Sie sind auf Seite 1von 13

Available online at www.sciencedirect.

com

The
Veterinary Journal
The Veterinary Journal 179 (2009) 2537
www.elsevier.com/locate/tvjl

Review

The pharmacokinetics and metabolism of ivermectin


in domestic animal species
Aranzazu Gonzalez Canga *, Ana M. Sahagun Prieto, M. Jose Diez Liebana,
Nelida Fernandez Martnez, Matilde Sierra Vega, Juan J. Garca Vieitez
Department of Biomedical Sciences, Veterinary Faculty, University of Leon, Spain

Accepted 17 July 2007

Abstract

The pharmacokinetic properties of drugs are closely related to their pharmacological ecacy. The kinetics of ivermectin are charac-
terised, in general terms, by a slow absorption process, a broad distribution in the organism, low metabolism, and slow excretion. The
kinetics vary according to the route of administration, formulation, animal species, body condition, age, and physiological status, all of
which contribute to dierences in drug ecacy. Characterisation of ivermectin kinetics can be used to predict and optimise the value of
the parasiticide eects and to design programmes for parasite control. This article reviews the pharmacokinetics of ivermectin in several
domestic animal species.
2007 Elsevier Ltd. All rights reserved.

Keywords: Ivermectin; Pharmacokinetics; Animal species; Absorption; Distribution; Metabolism; Excretion; Cattle; Sheep; Goat; Pig; Dog; Review

Introduction tin-B1a and >20% 22,23-dihydroavermectin-B1b (Fig. 1).


It is a highly lipophilic substance that dissolves in most
The rational use of a drug requires knowledge of its organic solvents, but is practically insoluble in water
basic pharmacokinetics in the target animal species, and (0.0004% m/v). Ivermectin was rst marketed in 1981 by
this helps to optimise clinical ecacy. Ivermectin is proba- Merck Sharp and Dohme as an antiparasitic agent (Steel,
bly one of the most widely used antiparasitic drugs world- 1993), and it remains the leading worldwide antiparasitic
wide, and its ecacy is well established. However, the agent for livestock. It has exceptional potency against
pharmacokinetic parameters of ivermectin vary extensively endo- and ectoparasites at extremely low doses (doses rec-
and in accordance with many factors that can all inuence ommended are expressed as lg/kg); this accounts for its
the drugs plasma concentration. These factors, which large margin of safety.
include the species, route of administration, vehicle used Ivermectin is highly active against a wide spectrum of
in the commercial formulation, bodyweight, body condi- nematode species, including most larvae and adult forms;
tion, physiological status, and amount and type of nutri- it is also highly eective against many arthropod parasites
tion, create diculties when extrapolating data from one of domestic animals (Table 1). All important gastrointesti-
species to another and should be considered in clinical nal and lung nematodes are susceptible to the drug, includ-
practice in order to achieve eective levels that will last ing sensitive mites, ticks, biting ies, and parasitic dipteran
as long as possible. larvae (Campbell and Benz, 1984; Campbell, 1989; McKel-
Ivermectin is a mixture of two chemically modied aver- lar and Benchaoui, 1996). In dogs, ivermectin is also active
mectins that contain at least 80% of 22,23-dihydroavermec- against developing larvae of Dirolaria immitis and is used
in heartworm prophylaxis.
*
Corresponding author. Tel.: +34 987 29 18 46; fax: +34 987 29 12 67. Toxicity to ivermectin is rare across animal species. The
E-mail address: agonc@unileon.es (A. Gonzalez Canga). signs of toxicosis are mydriasis and depression, followed by

1090-0233/$ - see front matter 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.tvjl.2007.07.011
26 A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537

instead of 200 lg/kg. Parenteral administration delays iver-


mectins absorption compared to the oral route, but leads
to an overall higher availability in plasma, a longer dura-
tion of activity, and better ecacy. Molento et al. (2004)
pointed out that the lower absorption of ivermectin after
oral administration could be inuenced by P-glycoprotein,
which is also present on the intestinal epithelium; when
ivermectin is co-administered with verapamil (a P-glyco-
protein blocker), the maximum plasma concentration
(Cmax) and bioavailability increased, leading to an
improvement in antiparasitic ecacy.
Ivermectins extremely low water solubility and its pre-
cipitation in SC tissues favour slow absorption from the
injection site, resulting in a prolonged presence in the
bloodstream. On the other hand, the erratic SC absorption
of ivermectin could relate to variability in pharmacokinetic
parameters.
In ruminant species, intraruminal (IR) administration
yields a lower systemic availability and could explain its
lesser ecacy against ectoparasites (Benz et al., 1989;
Fig. 1. Chemical structure of ivermectin. McKellar and Benchaoui, 1996) and shorter duration of
activity against gastrointestinal nematodes.
Small dierences in formulations may result in substan-
ataxia, recumbency, and death. It has no adverse eects on tial changes in the antiparasitic activity of ivermectin. This
breeding performance. Some Collie dogs and other herding property has been extensively studied in cattle. Absorption
breeds are remarkably susceptible, but even these animals is greater and faster with an aqueous vehicle than with pro-
will tolerate doses of 50 lg/kg, which are nearly 10-fold pylene glycol:glycerol-formal (60:40 v/v), and the drugs
greater than the therapeutic dose in dogs. The central ner- biological half-life is also longer, prolonging its clinical e-
vous system side-eects in sensitive Collie dogs have been cacy (Lo et al., 1985). Moreover, when using an oil-based
linked to the absence or functional deciency of P-glyco- formulation, absorption is faster after IM versus SC admin-
protein, which functions as a transmembrane eux pump istration due to greater blood ow in muscle. SC absorption
and plays a central role in limiting drug uptake by the is delayed with an oil-based vehicle compared with propyl-
brain, thereby protecting against ivermectin neurotoxicity. ene glycol:glycerol-formal due to slower release of the iver-
Many rumino-reticular delivery systems, as well as oral, mectin from the SC depot (Lifschitz et al., 1999b).
topical, and injectable formulations of ivermectin, are cur- Results do however vary. In one study, no dierences
rently available at the dosage recommended by manufac- were observed after SC administration of two commercial
turers, namely, 200 lg/kg in ruminants (500 lg/kg for formulations with the same vehicle (propylene glycol:glyc-
topical application) and equines, 300 lg/kg in pigs, and erol-formal 60:40 v/v) (Lifschitz et al., 1999a), whereas in
6 lg/kg in dogs. This paper reviews the most important another study from the same group there were signicant
aspects of ivermectin pharmacokinetics, including absorp- dierences in the absorption pattern (rate and extent)
tion, distribution, metabolism, and excretion (Fig. 2). between four formulations with the same vehicle (Lifschitz
et al., 2004).
General overview of ivermectin pharmacokinetics and Due to its high lipophilic nature, ivermectin is exten-
metabolism sively distributed with broad volumes of distribution (Vd)
in all species. It tends to accumulate in fat tissue, which acts
Since its introduction in 1981, there have been numerous as a drug reservoir and the highest levels of ivermectin are
pharmacokinetic studies of ivermectin. The drug can be found in liver and fat, and the lowest in brain tissue.
administered by oral, intramuscular (IM), subcutaneous Binding studies in dogs have shown that ivermectin binds
(SC), or topical routes, depending on the species. The phar- extensively to plasma albumin and lipoproteins (Rohrer
macokinetic properties are dose-dependent, with a linear and Evans, 1990), and this should be considered in
increase in the area under the curve (AUC) with increasing undernourished animals or in diseases in which plasma
dose. proteins decrease, as there would be a higher free fraction
The route of administration and the formulation of the drug. Ivermectin persists in the body for a prolonged
strongly aect ivermectins pharmacokinetics. The greatest period, due not only to low plasma clearance but also to
bioavailability is achieved with the SC injection, followed this accumulation in fat tissue. Plasma clearance appears
by the oral route. The lowest AUC values are obtained to be greater in pigs than in polygastric species
after topical administration, even if the dose is 500 lg/kg (goats > sheep > cattle).
A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537 27

Table 1
Ivermectin spectrum of activity in several domestic animals
Animal species Nematodes Arthropods Dose
Cattle Haemonchus spp. Hypoderma spp. 200 lg/kg subcutaneous and
Ostertagia spp. Sarcoptes bovis oral 500 lg/kg topical
Cooperia spp. Psoroptes ovis
Trichostrongylus spp. Linognathus spp.
Strongyloides papillosus; Haematopinus spp.
Bunostomum spp.
Nematodirus spp.
Trichuris spp.
Oesophagostomum spp.
Dictyocaulus viviparus
Sheep Haemonchus spp. Oestrus ovis 200 lg/kg subcutaneous and oral
Chabertia ovina Sarcoptes scabiei
Ostertagia spp. Psoroptes ovis
Cooperia spp. Melanophagus ovinus
Trichostrongylus spp.
Strongyloides papillosus
Bunostomum spp.
Nematodirus spp.
Trichuris ovis
Oesophagostomum spp.
Dictyocaulus laria
Goat Haemonchus spp. Sarcoptes spp. 200 lg/kg subcutaneous
Chabertia ovina Psoroptes ovis
Teladorsagia spp.
Cooperia spp.
Trichostrongylus spp.
Strongyloides papillosus
Oesophagostomum spp.
Dictyocaulus laria
Pig Ascaris suum Sarcoptes scabiei 300 lg/kg subcutaneous
Hyostrongylus rubidus
Strongyloides ransomi Haematopinus suis
Oesophagostomum spp.
Metastrongylus spp.
Stephanurus dentatus
Trichinella spiralis (intestinal)
Horse Strongylus spp. Gasterophilus spp. 200 lg/kg oral
Parascaris equorum Sarcoptes scabiei
Oxyuris equi
Draschia spp.
Habronema spp.
Trichostrongylus axei
Parascaris equorum (microlaria)
Strongyloides westeri
Dictyocaulus arneldi
Onchocerca spp.
Dog Dirolaria immitis Sarcoptes scabiei 6 lg/kg oral
(microlaria and fourth-stage larvae)
Toxocara canis Otodectes cynotis
Toxascaris leonine
Ancylostoma caninum
Uncinaria stenocephala
Trichuris vulpis

Ivermectin undergoes little metabolism; most of the dose H2B1a and 300 -O-desmethyl-H2B1b); 3-O-desmethyl
is excreted unchanged. Metabolic studies have been per- metabolite was found in goats (Alvinerie et al., 1994). In
formed in rats, cattle, sheep, goats, and pigs. The major sheep and cattle, less polar metabolites have been found
metabolites isolated in vivo are 24-OH-H2B1a and 24- in fat tissue, suggesting that in both species liver metabo-
OH-H2B1b in cattle, sheep, and rats (Chiu et al., 1986), lites are esteried with fatty acids and stored in fat as
whereas in pigs O-demethylation derivatives are the major non-polar entities (Chiu et al., 1988). These non-polar
metabolites that have been isolated (300 -O-desmethyl- metabolites have not been described in pigs, as their
28 A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537

Fig. 2. Pharmacokinetics of ivermectin.

hepatic metabolites lack a primary hydroxyl functional There are large interspecies and inter-individual varia-
group, and would be less favourable substrates for esteri- tions in ivermectin pharmacokinetics. Regarding interspe-
cation in fat. cies variability, the AUC values after SC and oral
Ivermectin is mainly eliminated in the faeces in all spe- administration are almost 2.5 times less in sheep compared
cies regardless of the route of administration, and faecal to horses, and the time to reach the maximum plasma con-
excretion accounts for 90% of the dose administered with centration (tmax) is longer (Marriner et al., 1987). On the
<2% of the dose excreted in urine. Bile is the main route other hand, the Vd could inuence plasma concentrations,
of excretion. As P-glycoprotein is also present in biliary as after IV administration the Vd increases in the following
canalicules, it could contribute to the drugs high faecal manner: cattle < sheep and pigs < goats. Inter-individual
excretion (Laont et al., 2002). Ivermectin is also excreted variation can also be attributed to dierences in body con-
by the mammary gland in dairy cows, sheep, and goats; dition, age, sex, and physiological status (McKellar and
this mode of excretion is related to its high lipophilicity. Marriner, 1987; Bogan and McKellar, 1988; Scott et al.,
After intravenous (IV) administration, the plasma elimina- 1990; McKellar et al., 1991; Scott and McKellar, 1992;
tion half-life appears to be longer for ruminant species than Lanusse et al., 1997; Gayrard et al., 1999; Cerkvenik
for monogastric animals. et al., 2002; Barber et al., 2003).
Excretion is also aected by the formulation and is The eect of malnutrition on ivermectin kinetics has
slower in cattle treated SC with non-aqueous vehicles com- been studied in cattle (Lifschitz et al., 1997). When admin-
pared with aqueous vehicles (Table 5); retention in the istered SC, plasma availability was greater in calves with a
body is also increased due to slow absorption from the restricted diet for 21 days compared to cattle fed ad libitum,
injection site (elimination half-lives are 2.0, 3.7, and 8.3 (undernourished: AUC = 443 ng day/mL, Cmax = 53.9 ng/
days with aqueous, aqueous:glycerol-formal 50:50 v/v, mL; ad libitum: AUC = 286 ng day/mL, Cmax = 48.5 ng/
and propylene glycol:glycerol-formal 60:40 v/v formula- mL). Lifschitz et al. (1997) hypothesised that due to the
tions, respectively) (Lo et al., 1985). Furthermore, excre- lipid solubility of ivermectin, the mobilisation of free fatty
tion is slower with an oily solvent compared with acids from adipose tissue could modify the plasma-adipose
propylene glycol:glycerol-formal (Lifschitz et al., 1999b). tissue exchange pattern. Moreover, ivermectin elimination
Even with the same vehicle, however, the elimination is delayed in undernourished (elimination half-life: 9.7
half-life varies depending on the pharmaceutical prepara- days; clearance: 0.465 L/kg day) versus regularly fed calves
tion (Lifschitz et al., 1999a). The half-life of the non-aque- (elimination half-life: 5.6 days; clearance: 0.733 L/kg day),
ous formulation of ivermectin is longer after SC than IV with signicant dierences in the elimination half-life and
administration, reecting the rate-limiting eect of the clearance. Lifschitz et al. (1997) proposed that dietary
absorption process on the drugs overall kinetics (Lo restrictions could reduce bile ow and, subsequently, bili-
et al., 1985; Echeverra et al., 1997). ary excretion of ivermectin.
A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537 29

Table 2
Pharmacokinetic models followed by ivermectin in dierent animal species after the administration of the drug by various routes
Animal species Route Model References
Cattle Intravenous Two-compartmental Lo et al. (1985); Bousquet-Melou et al. (2004);
Echeverria et al. (1997)
Subcutaneous One-compartmental Lifschitz et al. (1999b); Toutain et al. (1988)
Two-compartmental Lanusse et al. (1997)
Intraruminal Two-compartmental Alvinerie et al. (1998)
Topical One-compartmental Gayrard et al. (1999)
Sheep Intravenous Two-compartmental Lo et al. (1985); Gonzalez et al. (2007)
Subcutaneous One-compartmental Cerkvenik et al. (2002); Barber et al. (2003);
Echeverra et al. (2002)
Two-compartmental Marriner et al. (1987); Atta and Abo-Shihada (2000)
Goat Intravenous Two-compartmental Gonzalez et al. (2006)
Subcutaneous One-compartmental Alvinerie et al. (1993)
Intraruminal One or two-compartmentala Escudero et al. (1997)
Pig Intravenous Two-compartmental Craven et al. (2001)
Horse Subcutaneous Two-compartmental Marriner et al. (1987)
Oral Two-compartmental Perez et al. (2002)
Dog Intravenous Two-compartmental Lo et al. (1985)
a
Depending on the individual animal.

McKellar et al. (1991) showed that infestation with chitz et al., 1999b); plasma concentrations of 0.5 ng/mL
Nematodirus battus did not modify the kinetics of ivermec- also control Hypoderma spp. ies (Alvinerie et al., 1994).
tin administered SC or orally to sheep. Nevertheless, Ech- Intra-ruminal administration has also been tested in cat-
everra et al. (2002) observed that infestation with tle and results in a lower and earlier plasma peak concen-
Psoroptes spp. resulted in faster absorption with a higher tration and reduced bioavailability. With a single IR
Cmax value after SC treatment, probably due to the smaller dose, the bioavailability was 26% of that following SC
amount of body fat compared to healthy animals. administration (Chiu et al., 1990a). However, a sus-
The pharmacokinetic model describing the kinetics of tained-release bolus (SRB) that delivered 12 mg/day to
ivermectin varies according to the animal species and route the cattle rumen for 135 days yielded a high steady-state
of administration (Table 2). The choice of one model over concentration (20 ng/mL) between days 4 and 120 after
another involves a consideration of pharmacokinetic treatment, oering a desirable drug-release prole for par-
parameters and the ways in which they are calculated. asite control throughout an entire grazing season (Alviner-
ie et al., 1998).
Species considerations Pour-on formulations are used in cattle, as their applica-
tion is less stressful for handlers and animals. Bioavailabil-
Cattle ity is low and does not exceed 15% of that for SC injection
possibly due to wastage or the drug being trapped in the
Table 3 summarises the pharmacokinetic parameters skin and released very slowly over a longer period of time
calculated for dierent routes of administration in cattle. (Gayrard et al., 1999). Thus, the choice of a SC or pour-on
SC administration is the most studied, and the results show route could be important clinically, as topical formulations
a high degree of variability, which may be due to dier- (with a longer action) would be more eective against most
ences in breed, body condition, number of samples or data sensitive parasite species (D. viviparus or Oesophagostomum
points, methods of quantication, and kinetic treatment of radiatum), whereas SC administration should be considered
the data, or to erratic absorption from the injection site. for less sensitive nematodes (Nematodirus helvetianus or
Despite this variability, Campbell and Benz (1984) and Trichostrongylus colubriformis).
Benz et al. (1989) showed that the plasma concentrations With topical application, attention should be paid to the
achieved in cattle are clinically eective against some spe- animals licking behaviour. Laont et al. (2001) observed
cies of endo- and ectoparasites. It is known that ivermectin that the bioavailability of ivermectin was lower in calves
administered SC has persistent anthelmintic activity when licking was prevented (19%) compared to when it
against most gastrointestinal nematodes, lasting for was not (33%). Thus, with licking, a substantial amount
approximately 10 days, and it is active against Dictyocaulus of topically applied ivermectin could access the systemic
viviparus for 21 days (Barth, 1983; Bremner et al., 1983; circulation via oral consumption resulting in subtherapeu-
Armour et al., 1985). In cattle, plasma concentrations of tic concentrations in untreated and licked animals, which
0.51 ng/mL are required for optimal anthelmintic activity can contribute to the development of resistance. Bous-
against most gastrointestinal and lung nematodes (Lifs- quet-Melou et al. (2004) reported that 6.380.4 lg/kg
30 A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537

Table 3
Absorption pharmacokinetic parameters obtained after ivermectin administration to ruminants
Reference Route Cmax (ng/mL) tmax (h) AUC (ng day/mL) F (%)
Cattle
Lifschitz et al. (1999b)a IM 22.6p 54p 189n,p
Lanusse et al. (1997)e SC 42.8 96 459n
Lo et al. (1985)
Formulation Ab SC 84 24 246(04d) 55
Formulation Bc SC 25 48 186(04d) 41
Formulation Cd SC 13 48 149(04d) 33
Lifschitz et al. (1999b)
Formulation 1a SC 19.9o,p 96o,p 206n,o,p
Formulation 2d SC 35.4o 39.1o 207n,o
Lifschitz et al. (1999a)d
Formulation 3 SC 40.5 48 244n
Formulation 4 SC 46.4 50.9 266n
Lifschitz et al. (2004)d
Formulation D SC 23.6 27.4 231
Formulation E SC 32.7 62 308
Formulation F SC 22 103 262
Formulation G SC 28.4 44.6 242
Chiu et al. (1990a)f SC 133.2 24
Lifschitz et al. (2000) SC 40 24 278n
Echeverria et al. (1997)g SC 33.1 55.9 328.8
Toutain et al. (1988) SC 54.6 34.8
Alvinerie et al. (1998)r IR 28.5 364 247.6 (0160d)
Gayrard et al. (1999)g T 12.2 81.6 121.5(050d)
Laont et al. (2001)
Lickers T 39o 147 595.1o
Non-lickers T 16o 191 381.1o
Sheep
Prichard et al. (1985) IV 375
Gonzalez et al. (2007) IV 197
Bogan and McKellar (1988) SC 32.2 36
McKellar et al. (1991)
Healthy animals SC 30 46 101.7
Parasitized animals SC 35 38.2 175
Cerkvenik et al. (2002)g,h SC 11.9 40.8 64n
Barber et al. (2003)g SC 25.8 29.8 82.1(015d)
Lo et al. (1985)f SC 12 22
Echeverra et al. (2002)g
Healthy animals SC 24.1o 64.1o 207.5o
Parasitized animals SC 41.2o 21.6o 180o
Marriner et al. (1987)e SC 30.8 60 238
Atta and Abo-Shihada (2000)e SC 16.3 62.4 281n
Gonzalez et al. (2007) SC 19.6 3.1 190.7 98.2
Bogan and McKellar (1988)
Ewes O 14.7 24 36.3(07d)
Lambs O 23.6 36 93.7(07d)
McKellar et al. (1991)
Healthy animals O 29 19.3 74.6
Parasitized animals O 21 20 88.8
Mestorino et al. (2003)
Solution O 11.3 31.9 44.7
Tablets O 8.5 43.9 52
Marriner et al. (1987)e O 22 16.4 85 35.7l
Chiu et al. (1990a)f IR 12.5 24
Prichard et al. (1985) IR 17.6 23.5 94.2 25.1m
Prichard et al. (1985) IAB 60.6 4.8 440
Goat
Gonzalez et al. (2006)e IV 153
Gonzalez et al. (2006) SC 21.8 72 144 91.8
A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537 31

Table 3 (continued)
Reference Route Cmax (ng/mL) tmax (h) AUC (ng day/mL) F (%)
Alvinerie et al. (1993)g,h SC 6.1 68.4 60
Scott et al. (1990)h O 15.9 24 21.5
Escudero et al. (1997)h,p,i IR 9.3 31.2 34.4
Escudero et al. (1997)h,k IR 10.6 29 34.6
Scott et al. (1990)h T 3.9 48 13.2
Cmax = maximum plasma concentration; tmax = time to reach Cmax; AUC = area under the plasma concentrationtime curve; F = bioavailability;
d = day(s); = unknown data.
IM = intramuscular; SC = subcutaneous; IR = intraruminal; T = topical; IV = intravenous (200 lg/kg); O = oral; IAB = intra-abomasal. Doses are
always those recommended by manufacturers, except if indicated.
a
Oily vehicle.
b
Aqueous vehicle.
c
Aqueous-glycerol-formal vehicle (50:50, v/v).
d
Propyleneglycol:glycerol-formal vehicle (60:40, v/v).
e
Two-compartmental model.
f
300 lg/kg.
g
One-compartmental model.
h
Lactating animals.
i
Animals fasted for 36 h before ivermectin administration.
k
Animals fed ad libitum.
l
Relative to subcutaneous.
m
Relative to intra-abomasal.
n
AUC01; if other, it is indicated as superscript in brackets.
o
Signicant dierences within the study.
p
Signicant dierences within the study.
r
Sustained release bolus.

(1.316.1% of a pour-on dose) was ingested by untreated the mammary gland and because of these high concentra-
cattle licking treated cattle. tions milk from dairy cows treated with ivermectin must
The distribution of ivermectin is slow in cattle but be excluded from human consumption.
broad, as demonstrated by the high Vd and the high mean Chiu et al. (1990a) found that with IR treatment, the
residence time (MRT) (Table 5). When administered SC percentage of ivermectin excreted in faeces and urine 7 days
and IR, [3H]ivermectin was detected in all sampled tissues: after administration was 79.7% and 0.5%, respectively, and
the highest concentrations were found in liver and fat; high its concentration in bile was high (273 ng/mL). Alvinerie
levels were also recorded in the kidney and muscle (Chiu et al. (1998) reported that 8090% of drug delivered via
et al., 1990a). Availability was higher in tissues where par- SRB systems was excreted faecally and the drug was
asites usually reside than in plasma at 167%, 163%, and detected in faeces until day 160. This persistent excretion
244% in lungs, intestinal mucosae, and abomasal mucosae, could pose a threat to the ecosystem (through, for example,
respectively, and persisted in most organs for 48 days (Lifs- dung-breeding/dung-feeding invertebrates).
chitz et al., 2000). This could explain the strong ecacy of Following topical application, ivermectin has a longer
ivermectin against parasites in these locations. plasma half-life in cattle prevented from licking (15.1 days)
As in other species, ivermectin metabolism is minimal. than in those that are permitted to lick themselves (6.4
In studies using radiolabelled ivermectin, unchanged drug days); the half-life is also longer than after IV injection (6
represented 52% of the radioactivity in liver and fat (day days) reecting slow absorption through the skin, which
14 after SC treatment). Twenty-eight days after administra- limits later elimination. Laont et al. (2001) measured
tion, these levels decreased to 40% in liver and 19% in fat 69% of the dose in faeces from lickers after 28 days of treat-
(Chiu et al., 1986). Liver metabolites were hydroxylated ment, and only 6.6% of the dose in faeces from non-lickers.
derivatives of ivermectin (Chiu et al., 1986), whereas non- These results are consistent with ivermectin transiting
polar metabolites were detected in fat tissue (Chiu et al., directly through the digestive tract into faeces, which con-
1988). In another study, such non-polar derivatives repre- tributes greatly to the drugs faecal output (Laont et al.,
sented 64% of the radioactivity detected in fat on day 28 2001).
after treatment, and lengthened the depletion time of the
residues in fat compared to residues in liver (Chiu et al., Sheep
1990a).
After SC injection, Chiu et al. (1990a) found that on day Plasma levels of ivermectin are lower in sheep than in
7 after treatment, 1.5% and 62% of ivermectin was found in cattle. The SC bioavailability is highly variable, ranging
urine and faeces, respectively (Chiu et al., 1990a). Toutain from 22% (Lo et al., 1985) to 98.2% (Gonzalez et al.,
et al. (1988) found that 5.5% of a SC dose was secreted via 2007). Plasma concentrations are lower after oral versus
32 A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537

SC administration (McKellar and Marriner, 1987). Thus, (2002) found that only 2.1% of the dose was transferred
as in other animal species, oral administration yields by treated ewes; this is lower than the oral value (10%).
poorer ecacy and a shorter duration of action. Furthermore, the plasma concentration derived from trea-
As expected, absorption of ivermectin is faster after oral ted dams was only 4% of that found in the same lambs trea-
administration of a solution versus tablets (Mestorino ted orally. Although this seems low, it could have benecial
et al., 2003). The Cmax and AUC obtained after oral eects for lambs due to the high ecacy of ivermectin at a
administration was greater in lambs versus ewes, probably low dosage. On the other hand, treatment of ewes over the
reecting dierences in body composition (especially fat periparturient period has been recommended to reduce fae-
content) and to impaired elimination in lambs (Bogan cal egg output and after only one SC dose this reduced fae-
and McKellar, 1988). Ali and Hennessy (1996) demon- cal output persists for approximately 1 week.
strated that reducing feed intake for 24 h before IR admin-
istration could be a valid option to ensure the ecacy of Goats
ivermectin, as it should increase the drugs bioavailability
and extend its residence time. Studies in goats are limited but plasma levels tend to be
Distribution in the sheep (Table 5) is faster and broader lower than those obtained in cattle and in sheep (Table 3).
than in cattle or dogs (Lo et al., 1985) due to substantial The SC bioavailability is very high (91.8%) (Gonzalez
deposition into adipose tissue, which may act as a drug et al., 2006) and Scott et al. (1990) demonstrated that the
depot (Prichard et al., 1985). The larger fat reservoir in bioavailability of topically administered ivermectin was
sheep compared to cattle could contribute to not only the 61.5% of that found when the drug was orally adminis-
more extensive distribution but also the greater persistence tered; persistence in plasma was, however, more prolonged
in plasma at lower concentrations, probably because less following the percutaneous route.
blood is supplied to fatty tissues (Atta and Abo-Shihada, Ivermectin associates with lipoproteins in goats, prefer-
2000). Likewise, the higher Vd in sheep versus cattle corre- entially high density lipoprotein (88.1%), with binding per-
lates well with the lower plasma concentrations observed in centages of 7.3% for low density lipoprotein, 1.8% for very
sheep. low density lipoprotein, and 2.7% for albumin and a-1 gly-
As with cattle, Chiu et al. (1986) reported that coprotein (Bassissi et al., 2004). This extensive binding to
unchanged ivermectin represented >50% of the radioactiv- lipoproteins could aect the delivery of ivermectin to fat
ity in liver and fat, but in this case on day 3 after IR admin- tissue and consequently relate to its extended presence in
istration. Ivermectin levels decrease faster in sheep than in the body.
cattle (Chiu et al., 1986). The main metabolites isolated in Total body clearance after IV administration (Table 5)
the liver are the same in the two species and accounted for demonstrated the slow elimination process in goats (Gonz-
55% of the radioactivity on day 7 after IR treatment. A sig- alez et al., 2006). Excretion in milk is even lower than in
nicant rst-pass eect was not evident in sheep, as intra- sheep with only 0.31% of the dose recovered in milk 25
abomasal bioavailability was 100% (Prichard et al., 1985). days after SC treatment (Alvinerie et al., 1993). Scott
Prichard et al. (1985) reported that IR administration in et al. (1990) observed that the concentrations excreted in
sheep resulted in a low bioavailability (25%), similar to that milk were similar after oral and topical administration.
obtained in cattle. They also proposed that the ruminal
microora metabolise ivermectin, as 50% disappeared from Pigs
the rumen uid after 2-h incubation. Andrew and Halley
(1996) however attributed this disappearance to the high In pigs treated SC (Table 4), the Cmax and AUC are sig-
level of binding to solids and surfaces. More recently, Lifs- nicantly lower than in calves; this could be related to the
chitz et al. (2005) conrmed that ivermectin was thor- higher distribution and deposition of the drug in fat tissue,
oughly bound to solid ruminal contents (> 90%) without which diminishes plasma levels in this animal species (Lifs-
suering degradation. chitz et al., 1999a). The inuence of body fat levels on iver-
The IV half-life of ivermectin is similar in sheep and cat- mectin kinetics has been investigated in pigs, but the results
tle (Table 5); thus, the lower plasma levels in sheep are due are not clear. Craven et al. (2001) reported that fat content
to a broader distribution rather than to faster elimination had no detectable inuence on ivermectin disposition and
(Lo et al., 1985). In sheep, concentrations in milk are sim- they found no signicant dierences in the pharmacoki-
ilar to those in plasma (Bogan and McKellar, 1988), and netic parameters representative of the distribution process
only 0.71% of a SC dose was excreted through milk (less between two groups of pigs with dierent body conditions
than in cattle, probably due to species dierences in the vol- (Table 6). In another study, these workers compared two
ume and fat content of milk). However, Cerkvenik et al. groups of animals diering in back-fat thickness and
(2001) observed that ivermectin remains stable following weight (71.6 and 38.3 kg) and found that absorption was
thermal treatment, conrming that residues in dairy prod- slower and availability higher in fat pigs. Plasma levels
ucts would be an issue for consumers. were >2 ng/mL until 18 days after treatment in fat pigs
Indirect exposure of untreated sucking lambs to iver- and 11 days in thin pigs, suggesting a longer period of drug
mectin via milk ingestion is negligible; Cerkvenik et al. ecacy (Craven et al., 2002a). When animals with an inter-
A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537 33

Table 4 domestic species have been found in hepatic and fat metab-
Absorption pharmacokinetic parameters obtained after ivermectin admin- olism; O-demethylation products were the metabolites
istration to monogastric species
found in liver. In contrast to other species, the same deriv-
Reference Route Cmax tmax AUC atives were also present in fat, accounting for the similar
(ng/mL) (h) (ng d/mL)
elimination half-lives (5 days) of residues from liver and
Pigs fat in swine (Chiu et al., 1990).
Scott and McKellar (1992) SC 28.4 27.2 71.41
Lifschitz et al. (1999a)a
Taking into account the half-life, the disappearance of
Formulation 1 SC 33.3 66d 165c ivermectin from plasma (Table 6) is faster in pigs than in
Formulation 2 SC 39.6 22.6d 132c cattle or sheep (Lo et al., 1985), suggesting briefer protec-
Craven et al. (2002a) tion against parasites in this animal species. Clearance is
Animals weighing 38.3 kg SC 9.7 33.2d 85.7d also higher than in ruminant species, which correlates well
Animals weighing 71.6 kg SC 7.4 71.9d 111.7d with the shorter half-life in pigs. On the other hand, body
Craven et al. (2002b) condition does not aect clearance when ivermectin is
Animals weighing 51 kg SC 8 75.1 70.5 administered IV (Craven et al., 2001) or SC (Craven
Animals weighing 60 kg SC 7.2 48 87.7 et al., 2002a,b). Chiu et al. (1990b) reported that on day
7 after SC treatment, the concentrations excreted in faeces
Horse
and urine were half those found in cattle (30% and 0.6% of
Marriner et al. (1987)b SC 60.7 80 550.4
Marriner et al. (1987) O 82.3 3.1 200.9 the dose, respectively). One day after treatment, high con-
Gokbulut et al. (2001) O 21.4 7.9 centrations were found in bile (210 ng/mL) and faeces
Perez et al. (2002)b O 51.3 3.6 137.1(0-30d) (178.5 ng/g) (Scott and McKellar, 1992).
Donkey
Horses
Gokbulut et al. (2005) O 23.6 24.0 119.3

Dog In contrast to ruminants, the absorption process in


Daurio et al. (1992) horses is faster after oral versus SC administration. More-
Standard tablet O 2.97 5.3 4.5 over, although SC injection results in greater bioavailabil-
Chewable tablet O 3.37 8.5 5.5
ity than does oral administration (AUCoral = 36.5% of
Daurio et al. (1992)e
Standard tablet O 44.3 4.2 43.1 the AUCsc), the oral route is preferred, as parenteral
Modied tablet (crystalline) O 48.4 3.8 41.7 administration can produce local swelling and other
Cmax = maximum plasma concentration; tmax = time to reach Cmax; adverse reactions (Anderson, 1984). Plasma concentrations
AUC = area under the plasma concentrationtime curve; = unknown are higher and more rapidly achieved in horses compared
data. SC = subcutaneous; O = oral; d = day(s). Doses are always those to sheep (Marriner et al., 1987) (Table 4), probably because
recommended by manufacturers, except if indicated. the rumen delays absorption in ruminant species. Never-
a
Propyleneglycol:glycerol-formal vehicle.
b theless, the elimination half-lives after SC and oral treat-
Two-compartmental model.
c
AUC01; if other, it is indicated as superscript in brackets. ment are 3.7 and 2.8 days, respectively, and similar to
d
Signicant dierences within the study. sheep (Marriner et al., 1987).
e
100 lg/kg. In horses, the MRT is also longer after oral administra-
tion (4.2 days) (Perez et al., 2002) versus SC injection (3
days) (Gokbulut et al., 2001); it is longer still in donkeys
mediate bodyweight were used, no dierences in kinetic dis- treated orally with ivermectin (6.5 days) (Gokbulut et al.,
position were observed (Craven et al., 2002b). 2005), where the elimination of ivermectin is slower, with
A higher Vd after IV administration was observed in a half-life of 7.4 days (Gokbulut et al., 2005). In horses
pigs when compared to sheep or cattle. Ivermectin also dis- treated SC, most of the dose (90%) is faecally excreted in
tributed widely in this species after SC administration, with 4 days. The higher concentrations found in equine faeces
the highest levels in liver and fat (Chiu et al., 1990b). compared to cattle faeces have been attributed to a lower
Twenty-four hours after injection, a large amount remains production of more concentrated faeces (Perez et al., 2001).
at the injection site, indicating a slow release process. Iver-
mectin has been detected at all levels of the gastrointestinal Dogs
tract (contents and mucus), with high concentrations in the
lungs, skin, and also earwax (accounting for its eective- In dogs, the oral route is preferred for heartworm pre-
ness against ectoparasites, particularly ear mites) (Scott vention. Absorption of ivermectin is faster in dogs than
and McKellar, 1992). in ruminants and pigs, and similar to horses. Peak plasma
Chiu et al. (1990) reported that on day 7 the parent drug levels are attained in 35 h (Table 4). Oral bioavailability is
represented 45% of radioactivity in the liver and the per- greater if the tablets are chewable. The amount absorbed
centage was slightly higher in fat tissue (63%). These levels follows a linear dose-relationship, as Cmax and AUC
decreased to 30% and 35%, respectively, on day 14 after increase proportionally with dose (Daurio et al., 1992).
drug administration. Dierences with respect to other The Vc (volume of distribution in the central compart-
34 A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537

Table 5
Distribution and elimination pharmacokinetic parameters obtained after ivermectin administration to ruminants
Reference Route Vd (L/kg) t1/2a (d) t1/2b (d) MRT (d) t1/2 (d) Cl (L/kg d)
Cattle
Lo et al. (1985)a,b IV 1.9l 2.8
Echeverra et al. (1997)b IV 1.2m 3.4
Laont et al. (2001) IV 6 0.27
Bousquet-Melou et al. (2004)c IV 2.7m 8.1 7.8 0.35
Lifschitz et al. (1999b)d IM 5.2k
Chiu et al. (1990a)a SC 4.3
Echeverra et al. (1997)d SC 5.7
Lanusse et al. (1997)b SC 3.4m 4.2 17.2 7.4 0.48i
Lifschitz et al. (1999b)
Formulation 1d SC 5.9i
Formulation 2e SC 3.99i,k
Lifschitz et al. (1999a)e
Formulation 3 SC 5.3
Formulation 4 SC 6.3
Lifschitz et al. (2000) SC 5.8
Toutain et al. (1988)f,g SC 6.5
Chiu et al. (1990a) IR 3.7
Gayrard et al. (1999) T 8.4

Sheep
Lo et al. (1985)b IV 4.6l 2.7
Prichard et al. (1985) IV 5.3m 7.4 0.56
Gonzalez et al. (2007) IV 3.0l 0.7 9.6 10.3 1.11
Marriner et al. (1987)b SC 3.7
Atta and Abo-Shihada (2000)b SC 5.9 7
Cerkvenik et al. (2002)f,g SC 12.8m 5.2 2.9 3.24i
Echeverra et al. (2002)f
Healthy animals SC 8.8n 8.6 5.6
Parasitized animals SC 6.5n 6.7 5.5
Barber et al. (2003)f SC 1.7
Gonzalez et al. (2007) SC 17.6n 10.3 11 1.11
Marriner et al. (1987)b O 2.6
Atta and Abo-Shihada (2000) O 2.1
Mestorino et al. (2003)
Solution O 3.45 3.6
Tablets O 3.78 3.7
Chiu et al. (1990a)a IR 2.4
Prichard et al. (1985) IR 4.3

Goats
Gonzalez et al. (2006)b IV 2.8l 0.7 7.4 1.56
Gonzalez et al. (2006) SC 12.8n 8.3 5.6 1.43
Alvinerie et al. (1993)m,g SC 7.9 4.03
Escudero et al. (1997)g IR 2.62.8 1.181.24
Vd = volume of distribution; Vss = volume of distribution at steady state; t1/2a = half-life associated with a phase; t1/2b = half-life associated with b phase;
MRT = mean residence time; t1/2 = half-life; Cl = total body clearance; = unknown data. IV = intravenous; IM = intramuscular; SC = subcutaneous;
IR = intraruminal; T = topical; O = oral; d = day. Doses are always those recommended by manufacturers, except if indicated.
h
VSS/F.
a
300 lg/kg.
b
Two-compartment model.
c
70 lg/kg.
d
Oily vehicle.
e
Propyleneglycol:glycerol-formal vehicle (60:40, v/v).
f
One-compartment model.
g
Lactating animals.
i
Signicant dierences within the study.
k
Signicant dierences within the study.
l
Vc (volume of distribution in the central compartment).
m
Vss.
n
Va (volume of distribution of the area).
A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537 35

Table 6
Distribution and elimination pharmacokinetic parameters obtained after ivermectin administration to pigs
Reference Route Vc (L/kg) t1/2a (d) t1/2b (d) MRT (d) t1/2 (d) Cl (L/kg d)
a
Craven et al. (2001)
Animals weighing 28.5 kg IV 2.7 (5.1*) 0.14 1.18 0.5b 4.15
Animals weighing 41.7 kg IV 2.1 (5.3*) 0.15 1.33 0.7b 4.01
Lo et al. (1985) SC 0.5
Scott and McKellar (1992) SC 1.5
Lifschitz et al. (1999a) SC 3.53.8
Craven et al. (2002a)
Animals weighing 38.3 kg SC 8.1 3.55
Animals weighing 71.6 kg SC 9.8 2.75
Craven et al. (2002 b)
Animals weighing 50 kg SC 8.4 2.28 4.47
Animals weighing 60 kg SC 9.6 2.55 3.64
Vc; = volume of distribution in the central compartment; t1/2a = half-life associated with a phase; t1/2b = half-life associated with b phase; MRT = mean
residence time; t1/2 = half-life; Cl = total body clearance.
a
Two-compartmental model.
b
Signicant dierences within the study; = unknown data. IV = intravenous; SC = subcutaneous. Doses are always those recommended by man-
ufacturers, except if indicated.
*
Vss = volume of distribution at steady state.

ment) is 2.4 L/kg in dogs injected IV, intermediate between Andrew, N.W., Halley, B.A., 1996. Stability of ivermectin in rumen uids.
values obtained in cattle and sheep (Lo et al., 1985). On the Journal of Veterinary Pharmacology and Therapeutics 19, 295299.
Armour, J., Bairden, K., Batty, A.F., Davison, C.C., Ross, D.B., 1985.
other hand, excretion is more rapid in dogs versus cattle or Persistent anthelmintic activity of ivermectin in cattle. The Veterinary
sheep (IV elimination half-life = 1.8 days) (Lo et al., 1985). Record 116, 151153.
Atta, A.H., Abo-Shihada, M.N., 2000. Comparative pharmacokinetics of
Conclusions doramectin and ivermectin in sheep. Journal of Veterinary Pharma-
cology and Therapeutics 23, 4952.
Barber, S., Bowles, V., Lespine, A., Alvinerie, M., 2003. The comparative
Although the ecacy of ivermectin has been established serum disposition kinetics of subcutaneous administration of dora-
across a variety of domestic species, its pharmacokinetic mectin, ivermectin and moxidectin in the Australian merino sheep.
properties dier between them, and the factors responsible Journal of Veterinary Pharmacology and Therapeutics 26, 343348.
for modifying ivermectins pharmacokinetics should be Barth, D., 1983. Persistent anthelmintic eect of ivermectin in cattle. The
taken into account to ensure its clinical ecacy, prevent Veterinary Record 113, 300.
Bassissi, M.F., Alvinerie, M., Lespine, A., 2004. Macrocyclic lactones:
subtherapeutic levels, and minimise the development of distribution in plasma lipoproteins of several animal species including
resistance. humans. Comparative Biochemistry and Physiology 138, 437444.
Benz, G.W., Roncalli, R.A., Gross, S.J., 1989. Use of ivermectin in cattle,
References sheep, goats, and swine. In: Campbell, W.C. (Ed.), Ivermectin and
Abamectin. Springer-Verlag, New York, pp. 215229.
Ali, D.N., Hennessy, D.R., 1996. The eect of level of feed intake on Bogan, J.A., McKellar, Q.A., 1988. The pharmacodynamics of ivermectin
the pharmacokinetic disposition and ecacy of ivermectin in in sheep and cattle. Journal of Veterinary Pharmacology and Ther-
sheep. Journal of Veterinary Pharmacology and Therapeutics 19, apeutics 11, 260268.
8994. Bousquet-Melou, A., Mercadier, S., Alvinerie, M., Toutain, P.L., 2004.
Alvinerie, M., Sutra, J.F., Galtier, P., 1993. Ivermectin in goat plasma and Endectocide exchanges between grazing cattle after pour-on adminis-
milk after subcutaneous injection. Annales de Recherches Veterinaires tration of doramectin, ivermectin and moxidectin. International
24, 417421. Journal of Parasitology 34, 12991307.
Alvinerie, M., Sutra, J.F., Galtier, P., Lifschitz, A., Virkel, G., Sallovitz, Bremner, K.C., Berrie, D.A., Hotson, I.K., 1983. Persistence of the
J., Lanusse, C., 1998. Persistence of ivermectin in plasma and faeces anthelmintic activity of ivermectin in calves. The Veterinary Record
following administration of a sustained-release bolus to cattle. 113, 569.
Research in Veterinary Science 66, 5761. Campbell, W.C., 1989. Ivermectin and Abamectin. Springer-Verlag, New
Alvinerie, M., Sutra, J.F., Galtier, P., Toutain, P.L., 1994. Microdose York.
divermectine chez la vaca laitiere: concentrations plasmatiques et Campbell, W.C., Benz, G.W., 1984. Ivermectin: a review of ecacy
residus dans le lait. Recueil de Medecine Veterinaire 145, 761764. and safety. Journal of Veterinary Pharmacology and Therapeutics
Alvinerie, M., Tardieu, D., Sutra, J.F., Bojensen, G., Galtier, P., 1994. 7, 116.
Metabolic prole of ivermectin in goats: an in vivo and in vitro Cerkvenik, V., Doganoc, D.Z., Skubic, V., Beek, W.M.J., Keukens,
evaluation. In: Lees, P. (Ed.), European Association for Veterinary H.J., 2001. Thermal and long-term freezing stability of ivermectin
Pharmacology and Toxicology. Proceedings of the 6th International residues in sheep milk. European Food Research and Technology
Congress. Blackwell Scientic Publications, Edinburgh, p. 262. 213, 7276.
Anderson, R.R., 1984. The use of ivermectin in horses: research and Cerkvenik, V., Grabnar, I., Skubic, V., Doganoc, D.Z., Beek, W.M.,
clinical observations. Compendium on Continuing Education 6, S517 Keukens, H.J., Drobnic, M., Pogacnik, M., 2002. Ivermectin phar-
S520. macokinetics in lactating sheep. Veterinary Parasitology 104, 175185.
36 A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537

Chiu, S.H.L., Carlin, R., Taub, R., Sestokas, E., Zweig, J., Vandenheuvel, Lanusse, C., Lifschitz, A., Virkel, G., Alvarez, L., Sanchez, S., Sutra,
W.J., Jacob, T.A., 1988. Comparative metabolic disposition of J.F., Galtier, P., Alvinerie, M., 1997. Comparative plasma
ivermectin in fat tissues of cattle, sheep, and rats. Drug Metabolism disposition kinetics of ivermectin, moxidectin and doramectin in
and Disposition 16, 728736. cattle. Journal of Veterinary Pharmacology and Therapeutics 20,
Chiu, S.H.L., Green, M.L., Baylis, F.P., Eline, D., Rosegay, A., 9199.
Meriwether, H., Jacob, T.A., 1990a. Absorption, tissue distribu- Lifschitz, A., Murno, G., Pis, A., Sallovitz, J., Virkel, G., Lanusse, C.,
tion, and excretion of tritium-labeled ivermectin in cattle, sheep, 1997. Malnutrition modies the disposition kinetics of ivermectin in
and rat. Journal of Agricultural and Food Chemistry 38, 2072 calves. Journal of Veterinary Pharmacology and Therapeutics 20, 71
2078. 72.
Chiu, S.H.L., Sestokas, E., Taub, R., Buhs, R.P., Green, M., Sestokas, R., Lifschitz, A., Pis, A., Alvarez, L., Virkel, G., Sanchez, S., Sallovitz, J.,
Vandenheuvel, W.J., Arison, B.H., Jacob, T.A., 1986. Metabolic Kujanek, R., Lanusse, C., 1999a. Bioequivalence of ivermectin
disposition of ivermectin in tissues of cattle, sheep, and rats. Drug formulations in pigs and cattle. Journal of Veterinary Pharmacology
Metabolism and Disposition 14, 590600. and Therapeutics 22, 2734.
Chiu, S.H.L., Sestokas, E., Taub, R., Green, M.L., Baylis, F.P., Jacob, Lifschitz, A., Sallovitz, J., Imperiale, F., Pis, A., Lorda, J., Lanusse, C.,
T.A., Lu, A.Y.H., 1990b. Metabolic disposition of ivermectin in swine. 2004. Pharmacokinetic evaluation of four generic formulations in
Journal of Agricultural and Food Chemistry 38, 20792085. calves. Veterinary Parasitology 119, 247257.
Craven, J., Bjrn, H., Hennesy, D., Friis, C., Nansen, P., 2001. Lifschitz, A., Virkel, G., Ballent, M., Sallovitz, J., Pis, A., Lanusse, C.,
Pharmacokinetics of moxidectin and ivermectin following intravenous 2005. Moxidectin and ivermectin metabolic stability in sheep ruminal
injection in pigs with dierent body compositions. Journal of Veter- and abomasal contents. Journal of Veterinary Pharmacology and
inary Pharmacology and Therapeutics 24, 99104. Therapeutics 28, 411418.
Craven, J., Bjrn, H., Hennesy, D.R., Friis, C., 2002a. The eects of body Lifschitz, A., Virkel, G., Pis, A., Imperiale, F., Sanchez, S., Alvarez, L.,
composition on the pharmacokinetics of subcutaneously injected Kujanek, R., Lanusse, C., 1999b. Ivermectin disposition kinetics after
ivermectin and moxidectin in pigs. Journal of Veterinary Pharmacol- subcutaneous and intramuscular administration of an oil-based
ogy and Therapeutics 25, 227232. formulation to cattle. Veterinary Parasitology 86, 203215.
Craven, J., Hennessy, D.R., Friis, C., 2002b. Does the rate of fat Lifschitz, A., Virkel, G., Sallovitz, J., Sutra, J.F., Galtier, P., Alvinerie,
deposition inuence the pharmacokinetic disposition of subcutane- M., Lanusse, C., 2000. Comparative distribution of ivermectin and
ously administered moxidectin and ivermectin in pigs? Journal of doramectin to parasite location tissues in cattle. Veterinary Parasitol-
Veterinary Pharmacology and Therapeutics 25, 351357. ogy 87, 327338.
Daurio, C.P., Cheung, E.N., Jecoat, A.R., Skelly, B.J., 1992. Bioavail- Lo, P.K.A., Fink, D.W., Williams, J.B., Blodinger, J., 1985. Pharmaco-
ability of ivermectin administered orally to dogs. Veterinary Research kinetic studies of ivermectin: eects of formulation. Veterinary
Communications 16, 125130. Research Communications 9, 251258.
Echeverra, J., Mestorino, N., Errecalde, J., 2002. Comparative pharma- Marriner, S.E., McKinnon, I., Bogan, J.A., 1987. The pharmacokinetics
cokinetics of ivermectin after its subcutaneous administration in of ivermectin after oral and subcutaneous administration to sheep and
healthy sheep and sheep infected with mange. Journal of Veterinary horses. Journal of Veterinary Pharmacology and Therapeutics 10, 175
Pharmacology and Therapeutics 25, 159160. 179.
Echeverra, J., Mestorino, N., Giorgieri, S., Turic, E., Alt, M., Errecalde, McKellar, Q.A., Benchaoui, H.A., 1996. Avermectins and milbemyc-
J., 1997. Pharmacokinetics of ivermectin after its intravenous and ins. Journal of Veterinary Pharmacology and Therapeutics 19,
subcutaneous administration to cattle. Journal of Veterinary Pharma- 331351.
cology and Therapeutics 20, 7778. McKellar, Q.A., Jackson, F., Coop, R.L., Jackson, E., Scott, E., 1991.
Escudero, E., Carceles, C.M., Galtier, P., Alvinerie, M., 1997. Inuence of Eect of parasitism with Nematodirus battus on the pharmacokinetics
fasting on the pharmacokinetics of ivermectin in goats. Journal of of levamisole, ivermectin, and netobimin. Veterinary Parasitology 39,
Veterinary Pharmacology and Therapeutics 20, 7172. 123136.
Gayrard, V., Alvinerie, M., Toutain, P.L., 1999. Comparison of pharma- McKellar, Q.A., Marriner, S.E., 1987. Comparison of the anthelmintic
cokinetic proles of doramectin and ivermectin pour-on formulations ecacy of oxfendazole or ivermectin administered orally and ivermec-
in cattle. Veterinary Parasitology 81, 4755. tin administered subcutaneously to sheep during the periparturient
Gokbulut, C., Boyacioglu, M., Karademir, U., 2005. Plasma pharmaco- period. The Veterinary Record 18, 383386.
kinetics and faecal excretion of ivermectin (Eqvalan paste) and Mestorino, N., Turic, E., Pesoa, J., Echeverra, J., Errecalde, J.O., 2003.
doramectin (Dectomax, 1%) following oral administration in don- Pharmacokinetics in plasma of ivermectin after its oral (solution and
keys. Research in Veterinary Science 79, 233238. tablets) administration to sheep. Journal of Veterinary Pharmacology
Gokbulut, C., Nolan, A.M., McKellar, Q.A., 2001. Plasma pharmacoki- and Therapeutics 26, 307309.
netics and faecal excretion of ivermectin, doramectin and moxidectin Molento, M.B., Lifschitz, A., Sallovitz, J., Lanusse, C., Prichard, R., 2004.
following oral administration in horses. Equine Veterinary Journal 33, Inuence of verapamil on the pharmacokinetics of the antiparasitic
494498. drugs ivermectin and moxidectin in sheep. Parasitology Research 92,
Gonzalez, A., Sahagun, A., Diez, M.J., Fernandez, N., Sierra, M., Garcia, 121127.
J.J., 2007. Bioavailability of a commercial formulation of ivermectin Perez, R., Cabezas, I., Godoy, C., Rubilar, L., Munoz, L., Arboix, M.,
after subcutaneous administration to sheep. American Journal of Castells, G., Alvinerie, M., 2002. Pharmacokinetics of doramectin and
Veterinary Research 68, 101106. ivermectin after oral administration in horses. The Veterinary Journal
Gonzalez, A., Sahagun, A.M., Diez, M.J., Fernandez, N., Sierra, M., 163, 161167.
Garcia, J.J., 2006. Pharmacokinetics of a novel formulation of Perez, R., Cabezas, I., Sutra, J.F., Galtier, P., Alvinerie, M., 2001. Fecal
ivermectin after administration to goats. American Journal of Veter- excretion prole of moxidectin and ivermectin after oral administra-
inary Research 67, 323328. tion in horses. The Veterinary Journal 161, 8592.
Laont, C.M., Alvinerie, M., Bousquet-Melou, A., Toutain, P.L., 2001. Prichard, R.K., Steel, J.W., Lacey, E., Hennessy, D.R., 1985. Pharmaco-
Licking behaviour and environmental contamination arising from kinetics of ivermectin in sheep following intravenous, intra-abomasal
pour-on ivermectin for cattle. International Journal of Parasitology 3, or intraruminal administration. Journal of Veterinary Pharmacology
16871692. and Therapeutics 8, 8894.
Laont, C.M., Toutain, P.L., Alvinerie, M., Bousquet-Melou, A., 2002. Rohrer, S.P., Evans, D.V., 1990. Binding characteristics of ivermectin in
Intestinal secretion is a major route for parent ivermectin elimination plasma from Collie dogs. Veterinary Research Communications 14,
in the rat. Drug Metabolism and Disposition 30, 626630. 157165.
A. Gonzalez Canga et al. / The Veterinary Journal 179 (2009) 2537 37

Scott, E.W., Kinabo, L.D., McKellar, Q.A., 1990. Pharmacokinetics of Steel, J.W., 1993. Pharmacokinetics and metabolism of avermectins in
ivermectin after oral administration to adult milking goats. Journal of livestock. Veterinary Parasitology 48, 4547.
Veterinary Pharmacology and Therapeutics 13, 432435. Toutain, P.L., Campan, M., Galtier, P., Alvinerie, M., 1988. Kinetic
Scott, E.W., McKellar, Q.A., 1992. The distribution and some pharma- and insecticidal properties of ivermectin residues in the milk of
cokinetic parameters of ivermectin in pigs. Veterinary Research dairy cows. Journal of Veterinary Pharmacology and Therapeutics
Communications 16, 139146. 11, 288291.

Das könnte Ihnen auch gefallen