Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37674729

RESUMEN

Background: A significant overlap exists in the burden of Alcohol Use Disorders (AUDs) and the HIV epidemic in Sub-Saharan Africa. Over 60% of HIV infections occur in women, mostly through the cervical mucosa. Absorption and systemic circulation of alcohol induces global physiological and immune effects, including at the genital mucosa. Alcohol alters expression of cell surface receptors, mucosal barrier permeability, inflammatory responses, and lymphocyte trafficking and homing. However, a substantial knowledge gap exists on whether these cellular and or immunological effects of alcohol modify the consumers' CD4+ T cell susceptibility to HIV-1 entry at the cervical mucosa. HIV seronegative women, aged 18-49 years were recruited from Kasenyi and Kigungu fish landing sites of Lake Victoria. They were categorized as Alcohol Consumers (n=27) or non-Alcohol Consumers (n=26) based on the World Health Organization Alcohol-Use-Disorder-Test (WHO-AUDIT) at a cut-off score of >=8/40 and <8/40, respectively. Cytobrush-collected Cervical Mononuclear Cells [CMCs] and Peripheral Blood Mononuclear Cells [PBMCs] from heparinized whole-blood were surface stained for CD4+ T cell immunophenotyping. To measure susceptibility to HIV entry, CMCs and PBMCs were co-cultured overnight with equal amount of GFP-tagged HIV-1 pseudo-virus particles. Both immunophenotyping and HIV entry were measured on a BD LSR II flow cytometer. Results: There was no significant difference in the frequency of CD4+ T cells in blood (p=0.451) or mucosa (p=0.838) compartments across study groups. However, we observed a combined four-fold higher HIV entry (p=0.0001) into cervical versus blood-derived CD4+ T cells regardless of alcohol consumption status. More so, cervical-derived CD4+ T cells of alcohol-consumers showed a two-fold increase in susceptibility to HIV entry (P=0.0185) compared to the non-alcohol consumer group. Double positive α4ß7+CD4+T cells of alcohol consumers exhibited a higher HIV entry compared to those from alcohol non-consumers(p=0.0069). Conclusion: This study demonstrates that cervical CD4+ T cells are more susceptible to HIV entry than those from blood. Also, cervical CD4+ T cells of alcohol consumers are more susceptible than those of non-consumers. Differences in frequencies of α4ß7+ CD4+ T between alcohol consumers and non-consumers' cells may account for the increased susceptibility to HIV entry.

2.
Immunother Adv ; 3(1): ltad010, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37538934

RESUMEN

Control of schistosomiasis depends on a single drug, praziquantel, with variable cure rates, high reinfection rates, and risk of drug resistance. A vaccine could transform schistosomiasis control. Preclinical data show that vaccine development is possible, but conventional vaccine efficacy trials require high incidence, long-term follow-up, and large sample size. Controlled human infection studies (CHI) can provide early efficacy data, allowing the selection of optimal candidates for further trials. A Schistosoma CHI has been established in the Netherlands but responses to infection and vaccines differ in target populations in endemic countries. We aim to develop a CHI for Schistosoma mansoni in Uganda to test candidate vaccines in an endemic setting. This is an open-label, dose-escalation trial in two populations: minimal, or intense, prior Schistosoma exposure. In each population, participants will be enrolled in sequential dose-escalating groups. Initially, three volunteers will be exposed to 10 cercariae. If all show infection, seven more will be exposed to the same dose. If not, three volunteers in subsequent groups will be exposed to higher doses (20 or 30 cercariae) following the same algorithm, until all 10 volunteers receiving a particular dose become infected, at which point the study will be stopped for that population. Volunteers will be followed weekly after infection until CAA positivity or to 12 weeks. Once positive, they will be treated with praziquantel and followed for one year. The trial registry number is ISRCTN14033813 and all approvals have been obtained. The trial will be subjected to monitoring, inspection, and/or audits.

4.
Glob Bioeth ; 33(1): 78-102, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35814190

RESUMEN

Issues related to controlled human infection studies using Schistosoma mansoni (CHI-S) were explored to ensure the ethical and voluntary participation of potential CHI-S volunteers in an endemic setting in Uganda. We invited volunteers from a fishing community and a tertiary education community to guide the development of informed consent procedures. Consultative group discussions were held to modify educational materials on schistosomiasis, vaccines and the CHI-S model and similar discussions were held with a test group. With both groups, a mock consent process was conducted. Fourteen in-depth key informant interviews and three group discussions were held to explore perceptions towards participating in a CHI-S. Most of the participants had not heard of the CHI-S. Willingness to take part depended on understanding the study procedures and the consenting process. Close social networks were key in deciding to take part. The worry of adverse effects was cited as a possible hindrance to taking part. Volunteer time compensation was unclear for a CHI-S. Potential volunteers in these communities are willing to take part in a CHI-S. Community engagement is needed to build trust and time must be taken to share study procedures and ensure understanding of key messages.

6.
Arch Public Health ; 79(1): 120, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34225793

RESUMEN

BACKGROUND: Hookworm is a major contributor to worldwide disease burden with over 230 million people infected. It has been identified as one of the Neglected Tropical Diseases that can be controlled and even eliminated through mass drug administration and other effective interventions. Mathematical models have shown that hookworm can only be eliminated via a vaccine. Controlled Hookworm Human Infection (CHHI) models can facilitate rapid development of vaccines and drugs. METHODS: As a first step towards the establishment of CHHI in Africa, we held a stakeholders meeting in Lamberene, Gabon from 10 to 11 November 2019. RESULTS: Discussions revolved around the roles of the different regulatory institutions concerned; the need to strengthen existing regulatory capacity and the role of legislation; creating Gabon-specific ethical guidelines to govern Controlled Human Infection (CHI) studies; development of a study protocol; consideration of cultural and social peculiarities; the need for regular joint review meetings between interested parties throughout the process of protocol implementation; and participant compensation. Moreover, operational considerations concerning the introduction of CHHI in Gabon include the use of the local strain of hookworm for the challenge infections, capacity building for the local production of challenge material, and the establishment of adequate quality assurance procedures. CONCLUSION: The workshop addressed several of the anticipated hurdles to the successful implementation of CHHI in Gabon. It is our aim that this report will stimulate interest in the implementation of this model in the sub-Saharan African setting.

7.
Front Immunol ; 12: 635985, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746974

RESUMEN

Despite mass drug administration programmes with praziquantel, the prevalence of schistosomiasis remains high. A vaccine is urgently needed to control transmission of this debilitating disease. As some promising schistosomiasis vaccine candidates are moving through pre-clinical and clinical testing, we review the immunological challenges that these vaccine candidates may encounter in transitioning through the clinical trial phases in endemic settings. Prior exposure of the target population to schistosomes and other infections may impact vaccine response and efficacy and therefore requires considerable attention. Schistosomes are known for their potential to induce T-reg/IL-10 mediated immune suppression in populations which are chronically infected. Moreover, endemicity of schistosomiasis is focal whereby target and trial populations may exhibit several degrees of prior exposure as well as in utero exposure which may increase heterogeneity of vaccine responses. The age dependent distribution of exposure and development of acquired immunity, and general differences in the baseline immunological profile, adds to the complexity of selecting suitable trial populations. Similarly, prior or concurrent infections with other parasitic helminths, viral and bacterial infections, may alter immunological responses. Consequently, treatment of co-infections may benefit the immunogenicity of vaccines and may be considered despite logistical challenges. On the other hand, viral infections leave a life-long immunological imprint on the human host. Screening for serostatus may be needed to facilitate interpretation of vaccine responses. Co-delivery of schistosome vaccines with PZQ is attractive from a perspective of implementation but may complicate the immunogenicity of schistosomiasis vaccines. Several studies have reported PZQ treatment to induce both transient and long-term immuno-modulatory effects as a result of tegument destruction, worm killing and subsequent exposure of worm antigens to the host immune system. These in turn may augment or antagonize vaccine immunogenicity. Understanding the complex immunological interactions between vaccine, co-infections or prior exposure is essential in early stages of clinical development to facilitate phase 3 clinical trial design and implementation policies. Besides well-designed studies in different target populations using schistosome candidate vaccines or other vaccines as models, controlled human infections could also help identify markers of immune protection in populations with different disease and immunological backgrounds.


Asunto(s)
Desarrollo de Medicamentos , Enfermedades Endémicas/prevención & control , Vacunas Antiprotozoos/uso terapéutico , Schistosoma/inmunología , Esquistosomiasis/prevención & control , Animales , Coinfección , Diseño de Fármacos , Interacciones Huésped-Parásitos , Humanos , Inmunogenicidad Vacunal , Praziquantel/uso terapéutico , Vacunas Antiprotozoos/efectos adversos , Vacunas Antiprotozoos/inmunología , Schistosoma/patogenicidad , Esquistosomiasis/epidemiología , Esquistosomiasis/inmunología , Esquistosomiasis/transmisión , Esquistosomicidas/uso terapéutico
8.
AAS Open Res ; 2: 17, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31819922

RESUMEN

Schistosomiasis is a parasitic infection highly prevalent in sub-Saharan Africa, and a significant cause of morbidity; it is a priority for vaccine development. A controlled human infection model for Schistosoma mansoni (CHI-S) with potential to accelerate vaccine development has been developed among naïve volunteers in the Netherlands. Because responses both to infections and candidate vaccines are likely to differ between endemic and non-endemic settings, we propose to establish a CHI-S in Uganda where Schistosoma mansoni is endemic. As part of a "road-map" to this goal, we have undertaken a risk assessment. We identified risks related to importing of laboratory vector snails and schistosome strains from the Netherlands to Uganda; exposure to natural infection in endemic settings concurrently with CHI-S studies, and unfamiliarity of the community with the nature, risks and rationale for CHI. Mitigating strategies are proposed. With careful implementation of the latter, we believe that CHI-S can be implemented safely in Uganda. Our reflections are presented here to promote feedback and discussion.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...