Вы находитесь на странице: 1из 26

Veterinary Parasitology 261 (2018) 27–52

Contents lists available at ScienceDirect

Veterinary Parasitology
journal homepage: www.elsevier.com/locate/vetpar

Review article

Anthelmintic drugs used in equine species T


a,⁎ b
Cengiz Gokbulut , Quintin A. McKellar
a
Department of Pharmacology, Faculty of Medicine, Balikesir University, Balikesir, Turkey
b
University of Hertfordshire, Hatfield, Hertfordshire AL10 9AB, United Kingdom

A R T I C LE I N FO A B S T R A C T

Keywords: Internal parasites of horses comprise an intractable problem conferring disease, production and performance
Anthelmintics losses. Parasitism can rarely be controlled in grazing horses by management alone and anthelmintic drugs have
Equines formed the basis of therapy and prophylaxis for the last sixty years. The pharmacology of the anthelmintic drugs
Horse available dictate their spectrum of activity and degree of efficacy, their optimal routes of administration and
Pony
characteristics which prevent some routes of administration, their safety tolerance and potential toxicities and as
Donkey
a consequence of their persistence in the body at effective concentrations their use in epidemiological control
Benzimidazoles
Macrocyclic lactones programmes. Their use has also resulted in the selection of parasites with genetically controlled characteristics
Endectocides which reduce their susceptibility to treatment, characteristics which are often common to whole chemical classes
Tetrahydropyrimidines of anthelmintics.
Resistance Pharmacological properties also confer compatibility in terms of safety and persistence with other anthel-
mintic drugs and thus the potential of combinations to treat parasites from different phylogenetic groups such as
nematodes, cestodes and trematodes and also the potential by agency of their different molecular mechanisms of
action to delay the selection of resistant genes. The major groups of anthelmintics now available, the benzi-
midazoles (BZD), macrocyclic lactones (MLs) and tetrahydropyrimidines are all highly effective against their
targeted parasites (primarily nematodes for BZD’s and ML’s and cestodes for tetrahydropyrimidines) easily
administered orally to horses and are well tolerated with wide margins of safety. Nevertheless, some parasitic
stages are inherently less susceptible such as hypobiotic stages of the small strongyles (cyathostomins) and for
some such as the adult stages of cyathostomins resistance has developed. Furthermore, for some less common
parasites such as the liver fluke unlicensed drugs such as the salicylanilide, closantel have been used.
A deep understanding of the pharmacology of anthelmintic drugs is essential to their optimal use in equine
species.

1. Anthelmintic drugs used in equine species and compounds such as dichlorvos, trichlorvos, haloxan and closantel,
which are rarely used for the treatment of specific gastrointestinal
Modern anthelmintic drugs can be classified into seven distinct parasites or liver flukes. These anthelmintics are summarized in Tables
classes according to their chemical structures and pharmacological 1 and 3 according to their chemical groupings and their efficacy, re-
behaviours. Namely, benzimidazoles (BZDs), pro-benzimidazoles (pro- spectively. Relatively limited data are available on internal parasites
BZDs), tetrahydropyrimidines (THPs), imidazothiazoles (IMTs), iso- and the pharmacology of anthelmintic drugs in donkeys and mules
quinolines, salicylanilides (closantel) simple heterocycles, organopho- compared to ruminants and horses because donkeys and mules have
sphates and macrocyclic lactones (MLs, avermectins and milbemycins been considered to be minor species and have been neglected in such
or endectocides) (Table 1). Each class of anthelmintics has a different research. Most of the drugs in different classes used in horses and ru-
spectrum of activity and pharmacokinetic behaviour. The agents which minants have been extrapolated for use in donkeys and mules without
have a broad spectrum of anthelmintic activity can be classified into optimization of dosing regimens or the determination of pharmacoki-
three groups: Benzimidazoles and pro-BZDs, IMTs and THPs, and MLs. netic properties and have been used at the horse dose rate (Veneziano
Currently, these three classes of anthelmintic are licensed for use in et al., 2011). It has been reported that donkeys have a greater capacity
equine species against most important nematodes. The other anthel- to metabolize certain drugs, compared to horses; thus, higher dosages
mintic compounds with narrow-spectrum of activity include piperazine or shorter intervals could be required to maintain effective drug


Corresponding author.
E-mail address: cengizgokbulut@yahoo.com (C. Gokbulut).

https://doi.org/10.1016/j.vetpar.2018.08.002
Received 31 May 2018; Received in revised form 1 August 2018; Accepted 6 August 2018
0304-4017/ © 2018 Elsevier B.V. All rights reserved.
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Table 1 provide effective control against nematodes with single or multiple


Anthelmintics available for treatment of helminth infections in equine species. drug resistance, and (2) to prevent or decelerate the development of
Class Route of Commonly Formulations resistance to each component of the anthelmintic combinations
administration recommended (not all licensed (Bartram et al., 2012). Other combinations of two or more distinct
dosage (mg/kg) for equines) classes of anthelminthic compounds with a different spectrum of ac-
tivity and different mechanisms of action are used to broaden the
Benzimidazoles
Cambendazole PO 20 Paste
spectrum of activity of the anthelmintic treatment, for instance to
Parbendazole PO 2.5 Powder, susp. control both nematodes and tapeworms in horses. Double combinations
Thiabendazole PO 50 Paste, susp., including ML + Praziquantel, BZ + Pyrantel or BZ + Piperazine are
powder commercially available in Australia, New Zealand, UK, Canada and
Mebendazole PO 8.8 Paste, granule
USA, and triple combinations of ML + BZ + Praziquantel or ML +
Fenbendazoleb PO 7.5 Paste, susp.,
granule Pyrantel + Praziquantel are commercially available in Australia and
Oxfendazole PO 10 Paste, pellet, New Zealand to control parasites in horses (Table 2).
susp. In the UK the BZ Fenbendazole, the ML’s Ivermectin and
Oxibendazole PO 10 Paste, susp.
Moxidectin, the THP Pyrantel and the Isoquinoline Praziquantel are
Albendazolea PO 5 Paste, susp.,
tablet
licensed for use in the horse. Nevertheless, a wide range of drugs are
Triclabendazolea PO 12 Tablet, susp. used in other licensing authorities, several are used off-label for specific
Pro-benzimidazoles parasitic conditions and some have been utilised experimentally in
Netobimina PO 10 Solution horses. Furthermore, characteristics of some anthelmintics common to
Febantel PO 6 Paste, susp.,
a group but not specific to horses or donkeys have been included in this
granule
Thiophanatea PO 50 Powder review because they add understanding to those used in the horse.
Tetrahydopyrimidines There are also a number of anthelmintics which have been used ex-
Pyrantel pamoateb PO 6.6 Paste, susp. tensively in horses in the past but are not currently used and these have
Pyrantel tartrate PO 6.6 Powder, pellet been included to provide comprehensive cover but also because they
Morantel PO 7.5-12.5 Paste
Imidazothiazole
have properties which might prove valuable for the future.
Levamisolea PO 10 Solution
Salicylanilides 2. Benzimidazoles and Pro-benzimidazoles
Closantela PO 10-20 Solution
Isoquinoline
The Benzimidazoles (BDZs) were the first safe and effective broad-
Praziquantelb PO 1.0 Paste/oral gel
Simple heterocycles spectrum anthelmintics which were used widely in the treatment of a
Phenothiazine PO 75 Powder variety of gastrointestinal parasitic infections in veterinary and human
Piperazine PO 200 Powder medicine. The BZD and pro-BZDs are a large family of compounds that
Organophosphates are related to thiabendazole (TBZ) which was first released for use in
Dichlorvos PO 20 Paste, pellet
Haloxon PO 60 Paste, powder
horses as an anthelmintic in 1961 (Drudge et al., 1981). These drugs
Trichlorfon PO 40 Paste, powder have common features such as broad-spectrum anthelmintic activity
Macrocyclic lactones and relatively low mammalian toxicity. Benzimidazole anthelmintics
Avermectins licensed internationally for use in horses for the treatment of gastro-
Ivermectinb PO, Ta 0.2-0.5 Paste, drench,
intestinal helminthiases and for lungworm and migrating strongyle
tablet
Abamectin PO 0.2 Paste, oral gel infections, include TBZ, mebendazole (MBZ) oxibendazole (OXB) fen-
Doramectin PO 0.2 Oral gel bendazole (FBZ), oxfendazole (FBZSO), cambendazole (CBZ) and the
Eprinomectina T 0.5 Solution pro-BZD, febantel (FBT).
Milbemycins All BZDs have poor water solubility; consequently, the development
Moxidectinb PO 0.2-0.4 Oral gel
of formulations for parenteral administrations has been limited. For this
PO: per os; T: Topical; susp.: Suspension. reason, they are primarily administered as oral pharmaceutical forms
a
Not licensed use for horse. such as drenches, tablets, bolus, paste or feed additives. Only rico-
b
Contained in products licensed for use in horses in the UK. bendazole (the sulphoxide metabolite of ABZ) has been available as an
injectable formulation and licensed for use in ruminant species.
concentrations in donkeys (Matthews et al., 1997; Grosenbaugh et al.,
2011). 2.1. Chemistry and metabolism
In equine species, anthelmintics may be given orally or by the na-
sogastric route for treatment of gastrointestinal parasitic infections. A According to their chemical structure, the BZD and pro-BZD an-
wide variety of formulations are available, including drenches, gels, thelmintics can be classified into four groups (Lanusse and Prichard,
pastes, granules, and powders for feed additives. Parenteral adminis- 1993).
tration of anthelmintics including endectocides is not utilised in equine
species. After parenteral administration, rare adverse reactions such as 1 BZD thiazolyl, including thiabendazole (TBZ) and cambendazole
Clostridium spp. infections and anaphylaxis have been observed and (CBZ);
these undesirable effects were responsible for the withdrawal of the 2 BZD methylcarbamates, including OXB, FBZ and OFZ or FBZSO,
parenteral preparation of IVM for use in the horse in 1984 (Campbell MBZ, albendazole (ABZ), albendazole sulphoxide (ABZSO) or rico-
et al., 1989). bendazole (RBZ), parbendazole (PBZ), flubendazole (FLBZ), luxa-
Anthelmintic drug combinations are increasingly used to control bendazole (LXB);
parasites in domestic animals including equine species. Dual or triple 3 Halogenated BZD thiols, of which triclabendazole (TCBZ) is the only
combinations of anthelminthic compounds with a similar spectrum of one currently available,
activity and different mechanisms of action are widely used in various 4 Pro-Benzimidazoles, including febantel (FBT), thiophanate (TPT)
parts of the world for the control of horse parasites (summarized in and netobimin (NTB).
Table 2). Such combinations are made for two main purposes: (1) to
A 1,2-diaminobenzene ring forms the central ring structure of all the

28
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Table 2
The anthelmintic combinations commercially available for the treatment of helminth infections in equine species.
Combinations Route of administration Formulations

Double combinations
OFZ (20%) / PYR (26%) PO Paste/oral gel, tablet
OFZ (17.5%) / Morantel (24.5%) PO Paste
OFZ (10.72%) / Piperazine (21.44%) PO Paste/oral gel
IVM (1.87%) / Praziquantel (14.3%) PO Paste, tablet
ABM (0.4%) / Morantel (16.7%) PO Paste
ABM (0.37%) / Praziquantel (4.62%) PO Paste
MXD (2.0%) / Praziquantel (12.5%) PO Oral gel
DRM (1.78%) / Praziquantel (22.29%) PO Paste

Triple combinations PO Paste


IVM (0.4%) / FBZ (20%) / Praziquantel (5.0%) PO Paste
IVM (0.6%) / PYR (39%) / Praziquantel (4.3%) PO Paste
MXD (0.8%) / OFZ (20%) / Praziquantel (5.0%) PO Paste
ABM (0.4%) / OFZ (20%) / Praziquantel (5.0%) PO Paste

BZD anthelmintics. The difference between each agent is associated pharmaceutical formulations and better absorption from the gastro-
with different substitutions at the 2 and 5 substitute positions of the intestinal tract of hosts following oral administration. Febantel and NTB
central ring structure (Fig. 1). The pro-drugs enter the host body where are commercially available pro-BZD drugs. Their anthelmintic activity
they are metabolised into the active BZD compounds by the gastro- is due to the fact that they are metabolised in the animal to form the
intestinal microflora and liver. Febantel is metabolised into FBZ, TPT biologically active BZD carbamate nucleus. Febantel is a phenylguani-
into LXB and NTB into ABZ. The chemical structures of BZD and pro- dine that is hydrolysed and then cyclized to the active metabolite FBZ
BZD anthelmintics and metabolic pathways of FBZ, OFZ, ABZ and TCBZ (Delatour et al., 1982). Netobimin is a nitrophenylguinidine which
are shown in Figs. 1 and 2, respectively. undergoes both reduction and cyclization to form the active compound
Although xenobiotic metabolism occurs mainly in the liver, extra- ABZ (McDougall et al., 1985).
hepatic tissues including plasma, kidney, lungs and gastrointestinal
tract and microflora are also involved. Biotransformation of drugs 2.2. Efficacy and spectra of activity
generally results in metabolites of the parent drug which have reduced
or no activity (Baggot, 1974). Benzimidazoles and pro-BZDs are ex- The anthelmintic activities of BZDs and pro-BZDs against common
tensively metabolised in all animal species and biotransformation of gastrointestinal parasites in equine species are shown in Table 3. BZD
BZDs generally results in metabolites of the parent drug which have and pro-BZD drugs are used widely to treat gastrointestinal hel-
reduced or no activity although the less oxidised sulphide metabolites minthiasis including migrating strongyle larvae and lungworm infec-
of FBZSO (FBZ) and ABZSO (ABZ) are thought to be active and the BZD tions (Marriner and Bogan, 1985). Most of the BZDs (CBZ, OFZ, FBZ,
metabolites of the pro-BZD’s are the active moieties. The plasma MBZ and OXB) and the pro-BZD (FBT) are highly effective (above 90%)
elimination half-lives of the parent drugs are short, and the metabolic against adult forms of the large strongyles, cyathostomins, mature
moieties predominate in plasma and tissues and in excreta of the host as Oxyuris equi, the small pinworm (Probstmayria vivipara) and Trichos-
well as in parasites recovered from BZD-treated animals (Delatour and trongylus axei in horses (Courtney and Robertson,1997).
Parish, 1986). Albendazole, FBZ, TCBZ and the pro-BZs (FBT and NTB), There are some notable differences in the efficacy of BZDs against
which are commercially available thioether and sulphide BZs com- different parasite species. Thiabendazole needs to be given at a higher
monly undergo a sulphoxidation by gastrointestinal microflora and dosage for Parascaris equorum and TBZ, FBZSO and FBZ are only ef-
liver microsomes. They are reversibly metabolised to their sulphoxide fective against Strongyloides westeri when given at a higher dose. At
derivatives (Marriner and Bogan, 1981b). Irreversible sulphonation 7.5 mg/kg daily for five days, FBZ was shown to have 94.6% efficacy
follows sulphoxidation and is a slower oxidative step resulting in a against larval stages of cyathostomins in enteric mucosa; to be 80%
sulphone metabolite (Averkin et al., 1975). effective against migrating larvae of Strongylus vulgaris and fully effec-
Hydroxylation is another important metabolic pathway of the BZDs. tive against migrating stages of Strongylus edentatus (Duncan et al.,
In sheep, it was demonstrated that hydroxy OFZ (OH-FBZSO) and hy- 1980). It has been shown that FBZ was highly efficacious against P.
droxy FBZ (OH-FBZ) were major hepatic metabolites of FBZ, after in- equorum (faecal egg count reduction range: 97.5–99.9%), however, ef-
traruminal administration (Hennessy et al., 1993). These metabolites ficacy of FBZ was low against Strongylus spp (faecal egg count reduction
are excreted directly into the bile as free or conjugated metabolites range: 0.4–41%) at a dose of 7.5 mg/kg following oral administration in
(Hennessy et al., 1993). Lacey et al. (1987) reported that OH-FBZ was horses in the UK because of the development of BZD resistance (Relf
intrinsically more active than the parent FBZ in vitro in binding to et al., 2014).
parasite tubulin. Mebendazole and FLBZ undergo carbonyl reduction to Oxibendazole has little or no effect against migrating stages of S.
the corresponding alcohol (Vandenbossche et al., 1982). The alcohol vulgaris, T. axei, Habronema muscae and Drashia megastoma (Kates et al.,
metabolite of MBZ was found at higher concentrations than the parent 1975; Nawalinski and Theodorides, 1977) in horses and ponies. Al-
drug in sheep plasma (Behm et al., 1983). However, MBZ metabolites though, Parbendazole has not been approved for horses, it is fully ef-
are not thought to have an anthelmintic activity (Meuldermans et al., fective against migrating larvae of S. edentatus at 20 mg/kg BW, (Lyons
1976). et al., 1980), but ineffective against 7-day-old larvae of S. vulgaris
The pharmacokinetics and activity of BZD anthelmintics are parti- (Drudge and Lyons, 1970). Parbendazole has no activity against T. axei,
cularly influenced by the physicochemical properties and metabolic Habronema spp. D. megastoma and S. westeri (Lyons et al., 1980). Al-
pathways of the active molecules. These compounds are only sparingly bendazole is effective against fourth-stage larvae of S. vulgaris with
soluble in water and the absorption and pharmacokinetics of these minor signs of toxicity at a dosage rate of 50 mg/kg twice a day for 2
drugs are affected by their aqueous solubility (Ngomuo et al., 1984). days (Georgi et al., 1980).
Pro-BZD compounds are relatively much more soluble in water com- Although NTB is not licensed for use in any equine species, NTB
pared to other BZDs; and this provides an advantage to create different treatment resulted in a high reduction of patent strongyle infections in

29
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Fig. 1. Chemical structures of benzimidazole (BZ) and pro-benzimidazole (pro-BZD) anthelmintics.

horses at a dose of 10 mg/kg BW (Gokbulut et al., 2009). Thus, NTB cent 2 and 4 weeks, respectively after treatments in horses.
reduced the strongyle EPG by 100% for 4 weeks. This is in line with the Most common endoparasites that affect horses can also cause
earlier report of (Mirck and Vanmeurs, 1982), who indicated that orally parasitic infections in donkeys and generally, the anthelmintic drugs
administered ABZ reduced the faecal egg counts by 99.1 and 93.9 per licensed for use in horses are effective in donkeys. The anthelmintic

30
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Fig. 2. Metabolic pathways of fenbendazole (FBZ)\ oxfendazole (OFZ or FBZSO), albendazole (ABZ) and triclabendazole (TCBZ).

drugs with broad spectrum of activity including BZDs available for use disintegration of the cells. It was found that the BZDs competed for the
in horses have been shown to be effective against the common helminth attachment site on β-tubulin with colchicine, a substance known to
infections in donkeys at doses determined for horses (Malan and prevent cell division in the metaphase (Sangster et al., 1985; Lacey and
Reinecke, 1979; Taylor and Craig, 1993; Trawford and Tremlett, 1996; Gill, 1994).
Imam et al., 2010; Gokbulut et al., 2014). However, in donkeys, after The selective anthelmintic action of BZDs is due to differences in the
oral administration of FBZ at a dose of 50 mg/kg BW, neither the parent sensitivity of host and parasite cells to the tubulin binding effects of
molecule or it’s metabolites were detected in blood and this may cause these drugs (Friedman and Platzer, 1980). Benzimidazoles bind re-
the lack of efficacy against lungworm (Dictyocaulus arnfieldi) infection versibly to mammalian tubulin, whereas they bind irreversibly to ne-
(Taylor and Craig, 1993). Mebendazole, FBZ and OFZ are registered matode tubulin (Lacey and Gill, 1994) and thus prevent permanent
BZD compounds for use in donkeys. They have similar spectra of ac- tubulin polymerisation to microtubules in Nematodes. In addition, the
tivity in this species as in the horse. However, for treatment of D. selective toxicity may be due to differences between the structure of
arnfieldi in the donkey, higher dose of MBZ should be used (20 mg/kg) host and nematode microtubules. Mammalian cells have 13 micro-
and administered on 5 consecutive days (McKellar and Scott, 1990). tubule protofilaments whereas nematode cells have 11, 12 and 14
Recently, it has been shown that in donkeys, MBZ paste at a horse protofilaments (Chalfie and Thomson, 1979).
dosage (10 mg/kg BW) was effective (> 95% efficacy) and safe for the
treatment of cyathostomins in donkeys (Gokbulut et al., 2016a). Tri-
2.4. Safety and toxicity
clabendazole, at 12 mg/kg BW is effective against Fasciola hepatica in-
fection in donkeys following oral administration (Trawford and
The BZD and pro-BZ anthelmintics have a high therapeutic index
Tremlett, 1996).
and are extremely well tolerated by mammals. BZD compounds with
low-solubility may be relatively less toxic than the more water-soluble
2.3. Mode of action compounds because insufficient drug is absorbed to exert a systemic
toxic effect (McKellar and Scott, 1990).
Benzimidazoles and pro-BZDs are considered to have a similar mode In horses, including pregnant mares, FBZ at a dose of 100 mg/kg
of action. Differences in their efficacy largely reflect differences in produced no adverse effects (Becker, 1975) and reproductive function
bioavailability. The mode of action of BZD anthelmintics was initially of stallions was not affected (Squires et al., 1978). In sheep FBZ is
thought to be inhibition of various parasite metabolic enzymes in- generally safe at high doses but when FBZ was given at 45 mg/kg daily
cluding fumarate reductase and malate dehydrogenase (Tejada et al., for 30 days, myocardial lesions and vacuolation of the liver par-
1987). However, it is now established that BZD anthelmintic com- enchyma were observed (Booze and Oehme, 1982), and although no
pounds prevent polymerisation of the microtubules in eukaryotic cells such chronic toxicity studies have been carried out in the horse one
by selectively binding to parasite β-tubulin resulting in the destruction might expect similar organ and tissue susceptibility.
of cell structure and consequent death of the parasite (Lacey, 1990; The main toxic effect of BZDs is teratogenicity. This varies with the
Martin, 1997). Borgers et al. (1975) reported that after exposure of cells structure of the BZD compounds and the animal species (Delatour and
from nematodes to MBZ, the cytoplasmic microtubules disappeared, Parish, 1986). The teratogenic activity of FBZSO and ABZSO has been
and this caused disruption in the migration of subcellular organelles shown when administered to rats in early pregnancy (Delatour et al.,
with a failure of transport of secretory materials in the cells. Prolonged 1984). Sheep and rats are much more sensitive than other species of
storage of the secretory granules caused lysis of the cell cytoplasm and animals to the teratogenic activity of the BZDs and extensive studies

31
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

+++, 95–100% efficacy; ++, 80–100% efficacy; +, 0–100 efficacy; o, no efficacy; -, insufficient data. * MXD exhibited moderate efficacy (64% and 85%) against early third stage (EL3) and late third stage (LL3)/fourth
have been carried out in these species. Congenital malformations were
Trichostrongylus axei
reported during early gestation in ewes following administration of CBZ
(Delatour et al., 1976), FBZSO (Delatour et al., 1977) ABZ (Johns and
Philip, 1977) and FBT (Chement, 1982). The main congenital defects

+++

+++

+++
+++
identified in the lambs were skeletal malformations, occurring mainly

++
+
in the long bones, pelvis, joints and digits. The microtubular activity of
o
o
o

o
o

o



BZD compounds may lead to their teratogenic activity (McKellar and
megastoma

Scott, 1990). It is the unbound drug or metabolites, which are likely to


Draschia

exert toxic effects in mammals and the tightly protein-bound residues


that persist in the tissues for longer periods of time, are thought to be of
o
o
o
o
o
o

o








lower significance toxicologically (Delatour and Parish, 1986).
Habronema spp.

Teratogenic activity studies of the BZDs in early pregnancy in rats

stage (L4) encysted cyathostomins, respectively (Reinemeyer et al., 2015) whereas IVM was ineffective (0%) against LL3 and L4 stages of cyathostomin larvae (Xiao et al., 1994).
have shown that this species has similar sensitivity to sheep.
Teratogenic and embryo lethal effects have been reported for PBZ,

++
++
MBZ, CBZ, ciclobendazole, carbendazim, FBZSO, ABZ, FBT and NTB
o
o
o
o
o

o
o
o

o



(Delatour and Parish, 1986; Tsuchiya et al., 1987; Mantovani et al.,
Strongyloides

1995; El-Makawy et al., 2006). In male rats, testicular degeneration and


abnormal spermatogenesis were produced by high doses of carben-
+++
+++

+++

+++

+++
+++
westeri

dazim (Styles and Garner, 1974) and the mutagenic effects of carben-
o
o
o
o

o
o




dazim have also been shown in other species at high doses (Seiler,
equorum L5

1976).
Parascaris

+++

+++
+++
+++
+++
+++
+++

+++
+++
+++
+++
+++
+++
The activity of anthelmintic compounds used for the treatment of common gastrointestinal nematodes in equines (Adapted from Mirck, 1985).

++
++

2.5. Pharmacokinetics
+

+

equorum L4

The active BZD anthelmintics are thought to have a similar mode of


Parascaris

action (Coles, 1977). The different efficacies of the BZDs in vivo have
+++
+++
+++

+++
+++
+++

+++
+++

therefore been attributed largely to different pharmacokinetics within









the host (Bogan, 1983) and in vitro, to different solubility and therefore
absorption of the drug from culture media by the test parasites (Scott,
Strongylus
vulgaris

1988). Anthelmintic activity depends on the duration of parasite ex-


+++
+++
+++
+++
+++
+++
+++
+++
+++
+++
+++

+++
+++
+++
+++

posure to effective concentrations of the active compound (Baggot and


+
+

McKellar, 1994), thus observations on the pharmacokinetics of BZDs


are extremely important.
Strongylus
edentatus

Most of the BZD anthelmintics including methylcarbamate BZDs


+++
+++
+++

+++
+++
+++
+++
+++

+++
+++
++

++

have very limited absolute solubility in water and the dispositions of


+
+
+
o

o

these drugs are affected by their aqueous solubility. The relatively rapid
dissolution and consequent absorption from the gastrointestinal tract
Strongylus
equinus

+++
+++

+++
+++
+++
+++
+++

+++

+++
+++

and elimination of some BZDs explain their relatively short residence in


the body. Thus, more soluble compounds have a shorter residence in
+
o


comparison to less soluble BZDs, which are absorbed over prolonged


periods. Thiabendazole and CBZ, the most water-soluble of the ther-
strongyles

apeutic BZDs, are more extensively and rapidly dissolved in rumen fluid
+++
+++
+++
+++
+++
+++
+++
+++
+++
+++

+++
+++

+++

+++
+++
Small

++

and are thus absorbed more rapidly in ruminants (McKellar and Scott,
L5

1990). The less soluble BZD compounds remain as a slurry in the re-
ticulorumen and are absorbed over prolonged periods and thus remain
strongyles

+++

+++

+++
+++

in the plasma for a longer time. Since an equilibrium exists between the
Small

plasma and the gastrointestinal tract, the duration of exposure of the


L4

+
+

+
+

+




gut parasites to effective concentrations of the drug is extended. At the


extreme limits of solubility anthelmintics may be less effective since
Oxyuris
equi L5

+++
+++
+++
+++
+++
+++
+++
+++
+++

+++

+++
+++
+++
+++
+++
++

they may not be absorbed sufficiently and are excreted unchanged in


the faeces. This may explain the difference between the plasma con-
centrations of FBZSO achieved following oral administration of FBZSO
Oxyuris
equi L4

+++

+++

+++
+++
+++
+++

+++
+++
+++
+++

as the parent drug compared to its residence following administration


+

+
+

o

of its interconvertible metabolite FBZ (Ngomuo, 1983). A large pro-


portion of the less soluble FBZ is known to be excreted directly in the
Probstmayria

faeces (Prichard, 1978; McKellar et al., 2002; Gokbulut et al., 2006).


vivipara

The pharmacokinetics of BZD anthelmintics has been studied in


+++
+++

+++
+++

+++

+++

+++
+++

horses in which the metabolic interconversion of the sulphide and


o

o




sulphoxide BZDs (FBZ and OFZ respectively) appears to differ sub-


35-40

stantially from that in ruminants (Marriner and Bogan, 1985). Sulphide


(mg/
Dose

200
kg)

2.5
8.8

6.6

0.2
0.4
50
20

10
10

10
75

40
60

and sulphoxide BZDs are known to bind nematode tubulin (Lacey et al.,
5

5
6

1987) and therefore have activity against nematodes although sul-


Thiabendazole
Cambendazole

Phenothiazine
Fenbendazole

Oxibendazole
Parbendazole
Mebendazole

Oxfendazole

Moxidectin*
Albendazole

Ivermectin*
Trichlorfon
Levamisole

phides exert inhibitory activity on tubulin at lower concentrations than


Dichlorvos
Piperazine
Febantel

Haloxon
Pyrantel

sulphoxides. Nevertheless, in most species examined the sulphoxide


Table 3

Drugs

moiety predominates in plasma and is thought to confer activity against


gut-dwelling nematodes following secretion across the gastrointestinal

32
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

wall into the gut lumen where it may undergo sulpho-reduction. demonstrated that fasting or restricted feed intake increased the re-
There are significant differences in the pharmacokinetics of BZD lative bioavailability of ABZ metabolites (Sanchez et al., 1997). How-
anthelmintics between monogastric and ruminant species (Marriner ever, in the dog (McKellar et al., 1993a) feeding increased the bioa-
and Bogan, 1981b; McKellar and Scott, 1990). In contrast to other vailability of BZDs after oral administration. Although there were no
species (sheep, cattle, man), following treatment of FBZ or FBZSO statistically significant effects of feeding on the absorption of FBZ
equine species generate relatively higher concentrations of the more (10 mg/kg) in horses, the cumulative AUC for the active metabolic
oxidised sulphone metabolite than of sulphoxide. This may be due to moieties (FBZSO and FBZ) were four times as large in unfed than fed
the relatively rapid conversion of sulphoxide to the sulphone by the horses suggesting that FBZ might be more effective in the horse fol-
equine liver (Marriner and Bogan, 1985). The low plasma concentra- lowing food restriction (Table 6) (McKellar et al., 2002).
tions of active moieties (FBZ, FBZSO) following FBZ administration Parasitic infections of the gastrointestinal tract have been demon-
probably accounts for the higher and repeated dosage required for the strated to affect the bioavailability of anthelmintics in animals. In sheep
treatment of migrating larval and tissue stages of strongyles and lung infected with Ostertagia circumcincta and treated with FBZ, plasma
worms (Marriner and Bogan, 1981a). concentrations of both FBZ and FBZSO were reduced when compared
The BZD anthelmintic drugs have lower bioavailability in mono- with uninfected animals treated with the same dose of FBZ (Marriner
gastric animals (e.g. dog and horse), which have relatively faster gut et al., 1985). Moreover, the plasma concentration of FBZSO was re-
transit time than ruminants since rapid passage of food in the gut de- duced by 25% in goats infected with the same nematode compared to
creases the absorptive time for the drugs. Dougherty, (1992) reported uninfected goats given the same dose of FBZSO (Bogan et al., 1987). In
gut transit time of 8.47 h in horses. In ruminants, this time may range contrast, after intraruminal administration of ABZ, the AUC of the
from 30 to 80 h depending on the digestibility of food, since highly ABZSO was larger in sheep naturally and artificially infected with
digestible food has a shorter retention time (Warner, 1981). The sys- Haemonchus contortus than in non-infected animals (Alvarez et al.,
temic bioavailability of most drugs administered orally is lower in 1997). It has been shown that the pH of abomasal fluid was decreased
herbivorous species (horse and ruminants) than in dogs and cats. The by the presence of nematode parasites (Mostofa and McKellar, 1989;
greater “first-pass effect” and higher drug metabolising capacity of the McKellar et al., 1990). In addition, helminthiasis produces a constant
reticulo-rumen and liver account for this effect. In ruminants, the op- stimulus for gastrin secretion leading to hypergastrinaemia and pro-
posite is true for BZD drugs. After oral administration to ruminants, nounced hyperplastic changes in the abomasal mucosa (Anderson et al.,
greater bioavailability is observed than in monogastric species because, 1988). These changes probably affect absorption of BZDs in ruminants;
in ruminants, the relatively slower gut transit time conferred by the hence the pharmacokinetic behaviour and expected efficacy may be
forestomach reservoir means that BZDs are more extensively absorbed reduced by the presence of the parasites. An elevation in gastric pH
than in monogastric animals (Baggot and McKellar, 1994). In addition, causes a reduction in solubility of the BZDs and this would contribute to
FBZ or FBZSO were extensively absorbed when administered directly decreased bioavailability. It has been reported that there is a reduction
into the cecum in horses (McKellar et al., 2002). The anatomy of the in the bioavailability of the pro-BZD, FBT in lambs infected with Os-
equine ileo-cecal valve makes retrograde delivery of the drug into the tertagia, and in those infected with Trichostrongylus colubriformis there
distal ileum from the cecum unlikely, although not impossible. was a reduction in intestinal motility and mucosal villous atrophy, both
Previous studies in horses and donkeys indicated that the plasma of which could affect drug absorption (Debackere et al., 1993). The
concentrations of FBZSO and FBZ were relatively low and that they are pathophysiological changes associated with stomach parasites in the
extensively metabolised to their sulphone metabolite when each was horse have been less well studied and represent a less important group
administered at10 mg/kg BW. In horses, after oral administration of of parasites than those infecting the abomasum in ruminants. It is likely
FBZSO at a dose of 10 mg/kg BW, the area under the curve (AUC) for nevertheless, that stomach and intestinal parasites will have an effect
the parent drug and its metabolite FBZ were 4.36 and 1.40 μg h/mL on the absorption of BZD’s in horses. Parasitic infection may also alter
respectively (McKellar et al., 2002). In a similar study, carried out with drug metabolism, which could affect the disposition of the anthelmin-
FBZ at the same dose rate, lower plasma concentrations of parent drug, tics. It has been shown that the enzymatic activity of the hepatic mi-
FBZ, and its metabolite FBZSO were obtained, with FBZSO being below crosomal cytochrome P450-dependent monooxygenase system is de-
the limit of detection (< 0.04 μg/mL) and FBZ having an AUC of pressed in rats infected with F. hepatica (Tekwanl et al., 1988).
1.77 μg h/mL (Marriner and Bogan, 1985). This reflects the better ab- Similarly, in sheep, F. hepatica infection decreased sulphonation of ABZ
sorption and greater systemic availability of FBZSO compared to FBZ and this was related to a decline in liver microsomal P450-dependent
since each compound produces the same metabolites and these would monooxygenase activity (Galtier et al., 1986). In contrast to these ob-
be expected to have similar metabolic and excretory rates. It is of in- servations, there were no significant differences in the bioavailability of
terest that the AUC of the sulphide moiety was 2.3 times greater in anthelmintics (levamisole, Ivermectin, NTB and ABZ) in sheep infected
horses administered FBZSO than in horses given FBZ as the parent with Nematodirus battus and non-infected sheep (McKellar et al., 1991,
molecule. The much higher solubility of FBZSO (3.01 mg/L) than that 1993b). There does not appear to be available data on the effects of
of FBZ (0.07 mg/L) (Marriner and Bogan, 1985) may explain its greater parasitism on anthelmintic disposition in horses.
absorption at a rate exceeding its oxidative clearance and this may Oxibendazole is a member of BZD anthelmintic class with a broad
provide sufficient substrate for reductive metabolism to the sulphide in spectrum of activity against gastrointestinal helminths only, which has
horses. The limited adsorption rate of FBZ may be matched by rapid been marketed for use in horses for many years. Gokbulut et al. (2002)
oxidative metabolism with consequent low concentrations achieved in reported that the plasma concentrations of the parent OXB were rela-
plasma following its oral administration. tively very low following oral administration in horses (Table 4) and a
In animals, diet can substantially affect the bioavailability of drugs. liver microsome study showed that OXB was metabolised extensively to
Some non-steroidal anti-inflammatory drugs bind to food particles in unidentified metabolites in horses. Gottschall and Wang (1996) re-
horses which delay the absorption of the drugs significantly (Lees et al., ported that in swine OXB was quickly metabolised and liver was the
1988). Hennessy (1993) emphasised the role played by the adsorption only tissue which contained significant residue following oral admin-
of BZDs on the ruminal particulate material in the extension of their istration at a dose of 15 mg/kg BW. It was reported that OXB has little
bioavailability in ruminant species. The physicochemical nature of drug or no effect against migrating stages of S. vulgaris, or against T. axei, H.
association with particulate material is likely to be due to physical muscae and D. megastoma (Kates et al., 1975; Nawalinski and
adsorption rather than specific chemical binding (Lees et al., 1988). In Theodorides, 1977). Interestingly OXB has been shown to have activity
sheep, the relative bioavailability of BZDs decreased when administered against gastrointestinal parasites selected for resistance to other BZDs
concomitantly with food (Taylor et al., 1992). In cattle, it was (Uhlinger et al., 1986). High gastrointestinal concentrations of OXB

33
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Table 4
Pharmacokinetic parameters of benzimidazole (BZD) anthelmintics in equine species following oral administration.
Pharmacokinetic parameters References

2
Species Benzimidazoles Abbreviations Tmax (h) Cmax (μg/mL) AUClast (μg.h/mL) AUMClast (μg. h /mL) T1/2λz (h) MRTlast (h)
(10 mg/kg)

Horse Fenbendazole FBZ 8.00 ± 2.70 0.04 ± 0.01 0.61 ± 0.11 9.33 ± 2.89 – 14.21 ± 1.74 (McKellar et al., 2002)
FBZSO 9.50 ± 3.52 0.01 ± 0.00 0.17 ± 0.02 2.26 ± 0.46 – 12.90 ± 1.33
FBZSO2 10.50 ± 3.20 0.06 ± 0.01 1.12 ± 0.19 12.54 ± 2.43 – 16.50 ± 1.00
FBZ 10.00 ± 7.38 0.11 ± 0.05 – – – – (Marriner and Bogan,
FBZSO2 12.00 ± 6.20 0.16 ± 0.08 – – – – 1985)
Oxfendazole OFZ 8.88 ± 3.02 0.35 ± 0.07 4.36 ± 0.89 69.07 ± 21.85 – 14.77 ± 2.32 (McKellar et al., 2002)
FBZ 13.50 ± 1.99 0.09 ± 0.02 1.40 ± 0.32 23.45 ± 6.98 – 15.58 ± 1.01
OFZSO2 12.00 ± 3.63 0.76 ± 0.09 13.00 ± 1.55 210.24 ± 45.84 – 15.45 ± 2.10
OFZ 7.20 ± 3.91 0.43 ± 0.13 7.10 ± 2.65 139 ± 60.0 10.4 ± 3.84 16.8 ± 6.99 (Bruni et al., 2005)
OFZSO2 16.9 ± 5.37 0.27 ± 0.12 5.88 ± 2.52 116 ± 75 4.58 ± 0.94 17.6 ± 3.97
Oxibendazole OXB 0.81 ± 0.53 0.01 ± 0.01 0.07 ± 0.05 – – 14.17 ± 3.82 (Gokbulut et al., 2002)
Netobimin ABZSO 10.50 ± 3.66 0.53 ± 0.14 8.63 ± 1.01 129.12 ± 20.10 5.97 ± 1.59 15.08 ± 2.50 (Gokbulut et al., 2009)
ABZSO2 19.50 ± 3.96 0.36 ± 0.09 8.21 ± 2.87 177.55 ± 74.17 7.44 ± 1.06 21.33 ± 2.31
Triclabendazole TCBZSO 8.00 ± 00 4.57 ± 1.11 115.00 ± 25 – 9.74 ± 00 – (Kinabo and Bogan,
(12 mg/kg) TCBZSO2 25.33 ± 6.67 13.15 ± 1.56 594.00 ± 97 – 17.65 ± 00 – 1989)
Donkey Oxfendazole OFZ 5.67 ± 2.89 0.49 ± 0.11 5.17 ± 0.82 58.12 ± 14.18 4.49 ± 0.74 10.95 ± 1.92 (Gokbulut et al., 2006)
FBZSO2 8.00 ± 2.53 0.60 ± 0.09 10.33 ± 0.87 161.00 ± 22.99 7.53 ± 0.35 15.38 ± 1.50
Albendazole ABZSO 5.71 ± 0.87 0.08 ± 0.01 0.84 ± 0.14 8.27 ± 1.86 6.65 ± 3.22 9.15 ± 1.20
ABZSO2 8.00 ± 2.31 0.04 ± 0.00 0.50 ± 0.11 5.68 ± 1.93 7.44 ± 1.19 9.98 ± 1.58
Mebendazole MBZ 7.33 ± 3.93 0.04 ± 0.01 0.78 ± 0.35 19.49 ± 7.86 11.97 ± 4.38 20.34 ± 7.59 (Gokbulut et al.,
(10 mg/kg) 2016a)
Mebendazole MBZ 8.00 ± 0.00 0.07 ± 0.01 1.42 ± 0.20 33.78 ± 10.38 13.13 ± 3.85 23.43 ± 5.55
(20 mg/kg)
Triclabendazole TCBZSO 9.33 ± 1.33 4.34 ± 0.86 100.00 ± 16 – 9.40 ± 00 – (Kinabo and Bogan,
(12 mg/kg) TCBZSO2 12.00 ± 00 2.94 ± 0.44 63.00 ± 6.00 – 9.06 ± 00 – 1989)
Pony Triclabendazole TCBZSO 26.67 ± 2.67 4.01 ± 0.25 157.00 ± 20 – 10.91 ± 00 –
(12 mg/kg) TCBZSO2 29.33 ± 2.67 13.00 ± 2.30 710.00 ± 81. – 23.08 ± 08 –

Cmax: peak plasma concentration; Tmax: time to reach peak plasma concentration; AUClast: area under the (zero moment) curve from time 0 to the last detectable
concentration, MRT: mean residence time; T1/2λz: terminal half-life, AUMC: area under the first moment curve.

observed from a faecal study could be effective for adult stages of most environment of the gastrointestinal tract of horses. It has been shown
parasitic nematodes that inhabit in the gastrointestinal tract. Never- that following intraruminal or oral administration, NTB is reduced to
theless, low plasma concentrations of OXB are unlikely to be effective ABZ in the gastrointestinal tract of cattle and sheep (Lanusse and
against migrating larval and tissue stages of nematodes (Gokbulut et al., Prichard, 1990; Lanusse et al., 1993). Some pharmacokinetic para-
2002). It is likely that high first-pass metabolism decreases OXB meters of BZD anthelmintics (FBZ, FBZSO, OXB and NTB) previously
availability in the plasma of horses and it seems unlikely that phar- studied in horses are shown in Table 4. These studies reported that FBZ
macokinetic characteristics can explain the sustained activity of OXB or its active sulphoxide metabolite, FBZSO and OXB displayed relatively
against resistant nematodes since it has characteristics which ought to low plasma concentration, and were quickly metabolised to their in-
confer poorer activity against nematodes compared to other members active sulphone metabolites which predominated in plasma, whereas in
of this anthelmintic class. It is possible that the OXB exerts its effects on the NTB study, active ABZSO is predominant and displayed higher
resistant nematodes as a consequence of its affinity for tubulin (Lacey plasma disposition compared to ABZSO2 after NTB administration.
and Watson, 1985). The metabolic inhibition of OXB by co-adminis- These differences could be due to relatively slow oxidation of ABZSO to
tration with piperonyl butoxide (PB) previously indicated for FBZ and ABZSO2 in equine species (Gokbulut et al., 2006, 2009).
OFZ could be a strategy to extend the exposure of nematodes to the There is a paucity of data available on the pharmacokinetics or ef-
parent active moiety and thus to improve the efficacy of OXB in horses. ficacy of anthelmintics used in donkeys, as donkeys are an often-ne-
The gastrointestinal environment of equine species has a great re- glected species. Different classes of drugs used in horses and ruminants
ductive capacity for BZD compounds. It has been shown that the BZD are commonly extrapolated for use in donkeys without optimization of
anthelmintics licensed for use in horses in some countries (e.g. FBZ, dosing regimens and determination of pharmacokinetic and pharma-
OFZ and OXB) have relatively poor systemic availability, since they are codynamic properties. It has been reported that donkeys have a greater
extensively metabolised to their inactive metabolites by gastrointestinal capacity to metabolize certain drugs compared with horses; and al-
microflora and/or first-pass metabolism compared to ruminant species though there are some exceptions, higher dosage or shorter intervals
(Gokbulut et al., 2002; McKellar et al., 2002; Gokbulut et al., 2006). could be required in order to reach and maintain effective drug con-
The detailed characterization of the kinetic behaviour and metabolic centrations (Matthews et al., 1997; Kum et al., 2009; Grosenbaugh
pathways of a pro-BZD, NTB, has markedly contributed to its optimized et al., 2011; Sekkin et al., 2012). Gokbulut et al. (2006a) reported that
use in ruminants. Although, NTB is not licensed for use in equine spe- the systemic availability of FBZSO was significantly greater than that of
cies, the anthelmintic efficacy, plasma disposition and faecal excretion FBZ and ABZ following oral administration at same dose rate in don-
of NTB, and its metabolites (ABZ, ABZSO and ABZSO2), at 10 mg/kg keys. This probably reflects better water solubility and absorption of
were determined by Gokbulut et al. (2009) in horses after oral treat- FBZSO than FBZ and ABZ since the same metabolites are produced by
ment. This study indicated that NTB was not detectable in any plasma each compound and these would be expected to have similar metabolic
or faecal samples; however, ABZ was predominant in the faeces. Ne- and excretory rates. Although, the plasma dispositions of FBZSO and its
tobimin produced similar metabolites and disposition in horses com- sulphone metabolite in donkeys showed a similar profile to that in
pared with ruminant species. This suggested an efficient conversion of horses (Marriner and Bogan, 1985; McKellar et al., 2002). Unlike
the pro-drug NTB into the active form ABZ in the reductive horses, FBZ and its sulphoxide and sulphone metabolites were not

34
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

detected in the plasma of donkeys at any time. It is likely that FBZ dosage regimen of MBZ could be used for horses and donkeys (Gokbulut
absorption from the gastrointestinal track is substantially lower in et al., 2016a).
donkeys than horses and larger faecal excretion of FBZ than FBZSO and The plasma dispositions of MBZ (Gokbulut et al., 2016a) was dif-
ABZ supports this finding. The higher solubility of FBZSO (3.01 mg/L) ferent from those of FBZ, FBZSO and ABZ (Gokbulut et al., 2006) stu-
than of FBZ (0.07 mg/L) (Marriner and Bogan, 1985) may explain its died in donkeys at the same dosages (Table 4). The peak plasma con-
greater absorption in donkeys. It is also apparent that the sulphone and centrations and areas under the curve of OFZ (Cmax: 0.49 μg/mL, AUC:
sulphoxide metabolites were predominant in plasma following admin- 5.17 μg.h2/mL) and ABZ (Cmax: 0.08 μg/mL, AUC: 0.84 μg h2/mL) were
istration of FBZSO and ABZ in donkeys. It was demonstrated that horses higher than those of MBZ (Cmax: 0.04 μg/mL, AUC: 0.78 μg.h2/mL),
metabolize FBZ and FBZSO to their sulphone metabolites more quickly respectively. Nevertheless, the T1/2 (11.97 h) and MRT (20.34 h) values
than ruminants (Marriner and Bogan, 1985; McKellar et al., 2002). of MBZ are much longer compared with those of FBZSO (T1/2: 4.49 h,
Although the bioavailability of ABZ was lower than in ruminants, its MRT: 10.95 h) and ABZ (T1/2: 6.65 h, MRT: 9.15 h) at the same dosage
metabolic products displayed a similar profile to ruminants. Plasma (10 mg/kg). These differences probably reflect lower water solubility
dispositions of ABZ displayed a different profile compared to those of and subsequent lower absorption of MBZ compared with FBZSO and
FBZSO and FBZ in donkeys. Although, the parent ABZ was not detected ABZ but subsequent slower metabolism and excretion of the MBZ which
in plasma, its sulphoxide and sulphone metabolites appeared in plasma has been absorbed. Similarly, low systemic availability has been ob-
following oral ABZ administration. In addition, Cmax and AUC of the served with MBZ in humans and it has been suggested that this is a
sulphoxide metabolite (0.08 μg and 0.84 μg h/mL, respectively) which result of a very low level of absorption of MBZ from the gastrointestinal
is thought to be anthelmintically active were significantly higher and tract (Munst et al., 1980).
larger, respectively than that of the sulphone metabolite (0.04 μg/mL The plasma disposition of MBZ in donkeys also differs substantially
and 0.5 μg h/mL, respectively) which is thought to be anthelmintically from those observed in sheep and goats in which higher concentrations
inactive (Gokbulut et al., 2006). are observed (Galtier et al., 1994). The reason for the lower systemic
It has been reported that the time until the maximum faecal con- exposure in donkeys compared with that observed in ruminant species
centrations (32 h for FBZSO, 34 h for FBZ and 30 h for ABZ) were longer is probably related to differences in the physiological or anatomic
in donkeys compared to horses (24 h for FBZ and FBZSO) reflecting properties between the animal species (McKellar and Scott, 1990). The
slower gut transit time following oral administration in donkeys benzimidazole anthelmintic drugs have lower bioavailability in mono-
(McKellar et al., 2002; Gokbulut et al., 2006). It is likely that hay-based gastric animals (e.g. dog and horse), which have relatively faster gut
diet and different anatomic characteristics caused this difference transit time than ruminants since and rapid passage of food in the gut
(Gokbulut et al., 2006). In sheep and cattle, it was reported that the causes a decrease in the bioavailability of the drugs.
reduction of FBZSO to its sulphide (FBZ) occurred in ruminal fluid Kinabo and Bogan (Kinabo and Bogan, 1989) studied the plasma
(Beretta et al., 1987). The reductive environment of the gastrointestinal dispositions of TCBZ and its sulphone and sulphoxide metabolites in
tract of the donkey is likely to be responsible for similar metabolic re- horses, ponies and donkeys after oral administration of the drug at the
duction in that species. Following FBZ administration, the parent drug dose (12 mg/kg BW) recommended for ruminant species (Table 1). In
was predominant in faecal samples and its sulphoxide metabolite was this study, the AUC values of the sulphoxide (TCBZSO) were about 5
found in extremely low concentrations. Following ABZ administration times lower in equines compared to ruminant species (Kinabo and
its sulphoxide metabolites were detected at significantly greater con- Bogan, 1988). From this, it could be inferred that triclabendazole is less
centrations in faeces than FBZSO following FBZ administration. The well absorbed in equine than in ruminant species. This could be at-
extremely poor water solubility of FBZ may be the reason for this since tributed to factors such as variations in pH of the gastrointestinal
it is likely that absorption and oxidative metabolism are necessary to contents, gastrointestinal transit times and digestive tract capacities.
generate FBZSO and this will be restricted for drugs undergoing Because the sulphoxide concentrations may be a more accurate reflec-
minimal dissolution. In addition, following FBZSO administration, the tion of likely flukicidal activity, it would seem that a relatively higher
parent molecule was extensively metabolised to its sulphide metabolite dose of triclabendazole may be required in equine species (Kinabo and
(FBZ) which displayed a greater faecal excretion profile than that of the Bogan, 1989). Moreover, significant differences between donkeys and
parent FBZSO. Interestingly the sulphide metabolite was not detected in horses and ponies were observed for the sulphone metabolite of TCBZ
any plasma samples and it is possible that high first-pass metabolism whereby the Cmax and the AUC in donkeys were lower than in horses
decreased the sulphide metabolite bioavailability by converting back to and ponies (Table 4). It is probable that donkeys are capable of ex-
parent FBZSO. In other words, the rate of sulphoxidation in liver ex- creting the sulphone more efficiently than horses and ponies. Never-
ceeded the rate of gastroenteric reduction and absorption of the parent theless, these differences may be of little therapeutic significance be-
FBZSO. This may explain why the bioavailability of FBZSO was sig- cause the sulphone metabolite is generally devoid of anthelmintic
nificantly greater than that of FBZ and ABZ following oral administra- activity (Kinabo and Bogan, 1989).
tion at the same dose rate in donkeys.
Recently, the plasma disposition, milk excretion and anthelmintic 2.5.1. Potentiation of BZDs anthelmintics
efficacy of MBZ were investigated in donkeys following oral adminis- It is likely that metabolic inhibition will be more effective than
trations at 10 mg/kg and 20 mg/kg BW (Gokbulut et al., 2016a). A simply increasing the dose rate in order to improve the efficacy of BZDs,
dose-dependent plasma disposition of MBZ was observed (Table 4). At a since their pharmacokinetics in some monogastrics are dose-in-
dosage of 10 mg/kg BW, MBZ was not detected in any milk samples. dependent and absolute bioavailability cannot be increased by in-
However, MBZ reached peak milk concentration of 0.01 μg/mL at creasing the dose (McKellar et al., 1993a). The potentiation of the ac-
10.66 h and AUCmilk/AUCplasma value was 0.18 ± 0.02 following tivity of benzimidazoles and pro-benzimidazoles has been achieved by
20 mg/kg BW. Mebendazole appears to have a minimal disposition in the co-administration with metabolic inhibitors, which are thought to
milk suggesting that it may be used in lactating donkeys and that a zero act principally by improving the pharmacokinetic profiles of the active
milk-withdrawal period might be appropriate in this species, although moieties of the administered drug. The potentiating agents for the BZDs
further work on metabolism in this species would be necessary before a and pro-BZDs may be useful in practice since an increase in the ratio of
maximum residue limit could be set. The results of a faecal egg count sulphide + sulphoxide: sulphone metabolites occur, and the less oxi-
reduction (FECR) test for both MBZ dosages indicated high efficiency dised metabolites (sulphide) of the BZDs are considered to exhibit
(> 95%) until day 28 after treatment. The trial demonstrated that MBZ greater binding to nematode tubulin (Lacey et al., 1987).
oral paste at the horse dosage (10 mg/kg BW) was effective for the Two main microsomal enzymatic pathways have been proposed as
treatment of cyathostomins in donkeys and it is likely that a similar responsible for the sulphoxidation of sulphur containing BZDs including

35
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

ABZ, OFZ and TCBZ. The flavin-containing mono-oxygenase (FMO) BZD sulphides and sulphoxides when combined with PB. It is antici-
enzymatic system is responsible for sulphoxidation (Galtier et al., 1986) pated that the synergistic effects demonstrated could have major posi-
whereas the hepatic cytochrome P450 metabolic pathway is probably tive benefits for greater drug exposure of the target parasites and for the
responsible for the oxidation of BZ-SO to BZ-SO2 metabolite treatment of BZD resistant parasites in equine species. These findings
(Souhailielamri et al., 1988). Metabolic inhibitors such as methimazole have also indicated that PB had a more dramatic synergistic effect on
which inhibits flavin-containing mono-oxygenase and metyrapone the plasma dispositions of BZDs in horses compared to that in rumi-
which inhibits cytochrome P450 prevent the oxidative metabolism of nants, probably because equine species oxidise BZD sulphides to
NTB and ABZ sulphoxide thus improving the efficacy of these anthel- sulphoxide more rapidly than ruminants (McKellar et al., 2002).
mintics and pharmacokinetic profile of the active metabolites (Lanusse
and Prichard, 1992a,b). The bioavailability of anthelmintic metabolites 2.5.2. Stereospecific dispositions of BZD anthelmintics
was also improved when FBZ and FBZSO were co-administered with Enantioselective or chiral compounds are extensively used in ve-
methimazole (Lanusse et al., 1995). Co-administration of PBZ with terinary practice. Pharmacodynamic and pharmacokinetic properties of
FBZSO has been shown to increase the anthelmintic activity of FBZSO enantiospecific pairs are commonly different and are of major im-
and confer activity against BZD-resistant nematodes. This has been at- portance for their effective and safe therapeutic use (Delatour et al.,
tributed to a reduction in hepatic biotransformation and biliary secre- 1994).
tion of FBZSO. Parbendazole promotes an increase in extra-biliary Sulphur-containing BZD anthelmintics (FBZ, OFZ or FBZSO and
transfer of FBZSO into the intestinal lumen and thus exposure of the ABZ), form a chiral centre about the sulphur atom when administered
parasites to increased drug concentrations in the gastrointestinal lumen as sulphoxides (FBZSO) or when metabolised into their respective
(Hennessy et al., 1992). sulphoxides (FBZSO, ABZSO), they are subsequently metabolised into
Benchaoui and McKellar (1996) have shown that PB co-adminis- sulphones metabolites which are anthelmintically inactive, whereas
tration greatly improves the activity of FBZ against nematodes resistant sulphides and sulphoxides are both active. The stereospecific behaviour
to BZDs in sheep when administered at normal therapeutic doses. The of BZD sulphoxides has been investigated in the plasma of various
methylenedioxyphenyl compound PB is a potent inhibitor of the cyto- species following administration of the prochiral sulphide parent mo-
crome P450 oxidation of BZD sulphoxides to sulphones and it was lecule (Delatour et al., 1990, 1991b). In sheep, the ratio of FBZSO en-
shown to reduce the metabolism of FBZ in cultured rat hepatocytes by antiomers changed from 1.8 to 6.7 between 9 h and 120 h, and alben-
50% in 24 h (Benchaoui and McKellar, 1996). It was demonstrated that dazole sulphoxide (ABZSO) enantiomer ratio changed from 3.3 to 22.4
the major route of metabolism of PB involved the opening of the me- between 3 h and 36 h. The enantiospecific ratio of AUC values of
thylenedioxy ring followed by loss of the methylene group into the sulphoxide metabolites, following administration of sulphides, was
endogenous metabolic pool (Cockburn and Needham, 1999). This is 26:74 for FBSO and 14:86 for ABZSO enantiomers (Delatour et al.,
also believed to be the basis of the initial inhibition of the cytochrome 1990). The stereospecific behaviour of ABZSO was shown to be dif-
P450 enzyme system, which is essential for the compound’s efficacy as ferent between monogastrics and ruminants (Delatour et al., 1991a, b).
a synergist. Current evidence suggests that it is the carbene inter- The enantiospecific ratio (+/-) of plasma concentration of ABZSO
mediate formed with the methylene group that complex with the Fe++ changed over time in favour of (+) in all species examined, with the
ion of cytochrome P450 (Wilkinson et al., 1984; de Montellano and exception of rats, and the initial (+) / (−) plasma ratio was close to a
Reich, 1986) that induces its effect. Piperonyl butoxide has shown racemate (50:50) in monogastrics but was 75:25 in sheep. These results
isozyme specificity in rats using different probe drugs for hepatic drug- were confirmed by incubation of liver microsomes using ABZ as the
metabolising activity (Bachmann, 1989). In sheep, it was demonstrated substrate (Galtier et al., 1986; Moroni et al., 1995). It is thought that the
that co-administration of PB with FBZ significantly enhanced the FMO is mainly responsible for sulphoxidation, whereas cytochrome-
pharmacokinetic profile and potentiated the anthelmintic activity of the dependent monooxygenase is responsible for sulphonation (Benoit
BZDs. A dose-dependent inhibition of FBZ sulphoxidation by PB im- et al., 1992). The initial enantiospecific ratio was 30:70 when co in-
proved the efficacy of the drug against nematodes of sheep, including cubated with methimazole (an inhibitor of FMO) and 65:35 in the
BZD-resistant O. circumcincta and H. contortus (Benchaoui and McKellar, presence of clotrimazole (an inhibitor of cytochrome P450). These data
1996). The co-administration of PB significantly increased the AUC and indicate that the FMO is product stereoselective and produces (+)
Cmax of FBZ in fed and unfed horses (McKellar et al., 2002). In fed ABZSO, whereas cytochrome specifically uses (−) ABZSO as substrate
horses PB co-administration increased the AUC of the active moieties (Moroni et al., 1995). Both systems act equally in rats and probably in
FBZSO and FBZ by 13.9 times and 13.2 times respectively (Table 6). In other monogastrics (man, dog, horse), while the FMO system is pre-
unfed horses PB increased the AUC of the parent FBZ by 11 times but dominant in ruminants. Differences of interspecies stereoselectivity for
decreased the AUC of the FBZSO moiety by 2.3 times (McKellar et al., the BZD sulphoxides exist and may be explained by different relative
2002). It has also been shown that PB significantly inhibited the me- enzyme contributions.
tabolism of OFZ and the parent molecule and its sulphide metabolite The chiral dispositions of OFZ and ABZ have been studied in vitro
(FBZ) achieved much greater plasma concentrations (AUC and Cmax) and in vivo in equine species (McKellar et al., 2002; Bruni et al., 2005;
following intravenous administration of OFZ with PB in ponies Gokbulut et al., 2009). It has been confirmed that FBZSO displayed
(McKellar et al., 2002). Bruni et al. (2005) also showed that co-ad- enantioselective pharmacokinetics since the (+) FBZSO enantiomer
ministration with PB significantly improved the pharmacokinetic pro- predominated following administration of the racemate, and the (+)
file of OFZ in horses. The significantly greater AUC (4.25 times) and FBZSO / (−) FBZSO ratio was 60:40 throughout most of the disposition
Cmax (2.6 times) values of OFZ observed in the OFZ + PB treated horses period (McKellar et al., 2002). In addition, after oral administration of
correlate with a PB interference of OFZ sulphonation. Piperonyl but- NTB in horses, the enantiospecific ratio (−/+) of ABZSO in plasma
oxide also increased the AUC value for FBZSO2 by almost 2 times and remained in favour of (−) for the first 16 h but changed in favour of the
this could be related to the higher concentration of OFZ observed in the (+) enantiomer after 20 h following administration (Gokbulut et al.,
OFZ + PB treated horses, resulting in a greater substrate concentration 2009). This difference in the ratio may affect the anthelmintic activity
available for sulphonation when the sulphonating inhibitory effect of of the enantiomers, since it has been shown that the (-) ABZSO is
PB elapsed (Bruni et al., 2005). In addition, these outcomes also cor- quickly metabolised into the inactive sulphone metabolite (Goudah,
related with in vitro studies undertaken in liver microsomes from horses 2003) in sheep. Moreover, Alvarez et al. (1999, 2000) have reported
which indicated that PB inhibited significantly the metabolism of OFZ, that the main enantiomer taken up by F. hepatica was (+) ABZSO fol-
FBZ and OXB (Gokbulut et al., 2002; McKellar et al., 2002). lowing ABZ administration in infected sheep. For sulphur-containing
These studies demonstrated the very great potential for synergy of BZDs, the metabolism of sulphide to sulphoxide is thought to be

36
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

principally catalysed by the FMO system (Galtier et al., 1986) whereas mL). It differs from morantel by absence of a methyl group on the
metabolism of sulphoxide to sulphone is thought to be catalysed by thiophene ring (Fig. 3). Pyrantel pamoate is available as a ready-to-use
hepatic cytochrome P450 (Souhailielamri et al., 1988). suspension and paste for oral use in horses. It is poorly absorbed from
It has been shown that the co-administration of PB dramatically the gastrointestinal tract since 50%–70% of an ingested dose is excreted
altered the enantioselective pharmacokinetics of FBZSO since the (-) in the faeces (Arundel, 1983), and blood levels do not exceed 1 μg/mL
FBZSO enantiomer predominated for the first 12 h following adminis- (Davis, 1973). Reduced systemic absorption of PYR potentially in-
tration after which the ratio changed in favour of (+) FBZSO (McKellar creases availability in the lumen of the intestine (Bjorn et al., 1996).
et al., 2002). The in vitro studies with equine liver microsomes support Pyrantel pamoate is available in different formulations in different
much of the in vivo work described above. Piperonyl butoxide sig- jurisdictions as a paste for oral administration in horses and it is
nificantly inhibited the sulphonation of FBZSO and the sulphoxidation available as a suspension for oral or nasogastric administrations and
and sulphonation of FBZ. It was also apparent that (-) FBZSO was me- mixing with feed. A pellet formulation of PYR tartrate is available for
tabolised more rapidly than (+) FBZSO in equine liver microsomes and administration in feed (DiPietro and Todd Jr, 1987). The solid form of
that PB altered the metabolism such that the ratio of (+) FBZSO / (-) PYR salts are stable but their suspensions in water are degradable, re-
FBZSO remaining after incubation was much closer to unity. Moreover, sulting in a reduction of their potency.
Bruni et al. (2005) indicated that following administration of OFZ Pyrantel has been shown to be extensively metabolised to more
alone, the AUC value of (-) FBZSO was 2-fold higher than that obtained polar metabolites in cattle, sheep, dogs and rats by three metabolic
for (+) FBZSO (AUC ratio FBZSO (-) / (+) 2.03). However, after pathways; oxidation of the thiophene ring, oxidation of the tetra-
treatment with OFZ + PB the ratio decreased by 32% (1.37). The pyrimidine ring and mercapturic acid conjugation (Faulkner et al.,
combination produced a significant enhancement of (+) FBZSO en- 1972). Furthermore the, N-methyl-1,3-propanediamine skeleton of the
antiomer concentrations, which could correlate with the selective in- tetrahydropyrimidin ring has been shown to be relatively resistant to
hibition by PB of the CYP450 system associated with the formation of metabolism, as evidenced by its formation after hydrolysis in both urine
the (+) antipode (Bruni et al., 2005). The eudismic (potency) ratio of and body tissues, while hydroxylation of the thiophene ring is the main
FBZSO is unknown, however the alterations in enantiomer generation route of metabolism of PYR (Faulkner et al., 1972). It is thought that the
together with the alteration in achiral metabolism of BZDs by PB could polar metabolites of PYR excreted by urine and bile are anthelminti-
have a major impact on the efficacy of BZD sulphides and sulphoxides cally inactive (Lanusse and Prichard, 1993).
in the horse.
3.2. Efficacy and spectra of activity
3. Imidazothiazoles and tetrahydropyrimidines
In horses, PYR is the most widely used anthelmintic among the
The anthelmintic properties of imidazothiazole derivatives were tetrahydropyrimidines. The anthelmintic activity of PYR against
discovered firstly in the early 1960s. The racemic compound tetra- common gastrointestinal helminths in equine species is shown in
misole was synthesized in 1964 and then used widely for the treatment Table 3. Generally, PYR has a great efficacy against adult nematodes
of helminth infections in animals (Amery and Bruynseels, 1992). The in the gastrointestinal lumen and shows less activity against migrating
greater anthelmintic activity of the levo-isomer of tetramisole was or encysted larvae and developing forms (Bogan and Armour, 1987).
discovered in 1966 and the single isomer developed as the product Pyrantel is highly effective (95%–97%) against adult cyathostomins,
called levamisole (LVM) which has a broad spectrum anthelmintic ac- P. equorum and S. vulgaris, and has moderate activity against S.
tivity against a wide variety of gastrointestinal nematodes in domestic edentatus (70%) and O. equi (65%) (Mirck, 1985). PYR is not effective
animals. Recently, LVM has been categorised as a “multi-faceted drug” against Gasterophilus spp., but it has activity against the Anoploce-
(Gupta, 2016), and has been used not only for the treatment of gas- phalan tapeworms if used at 13.2 mg/kg (Theodorides, 1985). It was
trointestinal nematodes and lungworms in different animal species reported that PYR pamoate paste formulation was highly effective
(Miller, 1980), but also as an immunomodulator and adjuvant to treat a (95–98%) against mature infections of Anoplocephala perfoliata in
variety of diseases including cancer, rheumatoid arthritis and different horses and ponies following oral administration at a dosage of
dermatological disorders including viral and bacterial infections and 13.2 mg/kg, BW (Owen and Slocombe, 2004). Continuous low-level
inflammatory skin diseases in human medicine (Biswajit et al., 2014). daily administration of PYR tartrate to horses was highly effective
Early studies indicated that LVM was not a suitable anthelmintic against common gastrointestinal parasitic infections of horses, in-
compound for horses since it had a low efficacy against important ne- cluding large strongyles (S. vulgaris, S. edentatus and Triodontophorus
matodes and had a poor therapeutic ratio resulting in some adverse spp.), adult cyathostomins (Cyathostomum spp., Cylicocyclus spp., and
drug reactions including profuse sweating, lacrimation and hyper- Cylicostephanus spp.), and adult and fourth-stage P. equorum (Valdez
excitability (DiPietro and Todd Jr, 1987). et al., 1995). Recently, daily administration of PYR tartrate in feed (at
Tetrahydropyrimidines are imidazothiazole derivatives (Fig. 3) and a dose of 2.64 mg/kg/day) significantly reduced numbers of adult
include pyrantel (PYR), morantel and oxantel used to control gastro- cyathostomins in the gut lumen and early third-stage larvae in the
intestinal parasites in domestic animals. Among these compounds, only cecal mucosa and, increased the proportions of fourth-stage larvae in
PYR has been licensed and used extensively in horses as an oral paste or the gut contents in horses (Reinemeyer et al., 2014). Daily PYR tar-
granules at a therapeutic dose of 6.6–13.2 mg/kg following single oral trate administration in ponies was highly effective against strongyles
administration for the treatment of gastrointestinal parasites. Recently, and significantly reduced the strongyle egg and pasture larval counts
the use of PYR in horses has increased due to the worldwide spread of during a grazing season (Slocombe and Lake, 2007). In addition, PYR
anthelminthic resistance against benzimidazole agents. It has been pamoate was highly effective against O. equi (Reinemeyer et al.,
shown that the anthelmintic activity of PYR salts is directly related and 2010b) and an endectocide-resistant isolate of P. equorum
proportional to the presence of its free base amount (Sheehan et al., (Reinemeyer et al., 2010a) at a dose of 13.2 mg/kg following the paste
2016). formulation administration in horses. However, some investigations
from different countries have reported the emergence of PYR re-
3.1. Chemistry and metabolism sistance in horses (Brazik et al., 2006; Slocombe and de Gannes,
2006). Recently, it has been observed that the efficacy of PYR against
Pyrantel (1,4,5,6-tetrahydro-1-methyl-2-(2-(2-thienyl)ethenyl) pyr- Strongylus spp. reduced in five groups of yearlings (FECR range:
imidine) is available as a pamoate (syn. embonate) salt, which is almost 0–73%), but PYR was found to be highly efficacious in mares (FECR
insoluble in water, and as a tartrate, which is soluble in water (180 mg/ range: 98–99.4%) at a dose of 19 mg/kg following the oral

37
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Fig. 3. Chemical structures of imidazothiazole anthelmintic compounds (LVM and tetrahydropyrimidines).

administration in horses (Relf et al., 2014). However, PYR pamoate is 3.4. Safety and toxicity
not labelled for efficacy against luminal, fourth stage cyathostomin
larvae. The oral LD50 values are for PYR tartrate 175 mg/kg in mice and
It has been demonstrated that PYR pamoate had a good anthel- 170 mg/kg in rats (Bossche, 1985) and are much lower compared to
mintic efficacy (> 90%) against strongylidea in donkeys (Napoli et al., PYR pamoate which has an oral LD50 above 2000 mg/kg in rats, mice
2013). Recently, a therapeutic equivalence was demonstrated for paste and dogs (EMEA, 1998). First signs of toxicities associated with PYR
and granule formulations of PYR based on FECR test in donkeys tartrate were observed at a dose rate of 55 mg/kg and consisted of
(Gokbulut et al., 2014). PYR pamoate paste and granule formulations sweating, dyspnea and even death (Cornwell and Jones, 1968). In
were highly efficacious (> 95% efficacy) against intestinal strongylidae horses, no adverse effects occurred at doses up to 20 times greater than
following oral administration at a dosage of 6.94 mg/kg of BW in the therapeutic dose of PYR pamoate (embonate) (Slocombe and Smart,
donkeys. Moreover, the results obtained in both treated groups were 1975). No adverse effects were observed and PYR was well tolerated in
consistent with the currently approved label claim for PYR oral for- horses following a single oral administration of a paste and suspension
mulations against intestinal strongyle parasites of horses. Although, the formulation of PYR pamoate at the recommended dose or repeated
granule formulation displayed higher plasma concentrations and dosing at 19.1 mg/kg BW with a 2- to 4-week dosing interval (EMEA,
availability compared to the paste formulation, the difference in the 1998). The maximum tolerated oral dose of PYR tartrate in sheep was
formulations of PYR did not seem to alter the anthelmintic efficacy in 173 mg/kg (Austin et al., 1966) although the toxicity of PYR tartrate in
donkeys. water in adult sheep varies according to the method of administration
(Cornwell, 1966). It has been reported that no acute toxicity was ob-
served in nine ponies which received oral PYR tartrate at doses of
3.3. Mode of action 50 mg/kg and 75 mg/kg BW, but one of three ponies died at a dose of
100 mg/kg BW (Cornwell and Jones, 1968).
LVM and tetrahydropyrimidines including PYR share similar mode The reproductive performance of stallions and pregnant mares was
of action whereby they act selectively as agonists at synaptic and ex- not affected by PYR (Bentley et al., 1978). Teratogenicity studies con-
trasynaptic nicotinic acetylcholine (ACh) receptors on muscle cells of ducted in rabbits and rats indicated that there was no evidence of ter-
susceptible nematodes. They produce contractions and spastic paralysis atogenicity, fetotoxicity or maternal toxicity of PYR pamoate following
in the parasites, which serve to eliminate them from the host (Martin, daily oral administration at 9 mg/kg and 90 mg/kg BW (EMEA, 1998).
1997; Martin and Robertson, 2007). Simultaneous application of acet-
ylcholine and PYR showed that both agonists acted on the same nico-
tinic receptors (Harrow and Gration, 1985) and that PYR is up to 100 3.5. Pharmacokinetics
times more potent than acetylcholine in inducing muscular contraction
(Aubry et al., 1970). It has been shown that PYR increases membrane Although, PYR has been used as an anthelmintic drug for almost a
conductance and depolarizes the membrane by opening non-selective half century, there is a paucity of data available in the literature on the
ion channels that are permeable to both Na+ and K+ (Harrow and pharmacokinetics of different formulations in domestic animals in-
Gration, 1985). Moreover, the biophysical and some pharmacological cluding equine species. Different salts of PYR have different pharma-
properties of ACh receptors in nematode muscles are similar to the cokinetic properties and consequently different toxicities to the host.
neural receptor channels in mammalians, but, there are significant The pamoate (embonate) salt is almost insoluble in water and poorly
pharmacological differences between nematodes and mammals, LVM absorbed from the gastrointestinal tract and most passes unchanged in
and PYR display selective therapeutic effects on the nematode muscle the faeces (Arundel, 1983). Reduced systemic absorption of the
receptors, minimizing their toxic effects on host muscle (Martin and pamoate (embonate) form potentially increases availability in the
Robertson, 2007). lumen of the intestine (Bjorn et al., 1996). The tartrate salt of PYR is
soluble in water and absorbed rapidly and extensively from the

38
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Table 5 anthelmintic treatment in horses was unknown.


Pharmacokinetic parameters of pyrantel (PYR) in equine species following oral These studies demonstrate limited absorption of PYR following oral
administration. administration in horses and donkeys and it seems likely that a sub-
Species Horse Donkey stantial component of its dynamic effect will be associated with PYR
retained in the gastrointestinal tract where the adult stages of most
Formula paste paste granule parasitic nematodes reside. Any substantial effect of PYR on faecal in-
vertebrates is not known, however, it is apparent that faeces from
Dose (mg/kg) 13.3 6.94 6.94
Cmax (μg/mL) 0.09 ± 0.03 0.09 ± 0.02 0.21 ± 0.07 treated horses and donkeys will have high concentrations of PYR for at
Tmax (h) 7.50 ± 1.41 14.86 ± 5.52 14.00 ± 9.45 least 120 h for donkeys (Gokbulut et al., 2014) and 48 h for horses
AUClast (μg h/mL) 1.06 ± 0.24 2.65 ± 0.81 5.60 ± 0.59 (Gokbulut et al., 2001a).
T1/2λz (h) 13.43 ± 1.38 12.39 ± 5.35 14.86 ± 5.59
MRT (h) 11.99 ± 1.30 24.80 ± 5.54 25.44 ± 10.68
References (Gokbulut et al., 2001a) (Gokbulut et al., 2014)
4. Macrocyclic lactones (avermectins and milbemycins) in equine
species
Cmax: peak plasma concentration; Tmax: time to reach peak plasma concentra-
tion; AUClast: area under the (zero moment) curve from time 0 to the last de- Macrocyclic lactones (MLs, avermectins and milbemycins or en-
tectable concentration, MRT: mean residence time; T1/2λz: terminal half-life. dectosides) comprise a series of natural and semisynthetic molecules,
including ivermectin (IVM), abamectin (ABM), moxidectin (MXD),
intestine of monogastric animals (Faulkner et al., 1972). doramectin (DRM), eprinomectin (EPM), selamectin (SLM) and milbe-
The pharmacokinetic parameters of PYR in horses and donkeys are mycin oxime, which have some similar physicochemical properties, and
shown in Table 5. The pharmaceutical formulation of anthelmintic antiparasitic activity at very low dosage rates based on a common mode
drugs may affect their oral bioavailability, which depends on the rate of action (Lanusse and Prichard, 1993). They are highly effective
and extent of absorption of the drug from the gastrointestinal tract into against nematode and ectoparasitic arthropods in human and different
the bloodstream. In donkeys, pharmacokinetic results have indicated animal species. Therefore, MLs have been renamed as “endectocides”
that the paste formulation differed significantly from the granule for- since they are unique drugs effective against both endo- and ectopar-
mulation of PYR at the same dose rate after oral administration asites. However, these compounds are not effective against trematodes
(Gokbulut et al., 2014). The plasma concentrations of PYR were much or cestodes. Currently, IVM, MXD, DRM and ABM are licensed by some
lower following administration of the paste formulation than those authorities for use in horses for the treatment of endo- and ecto-para-
observed following granule administration. After oral administration, sites. They are usually administered orally as a paste or gel formulation,
Cmax (0.09 μg/mL) was significantly lower (P < 0.01) and AUC (2.65 although a liquid oral formulation of IVM for horses is also available in
μg.h/kg) smaller (P < 0.01) for PYR paste than PYR granule (Cmax: the market.
0.21 μg/mL, AUC: 5.60 μg h/kg) given at the same dose rates (6.94 mg/ The milbemycins were discovered in 1973 as acaricidal and in-
kg BW). It is likely that the poorer dissolution of the paste formulation secticidal compounds for crop protection (Takiguchi et al., 1980).
reduces its absorption compared with the granule formulation in don- However, the full potential of these chemical groups was not realized
keys (Gokbulut et al., 2014). until the acaricidal, insecticidal and nematocidal activity of the aver-
The pharmacokinetic properties of PYR in donkeys differed sub- mectins was documented in 1975 (Egerton et al., 1979). This discovery
stantially from those previously reported by Gokbulut et al. (2001a) in started a new era in the treatment of endo- and ecto-parasitic infections
horses using the same formulation and administration route. In horses, in animal and human medicine. The excellent microfilaricidal activity
PYR paste was administered at a dose of 13.3 mg/kg BW, whereas in of IVM against Dirofilaria immitis infections was discovered in dogs
donkeys, it was administered at a dose of 6.94 mg/kg BW. The lower during anthelmintic development studies (Blair and Campbell, 1979)
concentrations and shorter MRT of PYR in the plasma of horses reflect and Onchocerca cervicalis infections in horses (Herd and Donham,
lower bioavailability and shorter persistence compared with donkeys. 1983). Ivermectin was firstly introduced commercially as an animal
After correction for dose, assuming proportionality, Cmax (0.045 μg/ endectocide in 1981 in France (Shoop et al., 1995) and within 5 years,
mL), AUC (0.53 μg h/mL) and MRT (11.99 h) values in horses were it became the most popular anthelmintic worldwide. It was shown that
much lower than those observed in donkeys (Cmax: 0.09 μg/mL, AUC: avermectins had good activity against microfilaria of Onchocerca vol-
2.65 μg h/mL and MRT: 24.80 h). The gastrointestinal passage rate of vulus, and were well tolerated in man (Aziz et al., 1982). Abamectin was
digesta is affected by alteration in the quality and quantity of the feed introduced commercially as a veterinary endectoside in 1985 in Aus-
consumed and this could confer variable absorption time and therefore tralia (Tahir et al., 1986). Moxidectin was developed in the USA and
bioavailability of drugs administered orally. It has been shown that approved for marketing in Argentina in 1989 (Campbell, 2012). It has
variations in the quality (McKellar et al., 1993a; Gokbulut et al., 2007) been commercialized for use in cattle as a subcutaneous injectable so-
and quantity (Lifschitz et al., 1997; McKellar et al., 2002; Gokbulut lution (1%) and pour-on preparation (0.5%), in sheep as an oral drench
et al., 2010a) of diet could affect the bioavailability of anthelmintics in (0.1%) and in horses as an oral gel (2%) (at 0.4 mg/kg therapeutic dose
different animal species. Differences between studies referred to in level). An injectable formulation of MXD for equine species was de-
Table 5 may be associated with diet type in donkeys compared with veloped and its anthelmintic activity was evaluated but not marketed
horses. In the studies by Gokbulut et al. (2001a, 2014), donkeys were (Lyons et al., 1992). Milbemycin oxime has been licensed in the USA for
kept indoors and fed hay-based diet whereas, horses were yarded for use in dogs against adult Ancylostoma caninum as a therapeutic agent
the period during and immediately (4 h) after drug administration and and against D. immitis as a prophylactic drug since 1990 (Bowman et al.,
were then returned to a grass paddock. The grass-based diet probably 1990). Doramectin was developed in the UK and first marketed in 1993
decreased gut transit time for food, causing a decrease in the bioa- in Brazil and South Africa (Vercruysse, 1993). Doramectin formulation
vailability of PYR in horses compared with those in donkeys. Moreover, was developed with oil vehicles to enhance the persistence of efficacy in
anatomic features influence the passage of digesta, and the bioavail- treated animals (Conder and Baker, 2002). Like other avermectins, it
ability and pharmacokinetics of anthelmintics may be affected by dif- has therapeutic activity against gastrointestinal nematodes and ar-
ferent gut transit time in animal species (McKellar and Scott, 1990). thropod parasites (Conder and Campbell, 1995). Eprinomectin, the
Parasitism could have had an effect since donkeys were naturally in- most recently introduced avermectin, is a semi-synthetic avermectin. It
fected with intestinal parasites and such infections may have modest has been demonstrated that EPM has 2–3 times higher anthelmintic
effects on the absorption of anthelmintics (McKellar et al., 1991), un- efficacy compared to that of IVM in titration trials (Shoop et al., 1996b).
fortunately the parasitological status of the horses or the history of Pour-on formulations for topical administration and injectable

39
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

formulations for subcutaneous injection of EPM were licensed and embraced by the epi-acetyl amino substitution in EPM which retains/
made available in the market for use in cattle and deer only. enhances the potency and spectrum of activity of this avermectin yet
Macrocyclic lactones are widely used in horses for the treatment of enhancing its absorption from skin and conferring a very low milk ex-
gastrointestinal nematodes and ectoparasitic infections. The physico- cretion rate permitting its use in dairy cows (Shoop et al., 1996a).
chemical properties including high lipophilicity and stability allow a The milbemycins including MXD, are not only different from the
wide flexibility on their pharmaceutic formulations. There are a variety avermectins in lacking sugar groups in the 13-position, they also differ
of the formulations for the MLs, including formulations which combine from the avermectin aglycones by being protonated at this position in
the different member MLs with other anthelmintically active in- contrast to the avermectin aglycones which are hydroxylated at this
gredients to broaden the spectrum of activity of the combination. position. Milbemycins have an ethyl or a methyl at the 25-position and
Ivermectin was first marketed for horses as a micellar formulation differ from one to another by their substituents in the 5- and 25-posi-
containing 20 mg of IVM per millilitre of sterile aqueous solution (2%) tion. Moxidectin has a substituted olefinic side chain at the 25-position
for intramuscular injection. After parenteral administration, rare ad- and methoxime moiety at the 23-position which are both characteristics
verse reactions including Clostridium spp. infections and anaphylaxis specific for this drug and not found in other milbemycins or avermec-
were observed, and these undesirable effects were responsible for the tins.
withdrawal of the parenteral preparation of IVM for use in the horse in Macrocyclic Lactones are highly lipophilic substances and dissolve
1984 (Campbell et al., 1989). An oral paste formulation of IVM (1.87%) in most organic solvents, such as chloroform, acetone, toluene and
in titanium dioxide and propylene glycol is now available in graduated methylene chloride. Their solubility in water is very low
delivery syringes of which each part is designed to administer sufficient (0.006–0.009 mg/l) (Fisher and Mrozik, 1989). Moxidectin has a lower
IVM (at 200 μg/kg) for 100 kg of body weight. A liquid formulation of molecular weight (639.8 kDa) and higher water solubility (4.3 mg/l)
IVM is also marketed for administration by nasogastric intubation in than ABM and IVM (mol. wt. ∼870 kDa) (Lanusse and Prichard, 1993).
some countries. Moxidectin oral gel formulation (2%) is commercially In contrast with this, it was reported that MXD was 100 times more
available for antiparasitic treatment of horses. Another MXD (2%) oral lipophilic than IVM (Hayes, 1994). The water solubility and lipophili-
gel formulation combined with praziquantel (12.5%) is available to city of MXD are unusual for a drug since increased water solubility is
provide activity against cestodes (Cobb and Boeckh, 2009). Abamectin usually directly associated with decreased lipophilicity. Macrocyclic
(1.87%) paste in combination with praziquantel (14.03%) and DRM Lactones do not contain strongly acidic or basic functional groups and
(1.75%) oral gel formulations have been developed and are available in pH partitioning is not a major feature of their kinetic characteristics.
some markets for treatment of endo- and ecto-parasitic infections in These physicochemical differences may affect pharmacokinetic beha-
horses. viour, development of drug resistance and parasite uptake mechanism
(Lanusse and Prichard, 1993). All members of the MLs are thought to be
4.1. Chemistry and metabolism highly protein bound in plasma however their lipophilicity and asso-
ciation with circulating lipoproteins may be important determinants in
The chemical structures of MLs are closely related and have a 16- their exchange between tissue and bloodstream (Lespine et al., 2009).
membered ML ring, with a disaccharide substitution at C-13 (Fig. 4). Metabolic enzymes in nematodes (Alvinerie et al., 2001) and in
The major structural difference between the two groups is a bisolean- mammals (Chiu et al., 1987) can metabolize MLs. Cytochrome P450
drosyloxy substituent found at the C-13 position of the macrolide ring 3 A4 is the predominant isoform, which is primarily responsible for the
of the avermectins whereas that position is unsubstituted in milbemy- metabolism of avermectins, by liver microsomes in rats (Zeng et al.,
cins. The sugar moiety at C-13 together with the hydroxy group at C-5 1997) and humans (Zeng et al., 1998). However, metabolism is not a
may determine anthelmintic and insecticidal activity (Jackson, 1989). common pathway for the elimination of these compounds which are
Avermectins are derived from the mycelia of Streptomyces avermitilis primarily eliminated in faeces, with less than 2% of the total dose being
and milbemycins are produced from fermentation of Streptomyces hy- excreted in urine (Chiu et al., 1990). Liver and fat are two main tissue
groscopicus and Streptomyces cyanegriseus. Fermentation of S. avermitilis sites of IVM biotransformation. The unchanged parent compound is the
produces eight different components and they are named avermectin main liver residue in cattle, sheep and rats treated with IVM and the
A1a, A1b, A2a, A2b, B1a, B1b, B2a and B2b. The A-components have a major liver metabolite is a 24-hydroxy-methyl-H2-B1a and H2-B1b (Chiu
methoxy group at the 5-position, whereas the B-components have a et al., 1987). The monosaccharide and aglycone forms of these meta-
hydroxy group; the 1-components have a double bond between the 22- bolites were also identified in the liver. The major excretion routes of
and 23-position, whereas the 2-components have a single bond with a avermectins are bile and faeces in all animal species studied (Chiu and
hydroxy group at the 23-position; and a-components have a secondary Lu, 1989). Ivermectin has been shown to be excreted in high con-
butyl side chain at the 25-position, whereas the B-components have an centrations in the bile of ruminants (Bogan and McKellar, 1988).
isopropyl substituent at the 25-position. Doramectin is structurally si- Considerable amounts of IVM are also excreted via the mammary gland
milar to avermectin A1a but with cyclohexyl and methylpropyl sub- in animals during the milking period. N-deacetylation of AAB1 has been
stituents at Carbon 25. More radical alteration at C-25 has been shown to be the primary metabolic route of EPM in rats (Zeng and
achieved by mutational biosynthesis (Dutton et al., 1991) and the 25 – Andrew, 1999). C29-30- and C14-mono-hydroxy-methyl derivatives are
cyclohexyl derivative (DRM) has a longer elimination half-life and the main metabolic products of MXD (Zulalian et al., 1994) produced
slower clearance than unsubstituted dihydro avermectin B1a (the major by the cytochrome enzymes, P450 3 A and P450 2B (Dupuy et al.,
component of IVM) (Goudie et al., 1993). Abamectin (avermectin B1) 2001b). It has been reported that the absorption, distribution in tissues,
contains at least 80% of avermectin B1a and not more than 20% aver- metabolism, and elimination of MXD in the horse were similar to those
mectin B1b whereas, IVM, contains at least 80% of 22–23 dihy- reported for cattle, sheep, and rat (Afzal et al., 1997). Although minor
droavermectin B1a and less than 20% of the 22–23 dihydroavermectin metabolites were observed, parent MXD molecule was the principal
B1b. Eprinomectin (4″-epi-acetylamino-4″-deoxy-avermectin B1) is a excretory component and MXD was eliminated from the body mainly
semi-synthetic avermectin and is derived from avermectin B1 with via biliary excretion (Afzal et al., 1997) and probably intestinal excre-
modified terminal oleandrose moiety called 4″-epiacetylamino-4″- tion and subsequently voided with faeces.
deoxy-avermectin B1. Chemically, EPM is a mixture of two homologs,
EPM B1b and EPM B1a in a 9:1 ratio. The difference between the two 4.2. Efficacy and spectra of activity
groups is a single methylene group at C25-position (Fig. 4). Substitution
of the C-4″ hydroxy group with acyl, amino or thio groups improve the Ivermectin, MXD, DRM and ABM which are licensed MLs for use in
solubility and disposition of avermectins and this feature has been horses, are highly effective against bots, lungworms, thread worms,

40
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Fig. 4. Chemical structures of MLs (endectocides).

cutaneous onchocerciasis and ectoparasites. However, the major an- effective against adult Strongylus spp., arterial stages of S. vulgaris and
thelmintic activities of MLs against common gastrointestinal nematodes migratory S. edentatus, (Klei et al., 1984; Slocombe and Mccraw, 1984),
in equine species are shown in Table 3. Ivermectin, 0.2 mg/kg body S. westeri (Ryan and Best, 1985) and D. arnfieldi (Britt and Preston,
weight given orally, is highly active against adult and third-stage pul- 1985). In ponies, IVM is fully effective against oral and gastric stages of
monary larvae (L3) of P. equorum (French et al., 1988) but has variable Gasterophilus spp. after oral administration at 0.2 mg/kg body weight
activity against fourth-stage (L4) intestinal larvae (Campbell et al., (Bello, 1989). The previous studies indicated that IVM and MXD were
1989) in horses. It is more than 90% effective against adult cyathos- highly effective against adult cyathostomins but less effective against
tomins but has very little or no activity against hypobiotic early third- immatures (L4) in the intestinal mucosa (Lyons et al., 2010).
stage or mucosal late third-or fourth stage larvae (Eysker et al., 1992). It has been shown that MXD has excellent activity against equine
An efficacy study of IVM for ascarids indicated that the oral formulation nematodes (Lyons et al., 1992; DiPietro et al., 1997; Dorchies et al.,
of IVM is 90–100% effective against pulmonary and small intestinal 1998b). It has higher activity than IVM against tissue strongyle larvae
stages in foals and weanlings (Austin et al., 1991). Ivermectin is highly when given at an equal oral dose (DiPietro et al., 1997). In MXD treated

41
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

horses it was reported that strongyle mean egg per gram counts (EPG) lumbricoides, avermectins reduced inhibitory neuromuscular transmis-
remained low for longer than in IVM treated horses (Boersema et al., sion by a gamma-aminobutyric acid (GABA)-mediated mechanism at a
1998). The greater lipid solubility of MXD as compared to IVM may concentration of 5 × 10−6 M (Kass et al., 1984). At this concentration,
explain its increased activity against these parasites (Hayes, 1994; avermectins were thought to induce presynaptic GABA release or
Zulalian et al., 1994). The activity of MXD, given orally at 0.4 mg/kg, function as GABA agonists. At lower concentrations (2 × 10-12 M),
was 100% effective against T. axei, Triodontophorus spp., O. equi L5 and avermectins open GABA-independent chloride channels in A. suum
cyathostomins (adult and L5); 99% against O. equi L4 and 92% against S. muscle membranes and surprisingly, avermectins were shown to be
edentatus L5 and G. intestinalis (Dorchies et al., 1998b). One hundred per antagonistic to GABA-gated chloride ion channels at concentrations >
cent reduction of strongyle Faecal Egg Counts (FECs) was reported after 10−8 M (Martin and Pennington, 1989).
MXD treatment (Jacobs et al., 1995; Dorchies et al., 1998a). It was Although early reports suggested that the chloride channels were
shown that IVM and MXD were equally effective in reducing P. equorum associated with gamma aminobutyric acid (GABA) receptors (Turner
EPG (egg per gram counts) (DiPietro et al., 1997). After oral adminis- and Schaeffer, 1989), more recent evidence indicates that there is no
tration, MXD and IVM were fully effective in the control of Onchocerca GABA-gated Cl− channel association (Geary et al., 1992; Martin, 1997).
cervicalis microfilariae in horses (Mancebo et al., 1997). Moreover, Expression cloning experiments using Xenopus oocytes have suggested
MXD was highly effective against S. vulgaris (faecal egg count reduction an action of avermectins on avermectin-sensitive glutamate-gated Cl−
range, 99.9–100%) for 2 weeks after treatment (Tzelos et al., 2017). (GluCl) channels (Cully et al., 1994) and avermectins and milbemycin
Ivermectin and MXD provided acceptable efficacy (Lester et al., 2013; mediate their nematocidal effects on Caenorhabditis elegans via inter-
Relf et al., 2014; Stratford et al., 2014); however, strongyle EPG counts action with a common receptor molecule at glutamate-gated channels
results suggest that both drugs are less effective compared to the find- (Arena et al., 1995). Molecular studies showed that the Glu-Cl β-subunit
ings of previous studies done when these compounds were first mar- of the glutamate channel was expressed in the pharyngeal muscle of C.
keted (Lyons et al., 2011; Relf et al., 2014). Doramectin displayed a elegans (Laughton et al., 1995) and it was demonstrated that aver-
high efficacy of ≥96% in terms of reduction of faecal egg counts in mectin-sensitive glutamate-gated chloride channels were present on the
horses with strongylidae infections and egg reappearance period was 10 pharyngeal muscle of A. suum by using a microelectrode current clamp
weeks after intramuscular administration at a dose of 0.2 mg/kg (Cirak technique (Martin, 1996). Thus, while at high concentrations, aver-
et al., 2007). Doramectin was highly efficacious (99.6%) against mectins open the GABA-gated ion channel directly, at lower con-
strongyles (Strongylidae and Cyathostomins) and strongyle eggs re- centrations, they potentiate the effect of glutamate (Martin, 1997).
appeared 2 months after subcutaneous administration at a dose of Ivermectin has been shown to have excellent binding affinity for a
0.2 mg/kg BW in horses (Prokulewicz et al., 2014). Abamectin was P-glycoprotein (P-gp) which acts as a transmembrane efflux pump
highly effective against gastrointestinal parasites including Strongylus (Didier and Loor, 1996; Pouliot et al., 1997) and that the high tolerance
spp. Oxyuris equi and Parascaris equorum following subcutaneous ad- of mammals to IVM is abolished if the blood-brain barrier P-gp is de-
ministration at a dose of 0.2 mg/kg BW in horses (Mahfooz et al., 2008). leted (Schinkel et al., 1994). Thus, MLs probably have more than one
In donkeys, IVM was highly effective (96%) against strongyloidiasis mechanism of action against nematodes (Martin, 1997).
in terms of reduction of EPG following subcutaneous administration at In ectoparasites (Shistocerca gregaria) it has been reported that IVM
a dose of 0.2 mg/kg BW (Binev et al., 2005). In addition, it has been interacted with both a GABA receptor-Cl− ion channel (10-10 M) in a
reported that IVM was 100% effective against T. axei, P. equorum, O. reversible dose-dependent fashion and with GABA-insensitive neurones,
equi, Strongylus spp. and cyathostomins, however, it had moderate ef- where an irreversible increase in Cl- conductance was noted at 10-10 M
ficacy (62.71%) against larvae found in the cranial mesenteric artery (Duce and Scott, 1985). In contrast, in the ectoparasite, Periplaneta
aneurisms after oral treatment in donkeys (Imam et al., 2010). Iver- americans, avermectin B1a had no effect on GABA-independent neuronal
mectin was also effective in the treatment of psoroptic mange in don- conductance (Mellin et al., 1983), although avermectin-sensitive GABA
keys following subcutaneous administration at a dose of 0.2 mg/kg BW binding sites have been identified in insect nerve cord (Lummis and
(Abusamra et al., 1987) and had an efficacy of 96% in terms of re- Sattelle, 1985).
duction of faecal egg counts in donkeys with strongylidae infections The selective therapeutic effects of MLs may be explained by an
after subcutaneous administration at a dose of 0.2 mg/kg (Binev et al., action on a Glu Cl− ion-channel, present in nematodes and ectopar-
2005). The anthelmintic activity of pour-on EPM against the lungworm asites, but not in mammals (Martin, 1997). In addition, the pharma-
D. arnfieldi was 100% at a dose of 0.5 mg/kg BW in donkeys (Veneziano cokinetic properties of MLs may determine their selective activity in
et al., 2011). nematodes (reviewed by McKellar and Benchaoui, 1996). Ivermectin
It has been reported that MXD and IVM were 100% effective against distributes poorly into the brain of mammalian species (Chiu and Lu,
chorioptic and psoroptic mite infestations following oral doses of 1989) and the affinity of IVM for specific binding sites in C. elegans is
0.4 mg/kg and 0.2 mg/kg BW, respectively and complete clinical re- much higher (100-fold) than in rat brain (Turner and Schaeffer, 1989).
covery were observed within 2 weeks in both drug treatment groups The effects of avermectins are variable on different nematode spe-
(Osman et al., 2006). Ivermectin and DRM were highly effective against cies. While a slow rigid paralysis was observed in the free-living ne-
chorioptic mange in horses and donkeys following two oral doses of matode, C. elegans, a flaccid paralysis was reported following injection
either drug given 14 days apart at a dose of 0.2 mg/kg BW (Kotb and into A. suum (Turner and Schaeffer, 1989). It was also shown that
Abdel-Rady, 2013). Horses with psoroptic manage were effectively avermectins reduced the fecundity (amount of eggs in the uterus) of
cured with four pour-on EPM administrations with a one-week-intervals Cooperia curticei by 99% (McKellar et al., 1988) and the reproductive
at a dose of 0.5 mg/kg BW (Ural et al., 2008). In addition, five donkeys potential of Dermacentor albipictus and Amblyomma americanum
with psoroptic mange were successfully treated with subcutaneous IVM by > 96% (Wilkins et al., 1981).
injection at a dose of 0.2 mg/kg BW (Abusamra et al., 1987). In contrast
to these reports, horses with chorioptic mange were not cured with oral 4.4. Safety and toxicity
IVM paste at a dose rate of 0.1 mg/kg daily for 10 days or two doses of
0.2 mg/kg given two weeks apart (Littlewood et al., 1995). Avermectins and milbemycins have a substantial margin of safety
and high therapeutic indices when compared with other anthelmintic
4.3. Mode of action xenobiotics. Nevertheless, since GABA is a neurotransmitter limited to
the central nervous system in mammals, neurotoxicity may occur when
Although the mechanism of action of MLs anthelmintics is not fully MLs are administered at high doses. Neurotoxicity characterised by
understood, it is likely that it is common to the group. In Ascaris ataxia, tremors and coma was identified (Lankas and Gordon, 1989).

42
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Table 6
Pharmacokinetic parameters of endectocides in equine species.
Pharmacokinetic parameters References

Species MLs Route Dose Cmax Tmax AUC T1/2 MRT Vdss Cl
(μg/kg) (ng/mL) (h) (ng.h/mL) (h) (h) L/kg (L/h/kg)

Horse IVM IV 0.2 – – 8226.5 138.72 147.12 3.65 15.12 Gokbulut et al. (2010b)
SC 0.2 60.7 80 13209.6 88.2 114.7 – – Marriner et al. (1987)
IM 0.2 31.4 84 7245.6 108.5 206.4 – – Perez et al. (2003)
IM 0.2 25.9 67.2 8328 219.84 194.4 – – Saumell et al. (2017)
T 0.5 4.3 103.9 1438.3 172.3 283.2 – – Gokbulut et al. (2010b)
PO 0.2 46.3 7.0 2646 – – – – Scott (1997)
PO 0.2 25.8 5.5 1941.8 88.1 117.6 – – Gokbulut et al. (2016b)
PO 0.2 82.3 3.1 4821.6 66.3 – – – Marriner et al. (1987)
PO 0.2 21.4 7.9 1106.4 51.6 55.2 – – Gokbulut et al. (2001b)
PO 0.2 44.0 2.2 3184 102 114.7 – – Perez et al. (1999)
PO 0.2 51.3 3.6 3290.4 69.4 100.8 – – Perez et al. (2003)
PO 0.2 61.3 4.1 3959 156.7 176.2 – – Gokbulut et al. (2010b)
PO (Paste) 0.2 30.1 7.92 2736 131.52 122.4 – – Saumell et al. (2017)
PO (Sol.) 0.2 33.7 12 3312 151.68 124.8 – –
DRM IM 0.2 33.3 69.6 9446.4 223.2 319.2 – – Perez et al. (2010)
PO 0.2 51.6 4.8 4286.4 124.3 185.3 – –
PO 0.2 21.3 8.0 1279.2 93.8 72.0 – – Gokbulut et al. (2001b)
MXD IV 0.4 – – 11,400.0 81.0 92.3 4.14 36.0 Afzal et al. (1997)
PO 0.4 102.0 8.0 4550 82 – – –
PO 0.2 30.1 7.9 2227.0 534.5 420.0 – – Gokbulut et al. (2001b)
PO 0.4 70.4 8.9 8726.0 554.6 442.1 – – Perez et al. (1999)
ABM PO 0.2 34.82 36.0 3137.0 134.4 92.4 – – Echeverria et al. (2001)
EPM T 0.5 17.7 43.2 424.6 32.9 70.3 – – Gokbulut et al. (2016b)
Donkey IVM PO 0.2 23.6 24 2863.2 177.6 156.0 – – Gokbulut et al. (2005)
PO 0.3 43.2 8.0 1811 – – – – Scott (1997)
DRM PO 0.2 33.9 24.0 5493.6 266.4 218.4 – – Gokbulut et al. (2005)
EPM T 0.5 14.2 81.1 3106.4 151.9 214.8 – – Gokbulut et al. (2013)
T 0.5 6.1 113.0 1761.4 113.44 215.5 – – Gokbulut et al. (2011)

Cmax: peak plasma concentration; Tmax: time to reach peak plasma concentration; AUClast: area under the (zero moment) curve from time 0 to the last detectable
concentration, MRT: mean residence time; T1/2λz: terminal half-life, AUMC: area under the first moment curve, Vdss: Volume of distribution at steady state, Cl:
Clearance.

Avermectins cause the release of endogenous GABA from cerebral of IVM for 50% of a population (LD50) are 25, 50 and 80 mg/kg in
cortex synaptosomes of the rat (Pong et al., 1980) and bind to two mouse, rat and dog (beagle), respectively (Fisher and Mrozik, 1992).
different sites in the GABA-gated chloride channel, activating the Toxicity of MLs is rare in equine species. A few sporadic IVM and
channel on binding to the high-affinity site and blocking it on further MXD toxicosis in equine species have been reported in the literature
binding to the low-affinity site in rat cerebellar granule neurones and most of them are related to substantial over-dose administration. It
(Huang and Casida, 1997). The specific avermectin-binding sites may has been reported that a four-year-old thoroughbred gelding collapsed
be blocked by GABA and GABA agonists (Drexler and Sieghart, 1984). following the intravenous administration of 10 ml of a cattle formula-
Deficiency of P-gp, a transmembrane glycoprotein associated with tion of IVM (1.0% w/v) (Burrough, 1986). The CNS symptoms such as
multidrug resistance (Goldstein et al., 1989), was shown in the in- tremors, especially of the neck and forelimb, and periodic nystagmus
testinal epithelium and brain capillary endothelium in CF-1 mice which observed in this horse which became comatose were similar to those
were 100-fold more sensitive to avermectin neurotoxicity than other reported in collies affected by IVM. In addition, IVM toxicosis was re-
species and normal mice (Lankas et al., 1997). ported in three adult horses following administration of single dose
The toxicity of MLs has been correlated with their disposition in paste (1.87%) formulation (Swor et al., 2009). Clinically, bilateral
susceptible species and breeds of animals (McKellar, 1997a,b). The mydriasis, decreased pupillary light reflexes, and absent menace re-
distribution of IVM is restricted by the blood-brain barrier from the flexes were observed in these animals. A high concentration of IVM was
central nervous system of mammalian species (Chiu and Lu, 1989). The detected in the brain tissue at post-mortem examination of one of the
central nervous system of neonatal rats is more sensitive to toxicity than affected horses which were euthanized. An over-dose of IVM was ac-
that of adult rats due to the immature blood-brain barrier (Lankas and cidentally administered orally to a 3-month-old zebra (Hautekeete
Gordon, 1989). It has been shown that IVM was pumped as an efflux by et al., 1998) and in a separate instance to a 9-week-old miniature mule
P-gp in the blood-brain barrier of mice and this may prevent the ac- foal; similar clinical CNS symptoms occurred as those reported in
cumulation of IVM in normally tolerant animals (Schinkel et al., 1994, horses, including temporary blindness (Plummer et al., 2006). Mox-
1996). It is thought that the toxicity of MLs occurs due to inadequate idectin intoxications with CNS symptoms including coma, dyspnoea,
and deficient P-gp mediated drug transport in the blood–brain barrier. depression, ataxia, tremors, seizures, or weakness have been also re-
(Lespine et al., 2006, 2007). P-glycoproteins act as transport proteins ported in equine species following oral over-dose administrations
able to carry some drugs, including MLs, across cell membranes. Over- (Johnson et al., 1999; Khan et al., 2002).
or under-expression of P-gp can lead to accumulation or depletion of In horses, IVM at more than three times the therapeutic dose ad-
these drugs within cells. Moreover, there is a structure-affinity relation ministration and MXD at the therapeutic dose were not embryotoxic
for P-gp mediated drug transport of MLs and MXD is a poorer substrate and produced no adverse clinical abnormality in pregnant mares and
for P-gp, compared to other MLs, such as IVM and SLM (Lespine et al., new-born foals (Campbell and Benz, 1984; Mckissick et al., 1987).
2007). Although mammalian species have different sensitivity, similar Ivermectin did not adversely affect the fertility of mares or the orga-
signs of acute MLs toxicity, such as ataxia, tremors and coma are ob- nogenesis of their developing foals following oral treatment at a
served in most species (Lankas and Gordon, 1989). The oral lethal doses 0.6 mg/kg dose rate (therapeutic dose x 3) (Mckissick et al., 1987).

43
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Single oral IVM at doses of 0.2 or 0.6 mg/kg did not affect the sexual part be due to the differences in the methodology of the two different
behaviour of stallions or quality of their semen (Janett et al., 2001). intravenous studies. Male horses weighing 200–400 kg were used in the
Genotoxicity and carcinogenicity studies indicated that ABM and IVM MXD study (Afzal et al., 1997) whereas female horses weighing
had no genotoxic activity and no carcinogenic potential, respectively 510–600 kg were used in the IVM study (Gokbulut et al., 2010b). It has
(Lankas and Gordon, 1989). been observed that body weight and gender of the animals significantly
affect the plasma dispositions of MLs (McKellar and Gokbulut, 2012).
4.5. Pharmacokinetics The aqueous solubility of an active molecule may affect its bioa-
vailability, which depends on the rate and extent of absorption of the
Macrocyclic lactones are extremely lipophilic compounds that are drug from the site of application into the bloodstream (Baggot and
extensively distributed from the bloodstream to different tissues and McKellar, 1994). Although the antiparasitic spectrum and efficacy of
slowly eliminated from body compartments, especially liver and fat the different ML molecules are similar, different physicochemical
(Zulalian et al., 1994), and for this reason, large volumes of distribution properties of these drugs may account for differences in kinetic beha-
and longer persistence in the animals treated are observed for these viour, potency and persistence of their antiparasitic activity (Lifschitz
drugs compared to the other classes of anthelmintics. The pharmaco- et al., 2004) and in compatibility with excipients for formulation. The
kinetic dispositions of MLs are significantly affected by route of ad- vehicle in the pharmaceutical formulations of endectocides may play an
ministration, the formulation of the drug, and interspecies and inter- important role in their absorption from the site of administration and
individual variation (reviewed by McKellar and Benchaoui, 1996; bioavailability (Lo et al., 1985). In horses, it was demonstrated that
McKellar and Gokbulut, 2012). The studies on the pharmacokinetic time to the reach the peak plasma concentration (4–5 h) for the liquid
dispositions of MLs have been well documented in the last few years formulation of IVM was much shorter compared to that for the paste
and are summarized in Table 6. As in other animal species, the phar- formulation (15 h) following oral administrations at a dose of 0.2 mg/
macokinetic dispositions of IVM has been investigated more extensively kg BW. In addition, after treatment of horses with a liquid form of IVM
compared to that of the other members of the MLs since IVM was the via nasogastric tube, 20% higher bioavailability was obtained than for
first licensed and the most widely used endectocide in equine species. the oral paste form. Nevertheless, the anthelmintic activity of both IVM
In horses, the kinetic results for IVM obtained from eight studies formulations was similar when measured by reduced faecal egg count
(Table 10) display great variations (Cmax: 45.31 ± 20.16 ng/mL, tmax: (Asquith and Kivipelto, 1987; Asquith et al., 1988). In contrast with this
5.17 ± 2.24 h, AUC: 2960.65 ± 1149.18 ng.h/mL, T1/2: finding, recently, it has been reported that although, the other kinetic
95.06 ± 37.86 h, MRT: 114.48 ± 38.94 h following oral administra- parameters were similar, oral administration of a propylene glycol-
tion at the same dose rate. The reasons for these differences between based solution of IVM (2%) resulted in a later peak plasma concentra-
the studies are unknown but may in part be due to differences in tion (12 h) compared to the paste form (7.72 h) in horses (Saumell
methodology. It is unlikely that formulation differences could be re- et al., 2017). The differences between these two studies are probably
sponsible since all studies used the paste formulation of IVM. Feeding related to the different formulations of the oral solutions or the feeding
type or fasting could have caused differences in drug absorption, al- regime or body conditions of animals in each study.
though feeding is not considered to cause such substantive differences Limited information is available in the literature on the pharma-
in the oral absorption of anthelmintics in horses as it does in ruminants cokinetics of MLs after parenteral administration in horses, since the
(Ali and Hennessy, 1996; Gokbulut et al., 2007) and dogs (McKellar parenteral formulation of IVM has been shown to cause rare adverse
et al., 1993a). In some of the reported studies, the horses were kept reactions including Clostridium spp. infection and anaphylaxis
indoors and fed a hay-based diet whereas, in other studies, the horses (Campbell and Benz, 1984) and the parental formulation was with-
were yarded for the period during and immediately after drug admin- drawn in 1984 after a short period on the market (Lyons and Tolliver,
istration and were then returned to a grass paddock (Gokbulut et al., 2012). Although no authorised MLs are now available for parenteral
2001b). One of the most likely factors affecting the pharmacokinetics of administration, the plasma dispositions of IVM and DRM have been
IVM was the breed and size of the animals used. Perez et al. (1999) used investigated in horses after subcutaneous or intramuscular off-label
Chilean criollo horses weighing 380–496 kg whereas a mixed group of administration of the formulation licensed for use in cattle (Marriner
thoroughbreds weighing between 510–610 kg were administered IVM et al., 1987; Perez et al., 2003, 2010; Saumell et al., 2017). The pre-
in the study by Gokbulut et al. (2001b). Parasitic status of animals cipitation of MLs at the injection site also contributes to the slow ab-
could have had an effect, since the horses used by Perez et al. (1999) sorption phase of these drugs (Lo et al., 1985; Scott and McKellar,
were known to be infected with gastrointestinal parasites and such 1992). Parenteral administration of IVM and DRM resulted in greater
infections may have modest effects on absorption of anthelmintics availability than oral administration in horses (Table 6). It has been
(McKellar et al., 1991), unfortunately, the parasitological status of the reported that in horses, lower and later Cmax but much greater AUC, T1/
horses in most of these studies was unknown. 2 and MRT values were observed after subcutaneous or intramuscular
Moxidectin is much more lipophilic than the avermectins including injections of IVM (McKellar and Marriner, 1987; Perez et al., 2003;
IVM and DRM and remains longer in the treated animals which explains Saumell et al., 2017) and DRM (Perez et al., 2010) compared to those
the higher AUC and longer MRT which have been demonstrated in observed after oral administration (Table 6). However, although greater
ruminant species (Zulalian et al., 1994; Alvinerie et al., 1998; Carceles plasma availability of IVM was observed, a lower efficacy (36–64%)
et al., 2001) and following subcutaneous injection and oral adminis- was obtained after intramuscular administration compared to that ob-
tration in horses (Gokbulut et al., 2001b). However, the kinetic para- served following administration by the oral route (100% efficacy) in
meters observed after intravenous administrations of IVM at a dose of horses (Saumell et al., 2017). The greater IVM efficacy observed against
0.2 mg/kg BW (Gokbulut et al., 2010b) and MXD (at a dose of 0.4 mg/ adult cyathostomins following oral administration may be attributable
kg BW) (Afzal et al., 1997) are not in agreement with these findings in to the enhanced drug exposure of the worms located in the gastro-
horses. Although, the Vdss and oral bioavailability values are similar for intestinal lumen (Saumell et al., 2017). Similarly, it has been observed
IVM (Vdss:3.65 L/kg and Foral: 48%) and MXD (4.14 L/kg and Foral: that the higher activity of IVM was achieved against ascarids by the
40%), the AUC (8226.5 ng.h/mL), T1/2 (138.72 h) and MRT (147.12 h) paste formulation compared to the injectable administration (Lyons
values of IVM were much greater than those observed after MXD ad- et al., 1986).
ministration (AUC: 5700 ng.h/mL, dose corrected (0.2 mg/kg BW) as- Topical (pour-on) formulations of IVM, MXD, DRM and EPM have
suming proportionality, T1/2: 81.0 h and MRT: 92.3 h) in horses. The been developed and licensed for use in cattle. Nevertheless, off-label
reason for these differences in the kinetic dispositions of IVM and MXD administrations of pour-on IVM and EPM formulations have been in-
between oral and intravenous administrations is unclear but it might in vestigated to determine the potential for pour-on administration as an

44
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

alternative treatment route in equine species. Large variations in different physiological status of horses may alter the plasma disposi-
pharmacokinetics are evident among animal species after pour-on ad- tions of either molecules. In the study by Gokbulut et al. (2016b) the
ministration of IVM or EPM as a result of anatomic and physiologic horses were lactating and, the lactation status of the horses could be
variation, irregular absorption of drug from the site of application as- associated with faster plasma elimination (T1/2: 1.37 days) of EPM,
sociated with differences in coat length and hair density and the var- whereas the horses were not lactating (T1/2: 7.18 days) in the previous
iation of the environmental conditions to which the animals were ex- study using IVM (Gokbulut et al., 2010b).
posed during the studies. The pour-on formulations of MLs were Although IVM and DRM showed similar absorption patterns, the
administered at higher dose rates, their plasma availabilities via the plasma elimination and AUC of IVM were initially faster and larger,
topical route were much lower compared to oral or subcutaneous ad- respectively in comparison to DRM following oral administration in
ministration due to their poor or limited absorption through the skin, as donkeys (Gokbulut et al., 2005). Similar differences between IVM and
previously reported in different equine species including horses DRM were also observed in horses (Gokbulut et al., 2001b) and rumi-
(Gokbulut et al., 2010b, b) and donkeys (Gokbulut et al., 2011, 2013) nants (Lanusse et al., 1997; Toutain et al., 1997; Barber et al., 2003).
(Table 4) as it has been demonstrated in cattle (Gayrard et al., 1999). These differences may be associated with different physicochemical
In horses, it was shown that the plasma concentration and systemic properties between DRM and IVM. Both compounds are avermectins
availability of IVM were lower, but the plasma persistence was pro- but have a structural difference between in the dihydro modification at
longed with much longer terminal half-life after pour-on administration the 2223 position. The higher persistence of DRM compared to that of
(F%: 6.99) compared with the oral route (F%: 48.13) (Gokbulut et al., IVM could confer persistent efficiency against equine parasites due to
2016b). It is likely that after pour-on administration, MLs bind physi- its longer retention time in plasma.
cally to the haircoat at the application site further preventing absorp- The results from IVM in administered donkeys (Gokbulut et al.,
tion or that the molecules become trapped in the skin or precipitated at 2005) differ substantially from those reported for horses (Marriner
the skin surface at the site of application and are very slowly released et al., 1987; Perez et al., 1999; Gokbulut et al., 2001b; Perez et al.,
from there. Although the plasma concentration is much lower after 2003; Gokbulut et al., 2010b) at same dose rate and administration
pour-on administration in animals, the values of terminal half-life and route. In donkeys, larger AUC and longer MRT of both molecules in
MRT are significantly longer and these differences suggest slow ab- plasma and faeces reflect longer drug persistence and greater absorp-
sorption from the skin which limits later elimination of IVM. It has been tion from the gastrointestinal tract in donkeys compared to horses
shown that absorption was the rate-limiting kinetic process for pour-on (Gokbulut et al., 2001b). In contrast to this, Marriner et al. (1987);
formulations and the terminal half-life represents the half-life of ab- Perez et al. (1999, 2003) and Gokbulut et al (2010b) observed a much
sorption of MLs in cattle thus conferring a flip-flop effect in which the larger AUC with a higher Cmax for IVM in horses following oral ad-
disposition of the drug in the body and its elimination is controlled by ministration compared with IVM in donkeys. Reasons for the differ-
the absorption process (Gayrard et al., 1999). ences among the studies are unknown but may be associated with diet,
Moreover, the kinetic parameters of pour-on EPM in horses breed, parasitological status and anatomic characteristics in donkeys
(Gokbulut et al., 2016b) were different from those reported for pour-on compared with horses.
IVM following topical administration with the same dose rate (0.5 mg/ In two different studies, the plasma dispositions of EPM have been
kg BW) in horses (Gokbulut et al., 2010b). Thus, the plasma con- investigated in milking (Gokbulut et al., 2013) and non-milking don-
centration and persistence of pour-on EPM were much lower and keys (Gokbulut et al., 2011) following pour on administration at a dose
shorter than those of pour-on IVM. Furthermore, EPM was absorbed of 0.5 mg/kg BW. Although MRT values in both studies were similar,
and reached peak plasma concentration and was eliminated from Cmax and AUC values were almost 2 times smaller in the milking don-
plasma much faster, resulting in lower and shorter plasma availability keys compared with those in the non-milking donkeys. The reason for
compared with pour-on IVM (Table 4). This faster absorption process of the lower systemic exposure of EPM in milking-donkeys is unclear.
pour-on EPM could be correlated to different environmental or climatic These differences may in part be due to differences in methodology,
conditions. It was shown that the exposure of animals to rain or direct since different body size, length of haircoat and environmental condi-
UV-radiation changed the plasma dispositions of IVM in cattle after tions such as sun exposure can cause differences in drug absorption and
pour-on administration. It has been suggested that the exposure to rain systemic availability of drugs following pour-on administration
could result in the removal of active compound from body surface of (Gokbulut et al., 2011, 2012b). The faster elimination processes of EPM
animals and that the exposure to direct UV-radiation from sun could in this research compared to the previous study (Gokbulut et al., 2011)
cause increased cutaneous blood circulation at the administration site could be due to the different physiologic status of the study animals.
and ⁄ or decreased viscosity of the drug pour-on administered The lactation status of the donkeys could be associated with faster ex-
(Gokbulut et al., 2012a). However, the study with pour-on IVM cretion of EPM. In addition, the results observed in the milking donkeys
(Gokbulut et al., 2010b) was performed in December when the UV- (Gokbulut et al., 2013) indicated that the milk to plasma ratio (0.16) is
radiation is low, and the later study was performed in June when the similar to those reported in cattle (0.1) (Alvinerie et al., 1999). The milk
UV-radiation is relatively more intense. Moreover, the difference in the excretion of oral IVM and pour-on EPM have also been investigated in
pharmacokinetics between pour-on EPM and pour-on IVM could be due horses. Due to the poor systemic availability of pour-on EPM, its milk
to the different pharmaceutical formulations and physicochemical availability (AUCmilk: 8.51 ng.d/mL) was much lower compared with
properties of the drugs used. The rate of penetration of a drug will oral IVM (AUCmilk: 15.64 ng.d/mL). However, pour-on EPM displayed a
depend on its lipophilicity and EPM is the least lipophilic of the en- much greater milk partitioning (AUCmilk/plasma: 0.48) compared with
dectocides compounds (Sheriff et al., 2002). In addition, MLs are P-gp oral IVM (AUCmilk/plasma: 0.19) and compared with milk partitioning
substrates and their coefficients of lipophilicity and P-gp and Breast results obtained in different animal species (Shoop et al., 1996a; Dupuy
Cancer Resistance Protein (BCRP) effluxes determine their kinetic dis- et al., 2001a; Gokbulut et al., 2013). The origin of these differences is
positions or concentrations in different tissues of the host (Lespine unclear, although the anatomical and physiological variations of each
et al., 2012). The excipients in the pharmaceutical formulations of species do not appear to play an important role because it has been
endectocides may also alter their absorption from the application site shown that pour-on EPM displayed a similar extent of milk partitioning
and hence their plasma availability (Lo et al., 1985); The pharmaceu- in cows and donkeys (AUCmilk/plasma: 0.11 and 0.16, respectively)
tical formulation of pour-on EPM differs from pour-on IVM because the (Alvinerie et al., 1999; Gokbulut et al., 2013). Due to the high lipo-
IVM is formulated in crodamol, triethanolamine and isopropyl alcohol, philicity of the MLs, they are likely to partition into milk. However, the
whereas EPM formulation contains butylated hydroxytoluene, propy- fat content of mare’s milk (12.1 g/kg) is similar to donkey’s milk
lene glycol and octanoate decanoate as a vehicle. In addition, the (10.6 g/kg) and is nearly 3 times lower compared to cow’s milk (36.1 g/

45
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

kg) (Malacarne et al., 2002), suggesting that factors other than milk fat Closantel in horses it is known to have a very long terminal elimination
composition are involved in the excretion pattern of EPM and IVM in half-life in sheep with detectable concentrations measurable in plasma
horse milk. Transport proteins such as BCRP are located in mammary for up to approximately 100 days after oral administration of a ther-
glands during lactation, where they transport xenobiotic substrates apeutic dose (7.5 mg/kg) (Mohammed-Ali and Bogan, 1987). This
from plasma to milk (Jonker et al., 2005) and BCRP plays an important persistence in plasma has been attributed to strong binding to plasma
role in the milk partitioning of endectocides into milk (Lespine et al., albumin which has a turnover rate of about 16.6 days (Holmes et al.,
2012). In this respect, it would also be of great interest to understand 1968) and this may also explain it’s activity against parasites which
the different transportation patterns of these drugs in equine species. ingest host plasma proteins such as F. hepatica.
The mean highest drug concentrations in donkey milk (Cmax: 2.69 ng/ It has been demonstrated that in horses, closantel was highly ef-
mL for pour-on EPM) and mare’s milk (Cmax: 4.59 ng/mL for oral IVM fective against adult S. vulgaris, S. edentatus, A. perfoliata and
and 3.84 ng/mL for pour-on EPM) were much lower compared to the Triodontophorus spp., and larval stages of S. vulgaris and Gasterophilus
maximum residue limits (MRL), which were established as 20 μg/kg for intestinalis following oral administration at doses of 20 or 40 mg/kg BW
EPM and 10 μg/kg for IVM in bovine milk by the joint FAO/WHO Ex- (Guerrero et al., 1983). In addition, naturally infected horses with F.
pert Committee (EPMAR, 2014). These results indicated that both oral hepatica were successfully (100% efficacy) treated with closantel fol-
IVM and pour-on EPM have minimal disposition rates into milk, which lowing oral administration at a dose of 15 mg/mL BW (Alcaino and
is important if the donkey or mare’s milk is to be used for human Aguilar, 1985). Closantel displayed a high anthelmintic activity against
consumption. F. hepatica in experimentally infected ponies after oral administration at
Following pour-on administration, the depletion of IVM or EPM at a dose of 10 mg/kg BW (Dorchies et al., 1990).
the application and far from the application sites were also determined
in horses (Gokbulut et al., 2010b) and donkeys (Gokbulut et al., 2011), 5.2. Praziquantel
respectively. Relatively high concentrations and slow degradation of
IVM or EPM in hair samples collected from the application site was Praziquantel (Fig. 6) has been classified as an isoquinoline and has
observed in both studies. In addition, detectable concentrations were been marketed in animal and human medicine for its activity against
observed in the hair samples collected far away from the application cestodes and trematodes. It is metabolised to less active glucuronide
site and this was considered likely due to the excretion of compounds and sulfuric conjugates in laboratory animals (Diekmann and Buhring,
by sebum secretion (after absorption of a fraction of IVM or EMP). 1976). It has been shown to bind to parasite Glutathione S- transferase
Sebum secretion was previously reported in humans with scabies after (McTigue et al., 1995) and to alter parasite intracellular calcium con-
an oral treatment with IVM (Haas et al., 2002). These results indicate centrations (Harnett, 1988) and its effects are manifest as parasite te-
that IVM or EPM was available at the skin surface for a long period of gumental disruption and muscular contraction. Praziquantel is gen-
time after pour-on administration and this probably provides longer erally very well tolerated in all animals for which it has been used
persistence of efficacy for ectoparasites in horses and donkeys. More- therapeutically with a very large therapeutic index in laboratory ani-
over, differences in the physiological and histological structure of skin mals (Frohberg, 1984). There is no data on the pharmacokinetics of
or haircoat between donkeys and horses probably play an important Praziquantel in horses but in man it is known to be rapidly and largely
role for transfer of the drug from the surface of the skin into the sys- (90%) absorbed following administration and subsequently rapidly
temic circulation. eliminated with a plasma clearance of 28 L/kg (Leopold et al., 1978).
Clinical trials by Slocombe et al. (2007) showed that the efficacy of
praziquantel horse paste 9% was 90.9, 100 and 100% against A. per-
5. Other anthelmintics
foliata, A. magna and A. mamillana, respectively. and safe in horses.
Horses infected with A. perfoliata were successfully treated with prazi-
5.1. Closantel
quantel at a dose of 1.5 mg/kg BW (Abbott et al., 2008). Praziquantel in
combination with MLs such as IVM, DRM, ABM or MXD as double
Closantel is a salicylanilide (Fig. 5) which acts by uncoupling oxi-
combination have been developed in order to effectively treat equine
dative phosphorylation (Corbett and Goose, 1971; Prichard, 1978).
nematode and cestodes parasites. Triple combinations with addition of
Closantel is not thought to be extensively metabolized, although Glu-
OFZ, FBZ or PYR + have been developed by pharmaceutical companies
curonide metabolizes of other members of the salicylanilide group have
for the treatment of parasites in equine species and may extend the
been identified (Broome and Jones, 1966). Toxicity is thought to be
spectrum of activity and delay the development of drug resistance to
associated with uncoupling of phosphorylation in the mammalian host
equine parasites (Table 2).
and the salicylanilides generally have a rather narrow therapeutic index
of between 4 and 6. Specific signs of blindness and increased defecation
respectively have been recognized in sheep treated with the related 5.3. Trichlorfon
drugs Rafoxanide and Oxyclozanide (Mrozik et al., 1969; Boray et al.,
1967). Trichlorfon is an organophosphate compound (Fig. 7) and in
Although there is no information on the pharmacokinetics of common with other members of this group of compounds acts to inhibit
the enzyme acetyl cholinesterase. Acetyl cholinesterase is responsible

Fig. 5. Chemical structure of Closantel. Fig. 6. Chemical structure of Praziquantel.

46
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

piperazine causes a flaccid paralysis in ascarid nematodes by acting as


an antagonist at cholinoreceptors (Norton and De Beer, 1957). How-
ever, Martin (1982, 1985) has shown that it is a GABA agonist of low
potency acting on extra synaptic GABA receptors to cause increased Cl−
conductance and consequent hyperpolarisation. Piperazine has a high
therapeutic index with LD50 values of more than 4000 mg/kg in mice
for both the hexahydrate and adipate salts (Cross et al., 1954). At very
high dosages, incoordination, hyperaesthesia and diarrhoea have been
Fig. 7. Chemical structure of Trichlorfon. recorded in dogs and cats (Greenberg et al., 1958; Stoffman and
Braithwaite, 1976).
Piperazine is available as a powder for use in the horse and in zoo
canids and felines. It is used in the horse mainly because of its good
activity against Parascaris (Bogan and Duncan, 1984). In an earlier
study, it was reported that piperazine citrate at 88 mg/kg BW was fully
effective against mature and immature Parascaris in unweaned and
weaned foals, but the activity of piperazine dihydrochloride at doses of
44 mg/kg or 88 mg/kg BW was significant but incomplete against
Parascaris in yearlings (Drudge et al., 1960). The various dose levels of
the piperazine compounds were all successful in reducing the count of
Fig. 8. Chemical structure of Piperazine. strongylid eggs for 4 weeks after treatment. Recently, Lyons et al.
(2016) reported that piperazine has good but incomplete activity
for the degradation of the neurotransmitter acetyl choline and it is against strongyles and no activity against Strongyloides following oral
likely that this is the major effector mechanism for its antiparasitic administration at a dose of 112 mg/kg BW in horses. There is very little
activity (Bueding et al., 1972). It may be that the paralysis of nema- information available on the pharmacokinetics of Piperazine in horses.
todes associated with organophosphate activity is more effective when It is known to have low and variable absorption from the gastro-
it results in their removal (for example by gut peristalsis) from their intestinal tract in man (Fletcher et al., 1982) and it maybe the un-
predilection site. Trichlorfon inhibits acetylcholinesterase in mamma- absorbed gastrointestinal concentrations which confer activity.
lian plasma and brain but does not cause the polyneuritis associated
with some other organophosphate compounds when used at normal References
therapeutic dosages (Wolmstedt et al., 1978). During the interaction of
Abbott, J.B., Mellor, D.J., Barrett, E.J., Prolidman, C.J., Love, S., 2008. Serological
organophosphorus compounds with acetyl cholinesterase an initial
changes observed in horses infected with Anoplocephala perfoliata after treatment
weak bond is formed with a serine group on the enzyme. This bond can with praziquantel and natural reinfection. Vet. Rec. 162, 50–53.
be dephosphorylated using a nucleophilic competitor such as prali- Abusamra, M.T., Abbas, B., Ibrahim, K.E.E., 1987. 5 Cases of psoroptic mange in the
doxime and this forms the basis of treatment in intoxication. Ultimately domestic donkey (Equus asinus asinus) and treatment with ivermectin. Equine Vet. J.
19, 143–144.
if the bonds between the phosphate and its methyl, ethyl or amino Afzal, J., Burke, A.B., Batten, P.L., DeLay, R.L., Miller, P., 1997. Moxidectin: metabolic
groups are cleaved then the phosphorous becomes relatively negative fate and blood pharmacokinetics of C-14-labeled moxidectin in horses. J. Agric. Food
and thus insensitive to nucleophilic attack (it has “aged”). In this event Chem. 45, 3627–3633.
Alcaino, H., Aguilar, A., 1985. Actividad antihelmíntica del closantel y de la combinación
cholinesterase activity will only reappear if new enzyme is synthesized. febantel+ triclorfón en caballos fina sangre de carrera. Arch. Med. Vet. 17, 103–109.
In the absence of pralidoxime the antimuscarinic drug atropine is used Aldridge, W.M., 1954. Tricresylphosphates and cholinesterase. Biochem. J. 56, 185–189.
in the treatment of organophosphate toxicity to control muscarinic Ali, D.N., Hennessy, D.R., 1996. The effect of level of feed intake on the pharmacokinetic
disposition and efficacy of ivermectin in sheep. J. Vet. Pharmacol. Ther. 19, 89–94.
stimulation of the autonomic nervous system responsible for increased Alvarez, L.I., Sanchez, S.F., Lanusse, C.E., 1997. Modified plasma and abomasal dis-
secretions, bronchoconstriction, intestinal motility and miosis, but does position of albendazole in nematode-infected sheep. Vet. Parasitol. 69, 241–253.
not control muscle fasciculations associated with nicotinic stimulation Alvarez, L.I., Sanchez, S.P., Lanusse, C.E., 1999. In vivo and ex vivo uptake of albendazole
and its sulphoxide metabolite by cestode parasites: relationship with their kinetic
at neuromuscular junctions. (McKellar, 1997a). Organophosphates are behaviour in sheep. J. Vet. Pharmacol. Ther. 22, 77–86.
rapidly absorbed following oral administration and are metabolised by Alvarez, L.I., Imperiale, F.A., Sanchez, S.E., Murno, G.A., Lanusse, C.E., 2000. Uptake of
liver phosphatases which hydrolyse the parent molecules, and which albendazole and albendazole sulphoxide by Haemonchus contortus and Fasciola he-
patica in sheep. Vet. Parasitol. 94, 75–89.
have been shown to be host species specific (Aldridge, 1954), Meta-
Alvinerie, M., Escudero, E., Sutra, J.F., Eeckhoutte, C., Galtier, P., 1998. The pharma-
bolites are subsequently excreted in urine. cokinetics of moxidectin after oral and subcutaneous administration to sheep. Vet.
Efficacy and safety studies on trichlorfon reported that doses of Res. 29, 113–118.
40 mg/kg BW and less were generally well tolerated by horses (Drudge Alvinerie, M., Sutra, J.F., Galtier, P., Mage, C., 1999. Pharmacokinetics of eprinomectin in
plasma and milk following topical administration to lactating dairy cattle. Res. Vet.
et al., 1976). This investigation indicated that trichlorfon was highly Sci. 67, 229–232.
effective (efficacy between 97–100%) against G. intestinalis and G. na- Alvinerie, M., Dupuy, J., Eeckhoutte, C., Sutra, J.F., Kerboeuf, D., 2001. In vitro meta-
salis, P. equorum O. equi, S. vulgaris and S. edentatus. Higher dose rates bolism of moxidectin in Haemonchus contortus adult stages. Parasitol. Res. 87,
702–704.
(60 and 80 mg/kg BW) administered by stomach tube caused moder- Amery, W.K., Bruynseels, J.P., 1992. Levamisole, the story and the lessons. Int. J.
ately severe colic and diarrhea, whereas a dose of 80 mg/kg given in the Immunopharmacol. 14, 481–486.
feed resulted in only a transient softening of the faeces. More than two Anderson, N., Reynolds, G.W., Titchen, D.A., 1988. Changes in gastrointestinal mucosal
mass and mucosal and serum gastrin in sheep experimentally infected with ostertagia
thousand treatments of trichlorfon did not cause notable adverse clin- circumcincta. Int. J. Parasitol. 18, 325–331.
ical effects at a dose of 35–40 mg/kg in pregnant and nonpregnant Arena, J.P., Liu, K.K., Paress, P.S., Frazier, E.G., Cully, D.F., Mrozik, H., Schaeffer, J.M.,
mares, stallions, suckling and weanling foals, and yearlings (Drudge 1995. The mechanism of action of avermectins in Caenorhabditis elegans: correlation
between activation of glutamate-sensitive chloride current, membrane binding, and
et al., 1976). biological activity. Parasitol. 81, 286–294.
Arundel, J.H., 1983. Veterinary Anthelmintics. The University of Melbourne, Werribee,
Australia, pp. 90.
5.4. Piperazine Asquith, R.L., Kivipelto, J., 1987. The efficacy and acceptability of ivermectin liquid
compared to that of oral paste in horses. J. Equine Vet. Sci. 7, 353–355.
Asquith, R.L., Lane, T.J., Plue, R.E., Seward, R.L., Kivipelto, J., 1988. The bioavailability
Piperazine (Fig. 8) is a hexahydropyrazine and the citrate or dihy- of ivermectin in horses when administered in a liquid formulation by nasogastric
drochloride salts have been utilised in horses. It has been suggested that

47
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

intubation versus in an oral paste. J. Equine Vet. Sci. 8, 28–30. Broome, A.W., Jones, W.G.M., 1966. A new drug for the treatment of Fascioliosis in sheep
Aubry, M.L., Cowell, P., Davey, M.J., Shevde, S., 1970. Aspects of the pharmacology of a and cattle. Nature 310 745-744.
new anthelmintic: pyrantel. Br. J. Pharmacol. 38, 332–344. Bruni, S.F.S., Fuse, L.A., Moreno, L., Saumell, C.A., Alvarez, L.I., Fiel, C., McKellar, Q.A.,
Austin, W.C., Courtney, W., Danilewicz, J.C., Morgan, D.H., Conover, L.H., Howes, H.L., Lanusse, C.E., 2005. Changes to oxfendazole chiral kinetics and anthelmintic efficacy
Lynch, J.E., McFarland, J.W., Cornwell, R.L., Theodorides, V.J., 1966. Pyrantel tar- induced by piperonyl butoxide in horses. Equine Vet. J. 37, 257–262.
trate, a new anthelmintic effective against infections of domestic animals. Nature Bueding, Liu, G.L., Rogers, S.H., 1972. Inhibition by metriphonate and dichlorous of
212, 1273–1274. cholinesterases in schistosomes. Br. J. Pharmacol. 46, 480–487.
Austin, S.M., Dipietro, J.A., Foreman, J.H., Baker, G.J., Todd, K.S., 1991. Comparison of Burrough, S., 1986. Ivermectin toxicity in horses. N. Z. Vet. J. 34, 137–138.
the efficacy of ivermectin, oxibendazole, and pyrantel pamoate against 28-Day Campbell, W.C., 2012. History of avermectin and ivermectin, with notes on the history of
parascaris-equorum larvae in the intestine of pony foals. J. Am. Vet. Med. Assoc. 198, other Macrocyclic Lactone antiparasitic agents. Curr. Pharm. Biotechnol. 13,
1946–1949. 853–865.
Averkin, E.A., Beard, C.C., Dvorak, C.A., Edwards, J.A., Fried, J.H., Kilian, J.G., Schiltz, Campbell, W.C., Benz, G.W., 1984. Ivermectin: a review of efficacy and safety. J. Vet.
R.A., Kistner, T.P., Drudge, J.H., 1975. Methyl 5(6)-phenylsulfinyl-2-benzimidazo- Pharmacol. Ther. 7, 1–16.
lecarbamate, a new, potent anthelmintic. J. Med. Chem. 18, 1164–1166. Campbell, W.C., Leaning, W.H.D., Seward, R.L., 1989. Use of Ivermectin in Horses.
Aziz, M.A., Diallo, S., Diop, I.M., Lariviere, M., Porta, M., 1982. Efficacy and tolerance of Ivermectin and Abamectin. Springer, New York, pp. 234–244.
ivermectin in human onchocerciasis. Lancet 2, 171–173. Carceles, C.M., Diaz, M.S., Vicente, M.S., Sutra, J.F., Alvinerie, M., Escudero, E., 2001.
Bachmann, K.A., 1989. The use of single-sample clearance estimates to probe hepatic Milk kinetics of moxidectin and doramectin in goats. Res. Vet. Sci. 70, 227–231.
drug metabolism in rats. IV. A model for possible application to phenotyping xeno- Chalfie, M., Thomson, J.N., 1979. Organization of neuronal microtubules in the nematode
biotic influences on human drug metabolism. Xenobiotica 19, 1449–1459. Caenorhabditis elegans. J. Cell Biol. 82, 278–289.
Baggot, J.D., 1974. An introduction to principles of veterinary clinical pharmacology: the Chement, D., 1982. Essai de traitement des strongylidoses digestives des équidés par le
elimination of drugs. N. Z. Vet. J. 22, 63–71. fébantel. Ecole Nationale Vétérinaire d’Alfort. Thèse: Méd. Vét. Paris XII, 102.
Baggot, J.D., McKellar, Q.A., 1994. The absorption, distribution and elimination of an- Chiu, S.-H.L., Lu, A.Y., 1989. Metabolism and Tissue Residues Ivermectin and Abamectin.
thelmintic drugs: the role of pharmacokinetics. J. Vet. Pharmacol. Ther. 17, 409–419. Springer, pp. 131–143.
Barber, S., Bowles, V., Lespine, A., Alvinerie, M., 2003. The comparative serum disposi- Chiu, S.H.L., Taub, R., Sestokas, E., Lu, A.Y.H., Jacob, T.A., 1987. Comparative in vivo
tion kinetics of subcutaneous administration of doramectin, ivermectin and mox- and in vitro metabolism of ivermectin in steers, sheep, swine, and rat. Drug Metab.
idectin in the Australian Merino sheep. J. Vet. Pharmacol. Ther. 26, 343–348. Rev. 18, 289–302.
Bartram, D.J., Leathwick, D.M., Taylor, M.A., Geurden, T., Maeder, S.J., 2012. The role of Chiu, S.H.L., Green, M.L., Baylis, F.P., Eline, D., Rosegay, A., Meriwether, H., Jacob, T.A.,
combination anthelmintic formulations in the sustainable control of sheep nema- 1990. Absorption, tissue distribution, and excretion of tritium-labeled ivermectin in
todes. Vet. Parasitol. 186, 151–158. cattle, sheep, and rat. J. Agric. Food Chem. 38, 2072–2078.
Becker, W., 1975. Die Anwendung von Panacur bei trächtigen Tieren. Proceedings of the Cirak, V.Y., Gulegen, E., Yildirim, F., Durmaz, M., 2007. A field study on the efficacy of
Second European Multicolloquy on Parasitology, Trogir, Yugoslavia 105–106. doramectin against horse strongyles and determination its egg reappearance period.
Behm, C.A., Cornish, R.A., Bryant, C., 1983. Mebendazole concentrations in sheep Tierarztl. Wochenschr. 114, 64–66.
plasma. Res. Vet. Sci. 34, 37–41. Cobb, R., Boeckh, A., 2009. Moxidectin: a review of chemistry, pharmacokinetics and use
Bello, T.R., 1989. Efficacy of ivermectin against experimental and natural infections of in horses. Parasite. Vector. 2 S5.
Gasterophilus spp in ponies. Am. J. Vet. Res. 50, 2120–2123. Cockburn, A., Needham, D., 1999. The absorption, distribution, metabolism and excretion
Benchaoui, H.A., McKellar, Q.A., 1996. Interaction between fenbendazole and piperonyl of piperonyl butoxide in mammals. Piperonyl Butoxide. Elsevier, pp. 137–151.
butoxide: pharmacokinetic and pharmacodynamic implications. J. Pharm. Coles, G., 1977. The Mechanism of Action of Some Veterinary Anthelmintics Perspectives
Pharmacol. 48, 753–759. in The Control of Parasitic Disease In Animals in Europe. Royal College of Veterinary
Benoit, E., Cresteil, T., Riviere, J.L., Delatour, P., 1992. Specific and enantioselective Surgeons, pp. 53–63.
sulfoxidation of an aryl-trifluoromethyl sulfide by rat-liver Cytochromes-P-450. Drug Conder, G.A., Baker, W.J., 2002. Chemistry, pharmacology and safety: doramectin and
Metab. Dispos. 20, 877–881. selamectin. In: Vercruysse, J., Rew, R.S. (Eds.), Macrocyclic Lactones in Antiparasitic
Bentley, O., Burns, S., McDonald, D., Drudge, J., Lyons, E., Kruckenberg, S., Vaughn, J., Therapy. CABI Publishing, Wallingford, UK, pp. 30–50.
1978. Safety evaluation of pyrantel pamoate administered with trichlorfon as a Conder, G.A., Campbell, W.C., 1995. Chemotherapy of nematode infections of veterinary
broad-spectrum anthelmintic in horses. Vet. Med. Small Anim. Clin. 73, 70–73. importance, with special reference to drug resistance. Advances in Parasitology.
Beretta, C., Fadini, L., Stracciari, J.M., Montesissa, C., 1987. In vitro febantel transfor- Elsevier, pp. 1–84.
mation by sheep and cattle ruminal fluids and metabolism by hepatic subcellular Corbett, J.R., Goose, J., 1971. A possible biochemical mode of action of the fasciolicides
fractions from different animal species. Biochem. Pharmacol. 36, 3107–3114. nitroxynil, hexachlorophene and oxyclozanide. Pest Manag. Sci. 2, 119–121.
Binev, R., Kirkova, Z., Nikolov, J., Russenov, A., Stojanchev, K., Lazarov, L., Hristov, T., Cornwell, R., 1966. Field trials in sheep with the anthelmintic pyrantel tartrate.
2005. Efficacy of parenteral administration of ivermectin in the control of strongy- Comparative trials in the prevention of Nematodirus infection in lambs. Vet. Rec. 79,
lidosis in donkeys. J. S. Afr. Vet. Med. Assoc. 76, 214–216. 626–629.
Biswajit, D., Suvakanta, D., Chandra, C.R., Jashobir, C., 2014. Synergistic im- Cornwell, R., Jones, R., 1968. Critical tests in the horse with the anthelmintic pyrantel
munostimulatory activity of Terminalia bellerica gum polysaccharide with levami- tartrate. Vet. Rec. 82, 483–484.
sole. World. J. Pharm. Pharm. Sci. 3, 1367–1384. Courtney, C.H., Robertson, E.L., 1997. Chemotherapy of parasitic diseases. In: Adams,
Bjorn, H., Hennessy, D.R., Friis, C., 1996. The kinetic disposition of pyrantel citrate and H.R. (Ed.), Veterinary Pharmacology and Therapeutics. Iowa State University Press,
pamoate and their efficacy against pyrantel-resistant Oesophagostomum dentatum in Ames, Iowa, pp. 885–932.
pigs. Int. J. Parasitol. 26, 1375–1380. Cross, B.G., David, A., Vallance, D.K., 1954. Piperazine adipate: a new anthelmintic agent.
Blair, L.S., Campbell, W.C., 1979. Efficacy of avermectin B1a against microfilariae of Part II, toxicological and pharmacological studies. J. Pharm. Pharmacol. 6, 711–717.
Dirofilaria immitis. Am. J. Vet. Res. 40, 1031–1032. Cully, D.F., Vassilatis, D.K., Liu, K.K., Paress, P.S., Van der Ploeg, L.H.T., Schaeffer, J.M.,
Boersema, J.H., Eysker, M., van der Aar, W.M., 1998. The reappearance of strongyle eggs Arena, J.P., 1994. Cloning of an avermectin-sensitive glutamate-gated chloride
in the faeces of horses after treatment with moxidectin. Vet. Res. Forum 20, 15–17. channel from Caenorhabditis elegans. Nature 371, 707–711.
Bogan, J.A., 1983. Absorption and distribution. In: Bogan, J.A., Lees, P., Yoxall, A.T. Davis, A., 1973. Drug Treatment in Intestinal Helminthiases. World Health Organization,
(Eds.), Pharmacological Basis of Large Animal Medicine. Blackwell Scientific Geneva, Italy, pp. 125.
Publication, Oxford, UK, pp. 9–11. de Montellano, P.R.O., Reich, N.O., 1986. Inhibition of cytochrome P-450 enzymes. In:
Bogan, J., Armour, J., 1987. Anthelmintics for ruminants. Int. J. Parasitol. 17, 483–491. Ortiz de Montellano, P.R. (Ed.), Cytochrome P-450. Plenum, New York, pp. 273–314.
Bogan, J.A., Duncan, J.L., 1984. Anthelmintics for dogs, cats and horses. Br. Vet. J. 140, Debackere, M., Landuyt, J., Vercruysse, J., Mckellar, Q., 1993. The influence of Ostertagia
361–367. circumcincta and Trichostrongylus colubriformis infections on the pharmacokinetics of
Bogan, J.A., McKellar, Q.A., 1988. The pharmacodynamics of ivermectin in sheep and febantel in lambs. J. Vet. Pharmacol. Ther. 16, 261–274.
cattle. J. Vet. Pharmacol. Ther. 11, 260–268. Delatour, P., Parish, R., 1986. Benzimidazole anthelmintics and related compounds:
Bogan, J., Benoit, E., Delatour, P., 1987. Pharmacokinetics of oxfendazole in goats: a toxicity and evaluation of residues. In: Ed Rico, A.G. (Ed.), Drug Residues in Animals.
comparison with sheep. J. Vet. Pharmacol. Ther. 10, 305–309. Academic Press, New York, pp. 175–203.
Booze, T.F., Oehme, F.W., 1982. A Literature-Review of the Anthelmintic, Fenbendazole. Delatour, P., Lorgue, G., Lapras, M., Richard, Y., 1976. Embryotoxic and antimitotic
Vet. Hum. Toxicol. 24, 49–52. properties of parbendazole, mebendazole and cambendazole. C. R. Acad. Sci. Hebd.
Boray, J.C., Happich, F.A., Andrews, J.C., 1967. Comparative chemotherapeutical tests in Seances Acad. Sci. D. 282, 517.
sheep infected with immature and mature Fasciola hepatica. Vet. Rec. 80, 218–224. Delatour, P., Debroye, J., Lorgue, G., Courtot, D., 1977. Experimental embryotoxicity of
Borgers, M., De, S.N., De, M.B., Thienpont, D., 1975. Influence of the anthelmintic me- oxfendazole in the rat and sheep. Rec. Med. Vet. 153, 639–645.
bendazole on microtubules and intracellular organelle movement in nematode in- Delatour, P., Daudon, M., Martin, S., 1982. Febantel - metabolism-embryotoxicity re-
testinal cells. Am. J. Vet. Res. 36, 1153–1166. lationship. Teratology 25 A18-A18.
Bossche, H.V., 1985. Pharmacology of Anthelmintics Chemotherapy of Gastrointestinal Delatour, P., Garnier, F., Benoit, E., Longin, C., 1984. A correlation of toxicity of alben-
Helminths. Springer, Berlin Heidelberg, pp. 125–181. dazole and oxfendazole with their free metabolites and bound residues. J. Vet.
Bowman, D., Johnson, R., Hepler, D., 1990. Effects of milbemycin oxime on adult Pharmacol. Ther. 7, 139–145.
hookworms in dogs with naturally acquired infections. Am. J. Vet. Res. 51, 487–490. Delatour, P., Benoit, E., Garnier, F., Besse, S., 1990. Chirality of the sulfoxide metabolites
Brazik, E.L., Luquire, J.T., Little, D., 2006. Pyrantel pamoate resistance in horses re- of fenbendazole and albendazole in sheep. J. Vet. Pharmacol. Ther. 13, 361–366.
ceiving daily administration of pyrantel tartrate. J. Am. Vet. Med. Assoc. 228, Delatour, P., Benoit, E., Besse, S., Boukraa, A., 1991a. Comparative enantioselectivity in
101–103. the sulfoxidation of albendazole in man, dogs and rats. Xenobiotica 21, 217–221.
Britt, D., Preston, J., 1985. Efficacy of ivermectin against Dictyocaulus arnfieldi in ponies. Delatour, P., Garnier, F., Benoit, E., Caude, I., 1991b. Chiral behaviour of the metabolite
Vet. Rec. 116, 343–345. albendazole sulphoxide in sheep, goats and cattle. Res. Vet. Sci. 50, 134–138.

48
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Delatour, P., Benoit, E., & Soraci, A., 1994. Reckebusch Memorial Lecture: Drug chirality: doramectin and ivermectin pour-on formulations in cattle. Vet. Parasitol. 81, 47–55.
its significance in veterinary pharmacology and therapeutics. In: Lees, P. (ed), 6th Geary, T.G., Klein, R.D., Vanover, L., Bowman, J.W., Thompson, D.P., 1992. The nervous
International Congress of the European Association for Veterinary Pharmacology and systems of helminths as targets for drugs. Parasitol. 78, 215–230.
Toxicology. Blackwell Scientific Press, Edinburgh. p. 6-9. Georgi, J.R., Rendano, V.T., King, J.M., Bianchi, D.G., Theodorides, V.J., 1980. Equine
Didier, A., Loor, F., 1996. The abamectin derivative ivermectin is a potent P-glycoprotein verminous arteritis - efficiency and speed of larvicidal activity as influenced by do-
inhibitor. Anticancer Drugs 7, 745–751. sage of albendazole. Cornell Vet. 70, 147–152.
Diekmann, H.W., Buhring, K.U., 1976. Thye fate of praziquantel in the organism. III Gokbulut, C., Nolan, A.M., McKellar, Q.A., 2001a. Pharmacokinetic disposition and faecal
Metabolism in rat, beagle dog and rhesus monkey. Eur. J. Drug Metab. excretion of pyrantel embonate following oral administration in horses. J. Vet.
Pharmacokinet. 2, 107–112. Pharmacol. Ther. 24, 77–79.
DiPietro, J., Todd Jr, K., 1987. Anthelmintics used in treatment of parasitic infections of Gokbulut, C., Nolan, A.M., McKellar, Q.A., 2001b. Plasma pharmacokinetics and faecal
horses. Vet. Clin. North Am. Equine Pract. 3, 1–14. excretion of ivermectin, doramectin and moxidectin following oral administration in
DiPietro, J.A., Hutchens, D.E., Lock, T.F., Walker, K., Paul, A.J., Shipley, C., Rulli, D., horses. Equine Vet. J. 33, 494–498.
1997. Clinical trial of moxidectin oral gel in horses. Vet. Parasitol. 72, 167–177. Gokbulut, C., Nolan, A.M., McKellar, Q.A., 2002. Plasma disposition, faecal excretion and
Dorchies, P., de Lahitte, J.D., Donat, F., 1990. Treatment of experimental fascioliasis in in vitro metabolism of oxibendazole following oral administration in horses. Res. Vet.
horses with closantel. Rev. Med. Vet. 141, 383–387. Sci. 72, 11–15.
Dorchies, P., de Lahitte, J.D., Flochlay, A., Blond-Riou, F., 1998. Efficacy of moxidectin Gokbulut, C., Boyacioglu, M., Karademir, U., 2005. Plasma pharmacokinetics and faecal
2% equine gel against natural nematode infections in ponies. Vet. Parasitol. 74, excretion of ivermectin (Eqvalan (R) paste) and doramectin (Dectomax (R), (1%)
85–89. following oral administration in donkeys. Res. Vet. Sci. 79, 233–238.
Dougherty, C.T., 1992. Pasture management. In: Powel, D.G., Jackson, S.G. (Eds.), In The Gokbulut, C., Akar, F., McKellar, Q.A., 2006. Plasma disposition and faecal excretion of
Health of Horses. Longman Scientific and Technical Publication, Essex, UK, pp. oxfendazole, fenbendazole and albendazole following oral administration to donkeys.
99–120. Vet. J. 172, 166–172.
Drexler, G., Sieghart, W., 1984. Properties of a high affinity binding site for [3H]aver- Gokbulut, C., Karademir, U., Boyacioglu, M., Akar, F., 2007. The effect of diet type on the
mectin B1a. Eur. J. Pharmacol. 99, 269–277. plasma disposition of triclabendazole in goats. Res. Vet. Sci. 82, 388–391.
Drudge, J., Lyons, E., 1970. The chemotherapy of migrating strongyle larvae. Proc. 2nd Gokbulut, C., Cirak, V.Y., Senlik, B., Yildirim, F., McKellar, Q.A., 2009. Pharmacological
Int. Conf. Equine Infectious Diseases 310–322. assessment of netobimin as a potential anthelmintic for use in horses: plasma dis-
Drudge, J.H., Leland Jr, S.E., Wyant, Z.N., Elam, G.W., Hutzler, L.B., 1960. Field studies position, faecal excretion and efficacy. Res. Vet. Sci. 86, 514–520.
comparing piperazine-carbon disulfide complex with carbon disulfide for parasite Gokbulut, C., Boyacioglu, M., Karademir, U., Aksit, D., 2010a. The effect of fasting on the
control in the horse. Am. J. Vet. Res. 21, 397–402. plasma disposition of triclabendazole following oral administration in goats. Res. Vet.
Drudge, J.H., Lyons, E.T., Taylor, E.L., 1976. Critical tests and safety studies on tri- Sci. 89, 415–417.
chlorfon as an antiparasitic agent in the horse. Am. J. Vet. Res. 37, 139–144. Gokbulut, C., Cirak, V.Y., Senlik, B., Aksit, D., Durmaz, M., McKellar, Q.A., 2010b.
Drudge, J.H., Lyons, E.T., Tolliver, S.C., 1981. Parasite control in horses - a summary of Comparative plasma disposition, bioavailability and efficacy of ivermectin following
contemporary drugs. Vet. Med. Small Anim. Clin. 76, 1479–1489. oral and pour-on administrations in horses. Vet. Parasitol. 170, 120–126.
Duce, I.R., Scott, R.H., 1985. Actions of dihydroavermectin B1α on insect muscle. Br. J. Gokbulut, C., Di Loria, A., Gunay, N., Masucci, R., Veneziano, V., 2011. Plasma disposi-
Pharmacol. 85, 395–401. tion, concentration in the hair, and anthelmintic efficacy of eprinomectin after to-
Duncan, J.L., Mcbeath, D.G., Preston, N.K., 1980. Studies on the efficacy of fenbendazole pical administration in donkeys. Am. J. Vet. Res. 72, 1639–1645.
used in a divided dosage regime against strongyle infections in ponies. Equine Vet. J. Gokbulut, C., Sekkin, S., Aksit, D., Karagenc, T., Aysul, N., Tatli, O., Boyacioglu, M.,
12, 78–80. 2012a. The effects of simulated rain and sun exposure on the plasma disposition of
Dupuy, J., Chartier, C., Sutra, J.F., Alvinerie, M., 2001a. Eprinomectin in dairy goats: dose ivermectin following pour-on administration in heifers. J. Vet. Pharmacol. Ther. 35,
influence on plasma levels and excretion in milk. Parasitol. Res. 87, 294–298. 309–312.
Dupuy, J., Escudero, E., Eeckhoutte, C., Sutra, J.F., Galtier, P., Alvinerie, M., 2001b. In Gokbulut, G., Yalinkilinc, H.S., Naturali, S., Gallo, A., Veneziano, V., 2012b. Plasma
vitro metabolism of C-14-moxidectin by hepatic microsomes from various species. disposition and milk excretion of eprinomectin in lactating donkeys. J. Vet.
Vet. Res. Commun. 25, 345–354. Pharmacol. Ther. 35 131-131.
Dutton, C.J., Gibson, S.P., Goudie, A.C., Holdom, K.S., Pacey, M.S., Ruddock, J.C., Bulock, Gokbulut, C., Naturali, S., Rufrano, D., Anastasio, A., Yalinkilinc, H.S., Veneziano, V.,
J.D., Richards, M.K., 1991. Novel avermectins produced by mutational biosynthesis. 2013. Plasma disposition and milk excretion of eprinomectin following pour-on ad-
J. Antibiot. 44, 357–365. ministration in lactating donkeys. J. Vet. Pharmacol. Ther. 36, 302–305.
Echeverria, J., Mestorino, N., Turic, E., Pesoa, J., Errecalde, J.O., 2001. Pharmacokinetics Gokbulut, C., Aksit, D., Smaldone, G., Mariani, U., Veneziano, V., 2014. Plasma phar-
of abamectin in horses. J. Vet. Pharmacol. Ther. 24, 359–360. macokinetics, faecal excretion and efficacy of pyrantel pamoate paste and granule
Egerton, J.R., Ostlind, D.A., Blair, L.S., Eary, C.H., Suhayda, D., Cifelli, S., Riek, R.F., formulations following per os administration in donkeys naturally infected with in-
Campbell, W.C., 1979. Avermectins, new family of potent anthelmintic agents: effi- testinal strongylidae. Vet. Parasitol. 205, 186–192.
cacy of the b1a component. Antimicrob. Agents Chemother. 15, 372–378. Gokbulut, C., Aksit, D., Santoro, M., Roncoroni, C., Mariani, U., Buono, F., Rufrano, D.,
El-Makawy, A., Radwan, H.A., Ghaly, I.S., Abd El-Raouf, A., 2006. Genotoxical, ter- Fagiolo, A., Veneziano, V., 2016a. Plasma disposition, milk excretion and para-
atological and biochemical effects of anthelmintic drug oxfendazole Maximum sitological efficacy of mebendazole in donkeys naturally infected by Cyathostominae.
Residue Limit (MRL) in male and female mice. Reprod. Nutr. Dev. 46, 139–156. Vet. Parasitol. 217, 95–100.
EMEA, 1998. Prantel Embonate Summary Report. Committee for Veterinary Medicinal Gokbulut, C., Ozuicli, M., Aksit, D., Aksoz, E., Korkut, O., Yalcinkaya, M., Cirak, V.Y.,
Products. . http://www.ema.europa.eu/docs/en_GB/document_library/Maximum_ 2016b. Comparative plasma and milk dispositions, faecal excretion and efficacy of per
Residue_Limits_-_Report/2009/11/WC500015797.pdf. os ivermectin and pour-on eprinomectin in horses. J. Vet. Pharmacol. Ther. 39,
EPMAR, 2014. EMA/ CVMP/294840/2014, Committee for Medicinal Products for 584–591.
Veterinary Use. Maximum_Residue_Limits-Report/2014/05/WC500167329. Goldstein, L.J., Galski, H., Fojo, A., Willingham, M., Lai, S.L., Gazdar, A., Pirker, R.,
Eysker, M., Boersema, J.H., Kooyman, F.N.J., 1992. The effect of ivermectin treatment Green, A., Crist, W., Brodeur, G.M., Lieber, M., Cossman, J., Gottesman, M.M., Pastan,
against inhibited early third stage, late third stage and fourth stage larvae and adult I., 1989. Expression of multidrug resistance gene in human cancers. JNCI J. Natl.
stages of the cyathostomes in Shetland ponies and spontaneous expulsion of these Cancer Inst. 81, 116–124.
helminths. Vet. Parasitol. 42, 295–302. Gottschall, D.W., Wang, R., 1996. Depletion and bioavailability of C-14-oxibendazole
Faulkner, J.K., Figdor, S.K., Monro, A.M., Von Wittenau, M.S., Stopher, D.A., Wood, B.A., residues in swine tissues. Vet. Parasitol. 64, 83–93.
1972. The comparative metabolism of pyrantel in five species. J. Sci. Food Agric. 23, Goudah, A., 2003. Aspects of the pharmacokinetics of albendazole sulphoxide in sheep.
79–91. Vet. Res. Commun. 27, 555–566.
Fisher, M.H., Mrozik, H., 1989. Chemistry Ivermectin and Abamectin. Springer, New Goudie, A.C., Evans, N.A., Gration, K.A.F., Bishop, B.F., Gibson, S.P., Holdom, K.S., Kaye,
York, pp. 1–23. B., Wicks, S.R., Lewis, D., Weatherley, A.J., Bruce, C.I., Herbert, A., Seymour, D.J.,
Fisher, M.H., Mrozik, H., 1992. The chemistry and pharmacology of avermectins. Annu. 1993. Doramectin - a potent novel endectocide. Vet. Parasitol. 49, 5–15.
Rev. Pharmacol. Toxicol. 32, 537–553. Greenberg, J., Seymore, W.E., McEwan, A.F., 1958. Effects of varying doses of piperazine
Fletcher, K.A., Evans, D.A.P., Kelly, J.A., 1982. Urinary piperazine excretion in healthy citrate on puppies. Vet. Med. 53, 609–610.
caucasians. Ann. Trop. Med. Parasitol. 76, 77–82. Grosenbaugh, D., Reinemeyer, C., Figueiredo, M., 2011. Pharmacology and therapeutics
French, D.D., Klei, T.R., Taylor, H.W., Chapman, M.R., Wright, F.R., 1988. Efficacy of in donkeys. Equine Vet. Educ. 23, 523–530.
ivermectin in the oral paste formulation against naturally acquired adult and larval Guerrero, J., Michael, B.F., Rohovsky, M.W., Campbell, B.P., 1983. The activity of clo-
stages of parascaris-equorum in pony foals. Am. J. Vet. Res. 49, 1000–1003. santel as an equine antiparasitic agent. Vet. Parasitol. 12, 71–77.
Friedman, P.A., Platzer, E.G., 1980. Interaction of anthelmintic benzimidazoles with Gupta, M., 2016. Levamisole: a multi-faceted drug in dermatology. Indian J. Dermatol.
Ascaris suum embryonic tubulin. Biochimica et Biophysica Acta (BBA. - General Venereol. Leprol. 82, 230.
Subjects 630, 271–278. Haas, N., Lindemann, U., Frank, K., Sterry, W., Lademann, F., Katzung, W., 2002. Rapid
Frohberg, H., 1984. Results of toxicological studies on praziquantel. Arzneim. Forsch. 34, and preferential sebum secretion of ivermectin: a new factor that may determine drug
1137–1144. responsiveness in patients with scabies. Arch. Dermatol. 138, 1618–1619.
Galtier, P., Alvinerie, M., Delatour, P., 1986. In vitro sulfoxidation of albendazole by Harnett, W., 1988. The antiparasitic action of praziquantel. Parasitol. Today 4, 144–147.
ovine liver-microsomes - assay and frequency of various xenobiotics. Am. J. Vet. Res. Harrow, I.D., Gration, K.A.F., 1985. Mode of action of the anthelmintics morantel, pyr-
47, 447–450. antel and levamisole on muscle-cell membrane of the nematode Ascaris suum. Pestic.
Galtier, P., Coulet, M., Sutra, J.F., Birosauveur, B., Alvinerie, M., 1994. Fasciola hepatica - Sci. 16, 662–672.
mebendazole and thiabendazole pharmacokinetics in sheep. Exp. Parasitol. 79, Hautekeete, L.A., Khan, S.A., Hales, W.S., 1998. Ivermectin toxicosis in a zebra. Vet. Hum.
166–176. Toxicol. 40, 29–31.
Gayrard, V., Alvinerie, M., Toutain, P.L., 1999. Comparison of pharmacokinetic profiles of Hayes, P.W., 1994. Melbourne, AustraliaUnderstanding the Unique Persistent

49
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Anthelmintic Activity of These Second-Generation Macrocyclic Lactones Australian methimazole on the plasma disposition of fenbendazole and oxfendazole in sheep.
Veterinarians in Industry Conference (Annual AVA Conference)1994. Understanding Res. Vet. Sci. 58, 222–226.
the Unique Persistent Anthelmintic Activity of These Second-Generation Macrocyclic Lanusse, C., Lifschitz, A., Virkel, G., Alvarez, L., Sanchez, S., Sutra, J.F., Galtier, P.,
Lactones Australian Veterinarians in Industry Conference (Annual AVA Alvinerie, M., 1997. Comparative plasma disposition kinetics of ivermectin, mox-
Conference) 1–6. idectin and doramectin in cattle. J. Vet. Pharmacol. Ther. 20, 91–99.
Hennessy, D.R., 1993. Pharmacokinetic disposition of benzimidazole drugs in the rumi- Laughton, D.L., Wheeler, S.V., Lunt, G.G., Wolstenholme, A.J., 1995. The beta-subunit of
nant gastrointestinal tract. Parasitol. Today 9, 329–333. Caenorhabditis elegans avermectin receptor responds to glycine and is encoded by
Hennessy, D.R., Steel, J.W., Prichard, R.K., Lacey, E., 1992. The effect of co-administra- chromosome-1. J. Neurochem. 64, 2354–2357.
tion of parbendazole on the disposition of oxfendazole in sheep. J. Vet. Pharmacol. Lees, P., Taylor, J., Higgins, A., Sedgwick, A., 1988. In vitro and in vivo binding of
Ther. 15, 10–18. phenylbutazone and related drugs to equine feeds and digesta. Res. Vet. Sci. 44,
Hennessy, D.R., Steel, J.W., Prichard, R.K., 1993. Biliary-secretion and enterohepatic 50–56.
recycling of fenbendazole metabolites in sheep. J. Vet. Pharmacol. Ther. 16, Leopold, G., Ungethium, W., Groll, E., Diekmann, H.W., Nowak, H., Wenger, D.H.G.,
132–140. 1978. Clinical pharmacology in normal volunteers of praziquantel, a new drug
Herd, R.P., Donham, J.C., 1983. Efficacy of ivermectin against Onchocerca cervicalis mi- against schistosones and cestodes. Eur. J. Clin. Pharmacol. 14, 281–291.
crofilarial dermatitis in horses. Am. J. Vet. Res. 44, 1102–1105. Lespine, A., Dupuy, J., Orlowski, S., Nagy, T., Glavinas, H., Krajcsi, P., Alvinerie, M.,
Holmes, P.H., Dargie, J.D., Maclean, J.M., Mulligan, W., 1968. Albumin and globulin 2006. Interaction of ivermectin with multidrug resistance proteins (MRP1, 2 and 3).
turnover in chronic ovine fascioliosis. Vet. Rec. 83, 227–228. Chem. Biol. Interact. 159, 169–179.
Huang, J., Casida, J.E., 1997. Avermectin B-1a binds to high- and low-affinity sites with Lespine, A., Martin, S., Dupuy, J., Roulet, A., Pineau, T., Orlowski, S., Alvinerie, M., 2007.
dual effects on the gamma-aminobutyric acid-gated chloride channel of cultured Interaction of MLs with P-glycoprotein: structure-affinity relationship. Eur. J. Pharm.
cerebellar granule neurons. J. Pharmacol. Exp. Ther. 281, 261–266. Sci. 30, 84–94.
Imam, S.M.A., Seri, H.I., Hassan, T., Tigani, T.A., Zolain, H.B., Abakar, A.D., 2010. Lespine, A., Dupuy, J., Alvinerie, M., Comera, C., Nagy, T., Krajcsi, P., Orlowski, S., 2009.
Therapeutic efficacy evaluation of anthelmintics activity of albendazole and iver- Interaction of macrocyclic lactones with the multidrug transporters: the bases of the
mectin drench formulations in donkeys in Darfur, Sudan. Vet. Arh. 80, 585–595. pharmacokinetics of lipid-like drugs. Curr. Drug Metab. 10, 272–288.
Jackson, H.C., 1989. Ivermectin as a systemic insecticide. Parasitol. Today (Regul. Ed.) 5, Lespine, A., Menez, C., Bourguinat, C., Prichard, R.K., 2012. P-glycoproteins and other
146–156. multidrug resistance transporters in the pharmacology of anthelmintics: prospects for
Jacobs, D., Hutchinson, M., Parker, L., Gibbons, L., 1995. Equine cyathostome infection: reversing transport-dependent anthelmintic resistance. Int. J. Parasitol. Drugs Drug
suppression of faecal egg output with moxidectin. Vet. Rec. 137 545-545. Resist. 2, 58–75.
Janett, F., Thun, R., Ryhiner, A., Burger, D., Hassig, M., Hertzberg, H., 2001. Influence of Lester, H.E., Spanton, J., Stratford, C.H., Bartley, D.J., Morgan, E.R., Hodgkinson, J.E.,
Eqvalan (R) (ivermectin) on quality and freezability of stallion semen. Coumbe, K., Mair, T., Swan, B., Lemon, G., Cookson, R., Matthews, J.B., 2013.
Theriogenology 55, 785–792. Anthelmintic efficacy against cyathostomins in horses in Southern England. Vet.
Johns, D., Philip, J., 1977. Albendazole: safety in sheep. Proceedings of the Eighth Parasitol. 197, 189–196.
International Conference of the World Association for Advancement of Vet. Parasitol. Lifschitz, A., Virkel, G., Mastromarino, M., Lanusse, C., 1997. Enhanced plasma avail-
Sydney. City. p. 58. ability of the metabolites of albendazole in fasted adult sheep. Vet. Res. Commun. 21,
Johnson, P.J., Mrad, D.R., Schwartz, A.J., Kellam, L., 1999. Presumed moxidectin tox- 201–211.
icosis in three foals. J. Am. Vet. Med. Assoc. 214, 678–680. Lifschitz, A., Sallovitz, J., Imperiale, F., Pis, A., Jauregui Lorda, J., Lanusse, C., 2004.
Jonker, J.W., Merino, G., Musters, S., van Herwaarden, A.E., Bolscher, E., Wagenaar, E., Pharmacokinetic evaluation of four ivermectin generic formulations in calves. Vet.
Mesman, E., Dale, T.C., Schinkel, A.H., 2005. The breast cancer resistance protein Parasitol. 119, 247–257.
BCRP (ABCG2. Concentrates drugs and carcinogenic xenotoxins into milk. Nat. Med. Littlewood, J.D., Rose, J.F., Paterson, S., 1995. Oral ivermectin paste for the treatment of
11, 127–129. chorioptic mange in horses. Vet. Rec. 137, 661–663.
Kass, I.S., Stretton, A.O.W., Wang, C.C., 1984. The effects of avermectin and drugs related Lo, P.K.A., Fink, D.W., Williams, J.B., Blodinger, J., 1985. Pharmacokinetic studies of
to acetylcholine and 4-aminobutyric acid on neurotransmission in Ascaris suum. Mol. ivermectin - Effects of formulation. Vet. Res. Commun. 9, 251–268.
Biochem. Parasitol. 13, 213–225. Lummis, S., Sattelle, D., 1985. GABA and benzodiazepine binding sites in insect CNS.
Kates, K., Colglazier, M., Enzie, F., 1975. Oxibendazole: critical anthelmintic trials in Pestic. Sci. 16, 695–697.
equids. Vet. Rec. 97, 442–444. Lyons, E.T., Tolliver, S.C., 2012. Macrocyclic lactones for parasite control in equids. Curr.
Khan, S.A., Kuster, D.A., Hansen, S.R., 2002. A review of moxidectin overdose cases in Pharm. Biotechnol. 13, 1070–1077.
equines from 1998 through 2000. Vet. Hum. Toxicol. 44, 232–235. Lyons, E.T., Drudge, J.H., Tolliver, S.C., 1980. Anti-parasitic activity of parbendazole in
Kinabo, L.D.B., Bogan, J.A., 1988. Pharmacokinetics and efficacy of triclabendazole in critical tests in horses. Am. J. Vet. Res. 41, 123–124.
goats with induced Fascioliasis. J. Vet. Pharmacol. Ther. 11, 254–259. Lyons, E.T., Drudge, J.H., Tolliver, S.C., 1986. The ascaricidal efficacy of ivermectin paste
Kinabo, L.D.B., Bogan, J.A., 1989. Disposition of triclabendazole in horses, ponies and in horses - as good as injectable. Vet. Med. 81 1062-&.
donkeys. Equine Vet. J. 21, 305–307. Lyons, E.T., Tolliver, S.C., Drudge, J.H., Granstrom, D.E., Collins, S.S., Stamper, S., 1992.
Klei, T.R., Torbert, B.J., Chapman, M.R., Turk, M.A.M., 1984. Efficacy of ivermectin in Critical and controlled tests of activity of moxidectin (Cl 301,423) against natural
injectable and oral paste formulations against 8-week-old strongylus vulgaris larvae in infections of internal parasites of equids. Vet. Parasitol. 41, 255–284.
ponies. Am. J. Vet. Res. 45, 183–185. Lyons, E.T., Tolliver, S.C., Kuzmina, T.A., Collins, S.S., 2010. Critical tests evaluating
Kotb, S., Abdel-Rady, A., 2013. Epidemiological studies of equine mange with special efficacy of moxidectin against small strongyles in horses from a herd for which re-
reference to different therapeutic protocols for effective treatment of Chorioptic duced activity had been found in field tests in Central Kentucky. Parasitol. Res. 107,
mange. Assiut Univ. Bull. Environ. Res. 16, 29–52. 1495–1498.
Kum, C., Gokbulut, C., Sekkin, S., Boyacioglu, M., 2009. Pharmacokinetics of dano- Lyons, E.T., Tolliver, S.C., Collins, S.S., 2011. Reduced activity of moxidectin and iver-
floxacin following intravenous and intramuscular administration in donkeys. J. Vet. mectin on small strongyles in young horses on a farm (BC) in Central Kentucky in two
Pharmacol. Ther. 32, 105–108. field tests with notes on variable counts of eggs per gram of feces (EPGs). Parasitol.
Lacey, E., 1990. Mode of action of benzimidazoles. Parasitol. Today 6, 112–115. Res. 108, 1315–1319.
Lacey, E., Gill, J.H., 1994. Biochemistry of benzimidazole resistance. Acta Trop. 56, Lyons, E.T., Dorton, A.R., Tolliver, S.C., 2016. Evaluation of activity of fenbendazole,
245–262. oxibendazole, piperazine, and pyrantel pamoate alone and combinations against
Lacey, E., Watson, T.R., 1985. Structure-activity relationships of benzimidazole carba- ascarids, strongyles, and strongyloides in horse foals in field tests on two farms in
mates as inhibitors of mammalian tubulin, in vitro. Biochem. Pharmacol. 34, Central Kentucky in 2014 and 2015. Vet. Parasitol.: Regional Stud. Reports 3, 23–26.
1073–1077. Mahfooz, A., Masood, M.Z., Yousaf, A., Akhtar, N., Zafar, M.A., 2008. Prevalence and
Lacey, E., Brady, R.L., Prichard, R.K., Watson, T.R., 1987. Comparison of inhibition of anthelmintic efficacy of abamectin against gastrointestinal parasites in horses. Pak.
polymerisation of mammalian tubulin and helminth ovicidal activity by benzimida- Vet. J. 28, 76–78.
zole carbamates. Vet. Parasitol. 23, 105–119. Malacarne, M., Martuzzi, F., Summer, A., Mariani, P., 2002. Protein and fat composition
Lankas, G.R., Gordon, L.R., 1989. Toxicology Ivermectin and Abamectin. Springer, New of mare’s milk: some nutritional remarks with reference to human and cow’s milk.
York, pp. 89–112. Int. Dairy J. 12, 869–877.
Lankas, G.R., Cartwright, M.E., Umbenhauer, D., 1997. P-glycoprotein deficiency in a Malan, F., Reinecke, R., 1979. Anthelmintic efficiency of fenbendazole in equines. J. S.
subpopulation of CF-1 mice enhances avermectin-induced neurotoxicity. Toxicol. Afr. Vet. Assoc. 50, 255–258.
Appl. Pharmacol. 143, 357–365. Mancebo, O.A., Verdi, J.H., Bulman, G.M., 1997. Comparative efficacy of moxidectin 2%
Lanusse, C.E., Prichard, R.K., 1990. Pharmacokinetic behavior of netobimin and its me- equine oral gel and ivermectin 2% equine oral paste against Onchocerca cervicalis
tabolites in sheep. J. Vet. Pharmacol. Ther. 13, 170–178. (Railliet and Henry, 1910. microfilariae in horses with naturally acquired infections
Lanusse, C.E., Prichard, R.K., 1992a. Effects of methimazole on the kinetics of netobimin in Formosa (Argentina). Vet. Parasitol. 73, 243–248.
Metabolites in Cattle. Xenobiotica 22, 115–123. Mantovani, A., Ricciardi, C., Stazi, A.V., Macri, C., 1995. Effects observed on gestational
Lanusse, C.E., Prichard, R.K., 1992b. Methimazole increases the plasma-concentrations of day-13 in rat embryos exposed to albendazole. Reprod. Toxicol. 9, 265–273.
the albendazole metabolites of netobimin in sheep. Biopharm. Drug Dispos. 13, Marriner, S., Bogan, J., 1981a. Anthelmintic efficacy. Vet. Rec. 109, 477–478.
95–103. Marriner, S.E., Bogan, J.A., 1981b. Pharmacokinetics of fenbendazole in sheep. Am. J.
Lanusse, C.E., Prichard, R.K., 1993. Relationship between pharmacological properties and Vet. Res. 42, 1146–1148.
clinical efficacy of ruminant anthelmintics. Vet. Parasitol. 49, 123–158. Marriner, S.E., Bogan, J.A., 1985. Plasma concentrations of fenbendazole and oxfendazole
Lanusse, C.E., Gascon, L.H., Prichard, R.K., 1993. Gastrointestinal distribution of alben- in the horse. Equine Vet. J. 17, 58–61.
dazole metabolites following netobimin administration to cattle - relationship with Marriner, S.E., Evans, E.S., Bogan, J.A., 1985. Effect of parasitism with Ostertagia cir-
plasma disposition kinetics. J. Vet. Pharmacol. Ther. 16, 38–47. cumcincta on pharmacokinetics of fenbendazole in sheep. Vet. Parasitol. 17, 239–249.
Lanusse, C.E., Gascon, L.H., Prichard, R.K., 1995. Influence on the antithyroid compound Marriner, S.E., Mckinnon, I., Bogan, J.A., 1987. The pharmacokinetics of ivermectin after

50
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

oral and subcutaneous administration to sheep and horses. J. Vet. Pharmacol. Ther. Nawalinski, T., Theodorides, V., 1977. Efficacy of three forms of oxibendazole against
10, 175–179. gastrointestinal parasites of ponies [Nematodes]. Vet. Med. Small Anim. Clin. 72,
Martin, R.J., 1982. Electro physiological effects of piperazine and diethyl carbamazine on 96–101.
Ascaris suum somatic muscle. Br. J. Pharmacol. 77, 255–265. Ngomuo, A.J., 1983. Pharmacokinetic Studies of Fenbendazole and Oxfendazole in Cattle.
Martin, R.J., 1985. Gamma-aminobutyric acid and piperazine activated single-channel M.V.M., Thesis. University of Glasgow, UK.
currents from Ascaris suum body muscle. Br. J. Pharmacol. 84, 445–461. Ngomuo, A.J., Marriner, S.E., Bogan, J.A., 1984. The pharmacokinetics of fenbendazole
Martin, R.J., 1996. An electrophysiological preparation of Ascaris suum pharyngeal and oxfendazole in cattle. Vet. Res. Commun. 8, 187–193.
muscle reveals a glutamate-gated chloride channel sensitive to the avermectin ana- Norton, S., De Beer, E.J., 1957. Investigations on the action of piperazine on Acsacris
logue. Milbemycin D. Parasitol. 112, 247–252. Lumbricoides. Am. J. Trop. Med. Hyg. 6, 898–905.
Martin, R.J., 1997. Modes of action of anthelmintic drugs. Vet. J. 154, 11–34. Osman, S.A., Hanafy, A., Amer, S.E., 2006. Clinical and therapeutic studies on mange in
Martin, R.J., Pennington, A.J., 1989. Ivermectin induces cation channels in isolated horses. Vet. Parasitol. 141, 191–195.
patches of Ascaris muscle. J. Physiol.-Lond. 410 P83-P83. Owen, J., Slocombe, D., 2004. A modified critical test for the efficacy of pyrantel pamoate
Martin, R.J., Robertson, A.P., 2007. Mode of action of levamisole and pyrantel, for Anoplocephala perfoliata in equids. Can. J. Vet. Res. 68, 112.
Anthelmintic Resistance, E153 and Q57. Parasitol. 134, 1093–1104. Perez, R., Cabezas, I., Garcia, M., Rubilar, L., Sutra, J.F., Galtier, P., Alvinerie, M., 1999.
Matthews, N.S., Taylor, T., Hartsfield, S.M., 1997. Anaesthesia of donkeys and mules. Comparison of the pharmacokinetics of moxidectin (Equest (R)) and ivermectin
Equine Vet. Educ. 9, 198–202. (Eqvalan (R)) in horses. J. Vet. Pharmacol. Ther. 22, 174–180.
McDougall, J., Cameron, B., Lichtenwalner, D., 1985. The metabolism and pharmacoki- Perez, R., Godoy, C., Palma, C., Cabezas, I., Munoz, L., Rubilar, L., Arboix, M., Alvinerie,
netics of a novel anthelmintic (sch. 32481). Communication to the 3rd Conference of M., 2003. Plasma profiles of ivermectin in horses following oral or intramuscular
the European Association for Veterinary Pharmacology and Toxicology. administration. J. Vet. Med. A Physiol. Pathol. Clin. Med. 50, 297–302.
McKellar, Q.A., 1997a. Ecotoxicology and residues of anthelmintic compounds. Vet. Perez, R., Godoy, C., Palma, C., Munoz, L., Arboix, M., Alvinerie, M., 2010. Plasma dis-
Parasitol. 72, 413–426. position and fecal elimination of doramectin after oral or intramuscular adminis-
McKellar, Q.A., 1997. The use and optimization of anthelmintics in sustainable control of tration in horses. Vet. Parasitol. 170, 112–119.
internal parasites in ruminants ed. GK Barell, Toltec Print, Chistchurch, pp 107-128. Plummer, C.E., Kallberg, M.E., Ollivier, F.J., Brooks, D.E., Gelatt, K.N., 2006. Suspected
McKellar, Q.A., Benchaoui, H.A., 1996. Avermectins and milbemycins. J. Vet. Pharmacol. ivermectin toxicosis in a miniature mule foal causing blindness. Vet. Ophthalmol. 9,
Ther. 19, 331–351. 29–32.
McKellar, Q.A., Gokbulut, C., 2012. Pharmacokinetic features of the antiparasitic mac- Pong, S.S., Wang, C.C., Fritz, L.C., 1980. Studies on the mechanism of action of aver-
rocyclic lactones. Curr. Pharm. Biotechnol. 13, 888–911. mectin b1a - stimulation of release of gamma-aminobutyric acid from brain sy-
McKellar, Q., Marriner, S., 1987. Comparison of the anthelmintic efficacy of oxfendazole naptosomes. J. Neurochem. 34, 351–358.
or ivermectin administered orally and ivermectin administered subcutaneously to Pouliot, J.F., LHeureux, F., Liu, Z., Prichard, R.K., Georges, E., 1997. Reversal of P-gly-
sheep during the periparturient period. Vet. Rec. 120, 383–386. coprotein-associated multidrug resistance by ivermectin. Biochem. Pharmacol. 53,
McKellar, Q.A., Scott, E.W., 1990. The benzimidazole anthelmintic agents - a review. J. 17–25.
Vet. Pharmacol. Ther. 13, 223–247. Prichard, R.K., 1978. Anthelmintics. Vol. 1, Refresher Course for Veterinarians. The
McKellar, Q., Bogan, J., Horspool, L., Reid, K., 1988. Effect of ivermectin on the re- University of Sydney Postgraduate Committee in Veterinary Science, Sydney, City.
productive potential of Cooperia curticei. Vet. Rec. 122 444-444. pp. 421–463.
McKellar, Q.A., Mostofa, M., Eckersall, P.D., 1990. Effect of Ostertagia ostertagi secretions Prokulewicz, A., Pilarczyk, B., Tomza-Marciniak, A., 2014. Evaluation of the efficacy of
and various putative secretagogues and inhibitors on aminopyrine accumulation in doramectin in the control of Strongyle (Strongylidae, cyathostominae) Infestation in
dispersed bovine abomasal gland-cells. Res. Vet. Sci. 49, 323–326. Horses. Isr. J. Vet. Med. 69, 83–87.
McKellar, Q.A., Jackson, F., Coop, R.L., Jackson, E., Scott, E., 1991. Effect of parasitism Reinemeyer, C.R., Prado, J.C., Nichols, E.C., Marchiondo, A.A., 2010a. Efficacy of pyr-
with Nematodirus battus on the pharmacokinetics of levamisole, ivermectin and ne- antel pamoate against a macrocyclic lactone-resistant isolate of Parascaris equorum in
tobimin. Vet. Parasitol. 39, 123–136. horses. Vet. Parasitol. 171, 111–115.
McKellar, Q.A., Galbraith, E.A., Baxter, P., 1993a. Oral absorption and bioavailability of Reinemeyer, C.R., Prado, J.C., Nichols, E.C., Marchiondo, A.A., 2010b. Efficacy of pyr-
fenbendazole in the dog and the effect of concurrent ingestion of food. J. Vet. antel pamoate and ivermectin paste formulations against naturally acquired Oxyuris
Pharmacol. Ther. 16, 189–198. equi infections in horses. Vet. Parasitol. 171, 106–110.
McKellar, Q.A., Jackson, F., Coop, R.L., Baggot, J.D., 1993b. Plasma profiles of alben- Reinemeyer, C.R., Prado, J.C., Andersen, U.V., Nielsen, M.K., Schricker, B., Kennedy, T.,
dazole metabolites after administration of netobimin and albendazole in sheep - ef- 2014. Effects of daily pyrantel tartrate on strongylid population dynamics and per-
fects of parasitism and age. Br. Vet. J. 149, 101–113. formance parameters of young horses repeatedly infected with cyathostomins and
McKellar, Q.A., Gokbulut, C., Muzandu, K., Benchaoui, H., 2002. Fenbendazole phar- Strongylus vulgaris. Vet. Parasitol. 204, 229–237.
macokinetics, metabolism, and potentiation in horses. Drug Metab. Dispos. 30, Reinemeyer, C.R., Prado, J.C., Nielsen, M.K., 2015. Comparison of the larvicidal efficacies
1230–1239. of moxidectin or a five-day regimen of fenbendazole in horses harboring cyathos-
Mckissick, G.E., Sutherland, I.H., Foix, J., Olson, G., 1987. The safety of ivermectin ad- tomin populations resistant to the adulticidal dosage of fenbendazole. Vet. Parasitol.
ministered orally to pregnant mares. J. Equine Vet. Sci. 7, 357–367. 214, 100–107.
McTigue, M.A., Williams, D.R., Trainer, J.A., 1995. Crystal structures of a schistosomal Relf, V.E., Lester, H.E., Morgan, E.R., Hodgkinson, J.E., Matthews, J.B., 2014.
drug and vaccine target: gluthatione S- transferase from Schistosoma japonica and its Anthelmintic efficacy on UK Thoroughbred stud farms. Int. J. Parasitol. 44, 507–514.
complex with the leading antischistosomal drug praziquantel. J. Mol. Biol. 246, Ryan, W., Best, P., 1985. Efficacy of ivermectin paste against Strongyloides westeri in foals.
21–27. Vet. Rec. 117, 169–170.
Mellin, T.N., Busch, R.D., Wang, C.C., 1983. Postsynaptic inhibition of invertebrate Sanchez, S.F., Alvarez, L.I., Lanusse, C.E., 1997. Fasting-induced changes to the phar-
neuromuscular transmission by avermectin B1a. Neuropharmacology 22, 89–96. macokinetic behaviour of albendazole and its metabolites in calves. J. Vet.
Meuldermans, W.E.G., Hurkmans, R.M.A., Lauwers, W.F.J., Heykants, J.J.P., 1976. Thein Pharmacol. Ther. 20, 38–47.
vitro metabolism of mebendazole by pig, rat and dog liver fractions. Eur. J. Drug Sangster, N.C., Prichard, R.K., Lacey, E., 1985. Tubulin and benzimidazole-resistance in
Metab. Pharmacokinet. 1, 35–40. Trichostrongylus colubriformis (Nematoda). Parasitology 71, 645.
Miller, M., 1980. Use of levamisole in parasitic infections. Drugs 20, 122–130. Saumell, C., Lifschitz, A., Baroni, R., Fuse, L., Bistoletti, M., Sagues, F., Bruno, S., Alvarez,
Mirck, M.H., 1985. Chemotherapy of Gastrointestinal Nematodiasis in Equines G., Lanusse, C., Alvarez, L., 2017. The route of administration drastically affects
Chemotherapy of Gastrointestinal Helminths. Springer, Berlin Heidelberg, pp. ivermectin activity against small strongyles in horses. Vet. Parasitol. 236, 62–67.
443–462. Schinkel, A.H., Smit, J.J.M., van Tellingen, O., Beijnen, J.H., Wagenaar, E., van Deemter,
Mirck, M.H., Vanmeurs, G.K., 1982. Effect of albendazole and cambendazole on the egg L., Mol, C.A.A.M., van der Valk, M.A., Robanus-Maandag, E.C., te Riele, H.P.J., Berns,
output of strongylidae in horses. Tijdschr. 107, 731–736. A.J.M., Borst, P., 1994. Disruption of the mouse mdr1a P-glycoprotein gene leads to a
Mohammed-Ali, N.A.K., Bogan, J.A., 1987. The phamacodynamics of the flukicidal sali- deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell 77,
cylanides, rafoxanide, closantel and oxyclosanide. J. Vet. Pharmacol. Ther. 10, 491–502.
127–133. Schinkel, A.H., Wagenaar, E., Mol, C.A., van Deemter, L., 1996. P-glycoprotein in the
Moroni, P., Buronfosse, T., Longinsauvageon, C., Delatour, P., Benoit, E., 1995. Chiral blood-brain barrier of mice influences the brain penetration and pharmacological
sulfoxidation of albendazole by the flavin adenine dinucleotide-containing and cy- activity of many drugs. J. Clin. Invest. 97, 2517–2524.
tochrome p450-dependent monooxygenases from rat-liver microsomes. Drug Metab. Scott, E.W., 1988. Effects of Benzimidazole Anthelmintics on Nematode Parasites of
Dispos. 23, 160–165. Sheep. Ph.D. Thesis. University of Glasgow.
Mostofa, M., McKellar, Q.A., 1989. Effect of an Antimuscarinic drug on the plasma Scott, E.W., 1997. Pharmacokinetics of Ivermectin in Donkeys and Ponies 15. Association
pepsinogen activity of sheep infected with Ostertagia circumcincta. Res. Vet. Sci. 47, for Veterinary Clinical Pharmacology and Therapeutics Proceedings, Edinburgh, UK,
208–211. pp. 20–21.
Mrozik, H., Friedman, J., Schwartzkopf, G., Schardt, R., Patchett, A.A., Hoff, D.R., Yakstis, Scott, E.W., McKellar, Q.A., 1992. The distribution and some pharmacokinetic parameters
J.J., Riek, R.F., Ostlind, D.A., Plishker, G.A., Butler, R.W., Cuckler, D.C., Campbell, of ivermectin in pigs. Vet. Res. Commun. 16, 139–146.
W.C., 1969. A new agent for the treatment of liver fluke infection (fascioliasis). Seiler, J.P., 1976. The mutagenicity of benzimidazole and benzimidazole derivatives. VI.
Experientia 25 883-883. Cytogenetic effects of benzimidazole derivatives in the bone marrow of the mouse
Munst, G.J., Karlaganis, G., Bircher, J., 1980. Plasma-concentrations of mebendazole and the chinese hamster. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 40, 339–347.
during treatment of Echinococcosis - preliminary-results. Eur. J. Clin. Pharmacol. 17, Sekkin, S., Gokbulut, C., Kum, C., Karademir, U., 2012. Plasma disposition of enrofloxacin
375–378. following intravenous and intramuscular administration in donkeys. Vet. Rec. 171
Napoli, E., Gaglio, G., Falsone, L., Ferrara, M.C., Brianti, E., Giannetto, S., 2013. Use of 447-447.
eprinomectin and pyrantel pamoate in donkey: evaluation of anthelmintic efficacy. Sheehan, D., Sheehan, S., Marchiondo, A., 2016. Discovery and Chemistry of Pyrantel,
Large Anim. Rev. 19, 123–126. Morantel and Oxantel Pyrantel Parasiticide Therapy in Humans and Domestic

51
C. Gokbulut, Q.A. McKellar Veterinary Parasitology 261 (2018) 27–52

Animals. Elsevier, pp. 1–19. diseases. Parasitol. Today (Regul. Ed.) 4, 4–10.
Sheriff, J.C., Kotze, A.C., Sangster, N.C., Martin, R.J., 2002. Effects of macrocyclic lactone Theodorides, V.J., 1985. Antiparasitic drugs. In: Georgi, J.R. (Ed.), Parasitology for
anthelmintics on feeding and pharyngeal pumping in Trichostrongylus colubriformis in Veterinarians. W.B. Saunders Company, Philadelphia, USA, pp. 187–227.
vitro. Parasitol. 125, 477–484. Toutain, P.L., Upson, D.W., Terhune, T.N., McKenzie, M.E., 1997. Comparative phar-
Shoop, W.L., Mrozik, H., Fisher, M.H., 1995. Structure and activity of avermectins and macokinetics of doramectin and ivermectin in cattle. Vet. Parasitol. 72, 3–8.
milbemycins in animal health. Vet. Parasitol. 59, 139–156. Trawford, A.F., Tremlett, J.G., 1996. Efficacy of triclabendazole against Fasciola hepatica
Shoop, W.L., Demontigny, P., Fink, D.W., Williams, J.B., Egerton, J.R., Mrozik, H., Fisher, in the donkey (Equus asinus). Vet. Rec. 139, 142–143.
M.H., Skelly, B.J., Turner, M.J., 1996a. Efficacy in sheep and pharmacokinetics in Tsuchiya, T., Tanaka, A., Fukuoka, M., Sato, M., Yamaha, T., 1987. Metabolism of thia-
cattle that led to the selection of Eprinomectin as a topical endectocide for cattle. Int. bendazole and teratogenic potential of its metabolites in pregnant mice. Chem.
J. Parasitol. 26, 1227–1235. Pharm. Bull. 35, 2985–2993.
Shoop, W.L., Egerton, J.R., Eary, C.H., Haines, H.W., Michael, B.F., Mrozik, H., Eskola, P., Turner, M.J., Schaeffer, J.M., 1989. Mode of Action of Ivermectin Ivermectin and
Fisher, M.H., Slayton, L., Ostlind, D.A., Skelly, B.J., Fulton, R.K., Barth, D., Costa, S., Abamectin. Springer New York, pp. 73–88.
Gregory, L.M., Campbell, W.C., Seward, R.L., Turner, M.J., 1996b. Eprinomectin: a Tzelos, T., Barbeito, J.S.G., Nielsen, M.K., Morgan, E.R., Hodgkinson, J.E., Matthews, J.B.,
novel avermectin for use as a topical endectocide for cattle. Int. J. Parasitol. 26, 2017. Strongyle egg reappearance period after moxidectin treatment and its re-
1237–1242. lationship with management factors in UK equine populations. Vet. Parasitol. 237,
Slocombe, J.O.D., de Gannes, R.V.G., 2006. Cyathostomes in horses in Canada resistant to 70–76.
pyrantel salts and effectively removed by moxidectin. Vet. Parasitol. 140, 181–184. Uhlinger, C., Johnston, C., Fetrow, J., 1986. A field evaluation of oxibendazole in horses
Slocombe, J.O.D., Lake, M.C., 2007. Efficacy of Daily pyrantel tartrate (Strongid C. infected with benzimidazole resistant small strongyles. J. Equine Vet. Sci. 6, 11–14.
Against strongyles in ponies on pasture. J. Equine Vet. Sci. 27, 439–445. Ural, K., Ulutas, B., Kar, S., 2008. Eprinomectin treatment of psoroptic mange in hunter
Slocombe, J.O.D., Mccraw, B.M., 1984. Evaluation of ivermectin against later 4th-stage jumper and dressage horses: a prospective, randomized, double-blinded, placebo-
Strongylus vulgaris in ponies at 2 and 5 weeks after treatment. Can. J. Comp. Med. 48, controlled clinical trial. Vet. Parasitol. 156, 353–357.
343–348. Valdez, R.A., DiPietro, J.A., Paul, A.J., Lock, T.F., Hungerford, L.L., Todd, K.S., 1995.
Slocombe, J., Smart, J., 1975. Evaluation of pyrantel pamoate against strongyles in Controlled efficacy study of the bioequivalence of Strongid® C and generic pyrantel
horses. Can. Vet. J. 16, 310. tartrate in horses. Vet. Parasitol. 60, 83–102.
Slocombe, J.O.D., Heine, J., Barutzki, D., Slacek, B., 2007. Clinical trials of efficacy of Vandenbossche, H., Rochette, F., Horig, C., 1982. Mebendazole and related anthelmintics.
praziquantel horse paste 9% against tapeworms and its safety in horses. Vet. Adv. Pharmacol. Chemother. 19, 67–128.
Parasitol. 144, 366–370. Veneziano, V., Di Loria, A., Masucci, R., Di Palo, R., Brianti, E., Gokbulut, C., 2011.
Souhailielamri, H., Fargetton, X., Benoit, E., Totis, M., Batt, A.M., 1988. Inducing effect of Efficacy of eprinomectin pour-on against Dictyocaulus arnfieldi infection in donkeys
albendazole on rat-liver drug-metabolizing-enzymes and metabolite pharmacoki- (Equus asinus). Vet. J. 190, 414–415.
netics. Toxicol. Appl. Pharmacol. 92, 141–149. Vercruysse, J., 1993. Doramectin - a novel avermectin. Vet. Parasitol. 49, 1–3.
Squires, E.L., Amann, R.P., Pickett, B.W., Berndtson, W.E., Shideler, R.K., Voss, J.L., 1978. Warner, A., 1981. Rate of passage of digesta through the gut of mammals and birds. Nutr.
Effect of fenbendazole on reproductive function in stallions. Theriogenology 9, Abstr. Rev. 51, 789–820.
447–455. Wilkins, C., Conroy, B., Ho, P., O’Shanny, W., Capizzi, T., 1981. Effect of ivermectin on
Stoffman, E.E., Braithwaite, A., 1976. Piperazine overdoses in a kitten. Can. Vet. J. 17, the live mass period of attachment and percent control of ticks. Tick biology and
140–141. control: proceedings of an International Conference January 27–29, 1981/edited by
Stratford, C.H., Lester, H.E., Pickles, K.J., McGorum, B.C., Matthews, J.B., 2014. An in- GB Whitehead and JD Gibson.: Tick Research Unit, Rhodes University, Grahamstown,
vestigation of anthelmintic efficacy against strongyles on equine yards in Scotland. South Africa.
Equine Vet. J. 46, 17–24. Wilkinson, C., Murray, M., Marcus, C., 1984. Interactions of methylenedioxyphenyl
Styles, J.A., Garner, R., 1974. Benzimidazolecarbamate methyl ester-evaluation of its compounds with cytochrome P-450 and effects on microsomal oxidation. Rev.
effects in vivo and in vitro. Mutat. Res. Fund. Mol. M. 26, 177–187. Biochem. Toxicol. 6, 27–63.
Swor, T.M., Whittenburg, J.L., Chaffin, M.K., 2009. Ivermectin toxicosis in three adult Wolmstedt, B., Nordgrenm, I., Sandox, M., Sundwall, A., 1978. Metrifonate, Summary of
horses. J. Am. Vet. Med. Assoc. 235, 558–562. toxicological and pharmacological information available. Arch. Toxicol. 41, 3–29.
Tahir, M.S., Holroyd, G., Copeman, D.B., 1986. Treatment of beef calves with ivermectin Xiao, L., Herd, R.P., Majewski, G.A., 1994. Comparative efficacy of moxidectin and
and avermectin B 1 in dry tropical Australia. In 6th International Congress of ivermectin against hypobiotic and encysted cyathostomes and other equine parasites.
Parasitology 651 Abstract No. 651. Vet. Parasitol. 53, 83–90.
Takiguchi, Y.O., Mishima, H., Okuda, M., Terao, M., Aoki, A., Fukuda, R., 1980. Zeng, Z.P., Andrew, N.W., 1999. Characterization of eprinomectin N-deacetylase in rats.
Milbemycins, a new family of macrolide antibiotics: fermentation, isolation and Drug Metab. Dispos. 27, 269–273.
physico-chemical properties. J. Antibiot. 33, 1120–1127. Zeng, Z., Andrew, N.W., Halley, B.A., 1997. Identification of cytochrome P4503A as the
Taylor, T.S., Craig, T., 1993. Lungworms in donkeys: evaluation of anthelmintics under major enzyme sub-family responsible for the metabolism of 22,23-dihydro-13-O-[(2-
field conditions. J. Equine Vet. Sci. 13, 150–152. methoxyethoxy)methyl]-avermectin B-1 aglycone by rat liver microsomes.
Taylor, S., Mallon, T., Blanchflower, W., Kennedy, D., Green, W., 1992. Effects of diet on Xenobiotica 27, 985–994.
plasma concentrations of oral anthelmintics for cattle and sheep. Vet. Rec. 130, Zeng, Z., Andrew, N.W., Arison, B.H., Luffer-Atlas, D., Wang, R.W., 1998. Identification of
264–268. cytochrome P4503A4 as the major enzyme responsible for the metabolism of iver-
Tejada, P., Sanchez-Moreno, M., Monteoliva, M., Gomez-Banqueri, H., 1987. Inhibition of mectin by human liver microsomes. Xenobiotica 28, 313–321.
malate dehydrogenase enzymes by benzimidazole anthelmintics. Vet. Parasitol. 24, Zulalian, J., Stout, S.J., Dacunha, A.R., Garces, T., Miller, P., 1994. Absorption, tissue
269–274. distribution, metabolism, and excretion of moxidectin in cattle. J. Agric. Food Chem.
Tekwanl, B.L., Shukla, O.P., Ghatak, S., 1988. Altered drug metabolism in parasitic 42, 381–387.

52

Вам также может понравиться