Vous êtes sur la page 1sur 9

|

Received: 11 April 2017    Accepted: 11 December 2017

DOI: 10.1111/jvp.12484

ORIGINAL ARTICLE

Pharmacokinetic profiles of the active metamizole metabolites


after four different routes of administration in healthy dogs

M. Giorgi1  | B. Łebkowska-Wieruszewska2 | A. Lisowski3 | H. Owen4 | 


A. Poapolathep5 | T. W. Kim6  | V. De Vito7

1
Department of Veterinary
Sciences, University of Pisa, Pisa, Italy Metamizole (MT), an analgesic and antipyretic drug, is rapidly hydrolyzed to the active
2
Department of Pharmacology, University of primary metabolite 4-­methylaminoantipyrine (MAA) and relatively active secondary
Life Sciences, Lublin, Poland metabolite 4-­aminoantipyrine (AA). The aim of this study was to assess the pharma-
3
Department of Animal Hygiene and
cokinetic profiles of MAA and AA after dose of 25 mg/kg MT by intravenous (i.v.), in-
Environment, University of Life Sciences,
Lublin, Poland tramuscular (i.m.), oral (p.o.), and rectal (RC) routes in dogs. Six dogs were randomly
4
Department of Veterinary allocated to an open, single-­dose, four-­treatment, four-­phase, unpaired, crossover
Sciences, University of Queensland, Gatton,
Australia
study design. Blood was collected at predetermined times within 24 hr, and plasma
5
Department of Pharmacology, Faculty of was analyzed by a validated HPLC-­UV method. Plasma concentrations of MAA and AA
Veterinary Medicine, Kasetsart University, after i.v., i.m., p.o., and RC administrations of MT were detectable from 5 (i.v. and i.m.)
Bangkok, Thailand
6
or 30 (p.o. and RC) min to 24 hr in all dogs. The highest concentrations of MAA were
College of Veterinary Medicine, Chungnam
National University, Daejeon, South Korea found in the i.v., then i.m., p.o., and RC groups. Plasma concentrations of AA were simi-
7
Department of Veterinary lar for i.v., i.m., and RC, and the concentrations were approximately double those in the
Medicine, University of Sassari, Sassari, Italy
PO groups. The AUCEV/IV ratio for MAA was 0.75 ± 0.11, 0.59 ± 0.08, and 0.32 ± 0.05,
Correspondence for i.m., p.o., and RC, respectively. The AUCEV/IV ratio for AA was 1.21 ± 0.33,
Mario Giorgi, Department of Veterinary
Sciences, University of Pisa, Pisa, Italy.
2.17 ± 0.62, and 1.08 ± 0.19, for i.m., p.o., and RC, respectively. Although further stud-
Email: mario.giorgi@unipi.it ies are needed, rectal administration seems to be the least suitable route of adminis-

Funding information
tration for MT in the dog.
Ex 60% University of Pisa 2015

1 |  INTRODUCTION (Australia, USA, Sweden, Japan, Iran, and UK) because of safety con-
cerns in humans. Although MT is known to be a relatively safe drug
Metamizole (MT), or dipyrone, is one of the more effective nonopioid compared to other nonopioid analgesics, there are some reports, al-
analgesics available for the treatment of pain and fever in both human though not unanimously accepted, suggesting that after long-­term
and veterinary medicine (Baumgartner et al., 2009). It works as an ef- treatment, MT might cause hematopoietic pathology, agranulocyto-
ficacious COX-­3 inhibitor but also provides weak COX-­1 and COX-­2 sis, leukopenia, and even aplastic anemia in humans (Basak, Drozd-­
inhibition (Botting, 2000; Chandrasekharan et al., 2002). In partial ex- Sokołowska, & Wiktor-­Jedrzejczak, 2010; Bigal, Bordini, & Speciali,
planation for this action, MT has been shown to interact with both the 2002; Garcıa-­Martınez et al., 2003; Hedenmalm & Spigset, 2002;
N-­methyl-­D-­aspartate receptor and the opioid system; however, the Imagawa et al., 2011). However, pharmacovigilance veterinary reports
exact mechanism by which MT produces its analgesic action remains indicate that the incidence of adverse reactions in the target species
unclear (Khodai, 2008; Tortorici, Vasquez, & Vanegas, 1996). Recent is negligible (Committee for Veterinary Medicinal Products, 2003). For
studies have demonstrated that its analgesic action could partially be the veterinary market, MT is labeled for use in porcine, bovine, equine,
due to the interaction with the cannabinoid receptors (Crunfli, Vilela, & and canine species. It is administered parentally in the dose range of
Giusti-­Paiva, 2015; dos Santos et al., 2014). MT is available in human 20–50 mg/kg (package leaflet, Biovetalgin, Biowet, Drwalew, Poland) or
and veterinary markets in several countries (South America, Asia, orally 40–50 mg metamizole sodium/kg body weight (package leaflet,
and European countries) but has been withdrawn in other countries Metapyrin oral 100%, Serumwerk Bernburg AG, Bernburg, Germany).

J vet Pharmacol Therap. 2018;1–9. © 2018 John Wiley & Sons Ltd |  1
wileyonlinelibrary.com/journal/jvp  
|
2       GIORGI et al.

Metamizole is a prodrug which, in an aqueous environment, AA after MT oral administration in dogs (Kalchofner Guerrero et al.,
spontaneously breaks down into numerous metabolic products 2015; Loescher, 1993). This study was carried out to compare the
(Levy, Zylber-­Katz, & Rosenkranz, 1995; Vlahov, Badian, Verho, & pharmacokinetic profiles of MAA and AA after intravenous (i.v.), in-
Bacracheva, 1990) . In humans, MT is rapidly hydrolyzed to the tramuscular (i.m.), oral (p.o.), and rectal (RC) administrations of MT
main metabolite 4-­methylaminoantipyrine (MAA). MAA is then me- in healthy dogs.
tabolized to 4-­formylaminoantipyrine (FAA; an end-­metabolite) and
to 4-­aminoantipyrine (AA) (Levy et al., 1995). AA is further metabo-
2 | MATERIALS AND METHODS
lized to 4-­acetylaminoantipyrine (AAA) (Figure 1) (Levy et al., 1995;
Rogosch et al., 2012; Vlahov et al., 1990). MAA and AA are both active
2.1 | Animal experiments
metabolites (Vlahov et al., 1990; Weithmann & Alpermann, 1985). In
recent years, MT has experienced a revival for pain management in The animal study was carried out in accordance with the European
human and veterinary medicine, with pharmacokinetic studies being law (Directive 2010/63/EU). Six adult, intact female Labradors, aged
performed in food-­producing animals and pharmacodynamics studies between 3–6 years and weighing 36–42 kg, were used in the study.
on dogs (Aupanun et al., 2016; Burmańczuk, Kowalski, Giorgi, Owen, The dogs were determined to be clinically healthy based on physi-
& Grabowski, 2016; Giorgi et al., 2015; Giorgi et al., 2017; Imagawa cal examination, serum chemistry, and hematological analyses. Food
et al., 2011; Kalchofner Guerrero et al., 2015; Schütter, Tünsmeyer, & was withheld for 10 hr prior to the beginning of the experiment while
Kästner, 2016; Teixeira et al., 2013; Zanuzzo et al., 2015). water was given ad libitum. Feed was available from 4 hr after treat-
To the best of the authors’ knowledge, only two reports pres- ment. Animal health was evaluated daily by trained personnel (up to
ent limited data on the plasma concentration vs. time of MAA and 1 week after completion of the study) for visible adverse effects. Two

F I G U R E   1   Metabolic pathway of
metamizole (MT) reported in humans
(Geisslinger, Böcker, & Levy, 1996)
GIORGI et al. |
      3

weeks after completion of the experiment, the dogs underwent a (Como, Italy) consisting of a quaternary gradient system (PU 2089
health check for any abnormalities. PLUS), coupled with an ultraviolet detector (Jasco UV-­975) set at
Dogs were randomly assigned to four-­treatment groups (research 254 nm. The column was a Luna C18(2) (250 mm × 4.6 mm inner
randomizer software [www.randomizer.org]), using an open, single-­ diameter, 5-­μ particle size [Phenomenex, Bologna, Italy]) preceded
dose, four-­treatment, four-­phase, unpaired, crossover design (4 × 4 by a security guard column with the same stationary phase (C18(2)
Latin-­square). During the first phase, each subject in group 1 (n = 2) [Phenomenex, Bologna, Italy]). The HPLC system was kept at
was administered with a single dose of 25 mg/kg MT (Biovetalgin, 25°C. The analytes were eluted in isocratic mode with a mobile
injectable solution 500 mg/ml, BioWet, Drwalew, Poland) injected phase (ammonium acetate 20 mm:acetonitrile, pH 5, 80:20, v/v) at
i.v. into the jugular vein at a 1 ml/min injection rate. Group 2 (n = 1) a flow rate of 1 ml/min.
received an i.m. injection, at the same dose as the i.v. injection, in-
jected in the middle quadrant of the buttock muscle (Biovetalgin,
2.4 | Sample extraction
injectable solution 500 mg/ml, BioWet, Drwalew, Poland). Group
3 (n = 2) received the same dose (25 mg/kg) orally, in an immediate Sample extraction was carried out in a 15 ml polypropylene vial. IS
release (human) oral formulation (Pyralgina–Zakłady farmaceutyczne (100 μl, 25 μg/ml) was added to 0.5 ml of plasma. After 30 s vortexing,
Polpharma S.A. 500 mg/tablet, Poland). The doses were prepared sodium hydroxide (0.1 ml, 1 N) was added and the sample vortexed
by weighing and partitioning (by shaving) the marketed drug. Group again. Ethyl acetate:methylene chloride (4 ml, 3:7, v/v) was added,
4 (n = 1) was given MT at 25 mg/kg via the RC route (suppositories, and the sample was vortexed (30 s), shaken (60 osc/min, 10 min), and
Pyralgina, 750 mg, Zakłady farmaceutyczne Polpharma, Poland). The centrifuged at 10,956 g (rotor radius 5 cm) for 10 min at 10°C. Three
suppositories were dipped in cold water (10°C) for half an hour prior ml of the organic layer was collected in a new 15 ml screw cap vial.
to use in order to facilitate their division and insertion. The dogs were The organic phase was evaporated under a gentle stream of nitrogen
kept in individual cages and monitored for defecation for at least 2 hr (40°C) and reconstituted with 100 μl of mobile phase. Fifty μl of this
after insertion of the suppositories to ensure complete absorption of solution was injected onto the HPLC.
drug. Animals were given the suppository while in a prone position.
The dose was determined based on package leaflet recommendations
2.5 | Method revalidation
(injectable) and on an earlier study showing that 25 mg/kg was an ef-
fective dose in dogs (Imagawa et al., 2011). The HPLC method was revalidated using control dog plasma. In brief,
The washout period between the four phases (to ensure com- MAA and AA were linear in the range of 100–5000 ng/ml. LOD was
plete metabolism and excretion of MAA and AA) was 1 week. The 30 ng/ml, and LOQ was 50 ng/ml. When the metabolite concentrations
groups were rotated until the crossover study was completed. When in the samples exceeded the upper limit of the range, they were re-­
the study was completed each animal had received MT by all formu- analyzed after appropriate dilution. The intraday and interday precision
lations. Thirty minutes before the commencement of the study to coefficient of variations were lower than 5.9 and 6.7, and 8.1% and 7.9%
facilitate blood sampling, an 18-­gauge catheter was placed into the for MAA and AA, respectively. The relative error of accuracy in within-­
medial saphenous vein. Blood samples (2 ml) were collected into lith- day and between-­day assays was lower than 4.7 and 5.3, and 6.0%
ium heparin tubes at 5, 15, 30, and 45 min and 1, 1.5, 2, 4, 6, 8, 10, and 7.1% for MAA and AA, respectively. The recoveries of the extrac-
24, 36, and 48 hr after drug administration. Tubes were immediately tion procedure were between 93.5% and 96.7% for MAA and between
placed on ice and centrifuged at 2,200 g for 5 min within 30 min. The 95.1% and 98.0% for AA.
collected plasma was stored at −20°C until analysis, within 30 days
of collection.
2.6 | Pharmacokinetic analysis and
statistical analysis
2.2 | Standard compounds
The pharmacokinetic calculations were carried out using WinNonlin v
Pure AA and MAA analytical standard (>99.0% purity) were obtained 5.3.1 (Pharsight Corp). The curve fit was performed by a noncompart-
from Sigma-­Aldrich (St. Louis, MO, USA) and Toronto Research mental analysis. The percent of AUC that was extrapolated to infinity
Chemicals (Toronto, Canada), respectively. The Internal Standard (IS) (AUCExtrap%) was always <20% in all the subjects. The extraction ratio
was metoclopramide (powder > 99.0% purity) supplied by Sigma-­ was estimated by dividing total clearance for each animal after i.v. ad-
Aldrich (St. Louis, MO, USA). Dog control plasma samples were kindly ministration by cardiac output, where cardiac output was estimated as
supplied by the canine blood bank at the University of Pisa. 180 ×  body weight (kg)−0.19 (Toutain & Bousquet-­Mélou, 2004). The
results are presented as mean ± standard deviation.
To make comparisons across treatments, the different parameters
2.3 | HPLC-­UV analysis
were first tested for normal distribution and variance homogeneity.
The analytical method was approached based on previously de- Data were compared with the ANOVA or the nonparametric Wilcoxon
scribed methods with slight modifications (Domínguez-­Ramírez test, depending on whether the data passed a normality test. A p value
et al., 2012; Giorgi et al., 2015). The HPLC system was an LC Jasco less than .05 was considered statistically significant.
4       | GIORGI et al.

3 |  RESULTS concentration of MAA after the four routes of administration. Except
for the initial phase, the i.v. and i.m. concentrations vs. time profiles
No behavioral changes or alterations in health parameters were ob- were similar. The p.o. and RC concentrations vs. time profiles were also
served in the i.v., i.m., or RC groups of animals during or after (up similar in shape but not in MAA concentration.
to 7 days) the drug administration. Some salivation (self-­limiting) The highest concentrations of MAA were found in the
was noted in three subjects in the p.o. group 15–30 min after drug i.v., > i.m. > p.o. > RC group (Figure 3). The concentrations of MAA
administration. were higher than that of AA in the i.v. and i.m. groups (Figure 2 a, b).
Plasma concentrations of MAA and AA after i.v., i.m., p.o., and RC In contrast, the concentration of AA exceeded that of MAA 6 hr after
administrations of MT were detectable from 5 (i.v., i.m.) or 30 (p.o., RC) MT administration in the p.o. and RC groups (Figure 2 c, d). It is note-
min to 24 hr in all dogs (Figure 2). Figure 3 reports the average plasma worthy that the average plasma concentration of AA in the p.o. group

25

(a)
20
Concentration (µg/ml)

15

10

0
0 6 12 18 24
Time (hr)
15

12 (b)
Concentration (µg/ml)

0
0 6 12 18 24
Time (hr)
8

6 (c)
Concentration (µg/ml)

0
0 6 12 18 24
Time (hr)
6

(d)
4
Concentration (µg/ml)

F I G U R E   2   Mean plasma concentrations


2 of 4-­methylaminoantipyrine ( , MAA) and
4-­aminoantypyrine ( , AA) vs. time curves
following intravenous (a), intramuscular
0 (b), oral (c), and rectal (d) dose of 25 mg⁄kg
0 6 12 18 24 metamizole (MT) in healthy dogs (n = 6).
Time (hr) Bars represent the standard deviations
GIORGI et al. |
      5

F I G U R E   3   Mean plasma concentrations of 4-­methylaminoantipyrine (MAA) vs. time curves following intravenous ( ), and intramuscular
( ), oral ( ), and rectal ( ) dose of 25 mg⁄kg metamizole (MT) in healthy dogs (n = 6). Bars represent the standard deviations. The panel
focus on the relevant area of the curve where the IC50s (–) for the human COX enzymes are located

was higher (double) than that of AA in the other treatment groups or forgotten (Nikolova et al., 2012). MT acts as an effective pain-­
(Figure 4). relieving, antipyretic, and spasmolytic agent (Levy et al., 1995). It does
The main pharmacokinetic parameters of MAA are presented in not possess the contraindications or restrictions usually observed
Table 1. The ratios calculated based on the relative AUC0-last values with opioids or NSAIDs (Avellaneda et al., 2000; Baumgartner et al.,
AUCIM MAA/AUCIV MAA, AUCPO MAA/AUCIV MAA, and AUCRC MAA/AUCIV 2009; Edwards, Meseguer, Faura, Moore, & McQuay, 2010; Kemal,
MAA were 0.75 ± 0.11, 0.59 ± 0.08, and 0.32 ± 0.05, respectively. Sahin, & Apan, 2007; Zukowski & Kotfis, 2009). The analgesic efficacy
The main pharmacokinetic parameters of AA are presented in of MT in humans is abundantly reported in the literature (Avellaneda
Table 2. The ratios calculated based on the relative AUC0-last values et al., 2000; Edwards et al., 2010; Kemal et al., 2007; Korkmaz Dilmen
AUCIM AA/AUCIV AA, AUCPO AA/AUCIV AA, and AUCRC AA/AUCIV AA were et al., 2010; Olson, Sunshine, Zighelboim, & Lange, 1999; Zukowski &
1.21 ± 0.33, 2.17 ± 0.62, and 1.08 ± 0.19, respectively. Kotfis, 2009).
In contrast, the evidence is not as strong in veterinary medicine.
Some data regarding clinical and side effects of MT in horses, rab-
4 | DISCUSSION bits, rats, and dogs have been reported, and there is addition data
in relation to the pharmacokinetic profile of the metabolite MAA in
Metamizole has been used to treat pain and fever for almost a cen- horses, donkeys, pigs, rats, dogs, cats, and sheep (Aupanun et al., 2016;
tury in some countries, while in others, it is completely unknown Baumgartner et al., 2009; Burmańczuk et al., 2016; Christ, Kellner,

3
Concentraon (µg/ml)

F I G U R E   4   Mean plasma concentrations


of 4-­aminoantypyrine (AA) vs. time curves 1
following intravenous ( ), intramuscular
( ), oral ( ), and rectal ( ) dose of
25 mg⁄kg metamizole (MT) in healthy 0
dogs (n = 6). Bars represent the standard 0 6 12 18 24
deviations Time (hr)
|
6       GIORGI et al.

T A B L E   1   Main pharmacokinetic parameters of 4-­methylaminoantipyrine (MAA) following single intravenous (i.v.), intramuscular (i.m.), oral
(p.o), and rectal (RC) administrations of metamizole (MT) (25 mg/kg) in healthy dogs (n = 6)

Parameter Unit i.v. (Mean ± SD) i.m. (Mean ± SD) p.o. (Mean ± SD) RC (Mean ± SD)


2
R .92 ± .08 .94 ± .06 .90 ± .08 .88 ± .13
λz 1/hr 0.13 ± 0.04 0.11 ± 0.02 0.12 ± 0.02 0.12 ± 0.03
T1/2 λz# hr 5.94 ± 2.54 5.92 ± 1.04 5.59 ± 0.96 5.87 ± 1.44
Tmax hr 0.083bcd ± 0.000 0.625acd ± 0.130 1.910ab ± 0.200 1.910ab ± 0.200
Cmax μg/ml 21.80bcd ± 2.45 12.57acd ± 2.27 5.27abd ± 0.56 2.72abc ± 0.35
bcd acd abd
AUC0-last hr/μg/ml 45.34  ± 9.64 33.33  ± 4.83 26.62  ± 4.69 15.27abc ± 3.44
AUC0-∞ hr/μg/ml 46.79bcd ± 9.15 34.76acd ± 4.82 28.01abd ± 5.53 16.18abc ± 3.39
bcd acd abd
Vz/F ml/kg 4,946.33  ± 988.21 6,250.45  ± 1,409.44 7,449.71  ± 2,218.28 13,776.34abc ± 4,839.61
CL/F ml/hr/kg 552.43bcd ± 98.34 731.14acd ± 103.47 920.93abd ± 178.96 1,605.66abc ± 356.24
2 cd cd abd
AUMC0-∞ hr /μg/ml 246.57  ± 31.01 201.92  ± 53.035 202.93  ± 80.20 131.28abc ± 27.74
MRT0–∞ hr 5.37 cd ± 0.81 5.93cd ± 1.44 7.06ab ± 1.28 8.13ab ± 1.08
AUCEV/IV ratio % / 75.2cd ± 11.75 59.6bd ± 8.04 32.78ab ± 5.08

R2, correlation coefficient of the terminal portion of the curve; λz, terminal phase rate constant; T1/2λz, terminal half-­life; Tmax, time of peak; Cmax, peak
plasma concentration; Vz/F, apparent volume of distribution; CL/F, apparent clearance; AUC0-∞, area under the plasma concentration–time curve;
AUMC0-∞, area under the first moment curve; MRT0-∞, mean resident time; AUCEV/IV ratio, AUCextravascular/AUCIV.
Superscript letters indicate the statistically different values among the treatment groups.
#
Harmonic mean.

T A B L E   2   Main pharmacokinetic parameters of 4-­aminoantypyrine (AA) following single intravenous (i.v.), intramuscular (i.m.), oral (p.o.), and
rectal (RC) administrations of metamizole (MT) (25 mg/kg) in healthy dogs (n = 6)

Parameter Unit i.v. (Mean ± SD) i.m. (Mean ± SD) p.o. (Mean ± SD) RC (Mean ± SD)


2
R .97 ± .02 .98 ± .02 .98 ± .01 .98 ± .03
λz 1/hr 0.09 ± 0.03 0.08 ± 0.03 0.12 ± 0.04 0.10 ± 0.02
T1/2 λz# hr 8.05 ± 2.56 8.75 ± 2.58 6.12abd ± 2.08 7.25 ± 1.86
Tmax hr 5.33 ± 1.63 5.58 ± 2.49 6.33 ± 1.96 5.21 ± 1.67
Cmax μg/ml 1.29 ± 0.21 1.36 ± 0.38 2.64abd ± 0.44 1.43 ± 0.18
abd
AUC0-last hr/μg/ml 17.97 ± 2.91 21.28 ± 4.80 37.92  ± 7.60 19.08 ± 2.22
AUC0-∞ hr/μg/ml 21.08 ± 4.34 23.03 ± 7.31 42.37abd ± 11.78 21.87 ± 2.75
Vz/F ml/kg 13,855.67 ± 3,678.36 12,394.27 ± 3,898.61 5,146.44abd ± 393.45 11,978.01 ± 2,827.06
CL/F ml/hr/kg 1,224.03 ± 228.74 1,014.77 ± 242.02 628.43abd ± 168.32 1,156.87 ± 132.41
2 abd
AUMC0-∞ hr /μg/ml 273.97 ± 117.74 377.07 ± 169.98 489.45  ± 252.05 266.36 ± 69.61
MRT0–∞ hr 12.62 ± 3.13 14.07 ± 3.12 10.99abd ± 2.49 12.09 ± 2.73
bd
AUC EV/IV ratio % / 121.26 ± 33.59 217.55  ± 62.29 108.08 ± 19.93
2
R , correlation coefficient of the terminal portion of the curve; λz, terminal phase rate constant; T1/2λz, terminal half-­life; Tmax, time of peak; Cmax, peak
plasma concentration; Vz/F, apparent volume of distribution; CL/F, apparent clearance; AUC0-∞, area under the plasma concentration–time curve;
AUMC0-∞, area under the first moment curve; MRT0-∞, mean resident time; AUCEV/IV ratio, AUCextravascular/AUCIV.
Superscript letters indicate the statistically different values among the treatment groups.
#
Harmonic mean.

Ross, Rupp, & Schwarz, 1973; Flôr, Yazbek, Ida, & Fantoni, 2013; Giorgi pharmacokinetic–pharmacodynamic profiles of MT or its active me-
et al., 2015, 2017; Imagawa et al., 2011; Klaus, Schlingloff, Kleinitz, tabolites are limited or absent. In canine species, a single study only
Böttcher, & Hapke, 1997; Lebkowska-­Wieruszewska et al., 2017; reports basic pharmacokinetics of MAA and AA after a single 50 mg/
Loescher, 1993; Roelvink, Goossens, Kalsbeek, & Wensing, 1991; kg p.o. administration of MT (Kalchofner Guerrero et al., 2015).
Silva-­Moreno, Lopez-­Munoz, & Cruz, 2009; Teixeira et al., 2013; The pharmacokinetic profiles of MAA after i.v. and i.m. MT ad-
Zanuzzo et al., 2015). ministrations were comparable. The substantial differences detected
Due to MT’s striking pharmacological effect, low cost and safety in Cmax values were ascribable to the routes of administration of MT.
profile, there has been growing interest in its use in the veterinary field The complete/abrupt input of MT into the vascular compartment (i.v.
over the past decade. Although MT is widely used in clinical practice, injection) may have produced, in the initial minutes, a quicker drug
GIORGI et al. |
      7

transformation (MT to MAA) increasing the Cmax of MAA, compared higher IC50 values for AA (Hinz et al., 2007). If these minimal effective
to the i.m. group where an absorption phase is expected. MAA for- concentrations (MEC) apply to dogs, plasma concentration of MAA,
mation depends on a nonenzymatic, pH-­dependent hydrolysis of MT but not AA, was above the COX-­1/2 relevant IC50 values for a range of
and is immediate once MT reaches the circulation (Ergun, Frattarelli, time correlated to the route of drug administration (Figure 3). Hence,
& Aranda, 2004). Blood pH can accelerate its generation: the lower only MAA might generate a therapeutic effect in the dog. The dif-
the pH, the faster the hydrolysis (Ergun et al., 2004). Animals did not ferences in time MAA plasma concentrations were above the MECs
show any abnormal signs following i.v. administration despite the im- among i.v., i.m., and p.o. groups are small and might not be clinically
mediate peak of MAA concentration. The differences in Tmax values relevant. In contrast, the RC group showed a reduced time over the
may also be triggered by the absorption phase. The MAA AUC values MEC suggesting that this route of administration might be the least
(i.v. vs. i.m.) showed that the exposition to the active metabolite over effective among those tested in the dog.
time is statistically different but still high (75%) after i.m. administra- The pharmacokinetic trends detected for the AA after i.v., i.m., and
tion of MT. RC administrations of MT were similar. In contrast, the p.o. groups
The half-­life (t1/2λz) reported for MAA in the present study showed plasma concentrations and AUC0-last parameters double those
was similar to that previously reported in dogs (4–5 hr) and horses of the other groups. The explanation for this phenomenon is uncer-
(4.85 hr), higher to that reported in sheep (1.45–3 hr) and donkeys tain, but a similar difference in AA plasma concentrations has already
(1.81 hr) and lower than that reported in piglets (8.57 hr) (Aupanun been observed in horses after i.m. and i.v. administration of MT (Giorgi
et al., 2016; Burmańczuk et al., 2016; Giorgi et al., 2015; Klaus et al., et al., 2015) and could be due to first pass metabolism to AA in the
1997; Loescher, 1993). The reasons for this variation might be due to enteral route. As reported above, in humans, the analgesic effect of
diverse factors such as differences in administration route, animal spe- MT correlates with the concentration of MAA and AA, which differ
cies, age and pathophysiological conditions of animals, and sensitivity with regard to their time of onset (MAA < AA) and terminal half-­life
of the analytical method. (MAA and AA, 4–5 and 5–8 hr, respectively) (Nikolova et al., 2012).
Assuming that after i.v. administration of MT, all the drug is con- MAA is about 50 times more active than MT as an inhibitor of COX-­3
verted to MAA, Vd/F, and CL/F can be considered as absolute values. enzymes, while AA is less active than MT (Nikolova et al., 2012). It is
Vd/F is large, a finding consistent with MAA being detected in cere- important to specify that the metabolites which generate the anal-
brospinal fluid and CL/F shows an extraction ratio of 6%–9% (Cohen, gesic effect in dogs are still unknown. If it is assumed that analgesic
Zylber-­Katz, Caraco, Granit, & Levy, 1998). This finding suggests that effect is only attributable to MAA and AA metabolites as in humans,
the contribution of the liver and kidney (the main clearance organs) in the impact of AA to overall therapeutic analgesic effect might be neg-
the clearance of MAA is limited in the dog. This is also in line with the ligible due to both its weak activity and low plasma concentrations
small amount of AA produced. (below the MECs). Hence, it might be supposed that MAA is the main
The Cmax value of MAA after p.o. administration of MT, if normal- metabolite responsible for the overall analgesic effect. However, addi-
ized for the dose, was comparable to that shown in a previous study tional studies are essential, to evaluate whether the metabolic pattern
in dogs (Kalchofner Guerrero et al., 2015). However, although the known in humans matches that in dogs. In addition, pharmacodynamic
Kalchofner Guerrero study does not report the PK parameters, the studies determining the drug effectiveness in different types of pain
elimination phase of MAA seems slower than that in the present study. are required.
It is likely due to the different MT formulation used: immediate release This is the first study reporting the biopharmaceutics of MT and its
(present study) vs. new slow-­release formulation (Kalchofner Guerrero active metabolites MAA and AA after i.v., i.m., p.o., and RC administra-
et al., 2015). tions of MT in dogs. The MAA pharmacokinetic profiles were different
The pharmacokinetic trend of MAA after RC administration of MT with each route of administration, while the AA pharmacokinetic pro-
mirrors that of the p.o. group but with lower concentrations. RC ad- files were similar for i.v., i.m., and RC administrations. Even though addi-
ministration of MT resulted in the lowest Cmax of MAA (2.72 μg/ml) tional studies are warranted to understand the metabolism of MT along
and AUC RC/IV ratio (32.78%) among the formulations tested. A specific with its safety profile, rectal administration appears to be the least ef-
concern with RC administration of MT, and a likely reason for the low ficient route of administration for MT in the dog. However, to prepare
MAA concentrations detected in the present study, is the chance for the desired doses for p.o. and RC administration, it was assumed that
sequestration of drug in fecal matter, a general drawback of RC route the drug was completely uniform in its distribution in the formulations.
for drug administration. This phenomenon might explain the low AUC This was not analytically confirmed. It is likely therefore that the true
ratio obtained by this administration route. Previous studies reported doses administered could have varied from the calculated doses.
that RC administration of suppository marketed for humans showed
reduced bioavailability in dogs (De Vito et al., 2015; Giorgi, Del Carlo,
AC KNOW L ED G M ENTS
Saccomanni, Łebkowska-­Wieruszewska, & Kowalski, 2009) .
It is reported that the analgesic effect of MT correlates with The study was carried out using funds from University of Pisa
plasma concentration of its two active metabolites (Nikolova et al., (Athenaeum ex 60%, 2015). No external funding was used for prepa-
2012). Both metabolites have been shown to inhibit human COX-­1 ration of the manuscript. Authors thank Dr A Di Salvo (University of
and COX-­2 with an IC50 of 2.55 μm and 4.65 μm and 8.2-­ to 9-­fold Perugia) for the critical editing of the manuscript.
|
8       GIORGI et al.

CO NFLI CT OF I NTE RE S T Christ, O., Kellner, H. M., Ross, G., Rupp, W., & Schwarz, A. (1973).
Biopharmaceutical and pharmacokinetic studies on metamizol-­14C
None of the authors have any financial or personal relationships that (Novalgin 14C) given to rats, dogs and men. Arzneimittelforschung Drug
could inappropriately influence or bias the content of the manuscript. Research, 23, 1760–1767.
Cohen, O., Zylber-Katz, E., Caraco, Y., Granit, L., & Levy, M. (1998).
Cerebrospinal fluid and plasma concentrations of dipyrone me-
AUTHOR’S CONTRIBUTI O N tabolites after a single oral dose of dipyrone. European Journal
of Clinical Pharmacolgy, 54, 549–553. https://doi.org/10.1007/
MG and BLW conceived of the presented idea. MG and TWK devel- s002280050511
oped the theory and performed the computations. AL, BLW, and HO Committee for Veterinary Medicinal Products (2003). EMEA/
MRL/878/03-FINAL.
carried out the animal study. VDV and AP analyzed the samples. All
Crunfli, F., Vilela, F. C., & Giusti-Paiva, A. (2015). Cannabinoid CB1 re-
authors discussed the results and contributed to the interpretation of ceptors mediate the effects of dipyrone. Clinical and Experimental
them. MG, HO, and TWK took the lead in writing the manuscript. All Pharmacology and Physiology, 42, 246–255. https://doi.
authors read and approved the final manuscript. org/10.1111/1440-1681.12347
De Vito, V., Łebkowska-Wieruszewska, B., Shaban, A., Lisowski, A., Kowaski,
C. J., & Giorgi, M. (2015). Pharmacokinetic profiles of the analgesic flu-
O RCI D pirtine in dogs after the administration of four pharmaceutical formu-
lations. Veterinary Anaesthesia and Analgesia, 42, 629–637. https://doi.
M. Giorgi  http://orcid.org/0000-0003-3657-4703 org/10.1111/vaa.12235
Domínguez-Ramírez, A. M., Calzadilla, P. C., Cortés-Arroyo, A. R., Hurtado,
T. W. Kim  http://orcid.org/0000-0002-5239-3833
Y., de la Peña, M., López, J. R., … López-Muñoz, F. J. (2012). High-­
performance liquid chromatographic assay for metamizol metabo-
lites in rat plasma: Application to pharmacokinetic studies. Journal
of Pharmaceutical and Biomedical Analysis, 71, 173–178. https://doi.
REFERENCES
org/10.1016/j.jpba.2012.07.029
Aupanun, S., Laus, F., Poapolathep, A., Owen, H., Vullo, C., Faillace, V., & Edwards, J. E., Meseguer, F., Faura, C. C., Moore, R. A., & McQuay, H. J.
Giorgi, M. (2016). Pharmacokinetic Assessment of the Marker Active (2010). Single dose dipyrone for acute postoperative pain. Cochrane
Metabolites 4-­Methyl-­amino-­antipyrine and 4-­Acetyl-­amino-­antipyrine Database of Systematic Reviews, (9), CD003227.
After Intravenous and Intramuscular Injection of Metamizole (Dipyrone) Ergun, H., Frattarelli, D. A., & Aranda, J. V. (2004). Characterization of the
in Healthy Donkeys. Journal of Equine Veterinary Science, 47, 55–61. role of physicochemical factors on the hydrolysis of dipyrone. Journal
https://doi.org/10.1016/j.jevs.2016.08.005 of Pharmaceutical and Biomedical Analysis, 35, 479–487. https://doi.
Avellaneda, C., Gomez, A., Martos, F., Rubio, M., Sarmiento, J., & de la org/10.1016/j.jpba.2004.02.004
Cuesta, F. S. (2000). The effect of a single intravenous dose of metam- Flôr, P. B., Yazbek, K. V., Ida, K. K., & Fantoni, D. T. (2013). Tramadol plus
izol 2 g, ketorolac 30 mg and propacetamol 1 g on haemodynamic pa- metamizole combined or not with anti-­inflammatory drugs is clinically
rameters and postoperative pain after heart surgery. European Journal effective for moderate to severe chronic pain treatment in cancer pa-
of Anaesthesiology, 17, 85–90. tients. Veterinary Anaesthesia and Analgesia, 40, 316–327. https://doi.
Basak, G. W., Drozd-Sokołowska, J., & Wiktor-Jedrzejczak, W. (2010). org/10.1111/vaa.12023
Update on the incidence of metamizole sodium-­induced blood dyscra- Garcıa-Martınez, J. M., Fresno, V. J., Lastres, P., Bernabeu, C., Betes, P.
sias in Poland. Journal of International Medical Research, 38, 1374–1380. O., & Martın-Perez, J. (2003). Effect of metamizol on promyelocytic
https://doi.org/10.1177/147323001003800419 and terminally differentiated granulocytic cells. Comparative analysis
Baumgartner, C. M., Koenighaus, H., Ebner, J. K., Henke, J., Schuster, with acetylsalicylic acid and diclofenac. Biochemical Pharmacolgy, 65,
T., & Erhardt, W. D. (2009). Cardiovascular effects of dipyrone and 209–217.
propofol on hemodynamic function in rabbits. American Journal Geisslinger, G., Böcker, R., & Levy, M. (1996). High-­performance liquid chro-
of Veterinary Research, 70, 1407–1415. https://doi.org/10.2460/ matographic analysis of dipyrone metabolites to study their formation
ajvr.70.11.1407 in human liver microsomes. Pharmaceutical Research, 13, 1272–1275.
Bigal, M. E., Bordini, C. A., & Speciali, J. G. (2002). Intravenous dipyrone https://doi.org/10.1023/A:1016088925786
for the acute treatment of episodic tension-­type headache: A random- Giorgi, M., Aupanun, S., Lee, H. K., Poapolathep, A., Rychshanova,
ized, placebo-­controlled, double-­blind study. Brazilian Journal of Medical R., Vullo, C., … Laus, F. (2017). Pharmacokinetic profiles of the
and Biological Research, 35, 1139–1145. https://doi.org/10.1590/ active metamizole metabolites in healthy horses. Journal of
S0100-879X2002001000005 Veterinary Pharmacology and Therapeutics, 40, 165–171. https://doi.
Botting, R. M. (2000). Mechanism of action of acetaminophen: Is there a org/10.1111/jvp.12342
cyclooxygenase 3? Clinical Infectious Diseases, 31, S202–S210. https:// Giorgi, M., De Vito, V., Lee, H. K., Laus, F., Kowalski, C., Faillace, V., …
doi.org/10.1086/317520 Vullo, C. (2015). Pharmacokinetic investigations of the marker ac-
Burmańczuk, A., Kowalski, C., Giorgi, M., Owen, H., & Grabowski, T. (2016). tive metabolite-­4-­methylamino-­antipyrin after intravenous and
Pharmacokinetic investigations of the marker active metabolites intramuscular injection of metamizole in healthy sheep. Small
4-­methylamino-­antipyrine and 4-­amino-­antipyrine after intramuscular in- Ruminant Research, 132, 143–146. https://doi.org/10.1016/j.
jection of metamizole in healthy piglets. Journal of Veterinary Pharmacology smallrumres.2015.10.005
and Therapeutics, 39, 616–620. https://doi.org/10.1111/jvp.12317 Giorgi, M., Del Carlo, S., Saccomanni, G., Łebkowska-Wieruszewska,
Chandrasekharan, N. V., Dai, H., Roos, K. L., Evanson, N. K., Tomsik, J., B., & Kowalski, C. J. (2009). Pharmacokinetics of tramadol and its
Elton, T. S., & Simmons, D. L. (2002). COX-­3, a cyclooxygenase-­1 vari- major metabolites following rectal and intravenous administration
ant inhibited by acetaminophen and other analgesic/antipyretic drugs: in dogs. New Zealand Veterinary Journal, 57, 146–152. https://doi.
Cloning, structure, and expression. Proceedings of the National Academy org/10.1080/00480169.2009.36894
of Sciences of the United States of America, 99, 13926–13931. https:// Hedenmalm, K., & Spigset, O. (2002). Agranulocytosis and other blood
doi.org/10.1073/pnas.162468699 dyscrasias associated with dipyrone (metamizole). European Journal
GIORGI et al. |
      9

of Clinical Pharmacology, 58, 265–274. https://doi.org/10.1007/ Rogosch, T., Sinning, C., Podlewski, A., Watzer, B., Schlosburg, J., Lichtman,
s00228-002-0465-2 A. H., … Imming, P. (2012). Novel bioactive metabolites of dipyrone
Hinz, B., Cheremina, O., Bachmakov, J., Renner, B., Zolk, O., Fromm, M. F., (metamizol). Bioorganic & Medicinal Chemistry, 20, 101–107. https://doi.
& Brune, K. (2007). Dipyrone elicits substantial inhibition of peripheral org/10.1016/j.bmc.2011.11.028
cyclooxygenases in humans: New insights into the pharmacology of an dos Santos, G. G., Dias, E. V., Teixeira, J. M., Athie, M. C., Bonet, I. J., Tambeli, C.
old analgesic. FASEB Journal, 21, 2343–2351. https://doi.org/10.1096/ H., & Parada, C. A. (2014). The analgesic effect of dipyrone in peripheral
fj.06-8061com tissue involves two different mechanisms: Neuronal K(ATP) channel open-
Imagawa, V. H., Fantoni, D. T., Tatarunas, A. C., Mastrocinque, S., Almeida, T. F., ing and CB(1) receptor activation. European Journal of Pharmacology, 741,
Ferreira, F., & Posso, I. P. (2011). The use of different doses of metamizol 124–131. https://doi.org/10.1016/j.ejphar.2014.07.019
for post-­operative analgesia in dogs. Veterinary Anaesthesia and Analgesia, Schütter, A. F., Tünsmeyer, J., & Kästner, S. B. (2016). Influence of metam-
38, 385–393. https://doi.org/10.1111/j.1467-2995.2011.00617.x izole on 1) minimal alveolar concentration of sevoflurane in dogs and
Kalchofner Guerrero, K. S., Schwarz, A., Wuhrmann, R., Feldmann, S., 2) on thermal and mechanical nociception in conscious dogs. Veterinary
Hartnack, S., & Bettschart-Wolfensberger, R. (2015). Comparison Anaesthesia and Analgesia, 43(2), 215–226. https://doi.org/10.1111/
of a new metamizole formulation and carprofen for extended post-­ vaa.12289
operative analgesia in dogs undergoing ovariohysterectomy. Veterinary Silva-Moreno, A., Lopez-Munoz, F. J., & Cruz, S. (2009). D-­propoxyphene
Journal, 204, 99–104. https://doi.org/10.1016/j.tvjl.2015.01.028 and dipyrone co-­administration produces greater antinociception and
Kemal, S. O., Sahin, S., & Apan, A. (2007). Comparison of tramadol, fewer adverse effects than single treatments in rat. European Journal of
tramadol-­metamizol and tramadol-­lornoxicam administered by intra- Pharmacology, 607, 84–90. https://doi.org/10.1016/j.ejphar.2009.02.010
venous PCA in management of postoperative pain. The Journal of the Teixeira, R. C., Monteiro, E. R., Campagnol, D., Coelho, K., Bressan, T. F.,
Turkish Society of Algolgy, 19, 24–31. & Monteiro, B. S. (2013). Effects of tramadol alone, in combination
Khodai, L. (2008). Hemmung humaner NMDA-Rezeptoren durch Metamizol with meloxicam or dipyrone, on postoperative pain and the analgesic
und dessen Metabolit 4-Aminoantipyrin, Westfaalische Wilhelms requirement in dogs undergoing unilateral mastectomy with or without
Universitaat Munster (Dissertation). Germany, Munster. ovariohysterectomy. Veterinary Anaesthesia and Analgesia, 40, 641–
Klaus, A. M., Schlingloff, Y., Kleinitz, U., Böttcher, M., & Hapke, H. J. 649. https://doi.org/10.1111/vaa.12080
(1997). Pharmacokinetic study of dipyrone metabolite 4-­MAA in Tortorici, V., Vasquez, E., & Vanegas, H. (1996). Naloxone partial re-
the horse and possible implications for doping control. Journal of versal of the antinociception produced by dipyrone microinjected
Veterinary Pharmacology and Therapeutics, 20, 204–208. https://doi. into the periaqueductal gray of rats. Possible involvement of med-
org/10.1111/j.1365-2885.1997.tb00096.x ullary off-­ and on-­cells. Brain Research, 725, 106–110. https://doi.
Korkmaz Dilmen, O., Tunali, Y., Cakmakkaya, O. S., Yentur, E., Tutuncu, A. org/10.1016/0006-8993(96)00196-5
C., Tureci, E., & Bahar, M. (2010). Efficacy of intravenous paracetamol, Toutain, P. L., & Bousquet-Mélou, A. (2004). Plasma clearance. Journal of
metamizol and lornoxicam on postoperative pain and morphine con- Veterinary Pharmacology and Therapeutics, 27, 415–425. https://doi.
sumption after lumbar disc surgery. European Journal of Anaesthesiology, org/10.1111/j.1365-2885.2004.00605.x
27, 428–432. https://doi.org/10.1097/EJA.0b013e32833731a4 Vlahov, V., Badian, M., Verho, M., & Bacracheva, N. (1990). Pharmacokinetics
Lebkowska-Wieruszewska, B., Kim, T. W., Chea, B., Owen, H., Poapolathep, of metamizol metabolites in healthy subjects after a single oral dose of
A., & Giorgi, M. (2017). Pharmacokinetic profiles of the two major ac- metamizol sodium. European Journal of Clinical Pharmacolgy, 38, 61–65.
tive metabolites of metamizole (dipyrone) in cats following three dif- https://doi.org/10.1007/BF00314805
ferent routes of administration. Journal of Veterinary Pharmacology and Weithmann, K. U., & Alpermann, H. G. (1985). Biochemical and pharmaco-
Therapeutics, 1–6. https://doi.org/10.1111/jvp.12471 logical effects of dipyrone and its metabolites in model systems related
Levy, M., Zylber-Katz, E., & Rosenkranz, B. (1995). Clinical pharmacokinet- to arachidonic acid cascade. Arzneimittelforschung Drug Research, 35,
ics of dipyrone and its metabolites. Clinical Pharmacokinetics, 28, 216– 947–952.
234. https://doi.org/10.2165/00003088-199528030-00004 Zanuzzo, F. S., Teixeira-Neto, F. J., Teixeira, L. R., Diniz, M. S., Souza, V. L.,
Loescher, W. (1993). Pharmaka mit Wirkung auf das Zentralnervensystem. Thomazini, C. M., & Steagall, P. V. (2015). Analgesic and antihyperalge-
In W. Liischer, F. R. Ungemach & R. Kroker (Eds.), Grundlagen der sic effects of dipyrone, meloxicam or a dipyrone-­meloxicam combina-
Pharmakotherapie bei Haus-und Nutztieren (pp. 105–106). Berlin und tion in bitches undergoing ovariohysterectomy. Veterinary Journal, 205,
Hamburg, Germany: Verlag Paul Parey. 33–37. https://doi.org/10.1016/j.tvjl.2015.05.004
Nikolova, I., Tencheva, J., Voinikov, J., Petkova, V., Benbasat, N., & Danchev, Zukowski, M., & Kotfis, K. (2009). Safety of metamizol and paracetamol
N. (2012). Metamizole: A review profile of a well-­known “forgotten” for acute pain treatment. Anestezjologia Intensywna Terapia, 41,
drug. part i: Pharmaceutical and nonclinical profile. Biotechnology & 170–175.
Biotechnological Equipment, 26, 3329–3337. https://doi.org/10.5504/
BBEQ.2012.0089
Olson, N. Z., Sunshine, A., Zighelboim, I., & Lange, R. (1999). Analgesic effi-
How to cite this article: Giorgi M, Łebkowska-Wieruszewska B,
cacy of liquid ketoprofen compared to liquid dipyrone and placebo ad-
Lisowski A, et al. Pharmacokinetic profiles of the active
ministered orally as drops in postepisiotomy pain. International Journal
of Clinical Pharmacology and Therapeutics, 37, 168–174. metamizole metabolites after four different routes of
Roelvink, M. E., Goossens, L., Kalsbeek, H. C., & Wensing, T. (1991). administration in healthy dogs. J vet Pharmacol Therap.
Analgesic and spasmolytic effects of dipyrone, hyoscinen-­butylbromide 2018;00:1–9. https://doi.org/10.1111/jvp.12484
and a combination of the two in ponies. Veterinary Record, 26, 378–
380. https://doi.org/10.1136/vr.129.17.378

Vous aimerez peut-être aussi