Importantly, when mice were preimmunized with 17D, intratumoral 17D treatment achieved better local and distant antitumor immunity. Such beneficial effects of prevaccination are in part explained by the potentiation of CD4 and CD8 T-cell infiltration in the treated tumor. The repurposed use of a GMP-grade vaccine to be given via the intratumoral route in prevaccinated patients constitutes a clinically feasible and safe immunotherapy approach.


Asunto(s)
Inmunoterapia , Neoplasias/terapia , Vacuna contra la Fiebre Amarilla , Animales , Linfocitos T CD8-positivos/inmunología , Reposicionamiento de Medicamentos , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Vacuna contra la Fiebre Amarilla/uso terapéutico
15.
Emerg Microbes Infect ; 9(1): 520-533, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32116148

RESUMEN

The recent Zika virus (ZIKV) epidemic in the Americas, followed by the yellow fever virus (YFV) outbreaks in Angola and Brazil highlight the urgent need for safe and efficient vaccines against the ZIKV as well as much greater production capacity for the YFV-17D vaccine. Given that the ZIKV and the YFV are largely prevalent in the same geographical areas, vaccines that would provide dual protection against both pathogens may obviously offer a significant benefit. We have recently engineered a chimeric vaccine candidate (YF-ZIKprM/E) by swapping the sequences encoding the YFV-17D surface glycoproteins prM/E by the corresponding sequences of the ZIKV. A single vaccine dose of YF-ZIKprM/E conferred complete protection against a lethal challenge with wild-type ZIKV strains. Surprisingly, this vaccine candidate also efficiently protected against lethal YFV challenge in various mouse models. We demonstrate that CD8+ but not CD4+ T cells, nor ZIKV neutralizing antibodies are required to confer protection against YFV. The chimeric YF-ZIKprM/E vaccine may thus be considered as a dual vaccine candidate efficiently protecting mice against both the ZIKV and the YFV, and this following a single dose immunization. Our finding may be particularly important in the rational design of vaccination strategies against flaviviruses, in particular in areas where YFV and ZIKV co-circulate.


Asunto(s)
Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control , Virus de la Fiebre Amarilla/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Chlorocebus aethiops , Ratones , Linfocitos T/inmunología , Células Vero , Fiebre Amarilla/inmunología , Vacuna contra la Fiebre Amarilla/uso terapéutico
16.
Front Immunol ; 11: 1382, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32765496

RESUMEN

Yellow Fever (YF) vaccination is suggested to induce a large number of adverse events (AE) and suboptimal responses in patients with autoimmune diseases (AID); however, there have been no studies on 17DD-YF primary vaccination performance in patients with AID. This prospective non-interventional study conducted between March and July, 2017 assessed the safety and immunogenicity of planned 17DD-YF primary vaccination in patients with AID. Adult patients with AID (both sexes) were enrolled, along with healthy controls, at a single hospital (Vitória, Brazil). Included patients were referred for planned vaccination by a rheumatologist; in remission, or with low disease activity; and had low level immunosuppression or the attending physician advised interruption of immunosuppression for safety reasons. The occurrence of AE, neutralizing antibody kinetics, seropositivity rates, and 17DD-YF viremia were evaluated at various time points (day 0 (D0), D3, D4, D5, D6, D14, and D28). Individuals evaluated (n = 278), including patients with rheumatoid arthritis (RA; 79), spondyloarthritis (SpA; 59), systemic sclerosis (8), systemic lupus erythematosus (SLE; 27), primary Sjögren's syndrome (SS; 54), and healthy controls (HC; 51). Only mild AE were reported. The frequency of local and systemic AE in patients with AID and HC did not differ significantly (8 vs. 10% and 21 vs. 32%; p = 1.00 and 0.18, respectively). Patients with AID presented late seroconversion profiles according to kinetic timelines of the plaque reduction neutralization test (PRNT). PRNT-determined virus titers (copies/mL) [181 (95% confidence interval (CI), 144-228) vs. 440 (95% CI, 291-665), p = 0.004] and seropositivity rate (78 vs. 96%, p = 0.01) were lower in patients with AID after 28 days, particularly those with SpA (73%) and SLE (73%), relative to HC. The YF viremia peak (RNAnemia) was 5-6 days after vaccination in all groups. In conclusion, consistent seroconversion rates were observed in patients with AID and our findings support that planned 17DD-YF primary vaccination is safe and immunogenic in patients with AID.


