Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.454
Filter
Add more filters

Publication year range
1.
Cell ; 163(6): 1297-300, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638061

ABSTRACT

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.


Subject(s)
Drug Discovery , Nobel Prize , Parasitic Diseases/drug therapy , Artemisinins/chemistry , Artemisinins/therapeutic use , History of Medicine , History, 21st Century , Ivermectin/analogs & derivatives , Ivermectin/chemistry , Ivermectin/therapeutic use , Physiology/history
2.
PLoS Pathog ; 20(5): e1012245, 2024 May.
Article in English | MEDLINE | ID: mdl-38768235

ABSTRACT

Albendazole (a benzimidazole) and ivermectin (a macrocyclic lactone) are the two most commonly co-administered anthelmintic drugs in mass-drug administration programs worldwide. Despite emerging resistance, we do not fully understand the mechanisms of resistance to these drugs nor the consequences of delivering them in combination. Albendazole resistance has primarily been attributed to variation in the drug target, a beta-tubulin gene. Ivermectin targets glutamate-gated chloride channels (GluCls), but it is unknown whether GluCl genes are involved in ivermectin resistance in nature. Using Caenorhabditis elegans, we defined the fitness costs associated with loss of the drug target genes singly or in combinations of the genes that encode GluCl subunits. We quantified the loss-of-function effects on three traits: (i) multi-generational competitive fitness, (ii) fecundity, and (iii) development. In competitive fitness and development assays, we found that a deletion of the beta-tubulin gene ben-1 conferred albendazole resistance, but ivermectin resistance required the loss of two GluCl genes (avr-14 and avr-15). The fecundity assays revealed that loss of ben-1 did not provide any fitness benefit in albendazole conditions and that no GluCl deletion mutants were resistant to ivermectin. Next, we searched for evidence of multi-drug resistance across the three traits. Loss of ben-1 did not confer resistance to ivermectin, nor did loss of any single GluCl subunit or combination confer resistance to albendazole. Finally, we assessed the development of 124 C. elegans wild strains across six benzimidazoles and seven macrocyclic lactones to identify evidence of multi-drug resistance between the two drug classes and found a strong phenotypic correlation within a drug class but not across drug classes. Because each gene affects various aspects of nematode physiology, these results suggest that it is necessary to assess multiple fitness traits to evaluate how each gene contributes to anthelmintic resistance.


Subject(s)
Anthelmintics , Caenorhabditis elegans , Drug Resistance , Ivermectin , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/drug effects , Anthelmintics/pharmacology , Drug Resistance/genetics , Ivermectin/pharmacology , Alleles , Genetic Fitness/drug effects , Albendazole/pharmacology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Tubulin/genetics , Tubulin/metabolism , Selection, Genetic
3.
Bioessays ; 46(1): e2300143, 2024 01.
Article in English | MEDLINE | ID: mdl-37985957

ABSTRACT

Modeling metastasis in animal systems has been an important focus for developing cancer therapeutics. Xenopus laevis is a well-established model, known for its use in identifying genetic mechanisms underlying diseases and disorders in humans. Prior literature has suggested that the drug, ivermectin, can be used in Xenopus to induce melanocytes to convert into a metastatic melanoma-like state, and thus could be ideal for testing possible melanoma therapies in vivo. However, there are notable inconsistencies between ivermectin studies in Xenopus and the application of ivermectin in mammalian systems, that are relevant to cancer and melanoma research. In this review, we examine the ivermectin-induced phenotypes in Xenopus, and we explore the current uses of ivermectin in human research. We conclude that while ivermectin may be a useful drug for many biomedical purposes, it is not ideal to induce a metastatic melanocyte phenotype in Xenopus for testing the effects of potential melanoma therapeutics.


Subject(s)
Melanoma , Animals , Humans , Melanoma/drug therapy , Melanoma/genetics , Xenopus laevis , Ivermectin/pharmacology , Melanocytes/pathology , Mammals
4.
Proc Natl Acad Sci U S A ; 120(7): e2216179120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36753464

ABSTRACT

In the United States, liberals and conservatives disagree about facts. To what extent does expertise attenuate these disagreements? To study this question, we compare the polarization of beliefs about COVID-19 treatments among laypeople and critical care physicians. We find that political ideology predicts both groups' beliefs about a range of COVID-19 treatments. These associations persist after controlling for a rich set of covariates, including local politics. We study two potential explanations: a) that partisans are exposed to different information and b) that they interpret the same information in different ways, finding evidence for both. Polarization is driven by preferences for partisan cable news but not by exposure to scientific research. Using a set of embedded experiments, we demonstrate that partisans perceive scientific evidence differently when it pertains to a politicized treatment (ivermectin), relative to when the treatment is not identified. These results highlight the extent to which political ideology is increasingly relevant for understanding beliefs, even among expert decision makers such as physicians.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Humans , United States , COVID-19/epidemiology , COVID-19/therapy , Politics , Critical Care , Ivermectin
5.
N Engl J Med ; 386(18): 1721-1731, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35353979

ABSTRACT

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.).


Subject(s)
Anti-Infective Agents , COVID-19 Drug Treatment , Ivermectin , Adult , Ambulatory Care , Anti-Infective Agents/adverse effects , Anti-Infective Agents/therapeutic use , Bayes Theorem , Double-Blind Method , Hospitalization , Humans , Ivermectin/adverse effects , Ivermectin/therapeutic use , SARS-CoV-2 , Treatment Outcome
6.
N Engl J Med ; 387(7): 599-610, 2022 08 18.
Article in English | MEDLINE | ID: mdl-36070710

ABSTRACT

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.).


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Fluvoxamine , Ivermectin , Metformin , Adult , Aged , Aged, 80 and over , COVID-19/complications , COVID-19 Vaccines , Double-Blind Method , Female , Fluvoxamine/therapeutic use , Humans , Hypoxia/etiology , Ivermectin/therapeutic use , Male , Metformin/therapeutic use , Middle Aged , Obesity/complications , Overweight/complications , Pregnancy , Pregnancy Complications, Infectious/drug therapy , SARS-CoV-2
7.
PLoS Pathog ; 19(3): e1011188, 2023 03.
Article in English | MEDLINE | ID: mdl-36917600

ABSTRACT

Sea louse ectoparasitosis is a major threat to fish aquaculture. Avermectins such as ivermectin and emamectin have been effectively used against sea louse infestation, but the emergence of resistance has limited their use. A better understanding of the molecular targets of avermectins is essential to the development of novel treatment strategies or new, more effective drugs. Avermectins are known to act by inhibiting neurotransmission through allosteric activation of glutamate-gated chloride channels (GluCls). We have investigated the GluCl subunit present in Caligus rogercresseyi, a sea louse affecting aquaculture in the Southern hemisphere. We identify four new subunits, CrGluCl-B to CrGluCl-E, and characterise them functionally. CrGluCl-A (previously reported as CrGluClα), CrGluCl-B and CrGluCl-C all function as glutamate channel receptors with different sensitivities to the agonist, but in contrast to subunit -A and -C, CrGluCl-B is not activated by ivermectin but is rather antagonised by the drug. CrGluCl-D channel appears active in the absence of any stimulation by glutamate or ivermectin and CrGluCl-E does not exhibit any activity. Notably, the expression of CrGluCl-B with either -A or -C subunits gives rise to receptors unresponsive to ivermectin and showing altered response to glutamate, suggesting that coexpression has led to the preferential formation of heteromers to which the presence of CrGluCl-B confers the property of ivermectin-activation refractoriness. Furthermore, there was evidence for heteromer formation with novel properties only when coexpressing pairs E/C and D/B CrGluCl subtypes. Site-directed mutagenesis shows that three transmembrane domain residues contribute to the lack of activation by ivermectin, most crucially Gln 15' in M2, with mutation Q15'T (the residue present in ivermectin-activated subunits A and C) conferring ivermectin activation to CrGluCl-B. The differential response to avermectin of these Caligus rogercresseyi GluClsubunits, which are highly conserved in the Northern hemisphere sea louse Lepeophtheirus salmonis, could have an influence on the response of these parasites to treatment with macrocyclic lactones. They could serve as molecular markers to assess susceptibility to existing treatments and might be useful molecular targets in the search for novel antiparasitic drugs.


Subject(s)
Copepoda , Parasites , Phthiraptera , Animals , Ivermectin/pharmacology , Ivermectin/metabolism , Phthiraptera/metabolism , Parasites/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Glutamic Acid/pharmacology
8.
Proc Natl Acad Sci U S A ; 119(34): e2111932119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969762

ABSTRACT

Glutamate-gated chloride channels (GluCls) are unique to invertebrates and are targeted by macrocyclic lactones. In this study, we cloned an AVR-14B GluCl subunit from adult Brugia malayi, a causative agent of lymphatic filariasis in humans. To elucidate this channel's pharmacological properties, we used Xenopus laevis oocytes for expression and performed two-electrode voltage-clamp electrophysiology. The receptor was gated by the natural ligand L-glutamate (effective concentration, 50% [EC50] = 0.4 mM) and ivermectin (IVM; EC50 = 1.8 nM). We also characterized the effects of nodulisporic acid (NA) on Bma-AVR-14B and NA-produced dual effects on the receptor as an agonist and a type II positive allosteric modulator. Here we report characterization of the complex activity of NA on a nematode GluCl. Bma-AVR-14B demonstrated some unique pharmacological characteristics. IVM did not produce potentiation of L-glutamate-mediated responses but instead, reduced the channel's sensitivity for the ligand. Further electrophysiological exploration showed that IVM (at a moderate concentration of 0.1 nM) functioned as an inhibitor of both agonist and positive allosteric modulatory effects of NA. This suggests that IVM and NA share a complex interaction. The pharmacological properties of Bma-AVR-14B indicate that the channel is an important target of IVM and NA. In addition, the unique electrophysiological characteristics of Bma-AVR-14B could explain the observed variation in drug sensitivities of various nematode parasites. We have also shown the inhibitory effects of IVM and NA on adult worm motility using Worminator. RNA interference (RNAi) knockdown suggests that AVR-14 plays a role in influencing locomotion in B. malayi.


Subject(s)
Brugia malayi , Chloride Channels , Indoles , Animals , Brugia malayi/drug effects , Brugia malayi/genetics , Brugia malayi/metabolism , Chloride Channels/drug effects , Chloride Channels/genetics , Chloride Channels/metabolism , Glutamic Acid/metabolism , Indoles/pharmacology , Ivermectin/pharmacology , Ligands
9.
Clin Microbiol Rev ; 36(4): e0003323, 2023 12 20.
Article in English | MEDLINE | ID: mdl-37937980

ABSTRACT

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.


Subject(s)
Strongyloides stercoralis , Strongyloidiasis , Animals , Child , Humans , Strongyloidiasis/diagnosis , Strongyloidiasis/drug therapy , Strongyloidiasis/parasitology , Ivermectin/pharmacology , Ivermectin/therapeutic use , Immunocompromised Host , Immunosuppression Therapy
10.
J Cell Mol Med ; 28(14): e18539, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39046364

ABSTRACT

Chronic myeloid leukaemia (CML) is a haematological malignancy characterized by the constitutive tyrosine kinase activity of the BCR-ABL1 fusion protein. Flumatinib, a second-generation tyrosine kinase inhibitor, has exhibited superior clinical efficacy compared to its precursor, imatinib. However, with increased clinical use, resistance to flumatinib has emerged as a significant challenge. To investigate the mechanisms of flumatinib resistance in CML, we induced the human CML cell line K562 using a flumatinib concentration gradient method in vitro, successfully establishing a flumatinib-resistant K562/FLM cell line. This cell line exhibited cross-resistance to imatinib and doxorubicin, but remained sensitive to the antiparasitic agent ivermectin, which possesses antitumoural effects. Through cellular experimentation, we explored the resistance mechanisms, which indicated that K562/FLM cells evade flumatinib cytotoxicity by enhancing autophagy, increasing the expression of membrane transport proteins, particularly P-glycoprotein, ABCC1 and ABCC4, as well as enhancing phosphorylation of p-EGFR, p-ERK and p-STAT3 proteins. Moreover, it was found that ivermectin effectively suppressed the expression of autophagy and transport proteins in K562/FLM cells, reduced the activity of the aforementioned phosphoproteins, and promoted apoptotic cell death. Collectively, the increased autophagy, higher expression of drug-efflux proteins and hyperactivation of the EGFR/ERK/STAT3 signalling pathway were identified as pivotal elements promoting resistance to flumatinib. The significant effects of ivermectin might offer a novel therapeutic strategy to overcome flumatinib resistance and optimize the treatment outcomes of CML.


Subject(s)
Drug Resistance, Neoplasm , Ivermectin , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Drug Resistance, Neoplasm/drug effects , Ivermectin/pharmacology , K562 Cells , Autophagy/drug effects , Apoptosis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Imatinib Mesylate/pharmacology , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Cell Line, Tumor
11.
Clin Infect Dis ; 78(Supplement_2): S138-S145, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662693

ABSTRACT

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.


Subject(s)
Ivermectin , Macrolides , Onchocerciasis , Macrolides/therapeutic use , Macrolides/economics , Macrolides/administration & dosage , Onchocerciasis/drug therapy , Onchocerciasis/prevention & control , Onchocerciasis/economics , Onchocerciasis/epidemiology , Humans , Ivermectin/economics , Ivermectin/therapeutic use , Ivermectin/administration & dosage , Mass Drug Administration/economics , Disease Eradication/economics , Cost-Benefit Analysis
12.
Clin Infect Dis ; 78(Supplement_2): S131-S137, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662696

ABSTRACT

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.


Subject(s)
Albendazole , Diethylcarbamazine , Drug Therapy, Combination , Elephantiasis, Filarial , Filaricides , Ivermectin , Mass Drug Administration , Microfilariae , Elephantiasis, Filarial/drug therapy , Elephantiasis, Filarial/epidemiology , Elephantiasis, Filarial/prevention & control , Humans , Albendazole/therapeutic use , Albendazole/administration & dosage , Filaricides/therapeutic use , Diethylcarbamazine/therapeutic use , Diethylcarbamazine/administration & dosage , Ivermectin/therapeutic use , Ivermectin/administration & dosage , Animals , India/epidemiology , Microfilariae/drug effects , Adult , Prevalence
13.
Clin Infect Dis ; 78(Supplement_2): S93-S100, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38662701

ABSTRACT

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.


Subject(s)
Albendazole , Elephantiasis, Filarial , Filaricides , Ivermectin , Mass Drug Administration , Elephantiasis, Filarial/drug therapy , Elephantiasis, Filarial/prevention & control , Elephantiasis, Filarial/epidemiology , Elephantiasis, Filarial/transmission , Humans , Animals , Filaricides/therapeutic use , Filaricides/administration & dosage , Albendazole/administration & dosage , Albendazole/therapeutic use , Ivermectin/administration & dosage , Ivermectin/therapeutic use , Prevalence , Anopheles/parasitology , Disease Eradication/methods , Wuchereria bancrofti/drug effects , Diethylcarbamazine/administration & dosage , Diethylcarbamazine/therapeutic use , Drug Therapy, Combination
14.
Pflugers Arch ; 476(2): 211-227, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37979051

ABSTRACT

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.


Subject(s)
Asthma , COVID-19 , Mice , Animals , Anoctamin-1/metabolism , Ivermectin/pharmacology , Ivermectin/therapeutic use , Niclosamide/pharmacology , Niclosamide/therapeutic use , Anoctamins/metabolism , Lung/metabolism , Phospholipid Transfer Proteins/metabolism , Calcium/metabolism , Inflammation/drug therapy , Anti-Inflammatory Agents , Chloride Channels/metabolism
15.
J Neurosci Res ; 102(1): e25254, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37814994

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Glioma , Humans , Ivermectin/pharmacology , Ivermectin/therapeutic use , Ivermectin/history , Glioma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Nobel Prize , Apoptosis
16.
PLoS Pathog ; 18(6): e1010545, 2022 06.
Article in English | MEDLINE | ID: mdl-35696434

ABSTRACT

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.


Subject(s)
Anthelmintics , Haemonchus , Nematoda , Animals , Anthelmintics/pharmacology , Chlorides/metabolism , Chlorides/pharmacology , Drug Resistance/genetics , Female , Homeostasis , Ivermectin/metabolism , Ivermectin/pharmacology , Ivermectin/therapeutic use , Male , Nematoda/genetics , Neuronal Plasticity , Sheep/genetics , Transcriptome
17.
J Med Virol ; 96(3): e29552, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38511598

ABSTRACT

Ivermectin has broad-spectrum antiviral activities. Despite the failure in clinical application of COVID-19, it can serve as a lead compound for the development of more effective broad-spectrum antivirals, for which a better understanding of its antiviral mechanisms is essential. We thus searched for potential novel targets of ivermectin in host cells by label-free thermal proteomic profiling using Huh-7 cells. Inositol monophosphatase (IMPase) was found among the proteins with shifted thermal stability by ivermectin. Ivermectin could inhibit IMPase activity and reduce cellular myo-inositol and phosphatidylinositol-4-phosphate levels. On the other hand, inositol could impair the antiviral activity of ivermectin and lithium, an IMPase inhibitor with known antiviral activity. As phosphatidylinositol phosphate is crucial for the replication of many RNA viruses, inhibition of cellular myo-inositol biosynthesis may be an important antiviral mechanism of ivermectin. Hence, inhibition of IMPase could serve as a potential target for broad-spectrum antiviral development.


Subject(s)
5'-Nucleotidase , Ivermectin , Phosphoric Monoester Hydrolases , Humans , Ivermectin/pharmacology , Proteomics , Inositol/pharmacology , Antiviral Agents/pharmacology
18.
Electrophoresis ; 45(3-4): 212-217, 2024 Feb.
Article in English | MEDLINE | ID: mdl-36695087

ABSTRACT

A procedure to determine albendazole and ivermectin in veterinary formulations, like tablet, bolus, oral suspensions, and injections by micellar liquid chromatography, has been developed. Sample preparation was a batch solid-to-liquid extraction in mobile phase, consisting of a stirring step (15 min), followed by ultrasonication (15 min) and filtration of the obtained supernatant, to reach a target concentration of 2 mg/L for both analytes. Using a mobile phase of 0.15 M sodium dodecyl sulfate-6% 1-pentanol buffered at pH 3 with a 0.01 M phosphate salt, running at 1 mL/min through a C18 column, both drugs were resolved in less than 10 min. Absorbance detection wavelength was 292 nm. Procedure was validated by the guidelines of the International Council on Harmonization in terms of specificity, calibration range (0.025-5 mg/L), trueness (97.8%-102.6%), precision (<2.2%), and system suitability. The method was found easy-to-handle, low cost, safe, green, and with high sample-throughput, thus useful for routine analysis. Therefore, it represents a valuable alternative for quality control of veterinary formulations. It was applied to samples of veterinary formulations purchased from local chemists and veterinarians, and label claims were inside the acceptance criteria (95%-105%).


Subject(s)
Albendazole , Ivermectin , Micelles , Chromatography, Liquid/methods
19.
Br J Dermatol ; 190(2): 163-173, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-37625798

ABSTRACT

BACKGROUND: Treatment failure is considered to be an important factor in relation to the increase in scabies incidence over the last decade. However, the regional and temporal differences, in addition to the predictors of therapy failure, are unclear. OBJECTIVES: We aimed to conduct a systematic review of the prevalence of treatment failure in patients with scabies and investigation of associated factors. METHODS: We searched MEDLINE, EMBASE, CINAHL, Web of Science, Scopus, Global Health and the Cochrane Central Register of Controlled Trials from inception to August 2021 for randomized and quasi-randomized trials, in addition to observational studies that enrolled children or adults diagnosed with confirmed or clinical scabies treated with permethrin, ivermectin, crotamiton, benzyl benzoate, malathion, sulfur or lindane, and measured treatment failure or factors associated with treatment failure. We performed a random effects meta-analysis for all outcomes reported by at least two studies. RESULTS: A total of 147 studies were eligible for inclusion in the systematic review. The overall prevalence of treatment failure was 15.2% [95% confidence interval (CI) 12.9-17.6; I2 = 95.3%, moderate-certainty evidence] with regional differences between World Health Organization regions (P = 0.003) being highest in the Western Pacific region (26.9%, 95% CI 14.5-41.2). Oral ivermectin (11.8%, 95% CI 8.4-15.4), topical ivermectin (9.3%, 95% CI 5.1-14.3) and permethrin (10.8%, 95% CI 7.5-14.5) had relatively lower failure prevalence compared with the overall prevalence. Failure prevalence was lower in patients treated with two doses of oral ivermectin (7.1%, 95% CI 3.1-12.3) compared with those treated with one dose (15.2%, 95% CI 10.8-20.2; P = 0.021). Overall and permethrin treatment failure prevalence in the included studies (1983-2021) increased by 0.27% and 0.58% per year, respectively. Only three studies conducted a multivariable risk factor analysis; no studies assessed resistance. CONCLUSIONS: A second dose of ivermectin showed lower failure prevalence than single-dose ivermectin, which should be considered in all guidelines. The increase in treatment failure over time hints at decreasing mite susceptibility for several drugs, but reasons for failure are rarely assessed. Ideally, scabicide susceptibility testing should be implemented in future studies.


Subject(s)
Scabies , Adult , Child , Humans , Scabies/drug therapy , Ivermectin , Permethrin/therapeutic use , Hexachlorocyclohexane/therapeutic use , Malathion/therapeutic use , Administration, Oral
20.
Br J Dermatol ; 190(4): 486-491, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38112640

ABSTRACT

BACKGROUND: Scabies is a pruritic parasitic infestation of the skin. High-income countries have reported an increasing incidence over the last few years. Studies have indicated a reduction in the sensitivity of scabies mites to the standard treatment of choice, topical permethrin 5%. OBJECTIVES: To evaluate in a head-to-head manner the efficacy of two topical scabicides [permethrin 5% and benzyl benzoate 25% (BB)] in the treatment of scabies using the same administration modality; and to address potential confounding factors such as incorrectly performed treatment and hygiene measures. METHODS: In total, 110 patients with dermoscopy-verified scabies infestation were enrolled and randomized into two equally sized groups in a double-blinded manner. Fifty-five received topical permethrin 5% and 55 received topical BB 25%, both for daily use over a period of three consecutive days. Treatment outcome was evaluated by dermoscopy at a 3-week follow-up visit. RESULTS: Treatment resulted in a dermoscopy-verified cure rate of 27% in the permethrin group and 87% in the BB group. The tolerability and safety profile of permethrin 5% cream was excellent, while the BB emulsion produced a burning sensation in 43% of patients. CONCLUSIONS: Topical permethrin demonstrated a lack of efficacy in the majority of scabies cases, whereas BB demonstrated an excellent cure rate and reasonable tolerability. Considering the reduced sensitivity of scabies mites to permethrin 5%, our results suggest that BB is an appropriate first-line therapy in the treatment of scabies.


Subject(s)
Acaricides , Scabies , Animals , Humans , Acaricides/therapeutic use , Administration, Topical , Benzoates/therapeutic use , Ivermectin , Permethrin/therapeutic use , Sarcoptes scabiei , Scabies/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL