You are on page 1of 8

Paper 172 Disc

J. vet. Pharmacol. Therap. 22, 27±34, 1999. PHARMACOKINETICS

Bioequivalence of ivermectin formulations in pigs and cattle

A . L IF SC H IT Z * Lifschitz, A., Pis, A., Alvarez, L., Virkel, G., Sanchez, S., Sallovitz, J., Kujanek, R.,
Lanusse, C. Bioequivalence of ivermectin formulations in pigs and cattle. J. vet.
A. PIS*
Pharmacol. Therap. 22, 27±34.
L. ALVAREZ*
G . V IR K E L * The vehicle in which endectocide compounds are formulated plays a relevant
S . SAN CH E Z * role in their absorption kinetics and resultant systemic availability. The
J. S A L LO V IT Z * pharmaceutical bioequivalence and comparative plasma disposition kinetics of
ivermectin (IVM), following the subcutaneous administration of two injectable
R. K U J A N E K { &
formulations to pigs and cattle were investigated using parallel experimental
C. LANUSSE* designs. Sixteen parasite-free male Duroc Jersey-Yorkshire crossbred pigs (90±
110 kg) (Expt 1) and 16 parasite-free male Holstein calves (100±120 kg) (Expt
*Laboratorio de FarmacologõÂa, Departamento
2) were divided into two groups and treated subcutaneously at either 300 (pigs)
de FisiopatologõÂa, Facultad de Ciencias
or 200 (calves) mg/kg with two different propylene glycol/glycerol formal (60:
Veterinarias, Universidad Nacional del
40) based IVM formulations; in both experiments pigs or calves in Group A
Centro, Campus Universitario, 7000 Tandil,
Argentina; {Bayer Argentina, S.A., 1605 received the test (IVM-TEST) formulation and those in Group B were treated
Munro, Buenos Aires, Argentina with the reference formulation (IVM-CONTROL). Heparinized blood samples
Part of the data included in this article was were taken from 0 h up to either 20 (pigs) or 30 (calves) days post-treatment
Ahed
presented at the 7th International Congress of and plasma was extracted, derivatized and analysed by high performance liquid
Bhed
Ched
the European Association for Veterinary chromatography (HPLC) using fluorescence detection. Early detection of IVM
Pharmacology and Toxicology held in Madrid, (12 h) with a peak plasma concentration (Cmax) between 33 and 39 ng/mL was
Dhed
Spain, July 1997. observed in pigs. The drug was detected in plasma up to 20 days post-
Ref marker
Fig marker administration of either formulation, resulting in elimination half-lives between
Table mar- 3.47 and 3.80 days. There were no differences between the IVM-TEST and
ker IVM-CONTROL formulations in the kinetic parameters (except tmax) obtained in
Ref end pigs. IVM was detected in plasma between 12 h and 30 days post-
Ref start administration of both formulations under investigation in cattle. The plasma
disposition kinetics of IVM in calves was similar following treatment with both
formulations. Cmax values (between 40.5 and 46.4 ng/mL) were achieved at 2
days post-administration of both formulations. None of the estimated kinetic
parameters were statistically different between drug formulations. The
injectable IVM formulations investigated were bioequivalent after their sub-
cutaneous administration to both pigs and calves at recommended dose rates.
(Paper received 27 March 1998; accepted for publication 12 August 1998)
Dr Carlos E. Lanusse, Area FarmacologõÂa, Departamento de FisiopatologõÂa, Facultad
de Ciencias Veterinarias, Universidad Nacional del Centro, Campus Universitario
(7000) Tandil, Argentina. (E-mail: clanusse@vet.unicen.edu.ar)

INTRODUCTION available for pigs. IVM is highly effective against the adult,
developing and hypobiotic larvae of most gastrointestinal
Ivermectin (IVM) exhibits a broad-spectrum of activity against nematodes, lungworms and many arthropods in cattle (McKellar
gastrointestinal and lung nematodes (Egerton et al., 1981) as & Benchaoui, 1996). A high degree of efficacy of IVM against the
well as against external parasites of domestic animals (Campbell majority of adult and larval forms of gastrointestinal parasites in
et al., 1983). Since 1981, IVM has been released in over 60 pigs has been demonstrated (Benz et al., 1989).
countries for use in cattle, sheep, goats, horses, swine, dogs, cats, Ivermectin, a semisynthetic derivative of the avermectin
camels, reindeer, bison and humans (Shoop et al., 1995). IVM is family, is the generic name given to a mixture of two
commercially available as injectable and pour-on formulations chemically modified avermectins containing at least 80% of
for use in cattle. Injectable and premix formulations are also 22±23 dihydroavermectin B1a and less than 20% 22±23

#1999 Blackwell Science Ltd 27


Paper 172 Disc

28 A. Lifschitz et al.

dihydroavermectin B1b (Fisher & Mrozik, 1989). IVM is a large concentrate diet. Experimental animals had free access to water.
and highly lipophilic molecule that dissolves in most organic The experiments were conducted following internationally
solvents. It is relatively insoluble in water despite possessing accepted animal welfare guidelines.
two sugar rings containing two hydroxyl groups (Jackson,
1989). Owing to its high lipophilicity, IVM distributes largely Treatments: Pigs were randomly allocated into two groups of
to, and is eliminated slowly from, the body compartments eight animals each. Treatments were given as follows:
(McKellar & Benchaoui, 1996; Lanusse et al., 1997). The Group A: experimental animals were treated with the
plasma pharmacokinetic be-haviour of the drug differs accord- ivermectin test formulation (10 mg/mL) (IVM-TEST) provided
ing to the route of administration, formulation and animal by Bayer Argentina S.A. (Buenos Aires, Argentina) (Batch
species (Fink & Porras, 1989). LDB601). The treatment was given by subcutaneous injection in
The close relationship between disposition kinetics and clinical the neck immediately behind the ear at 300 mg/kg body weight.
efficacy for antiparasitic compounds is now well documented This formulation was considered as the test product in the
(Lanusse & Prichard, 1993; Baggot & McKellar, 1994). The current bioequivalence trial.
characterization of the plasma bioavailability and disposition Group B: experimental animals were treated with a commer-
kinetics of IVM can be used to predict and optimize its ecto- cially available formulation of ivermectin (Ivomec1, MSD Agvet,
endoparasiticide efficacy. The vehicle in which the endectocide NJ, USA) (Batch A0352Z) by subcutaneous injection at 300 mg/
compounds are formulated may play a relevant role in their kg body weight in the neck immediately behind the ear. This
absorption kinetics and plasma availability (Lo et al., 1985; preparation was considered as the reference (control) product in
Lanusse et al., 1997). Small differences in formulation and in the the trial (IVM-CONTROL).
resultant kinetic behaviour of IVM may account for important
changes to its efficacy against the most difficult to control
Experiment 2: Ivermectin bioequivalence in cattle
internal and external parasites in livestock. Pharmaceutical
technology has been applied to develop different drug formula- Animals: The study was conducted in 16 parasite-free male
tions and delivery systems to optimize the pharmacological Holstein calves weighing 100±120 kg. The health of the animals
potency of IVM and other endectocide molecules currently was monitored prior to and throughout the experimental period.
available. Alternative IVM formulations for use in several Animals were fed ad libitum with lucerne hay and concentrate
different species have been introduced to the market or are ration. They had free access to water.
under development since the expiration of the original patent for
the first approved IVM formulation (Ivomec1, MSD AgVet). Treatments: Calves were randomly allocated into two groups of
The goal of the studies reported here was to evaluate the eight animals each. Treatments were given as follows:
bioequivalence and comparative disposition of two injectable Group A: experimental animals were treated with the
formulations of IVM administered subcutaneously to pigs and ivermectin test formulation (10 mg/mL) (IVM-TEST) provided
cattle, using parallel experimental designs. The results reported by Bayer Argentina S.A (Batch LDB601). The treatment was
in this article contribute to further understanding of the plasma given by subcutaneous injection in the shoulder area at 200 mg/
disposition kinetics of IVM in both species. The outcome of these kg body weight. This formulation was considered as the test
trials ratifies the usefulness of the parallel design to evaluate product in the bioequivalence trial.
pharmaceutical bioequivalence of veterinary drugs when cross- Group B: experimental animals were treated with a commer-
over designs are not recommended, such as in the case of drugs cially available formulation of ivermectin (Ivomec1, MSD Agvet)
with prolonged half-lives (Toutain & Koritz, 1997). (Batch A0352Z) by subcutaneous injection in the shoulder area
at 200 mg/kg body weight. This preparation was considered as
the reference (control) product in the trial (IVM-CONTROL).

MATERIALS AND METHODS


Sampling: Heparinized blood samples were taken in both
experiments at 0, 12, 24 h and at 2, 3, 4, 5, 7, 9, 11, 15 and
Experimental design
20 days post-treatment; two extra samples on days 25 and 30
The bioequivalence of two IVM preparations, formulated in a post-treatment were obtained from treated calves. Blood samples
propylene glycol/glycerol formal (60: 40) vehicle for subcutaneous were centrifuged at 2000 g for 20 min and the recovered plasma
administration, was evaluated in pigs (Expt 1) and calves (Expt 2). was kept in labelled vials at ±20 8C until analysed. The samples
were analysed within 30 days of their collection.

Experiment 1: Ivermectin bioequivalence in pigs


Analytical procedures
Animals: The trial was conducted in 16 parasite-free male Duroc
Jersey-Yorkshire crossbred pigs weighing 90±110 kg. The health Chemical extraction and derivatization
of the animals was monitored prior to and throughout the The extraction of IVM from spiked and experimental samples was
experimental period. Animals were housed in individual pens carried out following adaptations of the technique described by
and fed a commercially produced 35% protein/grain (3: 1) De Montigny et al. (1990) and Alvinerie et al. (1993, 1995). The

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34


Paper 172 Disc

Ivermectin in pigs and cattle 29

adaptation introduced to that methodology included the use of The analyte was identified with the retention times of IVM
abamectin as internal standard to quantify IVM plasma pure reference standards. The linearity was determined by
concentrations. Basically, 1 mL-aliquot of plasma sample was injection of spiked IVM standards in plasma at different
combined with 100 mL of internal standard (abamectin) (100 concentrations (triplicate determinations). Calibration curves
ng/mL) and then mixed with 1 mL of acetonitrile-water (80: 20). were constructed using the least squares linear regression
After mixing for 20 min, the solvent-sample mixture was analysis and correlation coefficients (r) and coefficient of
centrifuged at 2000 g during 15 min. The supernatant was variations (CV) were calculated. Drug recovery was established
injected to a Supelclean LC18 cartridge (Supelco, Bellfonte, PA, by comparison of the detector responses (peak areas) obtained
USA) previously conditioned by passing 2 mL of methanol and 2 from IVM fortified plasma samples and those of the direct
mL deionized water. The cartridge was flushed with 2 mL of standards prepared in methanol carried through the derivatiza-
water/methanol (3:1). The analytes were eluted with 1 mL of tion process, using abamectin as internal standard. Percentages
methanol and concentrated to dryness under a stream of of IVM recovery from plasma samples were obtained in the range
nitrogen. The reconstitution was done using 100 mL of a between 0.25 and 50 ng/mL. The mean percentage of recovery
solution of N-methylimidazole (Sigma Chemical, St Louis, MO, and the coefficient of variation (CV) were calculated. The CV was
USA) in acetonitrile (1: 1). Derivatization was initiated adding obtained as the ratio between the standard deviations and the
150 mL of trifluoroacetic anhydride (Sigma Chemical, St Louis, mean recovery values (Bolton, 1984) of at least three
MO, USA) solution in acetonitrile (1: 2). A fluorescent IVM- determinations. The limit of drug detection was established with
derivative is formed by acetylation and dehydration reactions injection of plasma blanks fortified with the internal standard
when the drug reacts with trifluoroacetic anhydride in the and measuring the baseline noise at the retention time of the
presence of N-methylimidazole (De Montigny et al., 1990). After IVM peak. The mean baseline noise at the IVM retention time
completion of the reaction (5 30 s), an aliquot (100 mL) of this plus three standard deviations was defined as the detection limit.
solution was injected directly into a high performance liquid The mean baseline noise plus 10 standard deviations was defined
chromatograph (HPLC). as the theoretical quantification limit.

Drug analysis
Pharmacokinetic and statistical analyses of the data
IVM concentrations were determined by HPLC using a Shimadzu
10 A HPLC system (Shimadzu Corporation, Kyoto, Japan). HPLC The plasma concentration vs. time curves obtained after each
analysis was undertaken using a reverse phase C18 column treatment in each individual animal were fitted with the
(Supelco, 3 mm, 4.6 mm 6 150 mm) and an acetonitrile/ PKCALC computer program (Shumaker, 1986) coupled to an
methanol/water (60/35/5) mobile phase at a flow rate of 1.5 augmented copy of the stripping program ESTRIP (Brown &
mL/min. Fluorescence detection was conducted with a Shimadzu Manno, 1978). Pharmacokinetic parameters were determined
RF-10 Spectrofluorometric detector (Kyoto, Japan) reading at an using a model-independent method (Notari, 1987). The peak
excitation wavelength of 365 nm and an emission wavelength of concentration (Cmax) and time to peak concentration (tmax) were
475 nm. IVM concentrations were determined by the internal read from the plotted concentration±time curve in each
standard method using the Class LC 10 Software version 1.2 individual animal. The area under the concentration±time
(Shimadzu Corporation, Kyoto, Japan) on an IBM compatible AT curves (AUC) were calculated by the trapezoidal rule (Gibaldi &
486 computer. The IVM/abamectin peak area ratio was obtained Perrier, 1982) and further extrapolated to infinity by dividing
to estimate IVM concentrations in spiked (validation) and the last experimental concentration by the terminal slope (b).
experimental samples. There was no interference of endogenous Statistical moment theory was applied to calculate the mean
compounds in the chromatographic determinations. The sol- plasma residence time (MRT) for IVM, as follows:
vents (Baker, Phillipsburg, NJ, USA) used during the extraction
and drug analysis were HPLC grade. AUMC
MRT ˆ
AUC
where AUC is as defined previously and AUMC is the area under
Validation procedures
the curve of the product of time and drug concentration vs. time
A complete validation of the analytical procedures used for from zero to infinity (Gibaldi & Perrier, 1982).
extraction and quantification of IVM was performed before The pharmacokinetic parameters are reported as mean + SEM.
analysis of experimental samples from the bioequivalence trials. Mean pharmacokinetic parameters for IVM obtained after the
IVM (Batch # 95051 2BER01, purity 97.5%) and abamectin administration of both formulations in both species were
(Batch 9505 25BER01, purity 97.4%) pure reference standards statistically compared by analysis of variance (ANOVA) using
provided by Bayer Argentina S.A., were used to prepare Satistical Analysis Software (SAS). The normal distribution of the
calibration curves in the range between 0.25 and 10 ng/mL data was confirmed using the Bartlett test. The kinetic
and 10±100 ng/mL in pig and calf plasma, respectively. The parameters used for the bioequivalence comparison between
validation of the analytical method included the determination of formulations (Cmax, AUC) were compared by ANOVA after log
the linearity of the detector responses, recovery percentages and transformation (Steinijans et al., 1992). Mean values were
limits of detection and quantification. considered significantly different at P 5 0.05.

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34


Paper 172 Disc

30 A. Lifschitz et al.

RESULTS Table 2. Comparison of the plasma kinetic parameters for ivermectin


obtained after subcutaneous administration of the IVM-TEST and IVM-
CONTROL formulations to pigs at 300 mg/kg
The analytical procedures including chemical extraction, deri-
vatization and HPLC analysis of IVM were validated adequately. FORMULATION
The linear regression lines for IVM in plasma in the range 0.25± Kinetic Parameters IVM-TEST IVM-CONTROL P-value
10 ng/mL and 10±100 ng/mL showed high correlation Cmax (ng/mL) 33.3 + 4.10 39.6 + 3.84 0.290
coefficients for both pigs (from 0.994 to 0.999) and cattle (from tmax (days) 2.75 + 0.43 0.94 + 0.16 0.002
0.998 to 0.999), respectively. AUC0±20d (ng.d/mL) 162 + 22.6 127 + 6.89 0.282
The mean recovery of IVM from pigs and cattle plasma was AUCtotal (ng.d/mL) 165 + 21.6 132 + 7.23 0.241
87.1 and 93.2%, respectively. The limits of detection of the AUMCtotal (ng.d2/mL) 995 + 108 763 + 42 0.229
analytical techniques were 0.17 ng/mL (pigs) and 0.05 ng/mL t‰el (days) 3.47 + 0.50 3.80 + 0.19 0.324
MRT (days) 6.29 + 0.63 5.78 + 0.18 0.481
(cattle). The theoretical quantification limits of IVM in plasma
were 0.25 ng/mL (pigs) and 0.10 ng/mL (cattle). The results of Values are expressed as mean + SEM (n = 8).
the validation of the analytical technique to determine IVM in Cmax: peak plasma concentration; tmax: time to peak plasma concentra-
plasma obtained from pigs and calves are summarized in Table 1. tion; AUC0±20d: area under the concentration vs. time curve between
drug administration and 20 days post-treatment; AUCtotal: area under
Ivermectin was detected in plasma between 12 h and 20 days
the concentration vs. time curve extrapolated to infinity; AUMCtotal: area
post-administration of either formulation in pigs. The compara- under the first moment of the concentration vs. time curve extrapolated
tive mean plasma concentration of IVM obtained after the to infinity; t‰el: elimination half-life; MRT: mean residence time.
subcutaneous administration of the two formulations to pigs are
shown in Fig. 1. The mean pharmacokinetic parameters for IVM
obtained after the administration of IVM-TEST and IVM- The AUC0±20 days, AUCtotal, AUMC, Cmax, MRT and t‰el
CONTROL formulations to pigs are compared in Table 2. obtained for IVM-TEST and IVM-CONTROL formulations in pigs
were not statistically different. The peak plasma concentration
Table 1. Validation of the analytical methodology used to determine for the IVM-TEST formulation was achieved significantly later
ivermectin concentrations in pig and calf plasma
(P 5 0.002) than that of the IVM-CONTROL formulation. In
Pig Calf fact, the only difference observed between experimental groups
was that after the administration of the IVM-TEST formulation,
Limit of detection (ng/mL) 0.17 0.05
Limit of quantification (ng/mL) 0.25 0.10 the disposition of IVM was characterized by a slightly higher
Recovery (%) 87.1 (7.02) 93.2 (3.35) plasma concentration profile from day 2 to day 7 post-treatment,
Linearity (r) resulting in a mean Cmax value of 33.3 + 4.10 ng/mL achieved
(0.25±10 ng/mL) 0.994 0.999 at 2.75d post-administration. The highest plasma concentration
(10±100 ng/mL) 0.999 0.998 was achieved between 12 h and 2 days post-treatment
Coefficient of variation (%) 7.63 3.58 (Cmax = 39.6 + 3.84 ng/mL; tmax = 0.94d h) following admin-
The validation results were obtained as defined in the analytical istration of the IVM-CONTROL.
methodology section. Values in brackets represent coefficient of variation Thereafter, the plasma profiles of IVM declined, following a
(CV) for the recovery assays. r: coefficient of correlation obtained from very similar pattern, to a mean concentration between
the linear regression lines in the range between 0.25 and 10 ng/mL and 0.62 + 0.16 ng/mL (IVM-TEST) and 0.69 + 0.10 ng/mL
10±100 ng/mL, respectively.
(IVM-CONTROL) at 20 days post-treatment. IVM elimination
half-lives (3.47 and 3.80 days) and mean residence times (6.29
and 5.78 days) were similar between both formulations.
Ivermectin was detected in plasma between 12 h and 30 days
post-administration of both formulations under investigation to
cattle. The comparative mean plasma concentrations of IVM
obtained after the subcutaneous administration of the two
formulations to cattle are shown in Fig. 2. Mean pharmacoki-
netic parameters for IVM obtained after the administration of
IVM-TEST and IVM-CONTROL formulations to cattle are
compared in Table 3. The overall disposition kinetics of IVM
was similar following treatment with both formulations. None of
the kinetic parameters calculated were statistically different
between drug formulations. The comparison of the mean Cmax
and AUC values obtained for IVM following its subcutaneous
administration to pigs and calves at recommended dose rates, is
Fig. 1. Mean plasma (+ SEM) concentrations of ivermectin (IVM) shown in Fig. 3.
obtained after subcutaneous administration of the IVM-CONTROL (±.±) The statistical comparison (ANOVA), after log transformation of
and IVM-TEST (±!±) formulations (300 mg/kg) to pigs. the data, for AUC and Cmax and the bioequivalence range

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34


Paper 172 Disc

Ivermectin in pigs and cattle 31

Fig. 2. Mean plasma (+ SEM) concentrations of ivermectin (IVM)


Fig. 3. Comparative ivermectin peak plasma concentrations (Cmax) and
obtained after subcutaneous administration of the IVM-CONTROL (±.±)
area under the plasma concentration vs. time curves (AUC) obtained
and IVM-TEST (±!±) formulations (200 mg/kg) to calves.
after its subcutaneous administration (IVM-CONTROL formulation) to
pigs (300 mg/kg) ( ) and calves (200 mg/kg) ( ). Values express
means with n = 8. These dose-dependent parameters were statistically
different between species (P 5 0.001) when they were normalised by the
administered dose rate.

Table 3. Comparison of the plasma kinetic parameters obtained for


ivermectin after subcutaneous administration of the IVM-TEST and IVM-
CONTROL formulations to calves at 200 mg/kg
obtained for IVM-TEST and IVM-CONTROL after their subcuta-
FORMULATION neous administration to pigs and cattle are shown in Table 4.
Kinetic Parameters IVM-TEST IVM-CONTROL P-value

Cmax (ng/mL) 40.5 + 2.72 46.4 + 3.88 0.239


tmax (days) 2.00 + 0.50 2.12 + 0.14 0.590 DISCUSSION
AUC0±30d (ng.d/mL) 239 + 19.6 263 + 23.0 0.541
AUCtotal (ng.d/mL) 244 + 20.1 266 + 23.3 0.492 Understanding the pharmacokinetic behaviour of broad-spec-
AUMCtotal (ng.d2/mL) 1530 + 244 1452 + 181 0.802 trum antiparasitic drugs and the factors modulating that
t‰el (days) 6.30 + 0.81 5.50 + 0.39 0.359 behaviour is highly important for maximizing their clinical
MRT (days) 6.15 + 0.48 5.39 + 0.25 0.211
efficacy (Lanusse & Prichard, 1993). The pharmacokinetic
Values are expressed as mean + SEM (n = 8). Cmax: peak plasma behaviour of IVM in pigs has not been extensively investigated.
concentration; tmax: time to peak plasma concentration; AUC 0±30d: area The available information on the plasma profiles of IVM in pigs
under the concentration vs. time curve between drug administration and (Fink & Porras, 1989; Scott & McKellar, 1992) was obtained
30 days post-treatment; AUCtotal: area under the concentration vs. time
under experimental conditions different from those used in the
curve extrapolated to infinity; AUMCtotal: area under the first moment of
the concentration vs. time curve extrapolated to infinity; t‰el: elimina-
current trial. Thus, besides the characterization of the bioequi-
tion half-life; MRT: mean residence time. valence of both formulations under study, the results of the
current trial may contribute to the further understanding of the
plasma disposition kinetics of this endectocide molecule in pigs.
In agreement with a previous report (Scott & McKellar, 1992),
Table 4. Bioequivalence between IVM-
Kinetic Log Transformed Value Ratio* P-value
TEST and IVM-CONTROL formulations
Parameters IVM-TEST IVM-CONTROL mT/ mR

Cmax (ng/mL) 1.50 1.58 0.95 0.276


PIGS AUC0±20d (ng.d/mL) 2.18 2.10 1.04 0.246
AUCtotal (ng.d/mL) 2.19 2.11 1.04 0.284
Cmax (ng/mL) 1.60 1.66 0.96 0.256
CATTLE AUC0±30d (ng.d/mL) 2.37 2.41 0.98 0.547
AUCtotal (ng.d/mL) 2.38 2.41 0.99 0.510

(*) mT and mR represent the values of the kinetic parameter for the test and reference (control)
formulations, respectively, in pigs and cattle. Two pharmaceutical formulations are bioequivalent when
the ratio mT/mR for Cmax and AUC between test and reference formulations result between 0.80 and 1.25
(Steinijans et al., 1992).

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34


Paper 172 Disc

32 A. Lifschitz et al.

there were individual variations in the plasma profile of IVM in our laboratory (Lifschitz et al., 1997) using calves of the same
the present experiment. The mean Cmax of both formulations in breed, age and weight. It is important to note that higher IVM
our trial (between 33.3 and 39.6 ng/mL) was slightly greater plasma AUC values have been reported, using different cattle
compared to that reported by those authors (28.4 ng/mL); the breeds and much longer sampling periods (Lanusse et al., 1997).
tmax obtained for the IVM-CONTROL formulation was similar. In the current trial to evaluate the pharmaceutical bioequiva-
The mean IVM AUC values obtained after the administration of lence of the two formulations, the sampling period was extended
both IVM-TEST and IVM-CONTROL formulations in the current up to 30 days post-treatment, the period considered relevant to
trial were greater than those reported by those authors, which correlate IVM plasma concentration with the ecto-endoparasiti-
may be associated with the longer sampling period used in our cide activity of the drug.
trial. The mean elimination half-lives of IVM after the admin- The comparison of the IVM disposition kinetics in both species
istration of the two formulations were 3.47 and 3.80 days, was interesting. When the drug was given by subcutaneous
suggesting that the drug is cleared slowly from pigs, being injection at recommended dose rates, the Cmax and AUC values
detectable in plasma for up to 20 days post-treatment. obtained were higher in calves than in pigs (Fig. 3). Those dose-
The use of different experimental conditions may be one of dependent parameters were statistically different (P 5 0.001)
the key factors underlying the reported differences on kinetic between species, after their normalization by the administered
behaviour. In the current trial, the disposition of IVM in pigs dose rate. Differences in body composition may account for
was characterized with a sampling period that lasted for 20 changes in the pattern of IVM tissue distribution. A more
days post-treatment, which is longer than in the above extensive distribution and deposit of IVM in adipose tissue in pigs
referenced studies. The sensitivity of the analytical methodol- compared to cattle, may account for the lower plasma
ogy used to quantify IVM permitted the detection of the drug in availability obtained in this species.
plasma of treated animals over 20 days post-treatment. To minimize the different variables which may affect drug
Additionally, the pigs used in the current trial were heavier plasma concentrations, bioequivalence studies should be
(90±110 kg) than those used by Scott & McKellar (1992) (18± conducted in healthy animals that are as homogeneous as
26 kg), which may have some influence in the pharmacokinetic possible with respect to age, weight and sex. This is
behaviour of this lipophilic molecule. A greater proportion of fat particularly important for studies conducted as parallel design
in body composition may facilitate longer IVM persistence in (Toutain & Koritz, 1997). Bioequivalence of different formula-
the body, accounting for changes in the drug disposition. tions of the same active molecule comprises equivalence with
Differences in the bioavailability and half-life of IVM have been respect to the rate and extent of drug absorption (Steinijans &
reported following oral and subcutaneous administration to Hauschke, 1990). The basic assumption underlying this
pigs (Fink & Porras, 1989). Administration by the parenteral definition is that equivalent rates and extent of absorption
route produced slower drug absorption but an overall higher will lead to essentially the same drug plasma concentration±
drug availability in plasma. time profiles and therefore, to the same therapeutic or toxic
As demonstrated by Lo et al. (1985), Wicks et al. (1993) and effects (Toutain & Koritz, 1997).
Lanusse et al. (1997), the absorption rate of endectocide The area under the concentration curve (AUC) is of interest as
molecules in cattle is markedly influenced by their pharmaceu- it reflects the extent to which the active drug is absorbed and is
tical preparations. Following parenteral administration, the low independent of the rate of the absorption process. The
solubility of IVM in water and its deposition in subcutaneous pharmacokinetic parameters used to reflect the rate of drug
tissue favour a slow absorption from the injection site providing absorption are the maximum plasma concentration (Cmax) and
a prolonged residence in the bloodstream. In the current trial, the time at which that occurs following drug administration
the slow absorption of IVM resulted in a delayed tmax (between 2 (tmax) (Toutain & Koritz, 1997). The lack of statistical differences
and 2.12 days) after subcutaneous administration of IVM-TEST between test and reference formulations in these three variables
and IVM-CONTROL to calves. The clinical efficacy of an is sufficient to support bioequivalence. However, if the AUC and
antiparasitic compound relies on its disposition kinetics and Cmax values for test and reference formulations were not
pattern of plasma/tissue exchange in the host (Lanusse & statistically different, the plasma concentrations profiles are very
Prichard, 1993). IVM is a highly lipophilic molecule that is likely to be similar. The evaluation of the tmax parameter is less
extensively distributed into tissues (Bogan & McKellar, 1988; important and less reliable (Gettinby, 1994). This may be due to
Chiu et al., 1990). The distribution into adipose tissue is the fact that measurements are restricted to the sampled time
particularly relevant and may act as a drug reservoir, that points, so the data are less accurate and more variable (Gettinby,
contributes to the long residence of the drug in the bloodstream. 1994). This is in agreement with the Food and Drug Adminis-
The mean plasma elimination half-life of IVM obtained in this tration (1996), which has established the Cmax and AUC as the
trial after the administration of the two formulations (5.50 and pivotal kinetic parameters to determine bioequivalence. These
6.30 days) reflects the long persistence of IVM in the body. The kinetic parameters were not significantly different between the
IVM concentration profiles in plasma reflect those achieved in IVM-TEST and IVM-CONTROL formulations in either species in
different tissues, including those where target parasites are the current trials.
located. The disposition kinetic results obtained for IVM in cattle The present bioequivalence study was carried out using a
in the current trial were similar to those previously obtained in parallel design. In this type of design, the two groups of animals

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34


Paper 172 Disc

Ivermectin in pigs and cattle 33

are treated in parallel, whereas in the cross-over design each REFERENCES


formulation is administered to the same animals after a washout
period. The washout period is usually set at least 10 times the Alvinerie, M., Sutra, J., Badri, M. & Galtier, P. (1995) Determination of
terminal half-life of the active drug (Food & Drug Administration, moxidectin in plasma by high-performance liquid chromatography
with automated solid-phase extraction and fluorescence detection.
1996; Toutain & Koritz, 1997). Given the long residence time of
Journal of Chromatography B, 674, 119±124.
IVM in adipose tissue, an extremely long washout period would
Alvinerie, M., Sutra, J. & Galtier, P. (1993) Ivermectin in goat milk after
have been necessary to run a cross-over design in the current subcutaneous injection. Annales Recherches Veterinaires, 24, 417±421.
trials. Animal growth and age differences between the first and Baggot, J. & McKellar, Q. (1994) The absorption, distribution, and
second experimental phases would have worked against elimination of anthelmintic drugs: the role of pharmacokinetics.
standardized experimental conditions to evaluate IVM bioequi- Journal of Veterinary Pharmacology and Therapeutics, 17, 409±419.
valence between formulations. Benz, G., Roncalli, R. & Gross, S. (1989) Use of ivermectin in cattle,
sheep, goats and swine. In Ivermectin and Abamectin. Ed. Campbell, W.
For determining whether two pharmaceutical formulations
pp. 90±113. Springer-Verlag New York Inc., New York, USA.
are bioequivalent, regulatory agencies have set a statistical
Bogan, J. & McKellar, Q. (1988) The pharmacodynamics of ivermectin in
criterion. The criterion specifies that the mean AUC and Cmax sheep and cattle. Journal of Veterinary Pharmacology and Therapeutics,
values of the test formulation should not be more than 20% 11, 260±268.
different from the corresponding mean values of the reference Bolton, S. (1984) Pharmaceutical Statistics. Drugs and Pharmaceutical
formulation (Steinijans et al., 1992). The Guidelines on Sciences. pp. 1±31. Marcel Dekker Inc, New York, USA.
Bioequivalence from the European Committee for Proprietary Brown, R. & Manno, J. (1978) ESTRIP, a basic computer program for
obtaining initial polyexponential parameter estimates. Journal of
Medicinal Products specifies that the bioequivalence range,
Pharmaceutical Sciences, 67, 1687.
obtained as the ratio between the mean AUC and Cmax values,
Campbell, W.C., Fisher, M.H., Stapley, E.O., Albers-Schoenberg, G. &
for the test and reference formulations, should fall between 0.80 Jacob, T.A. (1983) Ivermectin: a potent new antiparasitic agent.
and 1.25 (Committee for Proprietary Medicinal Products, 1991). Science, 221, 823±828.
This can be expressed as 0.80 5 mT/mR 5 1.25, where mT and mR Chiu, S., Green, M., Baylis, F., Eline, D., Rosegay, A., Meriwether, H. &
denote the pharma-cokinetic parameters for test and reference Jacob, T. (1990) Absorption, tissue distribution, and excretion of
formulations, respectively. In order to reach harmonization, the tritium-labelled ivermectin in cattle, sheep, and rat. Journal of
Agricultural and Food Chemistry, 38, 2072±2078.
FDA criterion has been changed from (0.80, 1.20) to (0.80,
Committee for Proprietary Medicinal Products (1991) Working Party on
1.25) (Steinijans et al., 1992).
the Efficacy of Medicinal Products. Note for guidance: Investigation of
The logarithmic transformation of the AUC and Cmax values is bioavailability and bioequivalence, Commission of the European
recommended (Steinijans et al., 1992). The use of logarithmic Community.
transformation normalizes the distribution of the parameter, De Montigny, P., Shim, J.S.K. & Pivinichny, J.V. (1990) Liquid
stabilizes its variance and ensures the additivity of the statistical chromatographic determination of ivermectin with trifluoro-acetic
model (Steinijans & Hauschke, 1990; Toutain & Koritz, 1997). anhydride and N-methylimidazole as the derivatization reagent.
Journal of Pharmaceutical Biomedical Analysis, 8, 507±511.
The ratio mT/mR for Cmax and AUC for the formulations evaluated
Egerton, J., Eary, C. & Suhayda, D. (1981) The anthelmintic efficacy of
in the current trial were between 0.80 and 1.25, which confirms
ivermectin in experimentally infected cattle. Veterinary Parasitology, 8,
the lack of statistical differences between both formulations in 59±70.
both species (Table 4). Fink, D. & Porras, A. (1989) Pharmacokinetics of ivermectin in animals
In conclusion, the results reported in this article contribute to and humans. In Ivermectin and Abamectin. Ed. Campbell, W. pp. 90±
the further understanding of the plasma disposition kinetics of 113, Springer-Verlag New York Inc., New York, USA.
IVM in pigs and cattle. The usefulness of the parallel design to Fisher, M. & Mrozik, H. (1989) Chemistry. In Ivermectin and Abamectin.
Ed. Campbell, W. pp. 90±113. Springer-Verlag New York Inc., New
evaluate pharmaceutical bioequivalence of veterinary drugs
York, USA.
with prolonged half-lives was ratified. The pivotal bioequivalence
Food and Drug Administration (FDA) (1996) Bioequivalence Guidance,
parameters (Cmax and AUC) obtained after administration of the Center for Veterinary Medicine (CVM), Food and Drug Administration,
IVM-TEST and IVM-CONTROL formulations in either species pp. 1±26. Rockville MD, USA.
were not statistically different. Thus, the IVM formulations Gettinby, G. (1994) Experimental design for bioavailability and
investigated were bioequivalent after their subcutaneous admin- bioequivalence studies. 6th International Congress, European Associa-
istration to pigs and cattle at recommended dose rates. tion for Veterinary Pharmacology and Toxicology, Proceedings of the
Post-Congress Workshop, Edinburgh, UK.
Gibaldi, M. & Perrier, D. (1982) Pharmacokinetics, 2nd edn. pp. 45±109.
Marcel Dekker, Inc., New York, USA.
ACKNOWLEDGMENTS Jackson, H. (1989) Ivermectin as a systemic insecticide. Parasitology
Today, 5, 146±155.
The technical advice of Dr Michel Alvinerie (INRA, Toulouse, Lanusse, C., Lifschitz, A., Virkel, G., Alvarez, L., Sanchez, S., Sutra, J.F.,
Galtier, P. & Alvinerie, M. (1997) Comparative plasma disposition
France) for the development of the analytical techniques is
kinetics of ivermectin, moxidectin and doramectin in cattle. Journal of
gratefully acknowledged. Adrian Lifschitz is a recipient of a
Veterinary Pharmacology and Therapeutics, 20, 91±99.
fellowship from the Consejo Nacional de Investigaciones Lanusse, C. & Prichard, R. (1993) Relationship between pharmacological
CientõÂ ficas y TeÂcnicas (CONICET), Argentina. The financial properties and clinical efficacy of ruminant anthelmintics. Veterinary
support received from Bayer Argentina SA is acknowledged. Parasitology, 49, 123±158.

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34


Paper 172 Disc

34 A. Lifschitz et al.

Lifschitz, A., Murno, G., Pis, A., Sallovitz, J., Virkel, G. & Lanusse, C. Shoop, W., Mrozik, H. & Fisher, M. (1995) Structure and activity of
(1997) Malnutrition modifies the disposition kinetics of ivermectin in avermectins and milbemycins in animal health. Veterinary Parasitol-
calves. Journal of Veterinary Pharmacology and Therapeutics, (Supp. 1), ogy, 59, 139±156.
20, 102±103. Steinijans, V. & Hauschke, D. (1990) Update on the statistical analysis of
Lo, P., Fink, D., Williams, J. & Blodinger, J. (1985) Pharmacokinetics the bioequivalence studies. International Journal of Clinical Pharmacology
studies of ivermectin: effect of formulation. Veterinary Research Therapy and Toxicology, 28, 105±110.
Communications, 9, 251±268. Steinijans, V., Hauschke, D. & Jonkman, G. (1992) Controversies in
McKellar, Q. & Benchaoui, H. (1996) Avermectins and milbemycins. bioequivalence studies. Clinical Pharmacokinetics, 22, 247±253.
Journal of Veterinary Pharmacology and Therapeutics, 19, 331±351. Toutain, P. & Koritz, G. (1997) Veterinary drug bioequivalence
Notari, R. (1987) Biopharmaceutics and Clinical Pharmacokinetics, 4th edn. determination. Journal of Veterinary Pharmacology and Therapeutics,
pp. 45±129. Marcel Dekker, Inc., New York, USA. 20, 79±90.
Shumaker, R. (1986) PKCALC: a basic interactive computer program for Wicks, S., Kaye, B., Weatherley, A., Lewis, D., Davison, E., Gibson, S. &
statistical and pharmacokinetic analysis of data. Drug Metabolism Smith, D. (1993) Effect of formulation on the pharmacokinetics and
Reviews, 17, 331±348. efficacy of doramectin. Veterinary Parasitology, 49, 17±26.
Scott, E. & McKellar, Q. (1992) The distribution and some pharmaco-
kinetic parameters of ivermectin in pigs. Veterinary Research Commu-
nications, 16, 139±146.

#1999 Blackwell Science Ltd, J. vet. Pharmacol. Therap. 22, 27±34

You might also like