You are on page 1of 7

The

Veterinary Journal
The Veterinary Journal 170 (2005) 339–345
www.elsevier.com/locate/tvjl

Clindamycin bioavailability and pharmacokinetics following


oral administration of clindamycin hydrochloride capsules in dogs
a,*
Georgios C. Batzias , Georgios A. Delis a, Labrina V. Athanasiou b

a
Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
b
Veterin S.A., Aspropyrgos Attikis, Greece

Accepted 27 June 2004

Abstract

Oral bioavailability and pharmacokinetic behaviour of clindamycin in dogs was investigated following intravenous (IV) and oral
(capsules) administration of clindamycin hydrochloride, at the dose of 11 mg/kg BW. The absorption after oral administration was
fast, with a mean absorption time (MAT) of 0.87 ± 0.40 h, and bioavailability was 72.55 ± 9.86%. Total clearance (CL) of clinda-
mycin was low, after both IV and oral administration (0.503 ± 0.095 vs. 0.458 ± 0.087 L/h/kg). Volume of distribution at steady-
state (IV) was 2.48 ± 0.48 L/kg, indicating a wide distribution of clindamycin in body fluids and tissues. Elimination half-lives were
similar for both routes of administration (4.37 ± 1.20 h for IV, vs. 4.37 ± 0.73 h for oral).
Serum clindamycin concentrations following administration of capsules remained above the MICs of very susceptible microor-
ganisms (0.04–0.5 lg/mL) for 12 or 10 h, respectively. Time above the mean inhibitory concentration (MIC) is considered as the
index predicting the efficacy of clindamycin (T>MIC must be at least 40–50% of the dosing interval), so a once-daily oral adminis-
tration of 11 mg/kg BW of clindamycin can be considered therapeutically effective. For less susceptible bacteria (with MICs of 0.5–2
lg/mL) the same dose should be given but twice daily.
 2004 Elsevier Ltd. All rights reserved.

Keywords: Clindamycin; Capsules; Bioavailability; Pharmacokinetics; Dogs

1. Introduction of a hydroxyl group by chlorine at the C7 position led to


the formation of clindamycin.
Clindamycin is a semi-synthetic lincosamide that The new molecule has proved to be superior in terms
occupies a prominent place among the antimicrobials of antimicrobial activity against Gram+ aerobes (cocci
used against staphylococcal, anaerobic and some proto- and bacteria) and anaerobic bacteria with mean inhibi-
zoal infections in small animals (Prescott, 2000) and hu- tory concentrations (MICs) ranging generally from
mans (Lewis et al., 1999; Pfefferkorn and Borotz, 1994). 0.04 to 0.5 lg/mL for susceptible strains (Lamp et al.,
Lincomycin, the parent compound, a natural fermenta- 2002; Pankuch et al., 1993; Schaumann et al., 2000).
tion product of Streptomyces spp., is a monoglycoside Some protozoa, such as Toxoplasma gondii (Pfefferkorn
with an amino-like side chain. The synthetic substitution and Borotz, 1994) and Neospora caninum (Lindsay et al.,
1994) are also considered susceptible.
The metabolism of clindamycin (at least in humans, as
reported by Wynalda et al., 2003) includes hepatic bio-
transformation, mediated by cytochrome P450 enzymes
*
Corresponding author. Tel./fax: +30 2310999855. (primary CYP3A) towards the bioactive metabolites
E-mail address: batzias@vet.auth.gr (G.C. Batzias). clindamycin sulphoxide and N-desmethylclindamycin.

1090-0233/$ - see front matter  2004 Elsevier Ltd. All rights reserved.
doi:10.1016/j.tvjl.2004.06.007
340 G.C. Batzias et al. / The Veterinary Journal 170 (2005) 339–345

The parent compound and metabolites in humans are 2. Materials and methods
mainly excreted in faeces and, to a less extent, urine.
Clindamycin has usually been regarded primarily as 2.1. Experimental animals
a bacteriostatic agent (Dow, 1988). However, recent
studies have cast some doubt on this and reports sug- Six (n = 6) healthy, male, Beagle dogs weighing 12–17
gest that it displays concentration-independent bacteri- kg and aged 12 months, were used in this study. All dogs
cidal activity (Craig and Gudmundsson, 1996; Klepser were acclimatized in the experimental facilities for a per-
et al., 1997; Lewis et al., 1999) while exhibiting an in iod of two months before the onset of the experiment.
vitro post antibiotic effect (PAE) of 0.4–3.9 h, for Sta- They were fed on a commercial dry ration and had free
phylococcus aureus (Xue et al., 1996), and 2 h for access to drinking water, although they were fasted be-
Bacillus anthracis (Athamna et al., 2004). In vivo, fore drug administrations. Health condition was moni-
the PAE is generally longer and due to effects such tored continuously by clinical and laboratory
as post antibiotic leukocyte enhancement and post examinations. Body weight determination was con-
antibiotic sub-MIC effect (Xue et al., 1996). Vogelman ducted before drug administration.
et al. (1988) reported an in vivo PAE of clindamycin The in vivo experimental phase was carried out with
against S. aureus, in the neutropenic mouse thigh permission from the local bureau of the Hellenic Minis-
model, of 7.1 h. try of Agriculture (Veterinary Service) and in accord-
At the molecular level, clindamycin is a classical ance to EU directives (86/609/European Council) and
inhibitor of bacterial protein synthesis, by binding to state law (1197/81 and 2015/92).
the 23S ribosomal RNA of the 50S ribosomal subunit
(Prescott, 2000). However, it seems that the target site 2.2. Drugs
of clindamycin in Toxoplasma gondii is not the ribo-
somal RNA itself, but rather another rRNA molecule, Clindamycin hydrochloride was dissolved in phos-
encoded by a 35-kb circular extrachromosomal genome phate-buffered saline (pH 7.2) to obtain a 15% (w/v)
(Pfefferkorn and Borotz, 1994). clindamycin hydrochloride injectable solution. This
Clindamycin in companion animal medicine is mainly solution, sterilized through a 0.22 lm pore-size filter
prescribed for the treatment of superficial or deep pyo- (Millex-GV, Millipore S.A.), was used for the IV admin-
derma, primarily caused by Staphylococcus intermedius istration. Standard clindamycin hydrochloride CRS
(Bloom and Rosser, 2001; Harvey et al., 1993; Little- (86.8% clindamycin base) was purchased from European
wood et al., 1999; Scott et al., 1998), osteomyelitis (Bra- Pharmacopoeia. Commercially available clindamycin
den et al., 1987), infections of the oral cavity (Sarkiala hydrochloride capsules (Antirobe caps, 150 mg, Phar-
and Harvey, 1993) and other anaerobic infections (Berg macia and Upjohn) were used for oral administration.
et al., 1984; Boothe, 1990). Clindamycin has also been Eight capsules were analyzed using high performance
successfully used against toxoplasmosis (Prescott, liquid chromatography (HPLC) to confirm clindamycin
2000) and neosporosis (Ordeix et al., 2002). Clindamy- content, which was found to be in accordance with the
cin hydrochloride is commercially available in formula- value given by the manufacturer.
tions for oral administration (capsules, tablets and oral
solution), whereas injectable formulations containing 2.3. In vivo experimental phase
the phosphate ester, available for human medicine, are
also used (Bishop, 2001). The study involved IV as well as oral administration
The pharmacokinetics of clindamycin in the dog has of clindamycin hydrochloride to all dogs, according to a
been studied (Budsberg et al., 1992; Lavy et al., 1999) two-treatment, two-period design, using a four week
after intramuscular (IM) and subcutaneous (SC) admin- wash-out interval between the two periods. All animals
istration (Lavy et al., 1999). Two further studies (Brown were kept fasted for 12 h before each drug
et al., 1989, 1990) reported serum and tissue disposition administration.
of clindamycin in cats after oral administration of an
oral solution. However, despite considerable clinical 2.3.1. Period 1: intravenous administration
experience, which has established clindamycin among For the first experimental period, 11 mg/kg BW clin-
the most widely used antimicrobial agents, to the damycin hydrochloride were administered IV to all ani-
authorsÕ knowledge, only scarce data exist concerning mals (Day 0), after catheterisation of the left cephalic
the pharmacokinetics of clindamycin after oral adminis- vein. The catheters (18 G · 51 mm Abbocath-T, Abbott)
tration in the dog (Lavy et al., 2000). The objective of were removed shortly after administration of the drug.
the present study was to examine the pharmacokinetic Blood samples (3 mL) were collected into plastic
profile of clindamycin in dogs after oral administration tubes (S-Monovette, Sarstedt) by aspiration from the
of clindamycin hydrochloride capsules at the dose of catheterized lateral cephalic vein, prior to (t = 0 h) and
11 mg/kg BW. at 2, 5, 10, 15, 20, 30, 45 min, 1, 1.5, 2, 2.5, 3, 4, 5, 6,
G.C. Batzias et al. / The Veterinary Journal 170 (2005) 339–345 341