Asunto(s)
Enfermedades Autoinmunes/complicaciones , Vacuna contra la Fiebre Amarilla/inmunología , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Brasil , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Adulto Joven
17.
Transplant Proc ; 52(5): 1291-1293, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32505502

RESUMEN

Yellow fever (YF) is a vaccine-preventable disease, but live attenuated YF vaccine (YFV) is contraindicated in immunosuppressed patients due to the risk of life-threatening YFV-associated side effects. This study aimed to evaluate 1. the knowledge of renal transplant recipients (RTRs) about the contraindication and risks of YFV; 2. the prevalence of inadvertent vaccination of RTRs against YF; and 3. the outcome of these patients. A cross-sectional telephone contact study was conducted with 200 RTRs selected from the outpatient clinic of our transplantation unit. There were 116 successful telephone contacts (58%). A total of 11 vaccinated patients were identified: 5 received YFV in the pretransplant period and 6 in the post-transplant period. All patients received the full dose of the vaccine. Among those vaccinated after transplant, only 1 reported a mild adverse event (nausea) after receiving the vaccine. All vaccinated patients who were post-transplant did not know about vaccine contraindications as a result of their clinical condition. Among the unvaccinated patients, this rate was 12.4%. YFV is the main tool for disease prevention and control as there is no specific antiviral treatment for YF. Our results confirm the evidence that transplant recipients tolerate YFV well. However, data are not strong enough to recommend this vaccine in transplant recipients. Counseling RTRs on the contraindications of YFV is important to prevent inadvertent use of this vaccine in this population.


Asunto(s)
Trasplante de Riñón/efectos adversos , Complicaciones Posoperatorias/prevención & control , Vacunación/psicología , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Adulto , Contraindicaciones de los Medicamentos , Estudios Transversales , Femenino , Conocimientos, Actitudes y Práctica en Salud , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/virología , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Vacuna contra la Fiebre Amarilla/inmunología
18.
Front Immunol ; 11: 577751, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133096

RESUMEN

Introduction: Although effective live attenuated yellow fever (YF) vaccines have been available for over 9 decades sporadic outbreaks continue to occur in endemic regions. These may be linked to several factors including epidemiological factors such as vector and intermediate host distribution or vaccine coverage and efficacy. The World Health Organization's research priorities include gathering systematic evidence around the potential need for booster vaccination with YF vaccine whether this follows full or fractional doses in children. Knowledge on the longevity of response to YF vaccine and the implications of this response needs to be consolidated to guide future vaccination policy. Methods: We measured anti-YF IgG by microneutralization assay in a group of 481 African infants who had received YF vaccine as part of routine EPI programmes, to explore serological protection from YF 5-6 years post YF vaccination, as well as the effect of co variates. Findings: Notably, 22.2% of the cohort had undetectable antibody concentrations, with another 7.5% revealing concentrations below the threshold of seropositivity of 0.5 IU/mL. Sex, season, country and time since vaccination did not affect the longevity of antibody concentration or having antibody concentrations above a defined threshold. Conclusion: Roughly 30% of children in this cohort did not demonstrate anti-yellow fever antibody concentrations above the defined threshold of protection, with 20% having no demonstrable antibody. Knowledge on the longevity of response to YF vaccine and the implications needs to be consolidated to guide future vaccination policy.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Programas de Inmunización , Inmunogenicidad Vacunal , Inmunoglobulina G/sangre , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Virus de la Fiebre Amarilla/inmunología , Biomarcadores/sangre , Niño , Preescolar , Femenino , Gambia , Interacciones Huésped-Patógeno , Humanos , Esquemas de Inmunización , Lactante , Masculino , Malí , Pruebas Serológicas , Factores de Tiempo , Resultado del Tratamiento , Fiebre Amarilla/sangre , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Virus de la Fiebre Amarilla/patogenicidad
19.
Front Immunol ; 11: 575074, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193365

RESUMEN

Combined cellular and humoral host immune response determine the clinical course of a viral infection and effectiveness of vaccination, but currently the cellular immune response cannot be measured on simple blood samples. As functional activity of immune cells is determined by coordinated activity of signaling pathways, we developed mRNA-based JAK-STAT signaling pathway activity assays to quantitatively measure the cellular immune response on Affymetrix expression microarray data of various types of blood samples from virally infected patients (influenza, RSV, dengue, yellow fever, rotavirus) or vaccinated individuals, and to determine vaccine immunogenicity. JAK-STAT1/2 pathway activity was increased in blood samples of patients with viral, but not bacterial, infection and was higher in influenza compared to RSV-infected patients, reflecting known differences in immunogenicity. High JAK-STAT3 pathway activity was associated with more severe RSV infection. In contrast to inactivated influenza virus vaccine, live yellow fever vaccine did induce JAK-STAT1/2 pathway activity in blood samples, indicating superior immunogenicity. Normal (healthy) JAK-STAT1/2 pathway activity was established, enabling assay interpretation without the need for a reference sample. The JAK-STAT pathway assays enable measurement of cellular immune response for prognosis, therapy stratification, vaccine development, and clinical testing.


Asunto(s)
Virus del Dengue/inmunología , Inmunidad Celular , Orthomyxoviridae/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Rotavirus/inmunología , Vacunas Virales/uso terapéutico , Virosis/inmunología , Virus de la Fiebre Amarilla/inmunología , Biomarcadores/sangre , Dengue/sangre , Dengue/inmunología , Dengue/prevención & control , Dengue/virología , Vacunas contra el Dengue/uso terapéutico , Virus del Dengue/patogenicidad , Diagnóstico Diferencial , Interacciones Huésped-Patógeno , Humanos , Inmunogenicidad Vacunal , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/sangre , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Orthomyxoviridae/patogenicidad , Valor Predictivo de las Pruebas , ARN Mensajero/sangre , ARN Mensajero/genética , Infecciones por Virus Sincitial Respiratorio/sangre , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/patogenicidad , Rotavirus/patogenicidad , Infecciones por Rotavirus/sangre , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/virología , Vacunas contra Rotavirus , Transducción de Señal/genética , Virosis/sangre , Virosis/prevención & control , Virosis/virología , Fiebre Amarilla/sangre , Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control , Fiebre Amarilla/virología , Vacuna contra la Fiebre Amarilla/uso terapéutico , Virus de la Fiebre Amarilla/patogenicidad
20.
Antiviral Res ; 162: 1-4, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30529359

RESUMEN

Yellow fever is a serious disease caused by infection with the yellow fever virus (YFV). A live-attenuated YFV vaccine strain, 17D (YFV-17D) is the only virus strain available for the production of the YFV vaccine. This study evaluated the immunogenicity and immune persistence of vaccination with YFV-17D and identified their influencing factors in Chinese peacekeepers deployed to Africa. Serum specimens were collected before and ≥21 days after primary vaccination with YFV-17D in 349 Chinese peacekeepers who were subsequently deployed to Africa for the first time from 2016 to 2017 (population 1). Serum specimens were collected from 1 to 11 years after vaccination with YFV-17D in 2062 returned Chinese peacekeepers who were deployed to Africa from 2005 to 2015 (population 2). We found that YFV-17D exhibited an excellent protective effect in the Chinese peacekeepers deployed to Africa early following vaccination. In the Chinese peacekeepers one year after vaccination, the serum antibody titer against YFV increased with increasing age at vaccination; in those two or more years after vaccination, the serum antibody titer against YFV decreased over years and was similar to but greater than the minimum protective level 11 years after vaccination. The number of peacekeeping missions exhibited an almost negligible influence on the serum antibody titer against YFV. (This study has been registered at International Clinical Trials Registry Platform (http://www.who.int/ictrp/en/) under registration Nos. ChiCTR1800017024.).


Asunto(s)
Anticuerpos Antivirales/sangre , Inmunogenicidad Vacunal , Vacuna contra la Fiebre Amarilla/uso terapéutico , Fiebre Amarilla/prevención & control , Adolescente , Adulto , África , Factores de Edad , Pueblo Asiatico , China , Femenino , Humanos , Masculino , Persona de Mediana Edad , Personal Militar , Factores de Tiempo , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/uso terapéutico , Fiebre Amarilla/sangre , Fiebre Amarilla/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA