Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 232
Filtrar
1.
Zhonghua Yan Ke Za Zhi ; 59(7): 570-573, 2023 Jul 11.
Artigo em Chinês | MEDLINE | ID: mdl-37408429

RESUMO

A 32-year-old female patient presented with bilateral vision loss for 2 months following her intake of various antiparasitic drugs, including closantel, a veterinary drug, for a self-perceived intraocular parasitic infection. Swept-source optical coherence tomography revealed diffuse hyperreflectivity between the outer nuclear layer and the retinal pigment epithelium, as well as the largely indistinguishable outer retinal layers. This case was clinically diagnosed with veterinary closantel-induced toxic retinopathy and had a poor visual prognosis after nerve nutrition and circulation improvement therapy due to the long duration of the disease.


Assuntos
Antiparasitários , Doenças Retinianas , Humanos , Feminino , Adulto , Antiparasitários/efeitos adversos , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/diagnóstico , Retina , Salicilanilidas/efeitos adversos , Epitélio Pigmentado da Retina , Tomografia de Coerência Óptica/métodos , Angiofluoresceinografia
2.
Infection ; 51(4): 1127-1139, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36961623

RESUMO

PURPOSE: Neurocysticercosis is common in regions endemic for Taenia solium. Active-stage neurocysticercosis can be treated with antiparasitic medication, but so far no study on efficacy and safety has been conducted in Africa. METHODS: We conducted a prospective cohort study on treatment of neurocysticercosis in Tanzania between August 2018 and January 2022. Patients were initially treated with albendazole (15 mg/kg/d) for 10 days and followed up for 6 months. Additionally in July 2021, all participants who then still had cysts were offered a combination therapy consisting of albendazole (15 mg/kg/d) and praziquantel (50 mg/kg/d). Antiparasitic treatment was accompanied by corticosteroid medication and anti-seizure medication if the patient had experienced epileptic seizures before treatment. RESULTS: Sixty-three patients were recruited for this study, of whom 17 had a complete follow-up after albendazole monotherapy. These patients had a total of 138 cysts at baseline, of which 58 (42%) had disappeared or calcified by the end of follow-up. The median cyst reduction was 40% (interquartile range 11-63%). Frequency of epileptic seizures reduced considerably (p < 0.001). Three patients had all active cysts resolved or calcified and of the remaining 14, eight received the combination therapy which resolved 63 of 66 cysts (95%). Adverse events were infrequent and mild to moderate during both treatment cycles. CONCLUSION: Cyst resolution was unsatisfactory with albendazole monotherapy but was very high when it was followed by a combination of albendazole and praziquantel.


Assuntos
Anti-Helmínticos , Cistos , Neurocisticercose , Humanos , Neurocisticercose/tratamento farmacológico , Neurocisticercose/complicações , Neurocisticercose/parasitologia , Albendazol/efeitos adversos , Antiparasitários/efeitos adversos , Praziquantel/efeitos adversos , Tanzânia , Estudos Prospectivos , Cistos/induzido quimicamente , Cistos/complicações , Cistos/tratamento farmacológico , Convulsões/tratamento farmacológico , Convulsões/induzido quimicamente , Convulsões/complicações , Anti-Helmínticos/efeitos adversos
3.
PLoS Negl Trop Dis ; 16(2): e0010189, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35139080

RESUMO

The elimination of onchocerciasis through community-based Mass Drug Administration (MDA) of ivermectin (Mectizan) is hampered by co-endemicity of Loa loa, as individuals who are highly co-infected with Loa loa parasites can suffer serious and occasionally fatal neurological reactions from the drug. The test-and-not-treat strategy of testing all individuals participating in MDA has some operational constraints including the cost and limited availability of LoaScope diagnostic tools. As a result, a Loa loa Antibody (Ab) Rapid Test was developed to offer a complementary way of determining the prevalence of loiasis. We develop a joint geostatistical modelling framework for the analysis of Ab and Loascope data to delineate whether an area is safe for MDA. Our results support the use of a two-stage strategy, in which Ab testing is used to identify areas that, with acceptably high probability, are safe or unsafe for MDA, followed by Loascope testing in areas whose safety status is uncertain. This work therefore contributes to the global effort towards the elimination of onchocerciasis as a public health problem by potentially reducing the time and cost required to establish whether an area is safe for MDA.


Assuntos
Antiparasitários/uso terapêutico , Coinfecção/tratamento farmacológico , Ivermectina/uso terapêutico , Loa/efeitos dos fármacos , Loíase/tratamento farmacológico , Oncocercose/tratamento farmacológico , Animais , Anticorpos Anti-Helmínticos/sangue , Antiparasitários/efeitos adversos , Coinfecção/epidemiologia , Coinfecção/parasitologia , Feminino , Humanos , Ivermectina/efeitos adversos , Loa/genética , Loa/fisiologia , Loíase/epidemiologia , Loíase/parasitologia , Masculino , Administração Massiva de Medicamentos/efeitos adversos , Modelos Estatísticos , Onchocerca/efeitos dos fármacos , Onchocerca/genética , Onchocerca/fisiologia , Oncocercose/epidemiologia , Oncocercose/parasitologia
8.
Cochrane Database Syst Rev ; 7: CD015017, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34318930

RESUMO

BACKGROUND: Ivermectin, an antiparasitic agent used to treat parasitic infestations, inhibits the replication of viruses in vitro. The molecular hypothesis of ivermectin's antiviral mode of action suggests an inhibitory effect on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication in the early stages of infection. Currently, evidence on efficacy and safety of ivermectin for prevention of SARS-CoV-2 infection and COVID-19 treatment is conflicting. OBJECTIVES: To assess the efficacy and safety of ivermectin compared to no treatment, standard of care, placebo, or any other proven intervention for people with COVID-19 receiving treatment as inpatients or outpatients, and for prevention of an infection with SARS-CoV-2 (postexposure prophylaxis). SEARCH METHODS: We searched the Cochrane COVID-19 Study Register, Web of Science (Emerging Citation Index and Science Citation Index), medRxiv, and Research Square, identifying completed and ongoing studies without language restrictions to 26 May 2021. SELECTION CRITERIA: We included randomized controlled trials (RCTs) comparing ivermectin to no treatment, standard of care, placebo, or another proven intervention for treatment of people with confirmed COVID-19 diagnosis, irrespective of disease severity, treated in inpatient or outpatient settings, and for prevention of SARS-CoV-2 infection. Co-interventions had to be the same in both study arms.  We excluded studies comparing ivermectin to other pharmacological interventions with unproven efficacy. DATA COLLECTION AND ANALYSIS: We assessed RCTs for bias, using the Cochrane risk of bias 2 tool. The primary analysis excluded studies with high risk of bias. We used GRADE to rate the certainty of evidence for the following outcomes 1. to treat inpatients with moderate-to-severe COVID-19: mortality, clinical worsening or improvement, adverse events, quality of life, duration of hospitalization, and viral clearance; 2. to treat outpatients with mild COVID-19: mortality, clinical worsening or improvement, admission to hospital, adverse events, quality of life, and viral clearance; (3) to prevent SARS-CoV-2 infection: SARS-CoV-2 infection, development of COVID-19 symptoms, adverse events, mortality, admission to hospital, and quality of life. MAIN RESULTS: We found 14 studies with 1678 participants investigating ivermectin compared to no treatment, placebo, or standard of care. No study compared ivermectin to an intervention with proven efficacy. There were nine studies treating participants with moderate COVID-19 in inpatient settings and four treating mild COVID-19 cases in outpatient settings. One study investigated ivermectin for prevention of SARS-CoV-2 infection. Eight studies had an open-label design, six were double-blind and placebo-controlled. Of the 41 study results contributed by included studies, about one third were at overall high risk of bias.  Ivermectin doses and treatment duration varied among included studies.  We identified 31 ongoing and 18 studies awaiting classification until publication of results or clarification of inconsistencies. Ivermectin compared to placebo or standard of care for inpatient COVID-19 treatment We are uncertain whether ivermectin compared to placebo or standard of care reduces or increases mortality (risk ratio (RR) 0.60, 95% confidence interval (CI) 0.14 to 2.51; 2 studies, 185 participants; very low-certainty evidence) and clinical worsening up to day 28 assessed as need for invasive mechanical ventilation (IMV) (RR 0.55, 95% CI 0.11 to 2.59; 2 studies, 185 participants; very low-certainty evidence) or need for supplemental oxygen (0 participants required supplemental oxygen; 1 study, 45 participants; very low-certainty evidence), adverse events within 28 days (RR 1.21, 95% CI 0.50 to 2.97; 1 study, 152 participants; very low-certainty evidence), and viral clearance at day seven (RR 1.82, 95% CI 0.51 to 6.48; 2 studies, 159 participants; very low-certainty evidence). Ivermectin may have little or no effect compared to placebo or standard of care on clinical improvement up to 28 days (RR 1.03, 95% CI 0.78 to 1.35; 1 study; 73 participants; low-certainty evidence) and duration of hospitalization (mean difference (MD) -0.10 days, 95% CI -2.43 to 2.23; 1 study; 45 participants; low-certainty evidence). No study reported quality of life up to 28 days. Ivermectin compared to placebo or standard of care for outpatient COVID-19 treatment We are uncertain whether ivermectin compared to placebo or standard of care reduces or increases mortality up to 28 days (RR 0.33, 95% CI 0.01 to 8.05; 2 studies, 422 participants; very low-certainty evidence) and clinical worsening up to 14 days assessed as need for IMV (RR 2.97, 95% CI 0.12 to 72.47; 1 study, 398 participants; very low-certainty evidence) or non-IMV or high flow oxygen requirement (0 participants required non-IMV or high flow; 1 study, 398 participants; very low-certainty evidence). We are uncertain whether ivermectin compared to placebo reduces or increases viral clearance at seven days (RR 3.00, 95% CI 0.13 to 67.06; 1 study, 24 participants; low-certainty evidence). Ivermectin may have little or no effect compared to placebo or standard of care on the number of participants with symptoms resolved up to 14 days (RR 1.04, 95% CI 0.89 to 1.21; 1 study, 398 participants; low-certainty evidence) and adverse events within 28 days (RR 0.95, 95% CI 0.86 to 1.05; 2 studies, 422 participants; low-certainty evidence). None of the studies reporting duration of symptoms were eligible for primary analysis. No study reported hospital admission or quality of life up to 14 days. Ivermectin compared to no treatment for prevention of SARS-CoV-2 infection We found one study. Mortality up to 28 days was the only outcome eligible for primary analysis. We are uncertain whether ivermectin reduces or increases mortality compared to no treatment (0 participants died; 1 study, 304 participants; very low-certainty evidence). The study reported results for development of COVID-19 symptoms and adverse events up to 14 days that were included in a secondary analysis due to high risk of bias. No study reported SARS-CoV-2 infection, hospital admission, and quality of life up to 14 days. AUTHORS' CONCLUSIONS: Based on the current very low- to low-certainty evidence, we are uncertain about the efficacy and safety of ivermectin used to treat or prevent COVID-19. The completed studies are small and few are considered high quality. Several studies are underway that may produce clearer answers in review updates. Overall, the reliable evidence available does not support the use ivermectin for treatment or prevention of COVID-19 outside of well-designed randomized trials.


Assuntos
Antiparasitários/uso terapêutico , Antivirais/uso terapêutico , Tratamento Farmacológico da COVID-19 , Ivermectina/uso terapêutico , Antiparasitários/efeitos adversos , Antivirais/efeitos adversos , COVID-19/mortalidade , COVID-19/prevenção & controle , COVID-19/virologia , Causas de Morte , Humanos , Ivermectina/efeitos adversos , Placebos/uso terapêutico , Profilaxia Pós-Exposição , Ensaios Clínicos Controlados Aleatórios como Assunto , Respiração Artificial/estatística & dados numéricos , SARS-CoV-2/efeitos dos fármacos , Fatores de Tempo , Resultado do Tratamento
9.
Cochrane Database Syst Rev ; 6: CD013117, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34184757

RESUMO

BACKGROUND: Malaria is transmitted through the bite of Plasmodium-infected adult female Anopheles mosquitoes. Ivermectin, an anti-parasitic drug, acts by killing mosquitoes that are exposed to the drug while feeding on the blood of people (known as blood feeds) who have ingested the drug. This effect on mosquitoes has been demonstrated by individual randomized trials. This effect has generated interest in using ivermectin as a tool for malaria control. OBJECTIVES: To assess the effect of community administration of ivermectin on malaria transmission. SEARCH METHODS: We searched the Cochrane Infectious Diseases Group (CIDG) Specialized Register, CENTRAL, MEDLINE, Embase, LILACS, Science Citation index - expanded, the World Health Organization (WHO) International Clinical Trials Registry Platform, ClinicalTrials.gov, and the National Institutes of Health (NIH) RePORTER database to 14 January 2021. We checked the reference lists of included studies for other potentially relevant studies, and contacted researchers working in the field for unpublished and ongoing trials. SELECTION CRITERIA: We included cluster-randomized controlled trials (cRCTs) that compared ivermectin, as single or multiple doses, with a control treatment or placebo given to populations living in malaria-endemic areas, in the context of mass drug administration. Primary outcomes were prevalence of malaria parasite infection and incidence of clinical malaria in the community. DATA COLLECTION AND ANALYSIS: Two review authors independently extracted data on the number of events and the number of participants in each trial arm at the time of assessment. For rate data, we noted the total time at risk in each trial arm. To assess risk of bias, we used Cochrane's RoB 2 tool for cRCTs. We documented the method of data analysis, any adjustments for clustering or other covariates, and recorded the estimate of the intra-cluster correlation (ICC) coefficient. We re-analysed the trial data provided by the trial authors to adjust for cluster effects. We used a Poisson mixed-effect model with small sample size correction, and a cluster-level analysis using the linear weighted model to adequately adjust for clustering.  MAIN RESULTS: We included one cRCT and identified six ongoing trials.  The included cRCT examined the incidence of malaria in eight villages in Burkina Faso, randomized to two arms. Both trial arms received a single dose of ivermectin 150 µg/kg to 200 µg/kg, together with a dose of albendazole. The villages in the intervention arm received an additional five doses of ivermectin, once every three weeks. Children were enrolled into an active cohort, in which they were repeatedly screened for malaria infection.  The primary outcome was the cumulative incidence of uncomplicated malaria in a cohort of children aged five years and younger, over the 18-week study. We judged the study to be at high risk of bias, as the analysis did not account for clustering or correlation between participants in the same village. The study did not demonstrate an effect of Ivermectin on the cumulative incidence of uncomplicated malaria in the cohort of children over the 18-week study (risk ratio 0.86, 95% confidence interval (CI) 0.62 to 1.17; P = 0.2607; very low-certainty evidence). AUTHORS' CONCLUSIONS: We are uncertain whether community administration of ivermectin has an effect on malaria transmission, based on one trial published to date.


Assuntos
Antiparasitários/administração & dosagem , Ivermectina/administração & dosagem , Malária/transmissão , Controle de Mosquitos , Animais , Antiparasitários/efeitos adversos , Antiparasitários/sangue , Viés , Burkina Faso/epidemiologia , Pré-Escolar , Análise de Dados , Humanos , Incidência , Lactente , Ivermectina/efeitos adversos , Ivermectina/sangue , Malária/epidemiologia , Malária/prevenção & controle , Projetos Piloto , Prevalência , Ensaios Clínicos Controlados Aleatórios como Assunto
11.
Pharmacol Rep ; 73(5): 1473-1479, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33779964

RESUMO

OBJECTIVE: The effect of ivermectin on mortality in patients with novel coronavirus disease 2019 (COVID-19) has been investigated in many studies. We aimed to perform a meta-analysis of randomized controlled trials to investigate the overall effect of ivermectin on the risk of mortality in patients with COVID-19. METHODS: We systematically searched PubMed, Cochrane Central Register of Controlled Trials, Google Scholar, and preprint repository databases (up to February 28, 2021). Random-effects and inverse variance heterogeneity meta-analysis were used to pool the odds ratio of individual trials. The risk of bias was appraised using Version 2 of the Cochrane risk-of-bias tool for randomized trials. RESULTS: Six randomized controlled trials were included in this analysis with a total of 658 patients who were randomized to receive ivermectin and 597 patients randomized in the control group who did not receive ivermectin. Of six trials, four had an overall high risk of bias. The estimated effect of ivermectin indicated mortality benefits (pooled odds ratio = 0.21; 95% confidence interval 0.11-0.42, n = 1255), with some evidence against the hypothesis of 'no significant difference' at the current sample size. CONCLUSION: We observed a preliminary beneficial effect on mortality associated with ivermectin use in patients with COVID-19 that warrants further clinical evidence in appropriately designed large-scale randomized controlled trials.


Assuntos
Antiparasitários/efeitos adversos , COVID-19/mortalidade , Ivermectina/efeitos adversos , SARS-CoV-2 , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
12.
J Fish Dis ; 44(7): 899-912, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33606885

RESUMO

This retrospective descriptive study estimates cage-level mortality distributions after six immediate delousing methods: thermal, mechanical, hydrogen peroxide, medicinal, freshwater and combination of medicinal treatments. We investigated mortality patterns associated with 4 644 delousing treatment of 1 837 cohorts of farmed Atlantic salmon (Salmo salar) stocked in sea along the Norwegian coast between 2014 and 2017. The mortality is expressed as mortality rates. We found distributions of delta mortality rate within 1, 7 and 14 days after all six delousing treatments, using mortality rate within 7 days before treatments as baseline. The results show that we can expect increased mortality rates after all six delousing methods. The median delta mortality rates after thermal and mechanical delousing are 5.4 and 6.3 times higher than medicinal treatment, respectively, for the 2017 year-class. There is a reduction in the delta median mortality for thermal and freshwater delousing from 2015 to 2019. There is a wide variability in the mortality rates, in particular for thermal delousing. Our results suggest that the variability in delta mortality for thermal delousing has been reduced from the 2014 to 2017 year-class, indicating an improvement of the technique. However, a significant increase in the number of thermal treatments from 14 in 2015 to 738 in 2018 probably contributes to the overall increased mortality in Norwegian salmon farming.


Assuntos
Antiparasitários/uso terapêutico , Arguloida/efeitos dos fármacos , Ectoparasitoses/veterinária , Doenças dos Peixes/tratamento farmacológico , Salmo salar , Animais , Antiparasitários/efeitos adversos , Ectoparasitoses/tratamento farmacológico , Ectoparasitoses/mortalidade , Doenças dos Peixes/mortalidade , Água Doce , Temperatura Alta , Peróxido de Hidrogênio/efeitos adversos , Peróxido de Hidrogênio/uso terapêutico , Estudos Retrospectivos
13.
PLoS One ; 15(10): e0240894, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33119627

RESUMO

The salmon louse Lepeophtheirus salmonis has been a substantial obstacle in Norwegian farming of Atlantic salmon for decades. With a limited selection of available medicines and frequent delousing treatments, resistance has emerged among salmon lice. Surveillance of salmon louse sensitivity has been in place since 2013, and consumption of medicines has been recorded since the early 80's. The peak year for salmon lice treatments was 2015, when 5.7 times as many tonnes of salmonids were treated compared to harvested. In recent years, non-medicinal methods of delousing farmed fish have been introduced to the industry. By utilizing data on the annual consumption of medicines, annual frequency of medicinal and non-medicinal treatments, the aim of the current study was to describe the causative factors behind salmon lice sensitivity in the years 2000-2019, measured through toxicity tests-bioassays. The sensitivity data from 2000-2012 demonstrate the early emergence of resistance in salmon lice along the Norwegian coast. Reduced sensitivity towards azamethiphos, deltamethrin and emamectin benzoate was evident from 2009, 2009 and 2007, respectively. The annual variation in medicine consumption and frequency of medicinal treatments correlated well with the evolution in salmon louse sensitivity. The patterns are similar, with a relatively small response delay from the decline in the consumption of medicines in Norway (2016 and onward) to the decline in measured resistance among salmon louse (2017 and onward). 2017 was the first year in which non-medicinal treatments outnumbered medicinal delousing treatments as well as the peak year in numbers of cleanerfish deployed. This study highlights the significance of avoiding heavy reliance on a few substance groups to combat ectoparasites, this can be a potent catalyst for resistance evolution. Further, it demonstrates the importance of transparency in the global industry, which enables the industry to learn from poor choices in the past.


Assuntos
Antiparasitários/efeitos adversos , Resistência a Medicamentos/genética , Doenças dos Peixes/tratamento farmacológico , Salmo salar/crescimento & desenvolvimento , Animais , Antiparasitários/farmacologia , Aquicultura , Doenças dos Peixes/parasitologia , Pesqueiros , Humanos , Noruega , Organotiofosfatos/efeitos adversos , Organotiofosfatos/farmacologia , Ftirápteros/efeitos dos fármacos , Ftirápteros/patogenicidade , Salmo salar/parasitologia , Alimentos Marinhos
14.
Vet Rec ; 187(5): 197, 2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32887833

RESUMO

Rosemary Perkins argues that the environmental impact of veterinary parasiticide products is cause for concern and that more research needs to be done to fully appreciate their environmental effects.


Assuntos
Antiparasitários/efeitos adversos , Doenças do Gato/tratamento farmacológico , Doenças do Cão/tratamento farmacológico , Meio Ambiente , Doenças Parasitárias em Animais/tratamento farmacológico , Animais , Antiparasitários/uso terapêutico , Gatos , Cães , Humanos , Animais de Estimação
16.
Eur Rev Med Pharmacol Sci ; 24(13): 7412-7419, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32706080

RESUMO

OBJECTIVE: Vorinostat is a drug used to treat cutaneous T cell lymphoma whose action mechanism is based on Histone Deacetylase inhibition. Histone Deacetylases are a family of enzymes that remove acetyl groups from histone and non-histone proteins that control many crucial processes, such as gene regulation, cell cycle progression, differentiation, and apoptosis. Histone Deacetylase homologues are also expressed in parasites of the genus Plasmodium, Leishmania, Cryptosporidium, Schistosoma, Entamoeba, and others. In this way, antiparasitic properties of Vorinostat have been explored. The aim of this review is to report the current state knowledge of Vorinostat as antiparasitic drug against Plasmodium, Leishmania, Cryptosporidium, Schistosoma and Entamoeba in order to support future investigation in this field. MATERIALS AND METHODS: The authors revised the recent and relevant literature concerning the topic and discussed advances and limitations of studies on Vorinostat as potential drug to treat human parasitic diseases. RESULTS: Vorinostat has been efficient in vitro and, in some cases, in vivo, against parasites that cause parasitic diseases, such as malaria, leishmaniasis, cryptosporidiosis, amoebiasis, and schistosomiasis. CONCLUSIONS: In vitro and in vivo models have demonstrated the antiparasitic activity of Vorinostat, however, the challenge is to assay its activity in animal models and to evaluate if Vorinostat is safe for humans as new alternative to treat human parasitic infections.


Assuntos
Antiparasitários/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases , Parasitos/efeitos dos fármacos , Doenças Parasitárias/tratamento farmacológico , Proteínas de Protozoários/antagonistas & inibidores , Vorinostat/uso terapêutico , Animais , Antiparasitários/efeitos adversos , Reposicionamento de Medicamentos , Inibidores de Histona Desacetilases/efeitos adversos , Histona Desacetilases/metabolismo , Interações Hospedeiro-Parasita , Humanos , Parasitos/enzimologia , Parasitos/patogenicidade , Doenças Parasitárias/diagnóstico , Doenças Parasitárias/parasitologia , Proteínas de Protozoários/metabolismo , Vorinostat/efeitos adversos
17.
PLoS Negl Trop Dis ; 14(6): e0008298, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32511226

RESUMO

In Haiti, 22 communes still require mass drug administration (MDA) to eliminate lymphatic filariasis (LF) as a public health problem. Several clinical trials have shown that a single oral dose of ivermectin (IVM), diethylcarbamazine (DEC) and albendazole (ALB) (IDA) is more effective than DEC plus ALB (DA) for clearing Wuchereria bancrofti microfilariae (Mf). We performed a cluster-randomized community study to compare the safety and efficacy of IDA and DA in an LF-endemic area in northern Haiti. Ten localities were randomized to receive either DA or IDA. Participants were monitored for adverse events (AE), parasite antigenemia, and microfilaremia. Antigen-positive participants were retested one year after MDA to assess treatment efficacy. Fewer participants (11.0%, 321/2917) experienced at least one AE after IDA compared to DA (17.3%, 491/2844, P<0.001). Most AEs were mild, and the three most common AEs reported were headaches, dizziness and abdominal pain. Serious AEs developed in three participants who received DA. Baseline prevalence for filarial antigenemia was 8.0% (239/3004) in IDA localities and 11.5% (344/2994) in DA localities (<0.001). Of those with positive antigenemia, 17.6% (42/239) in IDA localities and 20.9% (72/344, P = 0.25) in DA localities were microfilaremic. One year after treatment, 84% percent of persons with positive filarial antigen tests at baseline could be retested. Clearance rates for filarial antigenemia were 20.5% (41/200) after IDA versus 25.4% (74/289) after DA (P = 0.3). However, 94.4% (34/36) of IDA recipients and 75.9% (44/58) of DA recipients with baseline microfilaremia were Mf negative at the time of retest (P = 0.02). Thus, MDA with IDA was at least as well tolerated and significantly more effective for clearing Mf compared to the standard DA regimen in this study. Effective MDA coverage with IDA could accelerate the elimination of LF as a public health problem in the 22 communes that still require MDA in Haiti.


Assuntos
Albendazol/administração & dosagem , Antiparasitários/administração & dosagem , Dietilcarbamazina/administração & dosagem , Ivermectina/administração & dosagem , Adolescente , Adulto , Albendazol/efeitos adversos , Animais , Antiparasitários/efeitos adversos , Criança , Pré-Escolar , Dietilcarbamazina/efeitos adversos , Quimioterapia Combinada , Filariose Linfática/tratamento farmacológico , Feminino , Haiti , Humanos , Ivermectina/efeitos adversos , Modelos Logísticos , Masculino , Administração Massiva de Medicamentos/efeitos adversos , Pessoa de Meia-Idade , Prevalência , Resultado do Tratamento , Adulto Jovem
19.
PLoS Negl Trop Dis ; 14(3): e0008150, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32196500

RESUMO

Parasitic infections are a major source of human suffering, mortality, and economic loss, but drug development for these diseases has been stymied by the significant expense involved in bringing a drug though clinical trials and to market. Identification of single compounds active against multiple parasitic pathogens could improve the economic incentives for drug development as well as simplifying treatment regimens. We recently performed a screen of repurposed compounds against the protozoan parasite Entamoeba histolytica, causative agent of amebic dysentery, and identified four compounds (anisomycin, prodigiosin, obatoclax and nithiamide) with low micromolar potency and drug-like properties. Here, we extend our investigation of these drugs. We assayed the speed of killing of E. histolytica trophozoites and found that all four have more rapid action than the current drug of choice, metronidazole. We further established a multi-institute collaboration to determine whether these compounds may have efficacy against other parasites and opportunistic pathogens. We found that anisomycin, prodigiosin and obatoclax all have broad-spectrum antiparasitic activity in vitro, including activity against schistosomes, T. brucei, and apicomplexan parasites. In several cases, the drugs were found to have significant improvements over existing drugs. For instance, both obatoclax and prodigiosin were more efficacious at inhibiting the juvenile form of Schistosoma than the current standard of care, praziquantel. Additionally, low micromolar potencies were observed against pathogenic free-living amebae (Naegleria fowleri, Balamuthia mandrillaris and Acanthamoeba castellanii), which cause CNS infection and for which there are currently no reliable treatments. These results, combined with the previous human use of three of these drugs (obatoclax, anisomycin and nithiamide), support the idea that these compounds could serve as the basis for the development of broad-spectrum anti-parasitic drugs.


Assuntos
Anisomicina/farmacologia , Antiparasitários/farmacologia , Reposicionamento de Medicamentos , Parasitos/efeitos dos fármacos , Prodigiosina/farmacologia , Pirróis/farmacologia , Animais , Anisomicina/efeitos adversos , Anisomicina/farmacocinética , Antiparasitários/efeitos adversos , Antiparasitários/farmacocinética , Linhagem Celular , Sobrevivência Celular , Fibroblastos/efeitos dos fármacos , Humanos , Indóis , Camundongos , Testes de Sensibilidade Parasitária , Prodigiosina/efeitos adversos , Prodigiosina/farmacocinética , Pirróis/efeitos adversos , Pirróis/farmacocinética , Ratos
20.
PLoS Negl Trop Dis ; 14(3): e0008106, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32176703

RESUMO

Lymphatic filariasis has remained endemic in Fiji despite repeated mass drug administration using the well-established and safe combination of diethylcarbamazine and albendazole (DA) since 2002. In certain settings the addition of ivermectin to this combination (IDA) remains a safe strategy and is more efficacious. However, the safety has yet to be described in scabies and soil-transmitted helminth endemic settings like Fiji. Villages of Rotuma and Gau islands were randomised to either DA or IDA. Residents received weight-based treatment unblinded with standard exclusions. Participants were actively found and asked by a nurse about their health daily for the first two days and then asked to seek review for the next five days if unwell. Anyone with severe symptoms were reviewed by a doctor and any serious adverse event was reported to the Medical Monitor and Data Safety Monitoring Board. Of 3612 enrolled and eligible participants, 1216 were randomised to DA and 2396 to IDA. Age and sex in both groups were representative of the population. Over 99% (3598) of participants completed 7 days follow-up. Adverse events were reported by 600 participants (16.7%), distributed equally between treatment groups, with most graded as mild (93.2%). There were three serious adverse events, all judged not attributable to treatment by an independent medical monitor. Fatigue was the most common symptom reported by 8.5%, with headache, dizziness, nausea and arthralgia being the next four most common symptoms. Adverse events were more likely in participants with microfilaremia (43.2% versus 15.7%), but adverse event frequency was not related to the presence of scabies or soil-transmitted helminth infection. IDA has comparable safety to DA with the same frequency of adverse events experienced following community mass drug administration. The presence of co-endemic infections did not increase adverse events. IDA can be used in community programs where preventative chemotherapy is needed for control of lymphatic filariasis and other neglected tropical diseases.


Assuntos
Albendazol/efeitos adversos , Antiparasitários/efeitos adversos , Dietilcarbamazina/efeitos adversos , Inseticidas/efeitos adversos , Ivermectina/efeitos adversos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Albendazol/administração & dosagem , Antiparasitários/administração & dosagem , Criança , Pré-Escolar , Infecções Comunitárias Adquiridas/tratamento farmacológico , Dietilcarbamazina/administração & dosagem , Quimioterapia Combinada/efeitos adversos , Quimioterapia Combinada/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Filariose Linfática/tratamento farmacológico , Feminino , Fiji , Helmintíase/tratamento farmacológico , Humanos , Lactente , Inseticidas/administração & dosagem , Ivermectina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Doenças Negligenciadas/tratamento farmacológico , População Rural , Escabiose/tratamento farmacológico , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...