8, 10, 12 and 24 h after administration. The intravenous oral administration, with the aid of WinNonlin soft-
catheters were flushed with 2 mL 1% heparinized normal ware, version 4.0.1 (Pharsight Corp).
saline after each sampling. The Area Under the Concentration–time curve from
time zero to the last sampling point that yielded a quan-
2.3.2. Period 2: oral administration tifiable concentration (AUC0–last) was calculated with
On Day 28, all dogs received one Antirobe capsule the Log/Linear trapezoidal method. The last five sam-
(150 mg clindamycin hydrochloride). Dose normalisa- pling points in each animal were used to determine the
tion from mg/animal to mg/kg BW was carried out in first-order constant associated with the terminal portion
each animal by dividing the total amount of clindamycin of the curve (kz). Terminal half life (t1/2kz) was derived as
received, by its body weight. Blood sample collection t1/2kz = ln(2)/kz. Thus, the AUC from time zero to infin-
was performed, following the technique described ity (AUC0–1) was calculated as AUC0–last + Clast/kz,
above, immediately before (t = 0 h) and at 15, 30, where Clast stands for the last measurable concentration.
45 min, 1, 1.5, 2, 2.5, 3, 4, 5, 6, 8, 10, 12 and 24 h after The Area Under the first Moment Curve from time zero
drug administration. to infinity ðAUMC0–1 ¼ AUMC0–last þ ðtlast  C last =kz Þ
All blood samples were allowed to stand in a dark þðC last =k2z Þ, divided by AUC0–1 provided the Mean
place for 20 min. After centrifugation at 1500g for 10 Residence Time (MRT). The apparent Mean Absorp-
min, at 4 C, the supernatant serum was transferred into tion Time (MAT) was the difference between the MRT
5-mL plastic tubes and was stored at 30 C, pending obtained after oral administration and the MRT after
analysis. IV administration.
Clindamycin oral bioavailability (F) was determined
in each animal as F = AUCoral/AUCIV · DoseIV/
2.4. Analytical experimental phase Doseoral · 100%. Further pharmacokinetic parameters
determined, include total clearance (CL = Dose/
Clindamycin concentrations in serum and capsules AUC0–1), volume of distribution at steady-state
were determined using reversed-phase HPLC with ultra (Vss = CL · MRT) and volume of distribution based
violet (UV) detection (Batzias et al., 2004). Briefly, dog on the terminal phase (Vz = Dose/kz/AUC0–1). CL and
serum (0.5 mL) was deproteinized with acetonitrile (1 Vz after oral administration were initially calculated by
mL) and extracted with dichloromethane (6 mL) under WinNonlin directly, as CL/F and Vz/F, respectively. To
alkaline conditions. A 5-mL aliquot of the organic layer obtain the real CL and Vz, they were corrected by F.
was evaporated and the dried residue was reconstituted
in 0.5 mL of mobile phase. The injection volume was 50 2.6. Statistical analysis
lL. Separation was achieved on a MZ-Analyzentechnik
Spherisorb ODS-2 (250 · 4 mm i.d., 5 lm particle size), Descriptive statistics spreadsheet (included in Win-
C18 reversed-phase analytical column. The mobile phase Nonlin software) provided mean values (±SD) for the
was a mixture of acetonitrile-phosphate buffer (19:81, pharmacokinetic parameters from all animals. Statisti-
v/v), pH 3.5, and contained 2.5 mM tetra-n-butylammo- cal evaluation of the pharmacokinetic parameters was
nium hydrogen sulfate (TBA), as a competing base. performed by using the SPSS software. A non-paramet-
Phosphate salts, TBA and ortho-phosphoric acid were ric Mann–Whitney test was employed to compare mean
all from Merck. Acetonitrile and dichloromethane were values of parameters derived from IV and oral adminis-
from J.T. Baker. Analyses were carried out on a Shim- tration. Means were considered significantly different at
adzu LC-10A series chromatographic system, which P < 0.05.
operated isocratically, at a flow-rate of 1.0 mL/min.
Isocratic elution was carried out at 40 C and UV
detection was performed at 195 nm. The limit of quan- 3. Results
tification (LOQ) was 80 ng/mL. Recovery ranged be-
tween 94.14% and 100.99%. Regression of accuracy Mean serum concentrations (±SD) of clindamycin
data yielded an overall mean recovery (±SEM) of after IV and oral administration are presented in detail
93.98 ± 0.42%, whereas precision values were better in Table 1. Clindamycin levels were below the LOQ of
than 4.41%. the analytical method at 24 h, in all animals. Maximum
concentration (Cmax) after oral administration, achieved
2.5. Pharmacokinetic analysis at 1 h (0.75–1.5 h in individual dogs), was 3.25 (±0.49)
lg/mL serum.
Non-Compartmental Analysis (NCA) based on the Serum concentration–time curves of clindamycin are
Statistical Moment Theory (Gibaldi and Perrier, shown in Fig. 1, whilst selected pharmacokinetic param-
1982b) was applied to determine the pharmacokinetic eters (mean values ± SD), describing drug disposition
parameters for each individual dog, after both IV and are presented in Table 2. During the terminal phase,
342 G.C. Batzias et al. / The Veterinary Journal 170 (2005) 339–345

Table 1 Table 2
Mean serum concentrations (±SD) of clindamycin in six dogs after a Selected pharmacokinetic (PK) parameters of clindamycin after a
single intravenous (IV) and oral (PO) administration of clindamycin single intravenous (IV) and oral (PO) administration of clindamycin
hydrochloride, at the dose of 11 mg/kg BW hydrochloride, at the dose of 11 mg/kg BW
Time after Mean serum PK Units IV PO
administration (h) concentrations (±SD) (lg/mL) parameter administration administration
IV administration PO administration AUC lg · h/mL 22.52 ± 4.30 16.19 ± 2.83
AUMC lg · h2/mL 119.04 ± 55.62 96.17 ± 32.01
0.033 9.40 ± 1.41 –
MRT h 5.10 ± 1.52 5.79 ± 1.05
0.083 6.64 ± 1.22 –
MATapparent h 0.64 ± 0.33
0.167 6.07 ± 1.04 –
MATtrue h 0.87 ± 0.40
0.25 5.94 ± 0.84 0.60 ± 0.56
0.333 5.66 ± 0.75 –
0.5 5.19 ± 0.50 1.89 ± 0.67 kz h1 0.169 ± 0.045 0.163 ± 0.031
0.75 4.52 ± 0.57 2.98 ± 0.40 t1/2kz h 4.37 ± 1.20 4.37 ± 0.73
1 4.10 ± 0.68 3.25 ± 0.49 CL L/h/kg 0.503 ± 0.095 0.458 ± 0.087
1.5 3.46 ± 0.53 3.09 ± 0.41 Vss L/kg 2.48 ± 0.48
2 2.91 ± 0.50 2.59 ± 0.33 Vz L/kg 3.08 ± 0.63 2.84 ± 0.44
2.5 2.48 ± 0.41 2.14 ± 0.33
3 2.14 ± 0.34 1.93 ± 0.31 F % 72.55 ± 9.86
4 1.67 ± 0.31 1.31 ± 0.25
5 1.31 ± 0.34 1.04 ± 0.22
6 0.94 ± 0.21 0.82 ± 0.16
8 0.66 ± 0.23 0.61 ± 0.15 of the first order constant associated with drug absorp-
10 0.51 ± 0.21 0.46 ± 0.14
12 0.41 ± 0.20 0.34 ± 0.12
tion (kabs), assuming first-order absorption process (Gi-
24 <LOQ <LOQ baldi and Perrier, 1982b). However, MAT calculated as
above, does not take into account the ‘‘elimination’’
from the absorption site, mirrored in the value of F.
To obtain the true MAT (1/kabs) in each animal, the
10 iv p.o individual apparent MAT values were divided by the
individual bioavailability factors. The mean value
Clindamycin concentration (µg/mL)

(±SD) calculated was 0.87 ± 0.40 h.

4. Discussion

The wide use of clindamycin in small animal medi-


0.1 cine, the favorable results derived from clinical studies
and the lack of pharmacokinetic data following oral
administration of clindamycin hydrochloride capsules,
provided the motivation for this study.
The rate of clindamycin absorption after oral admin-
0.01
0 2 4 6 8 10 12 14
istration of clindamycin hydrochloride capsules was
Time (h)
quite fast (MATtrue 0.87 ± 0.40 h). The time at which
Cmax was achieved (1 h) was comparable to values ob-
Fig. 1. Serum concentrations of clindamycin after intravenous and tained after SC and IM administration of a buffered
oral administration of clindamycin hydrochloride, at the dose of 11 20% aqueous solution of clindamycin hydrochloride at
mg/kg BW.
the dose of 10 mg/mL (Lavy et al., 1999). In the latter
study, Cmax was remarkably higher in the case of SC
the curves corresponding to the two administration administration (20.8 ± 6.2 lg/mL) than after IM injec-
routes declined in parallel (kz(IV) = 0.169 ± 0.045 h1, tion (4.4 ± 0.5 lg/mL), the last value being closer, but
kz(oral) = 0.163 ± 0.031 h1), yielding similar terminal still higher than our observations (3.25 ± 0.49 lg/mL).
half-lives (t1/2kz(IV) = 4.37 ± 1.20 h, t1/2kz(oral) = 4.37 ± In another study (Budsberg et al., 1992), clindamycin
0.73 h). Based on the similarity of the terminal half-lives, phosphate was administered IM at the dose of 11 mg/
a correction to the value of bioavailability (72.55 ± kg BW and Cmax (5.3 ± 1.0 lg/mL) was achieved at
9.86%), was not deemed necessary (Gibaldi and Perrier, 60.3 ± 23.9 min. Lavy et al. (2000) explored the pharma-
1982a). The apparent mean absorption time (MAT) of cokinetics of clindamycin after multiple oral dosing of
clindamycin (MATapparent = MRToral  MRTIV) was clindamycin hydrochloride gel 3% to dogs, at the dose
0.64 ± 0.33 h. MAT can be considered as the inverse of 11 mg/kg BW, b.i.d. for 10 consecutive days. The
G.C. Batzias et al. / The Veterinary Journal 170 (2005) 339–345 343

study displayed high between-days variability concern- of antimicrobial therapy. In general, MICs of aerobic
ing the values of all pharmacokinetic parameters deter- cocci and bacteria, with good susceptibility, range from
mined. A possible drawback of all of the above studies 0.04 lg/mL, for some susceptible strains of Staphylococ-
is that clindamycin serum concentrations were deter- cus aureus/intermedius to 0.5 lg/mL (Lavy et al., 1999,
mined by use of microbiological methods which, by def- Lamp et al., 2002). MIC50s for susceptible anaerobic
inition, could not discriminate between the parent bacteria generally range from 0.125 to 0.5 lg/mL. Inter-
compound and the bioactive metabolites (clindamycin mediately susceptible microorganisms, such as isolates
sulphoxide and N-desmethylclindamycin). of Fusobacterium spp and Clostridium perfingens, display
Drug absorption after oral administration of clinda- MIC50s as high as 4 lg/mL (Pankuch et al., 1993; Schau-
mycin capsules was significantly less in extent, in respect mann et al., 2000).
to bioavailability values (F = 72.55 ± 9.86%), than that It has been suggested that for time-dependent antimi-
after IM (115 ± 19%) and especially after SC crobial agents, such as clindamycin, serum concentra-
(310 ± 22%) injection of clindamycin hydrochloride tions should remain above the MIC of pathogen
(Lavy et al., 1999). The unfamiliarly high value in the microorganisms for at least 40–50% of the dosing inter-
case of SC administration is somewhat hard to explain. val (Craig, 1998). Clindamycin serum concentrations
Even lower clindamycin bioavailability (55.07–51.48%) after IV and oral administration remain above 0.5 lg/
has been reported in humans after oral administration mL approximately for 10 h (Table 1; Fig. 1).
of clindamycin hydrochloride capsules (Klepser et al., It would appear from our findings that for the most
1997). susceptible microorganisms that display MIC50s <0.5
An extensive distribution of the lipophilic clindamy- lg/mL, (especially Staphylococcus and Bacillus, against
cin has already been reported in other species, such as which a prolonged PAE has been demonstrated), a dos-
man (Panzer et al., 1972) and cats (Brown et al., 1990) age of 11 mg/kg BW PO s.i.d., using clindamycin hydro-
by direct measurement of tissue concentrations. To de- chloride capsules is likely to be therapeutically effective.
scribe drug disposition precisely without interference This dosage has been used empirically in clinical trials
from absorption processes, major pharmacokinetic with dogs suffering from superficial and deep pyoderma
parameters were also obtained after IV injection. The (Bloom and Rosser, 2001; Littlewood et al., 1999; Scott
volume of distribution found in the present study et al., 1998). To confirm our estimation, efficacy scores
(Vss = 2.48 ± 0.48 L/kg) confirmed the wide distribution from these studies were satisfactory (58.6–100%). On
of clindamycin in dogs. However, a major difference the other hand, for less susceptible bacteria (MIC50
exists between this value and the value (0.86 ± 0.35 L/ 0.5–2 lg/mL) we recommend the same dose but to be gi-
kg) provided by Lavy et al. (1999). Our findings con- ven b.i.d.
cerning CL (0.503 ± 0.095 L/h/kg) were also higher Whether the antimicrobial activity of clindamycin
than the value provided by Lavy et al. (1999) bioactive metabolites should be considered when
(6.10 ± 1.10 mL/min/kg, corresponding to 0.366 ± 0.066 performing a PK/PD evaluation has not yet been eluci-
L/h/kg). dated. In humans, the formation of N-desmethylclinda-
MRTIV and t1/2kz(IV) of clindamycin were 5.10 ± 1.52 mycin and its excretion in urine and faeces have been
and 4.37 ± 1.20 h, respectively. Lavy et al. (1999) re- confirmed (Wynalda et al., 2003), but the metabolite
ported significantly lower respective values of could not be detected in the plasma of patients treated
143.00 ± 34.00 min (2.38 ± 0.57 h) and 124.00 ± 57.00 with clindamycin (Gatti et al., 1998). In dogs, no data
min (2.07 ± 0.95 h). It is clear that the differences in exist on metabolite formation and kinetics, and any con-
the values of Vss and CL, which govern drug elimina- tribution to clindamycin efficacy is totally unknown.
tion, had an impact on the calculation of MRTIV and The currently suggested dosage regimens are based on
t1/2kz(IV). The similarity of terminal half lives after IV parent compound concentrations only, and the presence
and oral (4.37 ± 0.73 h) administration of clindamycin of metabolite activity (if any) would only amplify the
hydrochloride strongly indicates the absence of a flip- positive therapeutic outcome. More clinical and phar-
flop effect, since, during the terminal phase, elimination macokinetic/pharmacodynamic (PK/PD) trials (includ-
exclusively limited clindamycin disposition. ing data on metabolite activity) are required to
The reasons for the discrepancy in the major pharma- confirm the optimum clindamycin dose determination
cokinetic parameters between the present study and the in dogs.
study of Lavy et al. (1999) could include the nature of
the analytical methods employed, the different methods
used for pharmacokinetic analysis and the variation in Acknowledgement
the body weights and the breed of the animals (12–17
kg Beagle dogs versus 4–13 kg mixed breed dogs). The authors express their gratitude to Veterin S.A.
The susceptibility of target microorganisms to clinda- for kindly donating the experimental animals used in
mycin is a key factor for the prediction of the outcome this study.
344 G.C. Batzias et al. / The Veterinary Journal 170 (2005) 339–345

References Klepser, M.E., Nicolau, D.P., Quintiliani, R., Nightingale, C.H., 1997.
Bactericidal activity of low-dose clindamycin administered at 8-
Athamna, A., Athamna, M., Medlej, B., Bast, D.J., Rubinstein, E., 2004. and 12-hour intervals against Staphylococcus aureus, Streptococcus
In vitro post-antibiotic effect of fluoroquinolones, macrolides, b- pneumoniae, and Bacteroides fragilis. Antimicrobial Agents and
lactams, tetracyclines, vancomycin, clindamycin, linezolid, chloram- Chemotherapy 41, 630–635.
phenicol, quinupristin/dalfopristin and rifampicin on Bacillus Lamp, K., Lacy, M.K., Freeman, C., 2002. Metronidazole, clindamy-
anthracis. Journal of Antimicrobial Chemotherapy 53, 609–615. cin and streptogramin pharmacodynamics. In: Nightingale, C.H.,
Batzias, G.C., Delis, G.A., Koutsoviti-Papadopoulou, M., 2004. A Murakawa, T., Ambrose, P.G. (Eds.), Antimicrobial Pharmaco-
new HPLC/UV method for the determination of clindamycin in dynamics in Theory and Clinical Practice. Marcel Dekker Inc.,
dog blood serum. Journal of Pharmaceutical and Biomedical New York, pp. 227–231.
Analysis 35, 545–554. Lavy, E., Shem-Tov, M., Orbach, A., 2000. Oral absorption,
Berg, J.N., Scanlan, C.M., Buening, G.M., Fales, W.H., Schmidt, bioavailability and tolerance study of clindamycin gel 3% in dogs.
J.C., Wilson, J.C., 1984. Clinical models for anaerobic bacterial Journal of Veterinary Pharmacology and Therapeutics 23 (suppl. 1,
infections in dogs and their use in testing the efficacy of clindamy- on CD Rom).
cin and lincomycin. American Journal of Veterinary Research 45, Lavy, E., Ziv, G., Shem-Tov, M., Glickman, A., Dey, A., 1999.
1299–1306. Pharmacokinetics of clindamycin HCl administered intravenously,
Bishop, 2001. Drugs used in the treatment of bacterial, fungal, viral, intramuscularly and subcutaneously to dogs. Journal of Veterinary
and protozoal infections. In: The Veterinary Formulary, fifth ed. Pharmacology and Therapeutics 22, 261–265.
Pharmaceutical Press, London, pp. 135–218. Lewis, R.E., Klepser, M.E., Ernst, E.J., Lund, B.C., Biedenbach, D.J.,
Bloom, P.B., Rosser, E.J., 2001. Efficacy of once-daily clindamycin Jones, R.N., 1999. Evaluation of low-dose, extended-interval
hydrochloride in the treatment of superficial bacterial pyoderma in clindamycin regimens against Staphylococcus aureus and Strepto-
dogs. Journal of the American Animal Hospital Association 37, coccus pneumoniae using a dynamic in vitro model of infection.
537–542. Antimicrobial Agents and Chemotherapy 43, 2005–2009.
Boothe, D.M., 1990. Anaerobic infections in small animals. Veterinary Lindsay, D.S., Rippey, N.S., Cole, R.A., Parsons, L.C., Dubey, J.P.,
Medicine 2, 330–347. Tidwell, R.R., Blagburn, B.L., 1994. Examination of the activities of
Braden, T.D., Johnson, C.A., Gabel, C.L., Lott, G.A., Caywood, 43 chemotherapeutic agents against Neospora caninum tachyzoites in
D.D., 1987. Posologic evaluation of clindamycin, using a canine cultured cells. American Journal of Veterinary Research 55, 976–981.
model of posttraumatic osteomyelitis. American Journal of Veter- Littlewood, J.D., Lakhani, K.H., Paterson, S., Wood, J.L.N., Chanter,
inary Research 48, 1101–1105. N., 1999. Clindamycin hydrochloride and clavulanate-amoxycillin
Brown, S.A., Dieringer, T.M., Hunter, R.P., Zaya, M.J., 1989. Oral in the treatment of canine superficial pyoderma. The Veterinary
clindamycin disposition after single and multiple doses in normal Record 144, 662–665.
cats. Journal of Veterinary Pharmacology and Therapeutics 12, Ordeix, L., Lloret, A., Fondevila, D., Dubey, J.B., Ferrer, L., Fondati,
209–216. A., 2002. Cutaneous neosporosis during treatment of pemphigus
Brown, S.A., Zaya, M.J., Dieringer, T.M., Hunter, R.P., Nappier, foliaceus in a dog. Journal of the American Animal Hospital
G.A., Hoffman, G.A., Hornish, R.E., Yein, F.S., 1990. Tissue Association 38, 415–419.
concentrations of clindamycin after multiple oral doses in normal Pankuch, G.A., Jacobs, M.R., Appelbaum, P.C., 1993. Susceptibilities
cats. Journal of Veterinary Pharmacology and Therapeutics 13, of 428 gram-positive and -negative anaerobic bacteria to Bay y3118
270–277. compared with their susceptibilities to ciprofloxacin, clindamycin,
Budsberg, SC, Kemp, D.T., Wolski, N., 1992. Pharmacokinetics of metronidazole, piperacillin, piperacillin-tazobactam, and cefoxitin.
clindamycin phosphate in dogs after single intravenous and Antimicrobial Agents and Chemotherapy 37, 1649–1654.
intramuscular administrations. American Journal of Veterinary Panzer, J.D., Brown, D.C., Epstein, W.L., Lipson, R.L., Mahaffey,
Research 53, 2333–2336. W.H., Atkinson, W.H., 1972. Clindamycin levels in various body
Craig, W.A., 1998. Pharmacokinetic/pharmacodynamic parameters: tissues and fluids. Journal of Clinical Pharmacology-New Drugs
rationale for antibacterial dosing of mice and men. Clinical 12, 259–262.
Infectious Diseases 26, 1–12. Pfefferkorn, E.R., Borotz, S.E., 1994. Comparison of mutants of
Craig, W.A., Gudmundsson, S., 1996. Postantibiotic effect. In: Lorian, Toxoplasma gondii selected for resistance to azithromycin, spira-
V. (Ed.), Antibiotics in Laboratory Medicine, fourth ed. Williams mycin, or clindamycin. Antimicrobial Agents and Chemotherapy
& Wilkins, Baltimore, pp. 296–329. 38, 31–37.
Dow, S.W., 1988. Management of anaerobic infections. The Veteri- Prescott, J.F., 2000. Lincosamides, macrolides and pleuromutilins. In:
nary Clinics of North America - Small Animal Practice 18, 1167– Prescott, J.F., Baggot, J.D., Walker, R.D. (Eds.), Antimicrobial
1182. Therapy in Veterinary Medicine, third ed. Iowa State University
Gatti, G., Malena, M., Casazza, R., Borin, M., Bassetti, M., Cruciani, Press, Ames, pp. 229–262.
M., 1998. Penetration of clindamycin and its metabolite, N - Sarkiala, E., Harvey, C., 1993. Systemic antimicrobials in the
demethylclindamycin into cerebrospinal fluid following intrave- treatment of periodontitis in dogs. Seminars in Veterinary Med-
nous infusion of clindamycin phosphate in patients with AIDS. icine and Surgery-Small Animal 8, 197–203.
Antimicrobial Agents and Chemotherapy 42, 3014–3017. Schaumann, R., Ackermann, G., Pless, B., Claros, M.C., Goldstein,
Gibaldi., M., Perrier, D., 1982a. Absorption kinetics and bioavaila- A.C., Rodloff, A.C., 2000. In vitro activities of fourteen antimi-
bility. In: Pharmacokinetics. Marcel Dekker Inc., New York, pp. crobial agents against obligately anaerobic bacteria. International
145–198. Journal of Antimicrobial Agents 16, 225–232.
Gibaldi., M., Perrier, D., 1982b. Noncompartmental analysis based on Scott, D.W., Beningo, K.E., Miller, W.H., Jr, Rothstein, E., 1998.
statistical moment theory. In: Pharmacokinetics. Marcel Dekker Efficacy of clindamycin hydrochloride capsules for the treatment of
Inc., New York, pp. 409–424. deep pyoderma due to Staphylococcus intermedius infection in
Harvey, R.G., Noble, W.C., Ferguson, E.A., 1993. A comparison of dogs. Canadian Veterinary Journal 39, 753–756.
lincomycin hydrochloride and clindamycin hydrochloride in the Vogelman, B., Gudmundsson, S., Turnidge, J.D., Leggett, J., Craig,
treatment of superficial pyoderma in dogs. The Veterinary Record W.A., 1988. In vivo postantibiotic effect in a thigh infection in
132, 351–353. neutropenic mice. Journal of Infectious Diseases 157, 287–298.
G.C. Batzias et al. / The Veterinary Journal 170 (2005) 339–345 345

Wynalda, M.A., Hutzler, J.M., Koets, M.D., Podoll, T., Wienkers, Xue, I.B., Davey, P.G., Phillips, G., 1996. Variation in postantibiotic
L.C., 2003. In vitro metabolism of clindamycin in human liver and effect of clindamycin against clinical isolates of Staphylococcus
intestinal microsomes. Drug Metabolism and Disposition 31, 878– aureus and implications for dosing of patients with osteomyelitis.
887. Antimicrobial Agents and Chemotherapy 40, 1403–1407.

You might also like