Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Parasitology ; 147(9): 922-931, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32338232

RESUMO

Human African trypanosomiasis (HAT) is caused by Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense and caused devastating epidemics during the 20th century. Due to effective control programs implemented in the last two decades, the number of reported cases has fallen to a historically low level. Although fewer than 977 cases were reported in 2018 in endemic countries, HAT is still a public health problem in endemic regions until it is completely eliminated. In addition, almost 150 confirmed HAT cases were reported in non-endemic countries in the last three decades. The majority of non-endemic HAT cases were reported in Europe, USA and South Africa, due to historical alliances, economic links or geographic proximity to disease-endemic countries. Furthermore, with the implementation of the 'Belt and Road' project, sporadic imported HAT cases have been reported in China as a warning sign of tropical diseases prevention. In this paper, we explore and interpret the data on HAT incidence and find no positive correlation between the number of HAT cases from endemic and non-endemic countries. This data will provide useful information for better understanding the imported cases of HAT globally in the post-elimination phase.


Assuntos
Doenças Transmissíveis Importadas/epidemiologia , Doenças Endêmicas/estatística & dados numéricos , Tripanossomíase Africana/epidemiologia , Doenças Transmissíveis Importadas/parasitologia , Humanos , Incidência , Tripanossomíase Africana/parasitologia
2.
Fundam Clin Pharmacol ; 38(1): 72-83, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37479675

RESUMO

Human African trypanosomosis (HAT) which is also known as sleeping sickness is caused by Trypanosoma brucei gambiense that is endemic in western and central Africa and T. b. rhodesiense that is endemic in eastern and southern Africa. Drugs used for treatment against HAT first stage have limited effectiveness, and the second stage drugs have been reported to be toxic, expensive, and have time-consuming administration, and parasitic resistance has developed against these drugs. The aim of this study was to evaluate the anti-trypanosomal activity of nitrofurantoin-triazole hybrids against T. b. gambiense and T. b. rhodesiense parasites in vitro. This study screened 19 synthesized nitrofurantoin-triazole (NFT) hybrids on two strains of human trypanosomes, and cytotoxicity was evaluated on Madin-Darby bovine kidney (MDBK) cells. The findings in this study showed that an increase in the chain length and the number of carbon atoms in some n-alkyl hybrids influenced the increase in anti-trypanosomal activity against T. b. gambiense and T. b. rhodesiense. The short-chain n-alkyl hybrids showed decreased activity compared to the long-chain n-alkyl hybrids, with increased activity against both T. b. gambiense and T. b. rhodesiense. Incorporation of additional electron-donating substituents in some NFT hybrids showed increased anti-trypanosomal activity than to electron-withdrawing substituents in NFT hybrids. All 19 NFT hybrids tested displayed better anti-trypanosomal activity against T. b. gambiense than T. b. rhodesiense. The NFT hybrid no. 16 was among the best performing hybrids against both T. b. gambiense (0.08 ± 0.04 µM) and T. b.rhodesiense (0.11 ± 0.06 µM), and its activity might be influenced by the introduction of fluorine in the para-position on the benzyl ring. Remarkably, the NFT hybrids in this study displayed weak to moderate cytotoxicity on MDBK cells. All of the NFT hybrids in this study had selectivity index values ranging from 18 to greater than 915, meaning that they were up to 10-100 times fold selective in their anti-trypanosomal activity. The synthesized NFT hybrids showed strong selectivity >10 to T. b. gambiense and T. b. rhodesiense, which indicates that they qualify from the initial selection criteria for potential hit drugs.


Assuntos
Nitrofurantoína , Tripanossomíase Africana , Humanos , Animais , Bovinos , Nitrofurantoína/uso terapêutico , Trypanosoma brucei rhodesiense , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/epidemiologia , Tripanossomíase Africana/parasitologia , Trypanosoma brucei gambiense
3.
Trop Med Infect Dis ; 7(10)2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36288011

RESUMO

Human African trypanosomiasis (HAT) is a neglected tropical disease that has not received much attention in Zambia and most of the countries in which it occurs. In this study, we assessed the adequacy of the healthcare delivery system in diagnosis and management of rHAT cases, the environmental factors associated with transmission, the population at risk and the geographical location of rHAT cases. Structured questionnaires, focus group discussions and key informant interviews were conducted among the affected communities and health workers. The study identified 64 cases of rHAT, of which 26 were identified through active surveillance and 38 through passive surveillance. We identified a significant association between knowledge of the vector for rHAT and knowledge of rHAT transmission (p < 0.028). In all four districts, late or poor diagnosis occurred due to a lack of qualified laboratory technicians and diagnostic equipment. This study reveals that the current Zambian healthcare system is not able to adequately handle rHAT cases. Targeted policies to improve staff training in rHAT disease detection and management are needed to ensure that sustainable elimination of this public health problem is achieved in line with global targets.

4.
ChemistryOpen ; 10(9): 922-927, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34553828

RESUMO

This study identified the isoindolone ring as a scaffold for novel agents against Trypanosoma brucei rhodesiense and explored the structure-activity relationships of various aromatic ring substitutions. The compounds were evaluated in an integrated in vitro screen. Eight compounds exhibited selective activity against T. b. rhodesiense (IC50 <2.2 µm) with no detectable side activity against T. cruzi and Leishmania infantum. Compound 20 showed low nanomolar potency against T. b. rhodesiense (IC50 =40 nm) and no toxicity against MRC-5 and PMM cell lines and may be regarded as a new lead template for agents against T. b. rhodesiense. The isoindolone-based compounds have the potential to progress into lead optimization in view of their highly selective in vitro potency, absence of cytotoxicity and acceptable metabolic stability. However, the solubility of the compounds represents a limiting factor that should be addressed to improve the physicochemical properties that are required to proceed further in the development of in vivo-active derivatives.


Assuntos
Isoindóis/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma brucei rhodesiense/efeitos dos fármacos , Animais , Linhagem Celular , Estabilidade de Medicamentos , Feminino , Humanos , Isoindóis/síntese química , Isoindóis/metabolismo , Isoindóis/toxicidade , Camundongos , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Testes de Sensibilidade Parasitária , Solubilidade , Relação Estrutura-Atividade , Tripanossomicidas/síntese química , Tripanossomicidas/metabolismo , Tripanossomicidas/toxicidade
5.
BMC Med Genomics ; 13(1): 14, 2020 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-32000760

RESUMO

BACKGROUND: Rhodesiense sleeping sickness is caused by infection with T. b rhodesiense parasites resulting in an acute disease that is fatal if not treated in time. The aim of this study was to understand the global impact of active T. b rhodesiense infection on the patient's immune response in the early and late stages of the disease. METHODS: RNASeq was carried out on blood and cerebral spinal fluid (CSF) samples obtained from T. b. rhodesiense infected patients. The control samples used were from healthy individuals in the same foci. The Illumina sequenced reads were analysed using the Tuxedo suite pipeline (Tophat, Cufflinks, Cuffmerge, Cuffdiff) and differential expression analysis carried out using the R package DESeq2. The gene enrichment and function annotation analysis were done using the ToppCluster, DAVID and InnateDB algorithms. RESULTS: We previously described the transcriptomes of T. b rhodesiense from infected early stage blood (n = 3) and late stage CSF (n = 3) samples from Eastern Uganda. We here identify human transcripts that were differentially expressed (padj < 0.05) in the early stage blood versus healthy controls (n = 3) and early stage blood versus late stage CSF. Differential expression in infected blood showed an enrichment of innate immune response genes whereas that of the CSF showed enrichment for anti-inflammatory and neuro-degeneration signalling pathways. We also identified genes (C1QC, MARCO, IGHD3-10) that were up-regulated (log2 FC > 2.5) in both the blood and CSF. CONCLUSION: The data yields insights into the host's response to T. b rhodesiense parasites in the blood and central nervous system. We identified key pathways and signalling molecules for the predominant innate immune response in the early stage infection; and anti-inflammatory and neuro-degeneration pathways associated with sleep disorders in second stage infection. We further identified potential blood biomarkers that can be used for diagnosis of late stage disease without the need for lumbar puncture.


Assuntos
RNA-Seq , Transcriptoma , Trypanosoma brucei gambiense , Tripanossomíase Africana , Regulação para Cima , Adolescente , Adulto , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Criança , Feminino , Humanos , Masculino , Tripanossomíase Africana/sangue , Tripanossomíase Africana/líquido cefalorraquidiano
6.
Trop Med Infect Dis ; 5(1)2020 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-32012658

RESUMO

Human African Trypanosomiasis (HAT or sleeping sickness) is a life-threatening neglected tropical disease that is endemic in 36 sub-Saharan African countries. Until recently, treatment options were limited and hampered by unsatisfactory efficacy, toxicity, and long and cumbersome administration regimens, compounded by infrastructure inadequacies in the remote rural regions worst affected by the disease. Increased funding and awareness of HAT over the past two decades has led to a steady decline in reported cases (<1000 in 2018). Recent drug development strategies have resulted in development of the first all-oral treatment for HAT, fexinidazole. Fexinidazole received European Medicines Agency positive scientific opinion in 2018 and is now incorporated into the WHO interim guidelines as one of the first-line treatments for HAT, allowing lumbar puncture to become non-systematic. Here, we highlight the role of global collaborations in the effort to control HAT and develop new treatments. The long-standing collaboration between the WHO, Sanofi and the Drugs for Neglected Diseases initiative (Geneva, Switzerland) was instrumental for achieving the control and treatment development goals in HAT, whilst at the same time ensuring that efforts were led by national authorities and control programs to leave a legacy of highly trained healthcare workers and improved research and health infrastructure.

7.
Pathogens ; 8(3)2019 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-31470522

RESUMO

The recent massive reduction in the numbers of fresh Human African Trypanosomiasis (HAT) infection has presented an opportunity for the global elimination of this disease. To prevent a possible resurgence, as was the case after the reduced transmission of the 1960s, surveillance needs to be sustained and the necessary tools for detection and treatment of cases need to be made available at the points of care. In this review, we examine the available resources and make recommendations for improvement to ensure the sustenance of the already achieved gains to keep the trend moving towards elimination.

8.
Trends Parasitol ; 34(10): 818-827, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30181071

RESUMO

Human African trypanosomiasis (HAT) is responsible for around 3000 reported cases each year. Treatments for HAT are expensive and problematic to administer, and available drugs are old and less than ideal, some with high levels of toxicity that result in debilitating and, in some cases, fatal side effects. Treatment options are limited, with only one drug, eflornithine, introduced in the last 28 years. Here we examine the limitations of current chemotherapeutic approaches to manage HAT, the constraints to new drug development exploring drug failures and new drugs on the horizon, and consider the epidemiological, political, social, and economic factors influencing drug development.


Assuntos
Desenvolvimento de Medicamentos/tendências , Tripanossomicidas , Tripanossomíase Africana/tratamento farmacológico , Desenvolvimento de Medicamentos/economia , Desenvolvimento de Medicamentos/normas , Humanos , Tempo , Tripanossomicidas/normas
9.
Eur J Med Chem ; 150: 385-402, 2018 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-29544150

RESUMO

African trypanosomiasis is a neglected parasitic disease that is still of great public health relevance, and a severe impediment to agriculture in endemic areas. The pathogens possess certain unique metabolic features that can be exploited for the development of new drugs. Notably, they rely on an essential, mitochondrially-localized enzyme, Trypanosome Alternative Oxidase (TAO) for their energy metabolism, which is absent in the mammalian hosts and therefore an attractive target for the design of safe drugs. In this study, we cloned, expressed and purified the physiologically relevant form of TAO, which lacks the N-terminal 25 amino acid mitochondrial targeting sequence (ΔMTS-TAO). A new class of 32 cationic and non-cationic 4-hydroxybenzoate and 4-alkoxybenzaldehyde inhibitors was designed and synthesized, enabling the first structure-activity relationship studies on ΔMTS-TAO. Remarkably, we obtained compounds with enzyme inhibition values (IC50) as low as 2 nM, which were efficacious against wild type and multidrug-resistant strains of T. brucei and T. congolense. The inhibitors 13, 15, 16, 19, and 30, designed with a mitochondrion-targeting lipophilic cation tail, displayed trypanocidal potencies comparable to the reference drugs pentamidine and diminazene, and showed no cross-resistance with the critical diamidine and melaminophenyl arsenical classes of trypanocides. The cationic inhibitors 15, 16, 19, 20, and 30 were also much more selective (900 - 344,000) over human cells than the non-targeted neutral derivatives (selectivity >8-fold). A preliminary in vivo study showed that modest doses of 15 and 16 reduced parasitaemia of mice infected with T. b. rhodesiense (STIB900). These compounds represent a promising new class of potent and selective hits against African trypanosomes.


Assuntos
Benzaldeídos/farmacologia , Proteínas Mitocondriais/antagonistas & inibidores , Oxirredutases/antagonistas & inibidores , Parabenos/farmacologia , Proteínas de Plantas/antagonistas & inibidores , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma congolense/efeitos dos fármacos , Trypanosoma/efeitos dos fármacos , Benzaldeídos/síntese química , Benzaldeídos/química , Cátions/química , Cátions/farmacologia , Relação Dose-Resposta a Droga , Proteínas Mitocondriais/metabolismo , Estrutura Molecular , Oxirredutases/metabolismo , Parabenos/síntese química , Parabenos/química , Testes de Sensibilidade Parasitária , Proteínas de Plantas/metabolismo , Relação Estrutura-Atividade , Tripanossomicidas/síntese química , Tripanossomicidas/química , Trypanosoma/enzimologia
10.
Infect Dis Poverty ; 6(1): 150, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-29017597

RESUMO

BACKGROUND: Acute human African trypanosomiasis (rHAT) caused by Trypanosoma brucei rhodesiense is associated with high mortality and is fatal if left untreated. Only a few studies have examined the psychological, social and economic impacts of rHAT. In this study, mixed qualitative and quantitative research methods were used to evaluate the socio-economic impacts of rHAT in Mambwe, Rufunsa, Mpika and Chama Districts of Zambia. METHODS: Individuals diagnosed with rHAT from 2004 to 2014 were traced using hospital records and discussions with communities. Either they, or their families, were interviewed using a structured questionnaire and focus group discussions were conducted with affected communities. The burden of the disease was investigated using disability adjusted life years (DALYs), with and without discounting and age-weighting. The impact of long-term disabilities on the rHAT burden was also investigated. RESULTS: Sixty four cases were identified in the study. The majority were identified in second stage, and the mortality rate was high (12.5%). The total number of DALYs was 285 without discounting or age-weighting. When long-term disabilities were included this estimate increased by 50% to 462. The proportion of years lived with disability (YLD) increased from 6.4% to 37% of the undiscounted and un-age-weighted DALY total. When a more active surveillance method was applied in 2013-2014 the cases identified increased dramatically, suggesting a high level of under-reporting. Similarly, the proportion of females increased substantially, indicating that passive surveillance may be especially failing this group. An average of 4.9 months of productive time was lost per patient as a consequence of infection. The health consequences included pain, amnesia and physical disability. The social consequences included stigma, dropping out of education, loss of friends and self-esteem. Results obtained from focus group discussions revealed misconceptions among community members which could be attributed to lack of knowledge about rHAT. CONCLUSIONS: The social and economic impact of rHAT on rural households and communities is substantial. Improved surveillance and strengthening of local medical services are needed for early and accurate diagnosis. Disease prevention should be prioritised in communities at risk of rHAT, and interventions put in place to prevent zoonotic disease spill over from domestic animals and wildlife. Supportive measures to mitigate the long-term effects of disability due to rHAT are needed.


Assuntos
Efeitos Psicossociais da Doença , Pessoas com Deficiência , Anos de Vida Ajustados por Qualidade de Vida , Tripanossomíase Africana/economia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Pessoas com Deficiência/estatística & dados numéricos , Feminino , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Zâmbia
11.
Acta Trop ; 165: 216-229, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27570206

RESUMO

INTRODUCTION: Human African trypanosomiasis (HAT) comprises two fatal parasitic diseases. Uganda is home to both chronic T. b. gambiense (gHAT) and the acute zoonotic form T. b. rhodesiense (rHAT) which occur in two large but discrete geographical foci. The area affected by rHAT has been rapidly expanding due to importation of T. b. rhodesiense infected cattle into tsetse infested but previously HAT free districts. Migration of rHAT has resulted in a considerable human health burden in these newly affected districts. Here, we examined the impact of a single, district-wide, mass chemotherapeutic livestock intervention, on T. b. rhodesiense prevalence in cattle and on incidence and distribution of human rHAT cases in Kamuli and Soroti districts in eastern Uganda. METHODS: A single mass intervention in domestic cattle (n=30,900) using trypanocidal drugs was undertaken in November and December 2002 under the EU funded Farming in Tsetse Controlled Areas (FITCA) programme. The intervention targeted removal of the reservoir of infection i.e. human infective T. b. rhodesiense parasites in cattle, in the absence of tsetse control. Interventions were applied in high-risk sub-counties of Kamuli district (endemic for rHAT) and Soroti district (where rHAT has been recently introduced). The prevalence of T. brucei s.l. and the human infective subspecies, T. b. rhodesiense in cattle (n=1833) was assessed before and 3 and 12 months after intervention using PCR-based methods. A combination of descriptive statistical analysis and spatial scan statistics were applied to analyse rHAT cases reported over a 5-year period (January 2000-July 2005). RESULTS: A single intervention was highly effective at removing human infective T. b. rhodesiense parasites from the cattle reservoir and contributed to a significant decrease in human rHAT cases. Intervention coverage was higher in Kamuli (81.1%) than in Soroti (47.3%) district but despite differences in coverage both districts showed a reduction in prevalence of T. b. brucei s.l. and T. b. rhodesiense. In Kamuli, the prevalence of T. brucei s.l. decreased by 54%, from 6.75% to 3.11%, 3, months post-intervention, rising to 4.7% at 12 months. The prevalence of T. b. rhodesiense was 3% pre-intervention and no T. b. rhodesiense infections were detected 3 and 12, months post-treatment. In Soroti, the prevalence of T. brucei s.l. in cattle decreased by 38% (from 21% to 13%) 3 months after intervention decreasing to less than 10% at 12 months. The prevalence of T. b. rhodesiense was reduced by 50% at 12-months post-intervention (6%-3%). Most notably, was the impact of the intervention on the population dynamics between T. b. brucei and human infective T. b. rhodesiense. Before intervention in Kamuli district 56% of T. b. brucei s.l. circulating in cattle were T. b. rhodesiense; at both 3 and 12 months after intervention none of the re-infecting T. b. brucei s.l. were human infective, T. rhodesiense. For human rHAT cases, there was a seven-fold decrease in rHAT incidence after intervention in Kamuli district (5.54 cases/1,000 head of population 2000-2002 to 0.76 cases/1,000, 2003-2005). Incidence data suggests that the intervention had minimal impact on the number of rHAT cases in Soroti overall, but showed a significant decrease in the seasonal peak of cases in the year following treatment. CONCLUSION: A single intervention, targeted at cattle, introduced at district level, in the absence of tsetse control, was highly effective at removing human infective rHAT parasites from the cattle reservoir and contributed to a significant decrease in human rHAT cases. The differential impacts observed between the two districts are related to both the different stages of rHAT endemicity in the districts, and levels of intervention coverage achieved in the cattle population. Treatment of cattle to remove the reservoir of rHAT infection offers a promising and cost effective approach for the control of rHAT. It is important that cattle are treated before relocation to prevent possible merger of the two HAT foci, which would complicate diagnosis and treatment of both gHAT and rHAT.


Assuntos
Animais Domésticos/parasitologia , Gado/parasitologia , Tripanossomicidas/uso terapêutico , Trypanosoma brucei rhodesiense/efeitos dos fármacos , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/parasitologia , Zoonoses/prevenção & controle , Animais , Bovinos , Doenças dos Bovinos/epidemiologia , Feminino , Humanos , Incidência , Masculino , Vacinação em Massa , Reação em Cadeia da Polimerase/métodos , Prevalência , Tripanossomíase Africana/epidemiologia , Uganda/epidemiologia
12.
Infect Dis Poverty ; 6(1): 16, 2017 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-28162093

RESUMO

BACKGROUND: Uganda has suffered from a series of epidemics of Human African Trypanosomiasis (HAT), a tsetse transmitted disease, also known as sleeping sickness. The area affected by acute Trypanosoma brucei rhodesiense HAT (rHAT) has been expanding, driven by importation of infected cattle into regions previously free of the disease. These regions are also affected by African Animal Trypanosomiasis (AAT) demanding a strategy for integrated disease control. METHODS: In 2008, the Public Private Partnership, Stamp Out Sleeping Sickness (SOS) administered a single dose of trypanocide to 31 486 head of cattle in 29 parishes in Dokolo and Kaberamaido districts. This study examines the impact of this intervention on the prevalence of rHAT and AAT trypanosomes in cattle from villages that had (HAT+ve) or had not (HAT-ve) experienced a recent case of rHAT. Cattle herds from 20 villages were sampled and screened by PCR, pre-intervention and 6-months post-intervention, for the presence or absence of: Trypanosoma brucei s.l.; human infective T. b. rhodesiense; Trypanosoma vivax; and Trypanosoma congolense savannah. RESULTS: Post-intervention, there was a significant decrease in the prevalence of T. brucei s.l. and the human infective sub-species T. b. rhodesiense in village cattle across all 20 villages. The prevalence of T. b. rhodesiense was reduced from 2.4% to 0.74% (P < 0.0001), with the intervention showing greater impact in HAT-ve villages. The number of villages containing cattle harbouring human infective parasites decreased from 15/20 to 8/20, with T. b. rhodesiense infection mainly persisting within cattle in HAT+ve villages (six/eight). The proportion of T. brucei s.l. infections identified as human infective T. b. rhodesiense decreased after the intervention from 8.3% (95% CI = 11.1-5.9%) to 4.1% (95% CI = 6.8-2.3%). Villages that had experienced a recent human case (HAT+ve villages) showed a significantly higher prevalence for AAT both pre- and post-intervention. For AAT the prevalence of T. vivax was significantly reduced from 5.9% to 0.05% post-intervention while the prevalence of T. congolense increased from 8.0% to 12.2%. CONCLUSIONS: The intervention resulted in a significant decrease in the prevalence of T. brucei s.l., human infective T. b. rhodesiense and T. vivax infection in village cattle herds. The proportion of T. brucei s.l. that were human infective, decreased from 1:12 T. brucei s.l. infections before the intervention to 1:33 post-intervention. It is clearly more difficult to eliminate T. b. rhodesiense from cattle in villages that have experienced a human case. Evidence of elevated levels of AAT in livestock within village herds is a useful indicator of risk for rHAT in Uganda. Integrated veterinary and medical surveillance is key to successful control of zoonotic rHAT.


Assuntos
Bovinos/parasitologia , Tripanossomicidas , Trypanosoma brucei rhodesiense , Tripanossomíase Africana , Drogas Veterinárias , Animais , Humanos , Tripanossomicidas/administração & dosagem , Tripanossomicidas/uso terapêutico , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/prevenção & controle , Tripanossomíase Africana/transmissão , Tripanossomíase Africana/veterinária , Uganda/epidemiologia , Drogas Veterinárias/administração & dosagem , Drogas Veterinárias/uso terapêutico
13.
Res Rep Trop Med ; 4: 1-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-30100778

RESUMO

In 2001, the World Health Organization (WHO) established a public-private partnership to fight human African trypanosomiasis (HAT). As a result of this continuous collaboration, and in addition to the coordination with nongovernmental organizations and bilateral cooperation agencies, the number of new cases of HAT annually reported by the WHO has strikingly decreased. In 2012, HAT was included in WHO's roadmap on neglected tropical diseases with a 2020 target date for elimination. Although the prevalence of HAT is decreasing and its elimination is targeted, control approaches must be adapted to the different epidemiological patterns in order to adopt the most adequate strategies to maintain their cost-effectiveness. These strategies must be flexible and dynamic in order to be adapted to the disease progression, as well as to the changes affecting the existing health facilities in transmission areas, including their accessibility, their capabilities, and their involvement in the elimination process. Considering the different patterns of transmission (Trypanosoma brucei (T.b.) rhodesiense HAT) and transmission intensity (T.b. gambiense HAT), different settings have been defined. In the case of T.b. rhodesiense, this form exists primarily where wild animals are the main parasite reservoir, and where the main parasite reservoir is cattle. In T.b. gambiense, this form exists in areas with high intensity transmission, areas with moderate intensity transmission, and areas with low intensity transmission. Criteria and indicators must be established to monitor and evaluate the actions implemented toward the elimination of HAT.

14.
Eur J Med Chem ; 66: 450-65, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23831695

RESUMO

A whole organism high-throughput screen of approximately 87,000 compounds against Trypanosoma brucei brucei led to the recent discovery of several novel compound classes with low micromolar activity against this organism and without appreciable cytotoxicity to mammalian cells. Herein we report a structure-activity relationship (SAR) investigation around one of these hit classes, the 3-(oxazolo[4,5-b]pyridin-2-yl)anilides. Sharp SAR is revealed, with our most active compound (5) exhibiting an IC50 of 91 nM against the human pathogenic strain T.b. rhodesiense and being more than 700 times less toxic towards the L6 mammalian cell line. Physicochemical properties are attractive for many compounds in this series. For the most potent representatives, we show that solubility and metabolic stability are key parameters to target during future optimisation.


Assuntos
Anilidas/química , Anilidas/farmacologia , Tripanossomicidas/química , Tripanossomicidas/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos , Tripanossomíase Africana/parasitologia , Anilidas/toxicidade , Animais , Humanos , Camundongos , Mioblastos Esqueléticos/efeitos dos fármacos , Ratos , Especificidade da Espécie , Relação Estrutura-Atividade , Tripanossomicidas/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA