Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.927
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 163(6): 1297-300, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26638061

RESUMO

The 2015 Nobel Prize in Physiology or Medicine has been awarded to William C. Campbell, Satoshi Omura, and Youyou Tu for the discovery of avermectins and artemisinin, respectively, therapies that revolutionized the treatment of devastating parasite diseases. With the recent technological advances, a New Golden Age of natural products drug discovery is dawning.


Assuntos
Descoberta de Drogas , Prêmio Nobel , Doenças Parasitárias/tratamento farmacológico , Artemisininas/química , Artemisininas/uso terapêutico , História da Medicina , História do Século XXI , Ivermectina/análogos & derivados , Ivermectina/química , Ivermectina/uso terapêutico , Fisiologia/história
2.
N Engl J Med ; 386(18): 1721-1731, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35353979

RESUMO

BACKGROUND: The efficacy of ivermectin in preventing hospitalization or extended observation in an emergency setting among outpatients with acutely symptomatic coronavirus disease 2019 (Covid-19), the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is unclear. METHODS: We conducted a double-blind, randomized, placebo-controlled, adaptive platform trial involving symptomatic SARS-CoV-2-positive adults recruited from 12 public health clinics in Brazil. Patients who had had symptoms of Covid-19 for up to 7 days and had at least one risk factor for disease progression were randomly assigned to receive ivermectin (400 µg per kilogram of body weight) once daily for 3 days or placebo. (The trial also involved other interventions that are not reported here.) The primary composite outcome was hospitalization due to Covid-19 within 28 days after randomization or an emergency department visit due to clinical worsening of Covid-19 (defined as the participant remaining under observation for >6 hours) within 28 days after randomization. RESULTS: A total of 3515 patients were randomly assigned to receive ivermectin (679 patients), placebo (679), or another intervention (2157). Overall, 100 patients (14.7%) in the ivermectin group had a primary-outcome event, as compared with 111 (16.3%) in the placebo group (relative risk, 0.90; 95% Bayesian credible interval, 0.70 to 1.16). Of the 211 primary-outcome events, 171 (81.0%) were hospital admissions. Findings were similar to the primary analysis in a modified intention-to-treat analysis that included only patients who received at least one dose of ivermectin or placebo (relative risk, 0.89; 95% Bayesian credible interval, 0.69 to 1.15) and in a per-protocol analysis that included only patients who reported 100% adherence to the assigned regimen (relative risk, 0.94; 95% Bayesian credible interval, 0.67 to 1.35). There were no significant effects of ivermectin use on secondary outcomes or adverse events. CONCLUSIONS: Treatment with ivermectin did not result in a lower incidence of medical admission to a hospital due to progression of Covid-19 or of prolonged emergency department observation among outpatients with an early diagnosis of Covid-19. (Funded by FastGrants and the Rainwater Charitable Foundation; TOGETHER ClinicalTrials.gov number, NCT04727424.).


Assuntos
Anti-Infecciosos , Tratamento Farmacológico da COVID-19 , Ivermectina , Adulto , Assistência Ambulatorial , Anti-Infecciosos/efeitos adversos , Anti-Infecciosos/uso terapêutico , Teorema de Bayes , Método Duplo-Cego , Hospitalização , Humanos , Ivermectina/efeitos adversos , Ivermectina/uso terapêutico , SARS-CoV-2 , Resultado do Tratamento
3.
N Engl J Med ; 387(7): 599-610, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-36070710

RESUMO

BACKGROUND: Early treatment to prevent severe coronavirus disease 2019 (Covid-19) is an important component of the comprehensive response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. METHODS: In this phase 3, double-blind, randomized, placebo-controlled trial, we used a 2-by-3 factorial design to test the effectiveness of three repurposed drugs - metformin, ivermectin, and fluvoxamine - in preventing serious SARS-CoV-2 infection in nonhospitalized adults who had been enrolled within 3 days after a confirmed diagnosis of infection and less than 7 days after the onset of symptoms. The patients were between the ages of 30 and 85 years, and all had either overweight or obesity. The primary composite end point was hypoxemia (≤93% oxygen saturation on home oximetry), emergency department visit, hospitalization, or death. All analyses used controls who had undergone concurrent randomization and were adjusted for SARS-CoV-2 vaccination and receipt of other trial medications. RESULTS: A total of 1431 patients underwent randomization; of these patients, 1323 were included in the primary analysis. The median age of the patients was 46 years; 56% were female (6% of whom were pregnant), and 52% had been vaccinated. The adjusted odds ratio for a primary event was 0.84 (95% confidence interval [CI], 0.66 to 1.09; P = 0.19) with metformin, 1.05 (95% CI, 0.76 to 1.45; P = 0.78) with ivermectin, and 0.94 (95% CI, 0.66 to 1.36; P = 0.75) with fluvoxamine. In prespecified secondary analyses, the adjusted odds ratio for emergency department visit, hospitalization, or death was 0.58 (95% CI, 0.35 to 0.94) with metformin, 1.39 (95% CI, 0.72 to 2.69) with ivermectin, and 1.17 (95% CI, 0.57 to 2.40) with fluvoxamine. The adjusted odds ratio for hospitalization or death was 0.47 (95% CI, 0.20 to 1.11) with metformin, 0.73 (95% CI, 0.19 to 2.77) with ivermectin, and 1.11 (95% CI, 0.33 to 3.76) with fluvoxamine. CONCLUSIONS: None of the three medications that were evaluated prevented the occurrence of hypoxemia, an emergency department visit, hospitalization, or death associated with Covid-19. (Funded by the Parsemus Foundation and others; COVID-OUT ClinicalTrials.gov number, NCT04510194.).


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Fluvoxamina , Ivermectina , Metformina , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/complicações , Vacinas contra COVID-19 , Método Duplo-Cego , Feminino , Fluvoxamina/uso terapêutico , Humanos , Hipóxia/etiologia , Ivermectina/uso terapêutico , Masculino , Metformina/uso terapêutico , Pessoa de Meia-Idade , Obesidade/complicações , Sobrepeso/complicações , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , SARS-CoV-2
4.
Clin Microbiol Rev ; 36(4): e0003323, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-37937980

RESUMO

Strongyloidiasis is a World Health Organization neglected tropical disease usually caused by Strongyloides stercoralis, a parasitic worm with a complex life cycle. Globally, 300-600 million people are infected through contact with fecally contaminated soil. An autoinfective component of the life cycle can lead to chronic infection that may be asymptomatic or cause long-term symptoms, including malnourishment in children. Low larval output can limit the sensitivity of detection in stool, with serology being effective but less sensitive in immunocompromise. Host immunosuppression can trigger catastrophic, fatal hyperinfection/dissemination, where large numbers of larvae pierce the bowel wall and disseminate throughout the organs. Stable disease is effectively treated by single-dose ivermectin, with disease in immunocompromised patients treated with multiple doses. Strategies for management include raising awareness, clarifying zoonotic potential, the development and use of effective diagnostic tests for epidemiological studies and individual diagnosis, and the implementation of treatment programs with research into therapeutic alternatives and medication safety.


Assuntos
Strongyloides stercoralis , Estrongiloidíase , Animais , Criança , Humanos , Estrongiloidíase/diagnóstico , Estrongiloidíase/tratamento farmacológico , Estrongiloidíase/parasitologia , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Hospedeiro Imunocomprometido , Terapia de Imunossupressão
5.
Clin Infect Dis ; 78(Supplement_2): S138-S145, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38662693

RESUMO

BACKGROUND: Concerns that annual mass administration of ivermectin, the predominant strategy for onchocerciasis control and elimination, may not lead to elimination of parasite transmission (EoT) in all endemic areas have increased interest in alternative treatment strategies. One such strategy is moxidectin. We performed an updated economic assessment of moxidectin- relative to ivermectin-based strategies. METHODS: We investigated annual and biannual community-directed treatment with ivermectin (aCDTI, bCDTI) and moxidectin (aCDTM, bCDTM) with minimal or enhanced coverage (65% or 80% of total population taking the drug, respectively) in intervention-naive areas with 30%, 50%, or 70% microfilarial baseline prevalence (representative of hypo-, meso-, and hyperendemic areas). We compared programmatic delivery costs for the number of treatments achieving 90% probability of EoT (EoT90), calculated with the individual-based stochastic transmission model EPIONCHO-IBM. We used the costs for 40 years of program delivery when EoT90 was not reached earlier. The delivery costs do not include drug costs. RESULTS: aCDTM and bCDTM achieved EoT90 with lower programmatic delivery costs than aCDTI with 1 exception: aCDTM with minimal coverage did not achieve EoT90 in hyperendemic areas within 40 years. With minimal coverage, bCDTI delivery costs as much or more than aCDTM and bCDTM. With enhanced coverage, programmatic delivery costs for aCDTM and bCDTM were lower than for aCDTI and bCDTI. CONCLUSIONS: Moxidectin-based strategies could accelerate progress toward EoT and reduce programmatic delivery costs compared with ivermectin-based strategies. The costs of moxidectin to national programs are needed to quantify whether delivery cost reductions will translate into overall program cost reduction.


Assuntos
Ivermectina , Macrolídeos , Oncocercose , Macrolídeos/uso terapêutico , Macrolídeos/economia , Macrolídeos/administração & dosagem , Oncocercose/tratamento farmacológico , Oncocercose/prevenção & controle , Oncocercose/economia , Oncocercose/epidemiologia , Humanos , Ivermectina/economia , Ivermectina/uso terapêutico , Ivermectina/administração & dosagem , Administração Massiva de Medicamentos/economia , Erradicação de Doenças/economia , Análise Custo-Benefício
6.
Clin Infect Dis ; 78(Supplement_2): S131-S137, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38662696

RESUMO

Mass drug administration (MDA) of antifilarial drugs is the main strategy for the elimination of lymphatic filariasis (LF). Recent clinical trials indicated that the triple-drug therapy with ivermectin, diethylcarbamazine, and albendazole (IDA) is much more effective against LF than the widely used two-drug combinations (albendazole plus either ivermectin or diethylcarbamazine). For IDA-based MDA, the stop-MDA decision is made based on microfilariae (mf) prevalence in adults. In this study, we assess how the probability of eventually reaching elimination of transmission depends on the critical threshold used in transmission assessment surveys (TAS-es) to define whether transmission was successfully suppressed and triple-drug MDA can be stopped. This analysis focuses on treatment-naive Indian settings. We do this for a range of epidemiological and programmatic contexts, using the established LYMFASIM model for transmission and control of LF. Based on our simulations, a single TAS, one year after the last MDA round, provides limited predictive value of having achieved suppressed transmission, while a higher MDA coverage increases elimination probability, thus leading to a higher predictive value. Every additional TAS, conditional on previous TAS-es being passed with the same threshold, further improves the predictive value for low values of stop-MDA thresholds. An mf prevalence threshold of 0.5% corresponding to TAS-3 results in ≥95% predictive value even when the MDA coverage is relatively low.


Assuntos
Albendazol , Dietilcarbamazina , Quimioterapia Combinada , Filariose Linfática , Filaricidas , Ivermectina , Administração Massiva de Medicamentos , Microfilárias , Filariose Linfática/tratamento farmacológico , Filariose Linfática/epidemiologia , Filariose Linfática/prevenção & controle , Humanos , Albendazol/uso terapêutico , Albendazol/administração & dosagem , Filaricidas/uso terapêutico , Dietilcarbamazina/uso terapêutico , Dietilcarbamazina/administração & dosagem , Ivermectina/uso terapêutico , Ivermectina/administração & dosagem , Animais , Índia/epidemiologia , Microfilárias/efeitos dos fármacos , Adulto , Prevalência
7.
Clin Infect Dis ; 78(Supplement_2): S93-S100, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38662701

RESUMO

BACKGROUND: Mass drug administration (MDA) is the cornerstone for the elimination of lymphatic filariasis (LF). The proportion of the population that is never treated (NT) is a crucial determinant of whether this goal is achieved within reasonable time frames. METHODS: Using 2 individual-based stochastic LF transmission models, we assess the maximum permissible level of NT for which the 1% microfilaremia (mf) prevalence threshold can be achieved (with 90% probability) within 10 years under different scenarios of annual MDA coverage, drug combination and transmission setting. RESULTS: For Anopheles-transmission settings, we find that treating 80% of the eligible population annually with ivermectin + albendazole (IA) can achieve the 1% mf prevalence threshold within 10 years of annual treatment when baseline mf prevalence is 10%, as long as NT <10%. Higher proportions of NT are acceptable when more efficacious treatment regimens are used. For Culex-transmission settings with a low (5%) baseline mf prevalence and diethylcarbamazine + albendazole (DA) or ivermectin + diethylcarbamazine + albendazole (IDA) treatment, elimination can be reached if treatment coverage among eligibles is 80% or higher. For 10% baseline mf prevalence, the target can be achieved when the annual coverage is 80% and NT ≤15%. Higher infection prevalence or levels of NT would make achieving the target more difficult. CONCLUSIONS: The proportion of people never treated in MDA programmes for LF can strongly influence the achievement of elimination and the impact of NT is greater in high transmission areas. This study provides a starting point for further development of criteria for the evaluation of NT.


Assuntos
Albendazol , Filariose Linfática , Filaricidas , Ivermectina , Administração Massiva de Medicamentos , Filariose Linfática/tratamento farmacológico , Filariose Linfática/prevenção & controle , Filariose Linfática/epidemiologia , Filariose Linfática/transmissão , Humanos , Animais , Filaricidas/uso terapêutico , Filaricidas/administração & dosagem , Albendazol/administração & dosagem , Albendazol/uso terapêutico , Ivermectina/administração & dosagem , Ivermectina/uso terapêutico , Prevalência , Anopheles/parasitologia , Erradicação de Doenças/métodos , Wuchereria bancrofti/efeitos dos fármacos , Dietilcarbamazina/administração & dosagem , Dietilcarbamazina/uso terapêutico , Quimioterapia Combinada
8.
Clin Infect Dis ; 79(2): 354-363, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-38690892

RESUMO

BACKGROUND: Metformin has antiviral activity against RNA viruses including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The mechanism appears to be suppression of protein translation via targeting the host mechanistic target of rapamycin pathway. In the COVID-OUT randomized trial for outpatient coronavirus disease 2019 (COVID-19), metformin reduced the odds of hospitalizations/death through 28 days by 58%, of emergency department visits/hospitalizations/death through 14 days by 42%, and of long COVID through 10 months by 42%. METHODS: COVID-OUT was a 2 × 3 randomized, placebo-controlled, double-blind trial that assessed metformin, fluvoxamine, and ivermectin; 999 participants self-collected anterior nasal swabs on day 1 (n = 945), day 5 (n = 871), and day 10 (n = 775). Viral load was quantified using reverse-transcription quantitative polymerase chain reaction. RESULTS: The mean SARS-CoV-2 viral load was reduced 3.6-fold with metformin relative to placebo (-0.56 log10 copies/mL; 95% confidence interval [CI], -1.05 to -.06; P = .027). Those who received metformin were less likely to have a detectable viral load than placebo at day 5 or day 10 (odds ratio [OR], 0.72; 95% CI, .55 to .94). Viral rebound, defined as a higher viral load at day 10 than day 5, was less frequent with metformin (3.28%) than placebo (5.95%; OR, 0.68; 95% CI, .36 to 1.29). The metformin effect was consistent across subgroups and increased over time. Neither ivermectin nor fluvoxamine showed effect over placebo. CONCLUSIONS: In this randomized, placebo-controlled trial of outpatient treatment of SARS-CoV-2, metformin significantly reduced SARS-CoV-2 viral load, which may explain the clinical benefits in this trial. Metformin is pleiotropic with other actions that are relevant to COVID-19 pathophysiology. CLINICAL TRIALS REGISTRATION: NCT04510194.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , COVID-19 , Metformina , SARS-CoV-2 , Carga Viral , Humanos , Metformina/uso terapêutico , Metformina/farmacologia , Carga Viral/efeitos dos fármacos , Masculino , SARS-CoV-2/efeitos dos fármacos , Feminino , Pessoa de Meia-Idade , Método Duplo-Cego , Antivirais/uso terapêutico , Antivirais/farmacologia , Adulto , COVID-19/virologia , Ivermectina/uso terapêutico , Ivermectina/farmacologia , Fluvoxamina/uso terapêutico , Fluvoxamina/farmacologia , Idoso
9.
Pflugers Arch ; 476(2): 211-227, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37979051

RESUMO

Inflammatory airway diseases like cystic fibrosis, asthma and COVID-19 are characterized by high levels of pulmonary cytokines. Two well-established antiparasitic drugs, niclosamide and ivermectin, are intensively discussed for the treatment of viral inflammatory airway infections. Here, we examined these repurposed drugs with respect to their anti-inflammatory effects in airways in vivo and in vitro. Niclosamide reduced mucus content, eosinophilic infiltration and cell death in asthmatic mouse lungs in vivo and inhibited release of interleukins in the two differentiated airway epithelial cell lines CFBE and BCi-NS1.1 in vitro. Cytokine release was also inhibited by the knockdown of the Ca2+-activated Cl- channel anoctamin 1 (ANO1, TMEM16A) and the phospholipid scramblase anoctamin 6 (ANO6, TMEM16F), which have previously been shown to affect intracellular Ca2+ levels near the plasma membrane and to facilitate exocytosis. At concentrations around 200 nM, niclosamide inhibited inflammation, lowered intracellular Ca2+, acidified cytosolic pH and blocked activation of ANO1 and ANO6. It is suggested that niclosamide brings about its anti-inflammatory effects at least in part by inhibiting ANO1 and ANO6, and by lowering intracellular Ca2+ levels. In contrast to niclosamide, 1 µM ivermectin did not exert any of the effects described for niclosamide. The present data suggest niclosamide as an effective anti-inflammatory treatment in CF, asthma, and COVID-19, in addition to its previously reported antiviral effects. It has an advantageous concentration-response relationship and is known to be well tolerated.


Assuntos
Asma , COVID-19 , Camundongos , Animais , Anoctamina-1/metabolismo , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Niclosamida/farmacologia , Niclosamida/uso terapêutico , Anoctaminas/metabolismo , Pulmão/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Cálcio/metabolismo , Inflamação/tratamento farmacológico , Anti-Inflamatórios , Canais de Cloreto/metabolismo
10.
J Neurosci Res ; 102(1): e25254, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37814994

RESUMO

Ivermectin (IVM), a semi-synthetic macrolide parasiticide, has demonstrated considerable effectiveness in combating internal and external parasites, particularly nematodes and arthropods. Its remarkable ability to control parasites has earned it significant recognition, culminating in Satoshi Omura and William C. Campbell's receipt of the 2015 Nobel Prize in Physiology or Medicine for their contributions to the development of IVM. In recent years, investigations have revealed that IVM possesses antitumor properties. It can suppress the growth of various cancer cells, including glioma, through a multitude of mechanisms such as selective targeting of tumor-specific proteins, inducing programmed cell death, and modulation of tumor-related signaling pathways. Hence, IVM holds tremendous potential as a novel anticancer drug. This review seeks to provide an overview of the underlying mechanisms that enable IVM's capacity to suppress glioma. Furthermore, it aims to elucidate the challenges and prospects associated with utilizing IVM as a new anticancer agent.


Assuntos
Antineoplásicos , Glioma , Humanos , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Ivermectina/história , Glioma/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Prêmio Nobel , Apoptose
11.
PLoS Pathog ; 18(6): e1010545, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35696434

RESUMO

The antiparasitic drug ivermectin plays an essential role in human and animal health globally. However, ivermectin resistance is widespread in veterinary helminths and there are growing concerns of sub-optimal responses to treatment in related helminths of humans. Despite decades of research, the genetic mechanisms underlying ivermectin resistance are poorly understood in parasitic helminths. This reflects significant uncertainty regarding the mode of action of ivermectin in parasitic helminths, and the genetic complexity of these organisms; parasitic helminths have large, rapidly evolving genomes and differences in evolutionary history and genetic background can confound comparisons between resistant and susceptible populations. We undertook a controlled genetic cross of a multi-drug resistant and a susceptible reference isolate of Haemonchus contortus, an economically important gastrointestinal nematode of sheep, and ivermectin-selected the F2 population for comparison with an untreated F2 control. RNA-seq analyses of male and female adults of all populations identified high transcriptomic differentiation between parental isolates, which was significantly reduced in the F2, allowing differences associated specifically with ivermectin resistance to be identified. In all resistant populations, there was constitutive upregulation of a single gene, HCON_00155390:cky-1, a putative pharyngeal-expressed transcription factor, in a narrow locus on chromosome V previously shown to be under ivermectin selection. In addition, we detected sex-specific differences in gene expression between resistant and susceptible populations, including constitutive upregulation of a P-glycoprotein, HCON_00162780:pgp-11, in resistant males only. After ivermectin selection, we identified differential expression of genes with roles in neuronal function and chloride homeostasis, which is consistent with an adaptive response to ivermectin-induced hyperpolarisation of neuromuscular cells. Overall, we show the utility of a genetic cross to identify differences in gene expression that are specific to ivermectin selection and provide a framework to better understand ivermectin resistance and response to treatment in parasitic helminths.


Assuntos
Anti-Helmínticos , Haemonchus , Nematoides , Animais , Anti-Helmínticos/farmacologia , Cloretos/metabolismo , Cloretos/farmacologia , Resistência a Medicamentos/genética , Feminino , Homeostase , Ivermectina/metabolismo , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Masculino , Nematoides/genética , Plasticidade Neuronal , Ovinos/genética , Transcriptoma
12.
Crit Rev Toxicol ; 54(6): 345-358, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38860720

RESUMO

During the COVID-19 pandemic, several drugs were repositioned and combined to quickly find a way to mitigate the effects of the infection. However, the adverse effects of these combinations on the gastrointestinal tract are unknown. We aimed investigate whether Hydroxychloroquine (HD), Azithromycin (AZ), and Ivermectin (IV) used in combination for the treatment of COVID-19, can lead to the development of gastrointestinal disorders. This is a systematic review and network meta-analysis conducted using Stata and Revman software, respectively. The protocol was registered with PROSPERO (CRD42023372802). A search of clinical trials in Cochrane Library databases, Embase, Web of Science, Lilacs, PubMed, Scopus and Clinicaltrials.gov conducted on November 26, 2023. The eligibility of the studies was assessed based on PICO criteria, including trials that compared different treatments and control group. The analysis of the quality of the evidence was carried out according to the GRADE. Six trials involving 1,686 COVID-19 patients were included. No trials on the association of HD or AZ with IV met the inclusion criteria, only studies on the association between HD and AZ were included. Nausea, vomiting, diarrhea, abdominal pain and increased transaminases were related. The symptoms of vomiting and nausea were evaluated through a network meta-analysis, while the symptom of abdominal pain was evaluated through a meta-analysis. No significant associations with these symptoms were observed for HD, AZ, or their combination, compared to control. Low heterogeneity and absence of inconsistency in indirect and direct comparisons were noted. Limitations included small sample sizes, varied drug dosages, and potential publication bias during the pandemic peak. This review unveils that there are no associations between gastrointestinal adverse effects and the combined treatment of HD with AZ in the management of COVID-19, as compared to either the use of a control group or the administration of the drugs individually, on the other hand, highlighting the very low or low certainty of evidence for the evaluated outcomes. To accurately conclude the absence of side effects, further high-quality randomized studies are needed.


Assuntos
Azitromicina , Tratamento Farmacológico da COVID-19 , Quimioterapia Combinada , Gastroenteropatias , Hidroxicloroquina , Metanálise em Rede , SARS-CoV-2 , Azitromicina/uso terapêutico , Azitromicina/efeitos adversos , Humanos , Hidroxicloroquina/uso terapêutico , Hidroxicloroquina/efeitos adversos , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/epidemiologia , COVID-19 , Ivermectina/uso terapêutico , Ivermectina/efeitos adversos , Antibacterianos/uso terapêutico , Antibacterianos/efeitos adversos , Antivirais/uso terapêutico , Antivirais/efeitos adversos
13.
Arch Microbiol ; 206(2): 78, 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38277061

RESUMO

Varicellovirus bovinealpha 1 (formerly bovine alphaherpesvirus type 1, BoAHV-1) is associated with several syndromes in cattle, including respiratory disease and is one of the main agents involved in the bovine respiratory disease complex (BRDC). Its infectious cycle is characterized by latent infections with sporadic virus reactivation and transmission. Although the acute disease can be prevented by the use of vaccines, specific therapeutic measures are not available. Ivermectin (IVM) is a semi-synthetic avermectin with a broad-spectrum antiparasitic activity, which has previously shown to have potential as an antiviral drug. In this study, IVM antiviral activity against BoAHV-1 was characterized in two cell lines (MDBK [Madin Darby bovine kidney] and BT [bovine turbinate]), including the measurement of intracellular drug accumulation within virus-infected cells. IVM antiviral activity was assessed at three different drug concentrations (1.25, 2.5 and 5 µM) after incubation for 24, 48 and 72 h. Slight cytotoxicity was only observed with 5 µM IVM. Even the lowest IVM dose was able to induce a significant reduction in virus titers in both cell lines. These findings indicate that the antiviral effects of IVM were evident in our experimental model within the range of concentrations achievable through therapeutic in vivo administration. Consequently, additional in vivo trials are necessary to validate the potential utility of these results in effectively managing BoAHV-1 in infected cattle.


Assuntos
Ivermectina , Varicellovirus , Animais , Bovinos , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Antiparasitários/farmacologia , Antiparasitários/uso terapêutico , Antivirais/farmacologia
14.
Malar J ; 23(1): 54, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38383367

RESUMO

BACKGROUND: Mass Drug Administration (MDA) has become a mainstay for the control of several diseases over the last two decades. Successful implementation of MDA programmes requires community participation and can be threatened by systematic non-participation. Such concerns are particularly pertinent for MDA programmes against malaria, as they require multi-day treatment over several consecutive months. Factors associated with non-participation to the MDA campaign with ivermectin (IVM) and dihydroartemisinin-piperaquine (DHP) implemented within the MASSIV cluster randomized trial were determined. METHODS: Coverage data was extracted from the MASSIV trial study database, with every datapoint being a directly observed therapy (DOT). A complete month of MDA was classified as receiving all three daily doses of treatment. For both ivermectin and DHP, ordinal logistic regression was used to identify individual and household level variables associated with non-participation. RESULTS: For ivermectin, 51.5% of eligible participants received all 3 months of treatment while 30.7% received either one or two complete months. For DHP, 56.7% of eligible participants received all 3 months of treatment and 30.5% received either one or two complete months. Children aged 5-15 years and adults aged more than 50 years were more likely to receive at least one complete month of MDA than working age adults, both for ivermectin (aOR 4.3, 95% CI 3.51-5.28 and aOR of 2.26, 95% CI 1.75-2.95) and DHP (aOR 2.47, 95%CI 2.02-3.02 and aOR 1.33, 95%CI 1.01-1.35), respectively. Members of households where the head received a complete month of MDA were more likely to themselves have received a complete month of MDA, both for ivermectin (aOR 1.71, 95%CI 1.35-2.14) and for DHP (aOR 1.64, 95%CI 1.33-2.04). CONCLUSION: Personal and household-level variables were associated with participation in the MDA programme for malaria control. Specific strategies to (increase participation amongst some groups may be important to ensure maximum impact of MDA strategies in achieving malaria elimination. TRIAL REGISTRATION: The MASSIV trial is registered under NCT03576313.


Assuntos
Antimaláricos , Artemisininas , Malária , Piperazinas , Quinolinas , Adulto , Criança , Humanos , Ivermectina/uso terapêutico , Malária/prevenção & controle , Malária/tratamento farmacológico , Administração Massiva de Medicamentos , Quinolinas/uso terapêutico , Fatores de Risco , Pré-Escolar , Adolescente , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto
15.
BMC Infect Dis ; 24(1): 719, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39039459

RESUMO

BACKGROUND: Evidence on ivermectin as a treatment for Covid-19 is controversial. A Cochrane review concluded that the efficacy and safety of ivermectin is uncertain (evidence up to April 2022) and WHO recommended its use only in the setting of clinical trials. This study aimed to assess the efficacy and safety of oral ivermectin in hospitalized patients with mild to moderate Covid-19. TRIAL DESIGN AND METHODS: A double-blind, randomized placebo-controlled clinical trial was conducted among RT-PCR-confirmed, adults, hospitalised within the first four days of symptoms. Patients received oral ivermectin 24 mg or placebo daily for five days. RT-PCR was repeated on days five and ten. Clinical progression was monitored using the World Health Organization Clinical Progression Scale. Serum ivermectin levels were measured on days three, five, and seven. The primary outcome was the difference in the viral load between day zero and ten in the two groups. RESULTS: Out of 1699 patients screened, 249 underwent randomization and 127 received ivermectin, and 122 placebo. D10 median viral load for E gene (IQR) was 2,000 copies/mL (100 - 20,500) with ivermectin (n = 80) and 4,100 copies/mL (1,000-65,600) with placebo (n = 81, p = 0.028), per protocol analysis. The difference in Log viral load between day zero and ten between ivermectin and placebo was 3.72 and 2.97 respectively (p = 0.022). There was no significant difference in the WHO clinical progression scale or the adverse effects. Ivermectin blood levels taken before or with meals were not significantly different. Only 7 and 17 patients achieved blood levels above 160ng/ML and 100ng/ML respectively and they did not achieve a significantly lower viral load. CONCLUSION: Although ivermectin resulted in statistically significant lower viral load in patients with mild to moderate Covid-19, it had no significant effect on clinical symptoms. TRIAL REGISTRATION NUMBER: SLCTR/2021/020, Sri Lanka Clinical Trials Registry. 19/07/2021.


Assuntos
Tratamento Farmacológico da COVID-19 , Ivermectina , SARS-CoV-2 , Carga Viral , Humanos , Ivermectina/administração & dosagem , Ivermectina/efeitos adversos , Ivermectina/uso terapêutico , Método Duplo-Cego , Masculino , Feminino , Pessoa de Meia-Idade , Administração Oral , Carga Viral/efeitos dos fármacos , Adulto , SARS-CoV-2/genética , SARS-CoV-2/efeitos dos fármacos , Resultado do Tratamento , COVID-19/virologia , Idoso , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Antivirais/efeitos adversos
16.
Eur J Pediatr ; 183(6): 2527-2536, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38536458

RESUMO

Scabies is a human ectoparasitosis caused by Sarcoptes scabei var. hominis. World-wide around 300 million patients are affected. Infants and children have the highest incidence rates. Poverty and overcrowding are social factors contributing to a higher risk of transmission and treatment failure. The leading symptom of the infestation is itch. Complications are bacterial infections that are responsible for mortality. Diagnosis is clinical. Non-invasive imaging technologies like dermoscopy can be used. Polymerase chain reaction (PCR) is less sensitive and specific than microscopy of skin scrapings. Treatment of choice is topical permethrin 5%. Ivermectin is the only oral drug FDA-approved for scabies. It should be used in cases non-responsive to topical therapy and in case of high number of infested patients in addition to topical therapy. Pseudo-resistance to treatment is not uncommon. New drugs are on the horizon. What is Known: • Pruritus is the leading symptom causing sleep disturbances and scratching with the risk of secondary bacterial infections. • Treatment failure is related to inappropriate application of topical drugs and asymptomatic family members. What is New: • COVID-19 pandemic and migration are contributing to an increased incidence of scabies. • New compounds to treat scabies are on the horizon.


Assuntos
COVID-19 , Escabiose , Humanos , Escabiose/diagnóstico , Escabiose/tratamento farmacológico , Criança , Lactente , COVID-19/complicações , COVID-19/epidemiologia , COVID-19/diagnóstico , Permetrina/uso terapêutico , Ivermectina/uso terapêutico , Inseticidas/uso terapêutico , Antiparasitários/uso terapêutico , Pré-Escolar
17.
Exp Parasitol ; 256: 108670, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38092297

RESUMO

Ivermectin (IVM) resistance in parasitic nematodes such as Haemonchus contortus has spurred a search for substances that help to recover its efficacy. One potential agent is the natural product curcumin (CUR). In this study, CUR was combined with polyvinylpyrrolidone (PVP) (CUR/PVP) to improve its solubility and biological applicability. This study determined the effect of CUR preincubation on the effective concentration 50% (EC50) of IVM in three H. contortus isolates with different susceptibilities to IVM. The IVM EC50 was determined for three H. contortus isolates with different IVM susceptibilities using the larval migration inhibition (LMI) test. The three isolates were (i) PARAISO (IVM resistant), (ii) FMVZ-UADY (IVM susceptible), and (iii) CENID-SAI INIFAP (reference IVM susceptible). The L3 of each isolate were preincubated for 3 h with one of three concentrations of CUR (µg curcumin/mL): CONC-1 (3.67), CONC-2 (5.67), or CONC-3 (8.48). Corresponding controls were performed without CUR. The EC50 of IVM was determined for each isolate after they were exposed to the different CUR concentrations. The EC50 of IVM differed between the isolates PARAISO > FMVZ-UADY > CENID-SAI INIFAP (P < 0.05). The CUR preincubation at CONC-1 did not decrease the EC50 of IVM for any of the three isolates, suggesting a hormetic effect. By contrast, CUR preincubation at CONC-2 or CONC-3 decreased the IVM EC50 for the PARAISO isolate (P < 0.05) compared with the reference isolate and reduced the EC50 of IVM for the FMVZ-UADY and CENID-SAI INIFAP isolates below the EC50 for the CENID-SAI INIFAP isolate without CUR preincubation. In conclusion, preincubation of H. contortus L3 with CUR reduced the EC50 of IVM for field isolates classified as resistant and susceptible to IVM. The CUR preincubation reduced the IVM resistance factor in the different isolates tested.


Assuntos
Anti-Helmínticos , Curcumina , Hemoncose , Haemonchus , Animais , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Anti-Helmínticos/farmacologia , Anti-Helmínticos/uso terapêutico , Curcumina/farmacologia , Curcumina/uso terapêutico , Povidona/farmacologia , Povidona/uso terapêutico , Resistência a Medicamentos , Larva , Hemoncose/tratamento farmacológico , Hemoncose/veterinária
18.
Exp Parasitol ; 262: 108775, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38735518

RESUMO

The limited activity of the traditional medications against T. spiralis encysted larvae handicaps complete cure of trichinellosis till now due to decreased permeability and absorption through tissues. MOX is listed worldwide for prevention and treatment of several internal and external nematodes. Consequently, the aim of this work was to investigate the effect of moxidectin versus ivermectin on experimental acute and chronic trichinellosis and to illuminate the potential mechanisms of their effects. 105 Mice were divided into four groups; Group I: Uninfected healthy control; Group II: Infected untreated control; Group III: Infected and treated with IVM and Group IV: Infected and treated with MOX. The groups (II, III and IV) were later subdivided equally into three subgroups (a, b, and c) according to the stage of treatment. Parasitological counting of adults and larvae besides immune-histopathological examination of intestines and muscles were done. Results exhibited that both IVM and MOX succeeded in reducing adults and larvae counts with higher potential of MOX in both intestinal and muscle phase. The preeminence of MOX was indicated by decreased inflammation, a significant reduction in the microvascular density (CD31 immunostaining) as well as a reduction in the percentage of fibroblast activation protein (FAP) immunostaining in muscle tissues. Accordingly, the current work recommends moxidectin as an innovative treatment for trichinellosis.


Assuntos
Ivermectina , Macrolídeos , Triquinelose , Animais , Triquinelose/tratamento farmacológico , Triquinelose/prevenção & controle , Triquinelose/parasitologia , Macrolídeos/uso terapêutico , Macrolídeos/farmacologia , Camundongos , Ivermectina/uso terapêutico , Ivermectina/farmacologia , Doença Crônica , Trichinella spiralis/efeitos dos fármacos , Doença Aguda , Larva/efeitos dos fármacos , Feminino , Masculino , Anti-Helmínticos/uso terapêutico , Anti-Helmínticos/farmacologia , Músculo Esquelético/parasitologia , Músculo Esquelético/patologia
19.
Exp Parasitol ; 265: 108823, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39187057

RESUMO

Trichinosis is a common parasitic disease that affects the striated skeletal muscles, causing apoptotic and degenerative changes associated with myogenin expression in the affected myocytes. Hence, this study aimed to assess the ameliorative effects of stem cells and atorvastatin added to ivermectin on the infected myocytes during the muscular phase of murine trichinosis. 120 laboratory Swiss albino male mice were divided into 10 groups, and each group was subdivided into intestinal and muscular phases (each n = 6); uninfected control; untreated infected control; infected received ivermectin monotherapy; infected received atorvastatin monotherapy; infected received stem cells monotherapy; infected received ivermectin and atorvastatin dual therapy; infected received ivermectin and stem cells dual therapy; infected received atorvastatin and stem cells dual therapy; infected received ivermectin 0.2, atorvastatin 40, and stem cells triple therapy; and infected received ivermectin 0.1, atorvastatin 20, and stem cells triple therapy. Intestinal phase mice were sacrificed on the 5th day post-infection, while those of the muscular phase were sacrificed on the 35th day post-infection. Parasitological, histopathological, ultrastructural, histochemical, biochemical, and myogenin gene expression assessments were performed. The results revealed that mice that received ivermectin, atorvastatin, and stem cell triple therapies showed the maximum reduction in the adult worm and larvae burden, marked improvement in the underlying muscular degenerative changes (as was noticed by histopathological, ultrastructural, and histochemical Feulgen stain assessment), lower biochemical levels of serum NK-κB and tissue NO, and lower myogenin expression. Accordingly, the combination of stem cells, atorvastatin, and ivermectin affords a potential synergistic activity against trichinosis with considerable healing of the underlying degenerative sequel.


Assuntos
Apoptose , Atorvastatina , Ivermectina , Miogenina , Triquinelose , Animais , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , Masculino , Camundongos , Ivermectina/farmacologia , Ivermectina/uso terapêutico , Triquinelose/tratamento farmacológico , Triquinelose/parasitologia , Apoptose/efeitos dos fármacos , Miogenina/genética , Miogenina/metabolismo , Músculo Esquelético/parasitologia , Músculo Esquelético/patologia , Músculo Esquelético/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Transplante de Células-Tronco , Trichinella spiralis/genética , Trichinella spiralis/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
20.
Parasitol Res ; 123(4): 180, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38592544

RESUMO

Because of recent reports of praziquantel resistance in schistosome infections, there have been suggestions to employ ivermectin as a possible alternative, especially as its chemical composition is different from that of praziquantel, so cross-resistance is not expected. In order to ascertain possible damage and elimination of worms, we used ivermectin by oral gavage in infected mice, at a high dose (30.1 mg/kg, bordering toxicity). We also tested the efficacy of the drug at various times postinfection (PI), to check on possible effect on young and mature stages of the parasites. Thus, we treated mice on days 21 and 22 or on days 41 and 42 and even on days 21, 22, 41, and 42 PI. None of the treatment regimens resulted in cure rates or signs of lessened pathology in the mice. We also compared the effect of ivermectin to that of artemisone, an artemisinin derivative which had served us in the past as an effective anti-schistosome drug, and there was a stark difference in the artemisone's efficacy compared to that of ivermectin; while ivermectin was not effective, artemisone eliminated most of the worms, prevented egg production and granulomatous inflammatory response. We assume that the reported lack of activity of ivermectin, in comparison with praziquantel and artemisinins, originates from the difference in their mode of action. In wake of our results, we suggest that ivermectin is not a suitable drug for treatment of schistosomiasis.


Assuntos
Artemisininas , Schistosomatidae , Esquistossomose , Animais , Camundongos , Praziquantel/uso terapêutico , Ivermectina/uso terapêutico , Esquistossomose/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA