Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 402
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 17(3): e1009382, 2021 03.
Article in English | MEDLINE | ID: mdl-33730100

ABSTRACT

Mosquitoes are vectors of major diseases such as dengue fever and malaria. Mass drug administration of endectocides to humans and livestock is a promising complementary approach to current insecticide-based vector control measures. The aim of this study was to establish an insect model for pharmacokinetic and drug-drug interaction studies to develop sustainable endectocides for vector control. Female Aedes aegypti mosquitoes were fed with human blood containing either ivermectin alone or ivermectin in combination with ketoconazole, rifampicin, ritonavir, or piperonyl butoxide. Drug concentrations were quantified by LC-MS/MS at selected time points post-feeding. Primary pharmacokinetic parameters and extent of drug-drug interactions were calculated by pharmacometric modelling. Lastly, the drug effect of the treatments was examined. The mosquitoes could be dosed with a high precision (%CV: ≤13.4%) over a range of 0.01-1 µg/ml ivermectin without showing saturation (R2: 0.99). The kinetics of ivermectin were characterised by an initial lag phase of 18.5 h (CI90%: 17.0-19.8 h) followed by a slow zero-order elimination rate of 5.5 pg/h (CI90%: 5.1-5.9 pg/h). By contrast, ketoconazole, ritonavir, and piperonyl butoxide were immediately excreted following first order elimination, whereas rifampicin accumulated over days in the mosquitoes. Ritonavir increased the lag phase of ivermectin by 11.4 h (CI90%: 8.7-14.2 h) resulting in an increased exposure (+29%) and an enhanced mosquitocidal effect. In summary, this study shows that the pharmacokinetics of drugs can be investigated and modulated in an Ae. aegypti animal model. This may help in the development of novel vector-control interventions and further our understanding of toxicology in arthropods.


Subject(s)
Aedes/drug effects , Insecticides/pharmacokinetics , Ivermectin/pharmacokinetics , Animals , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions/physiology , Humans , Models, Animal , Mosquito Control/methods , Mosquito Vectors/drug effects , Ritonavir/pharmacokinetics
2.
Epilepsia ; 64(3): 553-566, 2023 03.
Article in English | MEDLINE | ID: mdl-36645121

ABSTRACT

There are only a few drugs that can seriously lay claim to the title of "wonder drug," and ivermectin, the world's first endectocide and forerunner of a completely new class of antiparasitic agents, is among them. Ivermectin, a mixture of two macrolytic lactone derivatives (avermectin B1a and B1b in a ratio of 80:20), exerts its highly potent antiparasitic effect by activating the glutamate-gated chloride channel, which is absent in vertebrate species. However, in mammals, ivermectin activates several other Cys-loop receptors, including the inhibitory γ-aminobutyric acid type A and glycine receptors and the excitatory nicotinic acetylcholine receptor of brain neurons. Based on these effects on vertebrate receptors, ivermectin has recently been proposed to constitute a multifaceted wonder drug for various novel neurological indications, including alcohol use disorders, motor neuron diseases, and epilepsy. This review critically discusses the preclinical and clinical evidence of antiseizure effects of ivermectin and provides several arguments supporting that ivermectin is not a suitable candidate drug for the treatment of epilepsy. First, ivermectin penetrates the mammalian brain poorly, so it does not exert any pharmacological effects via mammalian ligand-gated ion channels in the brain unless it is used at high, potentially toxic doses or the blood-brain barrier is functionally impaired. Second, ivermectin is not selective but activates numerous inhibitory and excitatory receptors. Third, the preclinical evidence for antiseizure effects of ivermectin is equivocal, and at least in part, median effective doses in seizure models are in the range of the median lethal dose. Fourth, the only robust clinical evidence of antiseizure effects stems from the treatment of patients with onchocerciasis, in which the reduction of seizures is due to a reduction in microfilaria densities but not a direct antiseizure effect of ivermectin. We hope that this critical analysis of available data will avert the unjustified hype associated with the recent use of ivermectin to control COVID-19 from recurring in neurological diseases such as epilepsy.


Subject(s)
Anticonvulsants , Antiparasitic Agents , Epilepsy , Ivermectin , Antiparasitic Agents/chemistry , Antiparasitic Agents/pharmacokinetics , Antiparasitic Agents/therapeutic use , Antiparasitic Agents/toxicity , Ivermectin/chemistry , Ivermectin/pharmacokinetics , Ivermectin/therapeutic use , Ivermectin/toxicity , Epilepsy/drug therapy , Humans , Cysteine Loop Ligand-Gated Ion Channel Receptors/agonists , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Anticonvulsants/toxicity , Brain/metabolism , Animals , Mice
3.
Can J Physiol Pharmacol ; 101(9): 475-480, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37235885

ABSTRACT

This study aimed to determine the effect of administration of oral vitamins A and E at different doses on plasma and brain concentrations of ivermectin in mice. The study was carried out on 174 Swiss Albino male mice aged 8-10 weeks. After leaving six mice for method validation, the remaining mice were randomly divided into seven groups with equal numbers of animals. Mice received ivermectin (0.2 mg/kg, subcutaneous) alone and in combination with low (vitamin A: 4000 IU/kg; vitamin E: 35 mg/kg) and high (vitamin A: 30 000 IU/kg; vitamin E: 500 mg/kg) oral doses of vitamins A and E. The plasma and brain concentrations of ivermectin were measured using high-performance liquid chromatography-fluorescence detector. We determined that high doses of vitamins A and E and their combinations increased the passing ratio of ivermectin into the brain significantly. The high-dose vitamin E and the combination of high-concentration vitamins E and A significantly increased the plasma concentration of ivermectin (P < 0.05). The high-dose vitamins E and A and their high-dose combination increased the brain concentration of ivermectin by 3, 2, and 2.7 times, respectively. This research is the first in vivo study to determine the interaction between P-gp substrates and vitamins E and A.


Subject(s)
Antiparasitic Agents , Brain , Ivermectin , Vitamin A , Vitamin E , Animals , Mice , Brain/metabolism , Ivermectin/blood , Ivermectin/pharmacokinetics , Vitamin A/administration & dosage , Vitamin E/administration & dosage , Vitamins , Antiparasitic Agents/blood , Antiparasitic Agents/pharmacokinetics
4.
Molecules ; 27(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35164028

ABSTRACT

Combination therapy of many anthelmintic drugs has been used to achieve fast animal curing. Q-DRENCH is an oral suspension, containing four different active drugs against GIT worms in sheep, commonly used in Australia and New Zeeland. The anti-parasitic drugs are Albendazole (ALB), Levamisole HCl (LEV), Abamectin (ABA), and Closantel (CLO). The main purpose of this study is to present a new simultaneous stability-indicting HPLC-DAD method for the analysis of the four drugs. The recommended liquid system was 1 mL of Triethylamine/L water, adjusting the pH to 3.5 by glacial acetic acid: acetonitrile solvent (20:80, v/v). Isocratic elusion achieved the desired results of separation at a 2 mL/min flow rate using Zorbax C-18 as a stationary phase. Detection was performed at 210 nm. The linearity ranges were 15.15 to 93.75 µg/mL for ALB, 25 to 150 µg/mL for LEV, 30 to 150 µg/mL for ABA, and 11.7 to 140.63 µg/mL for CLO. Moreover, the final greenness score was 0.62 using the AGREE tool, which reflects the eco-friendly nature. Moreover, the four drugs were determined successfully in the presence of their stressful degradation products. This work presents the first chromatographic method for simultaneous analysis for Q-DRENCH oral suspension drugs in the presence of their stressful degradation products.


Subject(s)
Albendazole/analysis , Ivermectin/analogs & derivatives , Levamisole/analysis , Salicylanilides/analysis , Administration, Oral , Albendazole/administration & dosage , Albendazole/chemistry , Albendazole/pharmacokinetics , Animals , Anthelmintics/administration & dosage , Anthelmintics/analysis , Anthelmintics/chemistry , Anthelmintics/pharmacokinetics , Australia , Chromatography, High Pressure Liquid/methods , Drug Stability , Evaluation Studies as Topic , Ivermectin/administration & dosage , Ivermectin/analysis , Ivermectin/chemistry , Ivermectin/pharmacokinetics , Levamisole/administration & dosage , Levamisole/chemistry , Levamisole/pharmacokinetics , Limit of Detection , New Zealand , Salicylanilides/administration & dosage , Salicylanilides/chemistry , Salicylanilides/pharmacokinetics , Sheep , Suspensions
5.
BMC Infect Dis ; 21(1): 411, 2021 May 04.
Article in English | MEDLINE | ID: mdl-33947344

ABSTRACT

BACKGROUND AND OBJECTIVES: An effective treatment option is not yet available for SARS-CoV2, which causes the COVID-19 pandemic and whose effects are felt more and more every day. Ivermectin is among the drugs whose effectiveness in treatment has been investigated. In this study; it was aimed to investigate the presence of gene mutations that alter ivermectin metabolism and cause toxic effects in patients with severe COVID-19 pneumonia, and to evaluate the effectiveness and safety of ivermectin use in the treatment of patients without mutation. MATERIALS AND METHODS: Patients with severe COVID19 pneumonia were included in the study, which was planned as a prospective, randomized, controlled, single-blind phase 3 study. Two groups, the study group and the control group, took part in the study. Ivermectin 200 mcg/kg/day for 5 days in the form of a solution prepared for enteral use added to the reference treatment protocol -hydroxychloroquine + favipiravir + azithromycin- of patients included in the study group. Patients in the control group were given only reference treatment with 3 other drugs without ivermectin. The presence of mutations was investigated by performing sequence analysis in the mdr1/abcab1 gene with the Sanger method in patients included in the study group according to randomization. Patients with mutations were excluded from the study and ivermectin treatment was not continued. Patients were followed for 5 days after treatment. At the end of the treatment and follow-up period, clinical response and changes in laboratory parameters were evaluated. RESULTS: A total of 66 patients, 36 in the study group and 30 in the control group were included in the study. Mutations affecting ivermectin metabolism was detected in genetic tests of six (16.7%) patients in the study group and they were excluded from the study. At the end of the 5-day follow-up period, the rate of clinical improvement was 73.3% (22/30) in the study group and was 53.3% (16/30) in the control group (p = 0.10). At the end of the study, mortality developed in 6 patients (20%) in the study group and in 9 (30%) patients in the control group (p = 0.37). At the end of the follow-up period, the average peripheral capillary oxygen saturation (SpO2) values of the study and control groups were found to be 93.5 and 93.0%, respectively. Partial pressure of oxygen (PaO2)/FiO2 ratios were determined as 236.3 ± 85.7 and 220.8 ± 127.3 in the study and control groups, respectively. While the blood lymphocyte count was higher in the study group compared to the control group (1698 ± 1438 and 1256 ± 710, respectively) at the end of the follow-up period (p = 0.24); reduction in serum C-reactive protein (CRP), ferritin and D-dimer levels was more pronounced in the study group (p = 0.02, p = 0.005 and p = 0.03, respectively). CONCLUSIONS: According to the findings obtained, ivermectin can provide an increase in clinical recovery, improvement in prognostic laboratory parameters and a decrease in mortality rates even when used in patients with severe COVID-19. Consequently, ivermectin should be considered as an alternative drug that can be used in the treatment of COVID-19 disease or as an additional option to existing protocols.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Ivermectin/therapeutic use , Pneumonia, Viral/drug therapy , ATP Binding Cassette Transporter, Subfamily B/genetics , Aged , Amides/therapeutic use , Antiviral Agents/pharmacokinetics , Azithromycin/therapeutic use , COVID-19/blood , COVID-19/mortality , Cytochrome P-450 CYP3A/genetics , Drug Therapy, Combination , Female , Humans , Hydroxychloroquine/therapeutic use , Ivermectin/pharmacokinetics , Male , Middle Aged , Pneumonia, Viral/blood , Pneumonia, Viral/virology , Prospective Studies , Pyrazines/therapeutic use , Single-Blind Method , Treatment Outcome
6.
Arch Toxicol ; 95(10): 3205-3221, 2021 10.
Article in English | MEDLINE | ID: mdl-34417632

ABSTRACT

Currently, the authorisation process for plant protection products (PPPs) relies on the testing of acute and topological toxicity only. Contrastingly, the evaluation of active substances includes a more comprehensive set of toxicity studies. Nevertheless, mixture effects of active ingredients and co-formulants may result in increased toxicity. Therefore, we investigated effects of surface active co-formulants on the toxicity of two PPPs focussing on qualitative and quantitative toxicokinetic effects on absorption and secretion. The respective products are based on the active substances abamectin and fluroxypyr-meptyl and were tested for cytotoxicity in the presence or absence of the corresponding surfactants and co-formulants using Caco-2 cells. In addition, the effect of co-formulants on increased cellular permeation was quantified using LC-MS/MS, while potential kinetic mixture effects were addressed by fluorescence anisotropy measurements and ATPase assays. The results show that surface active co-formulants significantly increase the cytotoxicity of the investigated PPPs, leading to more than additive mixture effects. Moreover, analytical investigations show higher efflux ratios of both active substances and the metabolite fluroxypyr upon combination with certain concentrations of the surfactants. The results further point to a significant and concentration-dependent inhibition of Pgp transporters by most of the surfactants as well as to increased membrane fluidity. Altogether, these findings strongly support the hypothesis that surfactants contribute to increased cytotoxicity of PPPs and do so by increasing the bioavailability of the respective active substances.


Subject(s)
Glycolates/toxicity , Herbicides/toxicity , Insecticides/toxicity , Ivermectin/analogs & derivatives , Biological Availability , Caco-2 Cells , Chromatography, Liquid , Fluorescence Polarization , Glycolates/administration & dosage , Glycolates/pharmacokinetics , Herbicides/administration & dosage , Herbicides/pharmacokinetics , Humans , Insecticides/administration & dosage , Insecticides/pharmacokinetics , Ivermectin/administration & dosage , Ivermectin/pharmacokinetics , Ivermectin/toxicity , Surface-Active Agents/chemistry , Tandem Mass Spectrometry
7.
J Vet Pharmacol Ther ; 44(1): 11-17, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33103252

ABSTRACT

The aim of this study was to compare the pharmacokinetics of ivermectin and its antiparasitic activity in two horse breeds. Eight Hutsul and 14 Toric horses were administered ivermectin orally at a dose of 0.2 mg/kg body weight. Blood samples were collected for 96 hr, and faecal samples were collected one day before and on days 14 and 21 after drug administration. Ivermectin concentrations in plasma samples were determined by high-performance liquid chromatography. Ivermectin concentration was significantly higher in Toric than in Hutsul horses 90 min after ivermectin administration and was maintained at higher level for up to 96 hr. The area under the concentration versus the time curve from 0 to the last sampling point (AUC0→t ) and the maximum plasma concentration (Cmax ) were significantly higher in Toric than in Hutsul horses (1792.09 ± 246.22 µg × hr/L vs. 716.99 ± 255.81 µg × hr/L and 62.72 ± 17.97 ng/ml vs. 35.34 ± 13.61 ng/ml, respectively). No parasitic eggs were found in the faecal samples collected from both groups of horses on days 14 and 21 after drug administration. The obtained results indicate that although the pharmacokinetics of ivermectin may differ significantly between horse breeds, these differences do not affect the effectiveness of therapy.


Subject(s)
Antiparasitic Agents/pharmacokinetics , Horse Diseases/drug therapy , Horses/metabolism , Ivermectin/pharmacokinetics , Parasitic Diseases, Animal/drug therapy , Animals , Antiparasitic Agents/therapeutic use , Area Under Curve , Feces/parasitology , Half-Life , Horse Diseases/parasitology , Horses/classification , Horses/genetics , Ivermectin/therapeutic use , Parasite Egg Count/veterinary
8.
Article in English | MEDLINE | ID: mdl-32660993

ABSTRACT

Previously, ivermectin (1 to 10 mg/kg of body weight) was shown to inhibit the liver-stage development of Plasmodium berghei in orally dosed mice. Here, ivermectin showed inhibition of the in vitro development of Plasmodium cynomolgi schizonts (50% inhibitory concentration [IC50], 10.42 µM) and hypnozoites (IC50, 29.24 µM) in primary macaque hepatocytes when administered as a high dose prophylactically but not when administered in radical cure mode. The safety, pharmacokinetics, and efficacy of oral ivermectin (0.3, 0.6, and 1.2 mg/kg) with and without chloroquine (10 mg/kg) administered for 7 consecutive days were evaluated for prophylaxis or radical cure of P. cynomolgi liver stages in rhesus macaques. No inhibition or delay to blood-stage P. cynomolgi parasitemia was observed at any ivermectin dose (0.3, 0.6, and 1.2 mg/kg). Ivermectin (0.6 and 1.2 mg/kg) and chloroquine (10 mg/kg) in combination were well-tolerated with no adverse events and no significant pharmacokinetic drug-drug interactions observed. Repeated daily ivermectin administration for 7 days did not inhibit ivermectin bioavailability. It was recently demonstrated that both ivermectin and chloroquine inhibit replication of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro Further ivermectin and chloroquine trials in humans are warranted to evaluate their role in Plasmodium vivax control and as adjunctive therapies against COVID-19 infections.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Ivermectin/pharmacology , Liver/drug effects , Malaria/drug therapy , Plasmodium cynomolgi/drug effects , Animals , Antimalarials/blood , Antimalarials/pharmacokinetics , Biological Availability , Chloroquine/blood , Chloroquine/pharmacokinetics , Drug Administration Schedule , Drug Combinations , Drug Synergism , Female , Hepatocytes/drug effects , Hepatocytes/parasitology , Ivermectin/blood , Ivermectin/pharmacokinetics , Liver/parasitology , Macaca mulatta , Malaria/parasitology , Male , Parasitemia/drug therapy , Plasmodium cynomolgi/growth & development , Plasmodium cynomolgi/pathogenicity , Primary Cell Culture , Schizonts/drug effects , Schizonts/growth & development
9.
J Antimicrob Chemother ; 75(2): 438-440, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31691813

ABSTRACT

BACKGROUND: Ivermectin is an older anthelminthic agent that is being studied more intensely given its potential for mass drug administration against scabies, malaria and other neglected tropical diseases. Its pharmacokinetics (PK) remain poorly characterized. Furthermore, the majority of PK trials are performed under fasted-state dosing conditions, and the effect of food is therefore not well known. To better plan and design field trials with ivermectin, a model that can account for both conditions would be valuable. OBJECTIVES: To develop a PK model and characterize the food effect with single oral doses of ivermectin. PATIENTS AND METHODS: We performed a population-based PK analysis of data pooled from two previous trials of a single dose of 12 mg ivermectin, one with dosing after a high-fat breakfast (n=12) and one with fasted-state dosing (n=3). RESULTS: The final model described concentration-time profiles after fed and fasted dosing accurately, and estimated the food effect associated with relative bioavailability to 1.18 (95% CI 1.10-1.67). CONCLUSIONS: In this analysis, the effect of a high-fat breakfast compared with a fasted-state administration of a single oral dose of 12 mg ivermectin was minimal.


Subject(s)
Food-Drug Interactions , Ivermectin/pharmacokinetics , Administration, Oral , Area Under Curve , Biological Availability , Cross-Over Studies , Humans
10.
J Vet Pharmacol Ther ; 43(5): 485-490, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32304335

ABSTRACT

We compared the pharmacokinetics of ivermectin premix and ivermectin microspheres in pigs after single and multiple administration regimes. In the single-dose experiments, 24 piglets were randomly divided into three groups and given ivermectin at 0.3 mg/kg using (a) 1.0% ivermectin administered subcutaneously, (b) 0.25% ivermectin premix orally, and (c) 0.25% ivermectin microspheres orally. In the multiple-dose experiment, 6 pigs in two equal groups received ivermectin premix and microspheres orally at 0.3 mg/kg for 7 consecutive days to monitor the valley plasma levels. The plasma samples were detected by fluorescence high-performance liquid chromatography, and concentration-time data were fitted to a noncompartmental model. After oral administration of ivermectin microspheres at a single dose, the elimination rate constant (Kel), the half-life (t1/2 ), the peak time (Tmax ), the mean residence time (MRT), and the peak concentration (Cmax ) were 0.012 ± 0.0031/hr, 59.94 ± 20.18 hr, 9.50 ± 0.93 hr, 55.96 ± 11.40 hr, and 37.75 ± 3.45 ng/ml, respectively. The Cmax of microspheres was not statistically different (p > .05) compared with that of premix groups (39.81 ± 5.83 ng/ml). Moreover, the AUC of the microcapsule groups was increased from 1,129.76 ± 245.62 to 1,607.33 ± 343.35 hr ng/ml compared with the premix groups, and the relative bioavailability increased by an average of 17.53% after oral administration with ivermectin microspheres. Multiple-dose administration also indicated pigs fed with ivermectin microspheres can get a higher minimum steady-state concentration and a longer maintenance time than ivermectin premix.


Subject(s)
Ivermectin/pharmacokinetics , Swine/metabolism , Administration, Oral , Animals , Antiparasitic Agents/administration & dosage , Antiparasitic Agents/pharmacokinetics , Area Under Curve , Biological Availability , Delayed-Action Preparations , Female , Half-Life , Ivermectin/administration & dosage , Male , Microspheres
11.
J Vet Pharmacol Ther ; 43(2): 123-128, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31660626

ABSTRACT

The objective of this research was to evaluate comparative pharmacokinetics of doramectin in alpacas, after subcutaneous administration of 0.2 mg/kg dose. Six healthy adult alpacas, mean age of 5 years ± 1, (three female and three gelded males) of mean bodyweight of 62 kg ± 16 kg with an average body condition scored 2.8 ± 1 out of five, were used in this study. Serial blood samples were collected from the jugular vein before the administration until day 21 afterwards to establish the pharmacokinetics of doramectin after its subcutaneous administration at 0.2 mg/kg dose. The blood samples were analysed using high-performance liquid chromatography (HPLC), fluorescence detection method with precolumn derivatisation, validated for alpacas. The pharmacokinetic parameters were calculated using a noncompartmental model, and results showed Cmax (6.05 ± 5.34 ng/ml), Tmax (3.83 ± 2.48 days), AUC (62.12 ± 18.86 ng/ml × d), terminal half-life (6.2 ± 4.9 days) and MRT (11.56 ± 4.43 days). The results of this study showed that the Cmax and AUC were much lower than in cattle and sheep at the same dosage. Tmax remained similar to cattle and sheep. This study presents valuable information about pharmacokinetics of doramectin in alpacas, which can be utilised in its future efficacy studies.


Subject(s)
Anthelmintics/pharmacokinetics , Camelids, New World/blood , Ivermectin/analogs & derivatives , Animals , Anthelmintics/administration & dosage , Area Under Curve , Female , Half-Life , Injections, Subcutaneous/veterinary , Ivermectin/administration & dosage , Ivermectin/pharmacokinetics , Male
12.
J Vet Pharmacol Ther ; 43(5): 477-484, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32270537

ABSTRACT

Ivermectin (IVM) is one of the most widely used antiparasitic drugs worldwide and has become the drug of choice for anthelmintic and tick treatment in beef cattle production. It is known that pharmacokinetic parameters are fundamental to the rational use of a drug and food safety and these parameters are influenced by different factors. The aim of this study was to evaluate the pharmacokinetic profile of IVM in Bos indicus, Bos taurus, and crossbreed cattle (B. indicus × B. taurus) kept under same field conditions and the possible impacts of sex and IVM formulation (1% and 3.15%). It was observed that IVM concentration was significantly affected by breed. The plasma concentrations of IVM, AUC, Cmax , and t1/2ß were significantly higher in B. indicus compared to B. taurus. Crossbreed animals showed an intermediate profile between European and Indian cattle. No alteration in pharmacokinetics parameters was detected when comparing different gender. Concerning the pharmacokinetic data of IVM formulation, it was verified that Tmax , AUC, and t1/2ß were higher in 3.15% IVM animals than those from 1% IVM formulation. The results clearly indicated that the IVM plasma concentrations in B. indicus were higher than that in B. taurus.


Subject(s)
Antiparasitic Agents/pharmacokinetics , Cattle/genetics , Cattle/physiology , Ivermectin/pharmacokinetics , Animals , Antiparasitic Agents/blood , Area Under Curve , Cattle/blood , Cattle/classification , Dose-Response Relationship, Drug , Female , Half-Life , Ivermectin/blood , Male , Sex Factors
13.
J Vet Pharmacol Ther ; 43(2): 171-178, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32030782

ABSTRACT

The aims of the present study were to evaluate the pharmacokinetic profile and efficacy of eprinomectin (EPM) against Rhipicephalus microplus in cattle of a new injectable form of EPM (Voss Performa®). The product was administered subcutaneously at a dose of 200 µg EPM/kg, in a single dose. The efficacy of EPM against R. microplus in cattle was evaluated through field and stall tests. Studies were performed to estimate the pharmacokinetic parameters of EPM with the purpose of better understanding the kinetics of the formulation. The formulation was effective in controlling R. microplus in both naturally and artificially infested cattle, providing efficacy greater than 95%. The results of pharmacokinetic study were Cmax of 47.15 ± 22.20 ng/ml, Tmax of 1.33 ± 0.492 days, T1/2 of 2.96 ± 1.212 days, AUC0-t of 228.08 ± 57.30 ng day ml-1 , and AUC0-∞ of 240.50 ± 58.44 ng day ml-1 . Therefore, the new injectable EPM formulation becomes an important alternative for the control of cattle tick in Brazil.


Subject(s)
Cattle Diseases/drug therapy , Insecticides/therapeutic use , Ivermectin/analogs & derivatives , Rhipicephalus/drug effects , Tick Infestations/veterinary , Animals , Area Under Curve , Cattle , Cattle Diseases/parasitology , Female , Half-Life , Insecticides/pharmacokinetics , Ivermectin/pharmacokinetics , Ivermectin/therapeutic use , Male , Tick Infestations/drug therapy
14.
J Environ Sci Health B ; 55(6): 517-524, 2020.
Article in English | MEDLINE | ID: mdl-32065572

ABSTRACT

The current study estimated the dissipation rates of abamectin, chlorfenapyr and pyridaben acaricides in pods of green beans (Phaseolus vulgaris L.) under field conditions in Egypt. Pesticides were extracted and cleaned-up by QuEChERS method and were analyzed by HPLC. The dissipation of these acaricides followed the first order kinetics model with half-life (t1/2) values 1.00, 3.50 and 1.50 days for abamectin, chlorfenapyr and pyridaben, respectively. The lowest residues, at different time intervals of field application rate of each pesticide, were observed with abamectin followed by pyridaben and then chlorfenapyr. Pre-harvest intervals (PHIs) were 10.00, 13.50 and 6.00 days for abamectin, chlorfenapyr and pyridaben, respectively and were below the established European maximum residue limits (EU MRLs) 10-14, 14-21 and 7-10 days after application, respectively. If the fresh pods will be consumed after harvest, it is expected that the presence of these pesticides in the food will have a negative impact on human health. Therefore, the elimination of the residues of these harmful pesticides must be carried out.


Subject(s)
Acaricides/pharmacokinetics , Ivermectin/analogs & derivatives , Phaseolus/drug effects , Pyrethrins/pharmacokinetics , Pyridazines/pharmacokinetics , Acaricides/analysis , Chemical Fractionation , Chromatography, High Pressure Liquid , Egypt , Food Contamination/analysis , Humans , Ivermectin/analysis , Ivermectin/pharmacokinetics , Kinetics , Pesticide Residues/analysis , Phaseolus/metabolism , Pyrethrins/analysis , Pyridazines/analysis
15.
Clin Infect Dis ; 69(7): 1112-1119, 2019 09 13.
Article in English | MEDLINE | ID: mdl-30590537

ABSTRACT

BACKGROUND: Ivermectin is being considered for mass drug administration for malaria, due to its ability to kill mosquitoes feeding on recently treated individuals. In a recent trial, 3-day courses of 300 and 600 mcg/kg/day were shown to kill Anopheles mosquitoes for at least 28 days post-treatment when fed patients' venous blood using membrane feeding assays. Direct skin feeding on humans may lead to higher mosquito mortality, as ivermectin capillary concentrations are higher. We compared mosquito mortality following direct skin and membrane feeding. METHODS: We conducted a mosquito feeding study, nested within a randomized, double-blind, placebo-controlled trial of 141 adults with uncomplicated malaria in Kenya, comparing 3 days of ivermectin 300 mcg/kg/day, ivermectin 600 mcg/kg/day, or placebo, all co-administered with 3 days of dihydroartemisinin-piperaquine. On post-treatment day 7, direct skin and membrane feeding assays were conducted using laboratory-reared Anopheles gambiae sensu stricto. Mosquito survival was assessed daily for 28 days post-feeding. RESULTS: Between July 20, 2015, and May 7, 2016, 69 of 141 patients participated in both direct skin and membrane feeding (placebo, n = 23; 300 mcg/kg/day, n = 24; 600 mcg/kg/day, n = 22). The 14-day post-feeding mortality for mosquitoes fed 7 days post-treatment on blood from pooled patients in both ivermectin arms was similar with direct skin feeding (mosquitoes observed, n = 2941) versus membrane feeding (mosquitoes observed, n = 7380): cumulative mortality (risk ratio 0.99, 95% confidence interval [CI] 0.95-1.03, P = .69) and survival time (hazard ratio 0.96, 95% CI 0.91-1.02, P = .19). Results were consistent by sex, by body mass index, and across the range of ivermectin capillary concentrations studied (0.72-73.9 ng/mL). CONCLUSIONS: Direct skin feeding and membrane feeding on day 7 resulted in similar mosquitocidal effects of ivermectin across a wide range of drug concentrations, suggesting that the mosquitocidal effects seen with membrane feeding accurately reflect those of natural biting. Membrane feeding, which is more patient friendly and ethically acceptable, can likely reliably be used to assess ivermectin's mosquitocidal efficacy. CLINICAL TRIALS REGISTRATION: NCT02511353.


Subject(s)
Antiparasitic Agents/administration & dosage , Culicidae/drug effects , Insecticides/administration & dosage , Ivermectin/administration & dosage , Adult , Animals , Anopheles/drug effects , Antiparasitic Agents/pharmacokinetics , Feeding Behavior , Female , Humans , Ivermectin/pharmacokinetics , Malaria/parasitology , Malaria/prevention & control , Male , Mosquito Control , Young Adult
16.
Article in English | MEDLINE | ID: mdl-30323047

ABSTRACT

Soil-transmitted helminth (STH) infections still remain a major health problem in poor rural settings. The lack of efficacious drugs against all STH species raises interest in drug combinations. Drug-drug interactions (DDIs) are, however, of major concern, so careful in vitro and in vivo characterization is needed. The combination of tribendimidine with either ivermectin or oxantel pamoate targets a broad range of STHs and thus represents a promising treatment alternative. Drug-drug interactions, however, have not yet been investigated. Therefore, the effects of combinations of ivermectin, oxantel pamoate, and tribendimidine's active metabolite deacylated amidantel (dADT) on cytochrome P450 (CYP450) metabolism were evaluated, followed by a pharmacokinetic analysis of tribendimidine and ivermectin alone and in combination in healthy rats. Oxantel pamoate is only poorly absorbed and was therefore excluded from pharmacokinetic analysis. No evident effect was observed for tribendimidine-oxantel pamoate at the CYP450 metabolism level, whereas a combination of tribendimidine and ivermectin led to moderately increased CYP2D6 inhibition compared to ivermectin or tribendimidine alone. Coadministration of tribendimidine with ivermectin altered neither the time to maximum concentration of drug in plasma (Tmax) nor the elimination half-lives of dADT, the acetylated derivative of amidantel (adADT), and ivermectin. While the area under the concentration-versus-time curve (AUC) and maximum concentration of drug in plasma (Cmax) values of dADT, adADT, and ivermectin are reduced by coadministration, the change is insufficient to declare that a DDI has been detected. Further studies are necessary to understand the observed interaction of tribendimidine and ivermectin, which is not related to P450 metabolism, and its significance for the situation in humans.


Subject(s)
Anthelmintics/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Ivermectin/pharmacokinetics , Phenylenediamines/pharmacokinetics , Pyrantel Pamoate/analogs & derivatives , Animals , Anthelmintics/pharmacology , Area Under Curve , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Drug Interactions , Drug Therapy, Combination , Helminthiasis, Animal/drug therapy , Helminths/drug effects , Ivermectin/pharmacology , Male , Phenylenediamines/pharmacology , Pyrantel Pamoate/pharmacokinetics , Pyrantel Pamoate/pharmacology , Rats
17.
J Antimicrob Chemother ; 74(6): 1642-1647, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30859185

ABSTRACT

BACKGROUND: Yearly, millions of children are treated globally with ivermectin mainly for neglected tropical diseases. Anatomical, physiological and biochemical differences between children and adults may result in changes in pharmacokinetics. However, paediatric pharmacokinetic data of ivermectin are lacking. METHODS: In the framework of a randomized controlled dose-finding trial in rural Côte d'Ivoire, Trichuris trichiura-infected pre-school-aged children (PSAC, 2-5 years) and school-aged children (SAC, 6-12 years) were assigned to 100 or 200 µg/kg and 200, 400 or 600 µg/kg ivermectin, respectively (ISRCTN registry no. ISRCTN15871729). Capillary blood was collected on dried blood spot cards until 72 h post-treatment. Ivermectin was quantified by LC-MS/MS, and pharmacokinetic parameters were evaluated by non-compartmental analysis. RESULTS: C max and AUC increased in PSAC and SAC with ascending doses and were similar in both age groups when the current standard dose (200 µg/kg) was administered (∼23 ng/mL and ∼350 ng×h/mL, respectively). PSAC with lower BMI were associated with significantly higher AUCs. AUC and Cmax were ∼2-fold lower in children compared with parameters previously studied in adults, whereas body weight-adjusted CL/F (∼0.35 L/h/kg) was significantly higher in children. Tmax (∼6 h), t1/2 (∼18 h), mean residence time (MRTINF) (∼28 h) and V/F (∼8 L/kg) were similar in all paediatric treatment arms. CONCLUSIONS: A positive association of AUC or Cmax with dose was observed in both age groups. Undernutrition might influence the AUC of ivermectin in PSAC. Ivermectin shows a lower exposure profile in children compared with adults, highlighting the need to establish dosing recommendations for different age groups.


Subject(s)
Antiparasitic Agents/administration & dosage , Antiparasitic Agents/pharmacokinetics , Ivermectin/administration & dosage , Ivermectin/pharmacokinetics , Trichuriasis/drug therapy , Trichuriasis/parasitology , Trichuris/drug effects , Animals , Area Under Curve , Child , Child, Preschool , Chromatography, Liquid , Drug Monitoring , Female , Humans , Male , Tandem Mass Spectrometry , Treatment Outcome
18.
Br J Clin Pharmacol ; 85(3): 626-633, 2019 03.
Article in English | MEDLINE | ID: mdl-30566757

ABSTRACT

AIMS: The anthelminthic ivermectin is receiving new attention as it is being repurposed for new indications such as mass drug administrations for the treatment of scabies or in malaria vector control. As its pharmacokinetics are still poorly understood, we aimed to characterize the population pharmacokinetics of ivermectin in plasma and dried blood spots (DBS), a sampling method better suited to field trials, with special focus on the influence of body composition and enterohepatic circulation. METHODS: We performed a clinical trial in 12 healthy volunteers who each received a single oral dose of 12 mg ivermectin, and collected peripheral venous and capillary DBS samples. We determined ivermectin concentrations in plasma and DBS by liquid chromatography tandem mass spectrometry using a fully automated and scalable extraction system for DBS sample processing. Pharmacokinetic data were analysed using non-linear mixed effects modelling. RESULTS: A two-compartment model with a transit absorption model, first-order elimination, and weight as an influential covariate on central volume of distribution and clearance best described the data. The model estimates (inter-individual variability) for a 70 kg subject were: apparent population clearance 7.7 (25%) l h-1 , and central and peripheral volumes of distribution 89 (10%) l and 234 (20%) l, respectively. Concentrations obtained from DBS samples were strongly linearly correlated (R2  = 0.97) with plasma concentrations, and on average 30% lower. CONCLUSION: The model accurately depicts population pharmacokinetics of plasma and DBS concentrations over time for oral ivermectin. The proposed analytical workflow is scalable and applicable to the requirements of mass drug administrations.


Subject(s)
Antiparasitic Agents/pharmacokinetics , Dried Blood Spot Testing , Ivermectin/pharmacokinetics , Administration, Oral , Adult , Antiparasitic Agents/administration & dosage , Drug Repositioning/standards , Feasibility Studies , Female , Healthy Volunteers , Humans , Ivermectin/administration & dosage , Malaria/prevention & control , Male , Mass Drug Administration/standards , Models, Biological , Mosquito Vectors/drug effects , Scabies/drug therapy , Time Factors , Young Adult
19.
Ecotoxicol Environ Saf ; 169: 944-949, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30597795

ABSTRACT

Ivermectin (IVM), a macrocylic lactone from the avermectin family, is a potent broad-spectrum anthelmintic drug widely used in veterinary as well as human medicine. Although the health benefits of IVM treatment are particularly important, this drug also represents an environmental pollutant with potentially negative effects on many non-target species. To evaluate the ecotoxicological risk of IVM administration to livestock, information evaluating achievable environment-reaching concentration is needed. Therefore, the present study was designed to determine the excretion profile of subcutaneously administered IVM in sheep. The standard recommended dose of IVM (0.2 mg kg-1 b.w.) was used. UHPLC/MS/MS was used for the analysis of IVM faecal concentration. In addition, the effect of IVM on seed germination and early roots growth of white mustard (Sinapis alba L.) was evaluated in order to estimate the potential phytotoxic effect of IVM. Based on the obtained results, the parameters of IVM pharmacokinetics (maximum concentration (cmax), time to achieve maximum concentration (tmax), mean residence time (MRT), area under the curve (AUC)) were calculated. IVM elimination in sheep was slow, but faster than the elimination reported previously in cattle. Great interindividual differences were also observed. A two-peak profile of concentration curves indicate the importance of the active efflux of IVM via enterocytes. A "seed germination and early roots growth" test revealed significant IVM phytotoxicity (20% inhibition of root growth) even at 50 nM concentration, a level which may be found in the environment. This newly demonstrated phytotoxicity of IVM together with its well-known toxicity to invertebrates should be taken into account, and thus animals treated with IVM should not be kept in pastures, especially not in sites with high ecological value.


Subject(s)
Anthelmintics/pharmacokinetics , Anthelmintics/toxicity , Environmental Pollution/adverse effects , Ivermectin/pharmacokinetics , Ivermectin/toxicity , Sinapis/drug effects , Animals , Area Under Curve , Cattle , Ecotoxicology , Environmental Pollution/analysis , Feces/chemistry , Injections, Subcutaneous , Sheep , Sinapis/growth & development
20.
J Vet Pharmacol Ther ; 42(5): 497-504, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31183888

ABSTRACT

The purpose of this study was to determine the pharmacokinetic interaction between ivermectin (0.4 mg/kg) and praziquantel (10 mg/kg) administered either alone or co-administered to dogs after oral treatment. Twelve healthy cross-bred dogs (weighing 18-21 kg, aged 1-3 years) were allocated randomly into two groups of six dogs (four females, two males) each. In first group, the tablet forms of praziquantel and ivermectin were administered using a crossover design with a 15-day washout period, respectively. Second group received tablet form of ivermectin plus praziquantel. The plasma concentrations of ivermectin and praziquantel were determined by high-performance liquid chromatography using a fluorescence and ultraviolet detector, respectively. The pharmacokinetic parameters of ivermectin following oral alone-administration were as follows: elimination half-life (t1/2λz ) 110 ± 11.06 hr, area under the plasma concentration-time curve (AUC0-∞ ) 7,805 ± 1,768 hr. ng/ml, maximum concentration (Cmax ) 137 ± 48.09 ng/ml, and time to reach Cmax (Tmax ) 14.0 ± 4.90 hr. The pharmacokinetic parameters of praziquantel following oral alone-administration were as follows: t1/2λz 7.39 ± 3.86 hr, AUC0-∞ 4,301 ± 1,253 hr. ng/ml, Cmax 897 ± 245 ng/ml, and Tmax 5.33 ± 0.82 hr. The pharmacokinetics of ivermectin and praziquantel were not changed, except Tmax of praziquantel in the combined group. In conclusion, the combined formulation of ivermectin and praziquantel can be preferred in the treatment and prevention of diseases caused by susceptible parasites in dogs because no pharmacokinetic interaction was determined between them.


Subject(s)
Antiparasitic Agents/pharmacokinetics , Dogs/blood , Ivermectin/pharmacokinetics , Praziquantel/pharmacokinetics , Administration, Oral , Animals , Antiparasitic Agents/administration & dosage , Area Under Curve , Drug Interactions , Female , Half-Life , Ivermectin/administration & dosage , Ivermectin/blood , Male , Praziquantel/administration & dosage , Praziquantel/blood
SELECTION OF CITATIONS
SEARCH DETAIL