Clinical Pharmacology of Methadone in Dogs.

download Clinical Pharmacology of Methadone in Dogs.

of 10

Transcript of Clinical Pharmacology of Methadone in Dogs.

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    1/10

    R E S E A R C H P A P E R

    Clinical pharmacology of methadone in dogs

    Carina Ingvast-Larsson*, Anja Holgersson*, Ulf Bondesson, Anne-Sofie Lagerstedt & Kerstin Olsson

    *Division of Pathology, Pharmacology and Toxicology, Department of Biomedical Sciences and Veterinary Public Health,

    Swedish University of Agricultural Sciences, Uppsala, Sweden

    Division of Analytical Pharmaceutical Chemistry, Biomedical Center, Uppsala University, Uppsala and Department of

    Chemistry, National Veterinary Institute, Uppsala, Sweden

    Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden

    Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden

    Correspondence: Carina Ingvast-Larsson, Division of Pathology, Pharmacology and Toxicology, Department of Biomedical Sciences and

    Veterinary Public Health, Swedish University of Agricultural Sciences, P.O. Box 7028, SE-750 07 Uppsala, Sweden. E-mail: carina.

    [email protected]

    Abstract

    ObjectiveTo investigate the pharmacokinetics and

    effects of methadone on behaviour and plasma

    concentrations of cortisol and vasopressin in

    healthy dogs.

    Study design Randomized, cross-over, experimentaltrial.

    Animals Nine adult dogs (beagle and beagle cross

    breeds), four males and five females.

    MethodsMethadone hydrochloride, 0.4 mg kg)1,

    was administered intravenously (IV) and subcuta-

    neously (SC) with a crossover design. Drug and

    hormone analyses in plasma were performed using

    Liquid ChromatographyElectrospray Ionization

    Tandem Mass Spectrometry and radioimmunoassay

    respectively. Behavioural data were collected using

    a standardized protocol.

    Results After IV administration, the plasma con-

    centration of methadone at 10 minutes was

    82.1 9.2 ng mL)1 (mean SD), the terminal

    half-life was 3.9 1.0 hours, the volume of distri-

    bution 9.2 3.3 L kg)1 and plasma clearance

    27.9 7.6 mL minute)1 kg)1. After SC adminis-

    tration, time to maximal plasma concentration was

    1.26 1.04 hours and maximal plasma concen-

    tration of methadone was 23.9 14.4 ng mL)1,

    the terminal half-life was 10.7 4.3 hours and

    bioavailability was 79 22%. Concentrations of

    both cortisol and vasopressin were increased for an

    hour following IV methadone. The observed

    behavioural effects of methadone were decreased

    licking and swallowing and an increase in whiningafter SC administration. The latter finding is notable

    as it can be misinterpreted as pain when methadone

    is used as an analgesic.

    Conclusion and clinical relevance When methadone

    was administered by the SC route, the half-life was

    longer, but the individual variation in plasma

    concentrations was greater compared with IV admin-

    istration. Increased frequency of whining occurred

    after administration of methadoneand maybe a drug

    effect and not a sign of pain. Cortisol and vasopressin

    concentrations in plasma may not be suitable for

    evaluating analgesia after methadone treatment.

    Keywords behaviour, cortisol, dog, methadone,

    pharmacokinetics, vasopressin.

    Introduction

    Methadone is an opioidl-receptor agonist, which is

    frequently used both in companion animals as well

    48

    Veterinary Anaesthesia and Analgesia, 2010, 37, 4856 doi:10.1111/j.1467-2995.2009.00476.x

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    2/10

    as humans for the relief of pain. The use in animals

    is off-label as the drug is only approved for use in

    humans. Methadone is of higher relative intrinsic

    efficacy at the l-receptor than morphine (Selley

    et al. 2001). In human medicine, there are several

    reports of methadone effectiveness when treatingpain resistant to other opioids like morphine and

    hydromorphone and it has a high oral bioavail-

    ability and a long terminal half-life compared with

    many other opioids (Dale et al. 2002, 2004). In

    dogs, however, the oral bioavailability is low and

    terminal half-lives range from 1.754 hours and

    212 hours following intravenous (IV) and subcu-

    taneous (SC) administration, respectively (Schlitt

    et al. 1978; Garrett et al. 1985; Schmidt et al. 1994;

    Kukanich et al. 2005; Kukanich & Borum 2008).

    The effect of opioids on the release of cortisol

    seems to be highly species-dependent (Pfeiffer &

    Herz 1984; Pechnick 1993). An increase in

    plasma cortisol after single administration of mor-

    phine and butorphanol was reported in cats

    (Borrell et al. 1975) and dogs (Fox et al. 1998)

    respectively, but in cows (Nanda et al. 1992)

    and in humans, opioids usually decreased the

    concentrations of cortisol in plasma. Nevertheless,

    increases of cortisol concentrations in blood are

    often used as an indicator of distress and pain and

    also to evaluate analgesia in animals (Minton

    1994; Molony & Kent 1997; Devitt et al. 2005;

    Sibanda et al. 2006; Vinuela-Fernandez et al.

    2007). Hellebrekers et al. (1989) reported anincrease in concentrations of vasopressin in plasma

    after administration of methadone in dogs. There

    are also indications that both plasma cortisol and

    vasopressin become increased in dogs subjected to

    stress (Hydbring-Sandberg et al. 2004).

    The aim of the study was to describe the

    pharmacokinetic profile of methadone after IV and

    SC administration in dogs as well as to assess the

    effects on behaviour, cortisol and vasopressin

    plasma concentrations.

    Material and methods

    Animals and experimental procedure

    Ten clinically healthy Beagle and Beagle cross-bred

    dogs, AJ, five males and five females weighing

    17.0 3.1 kg (mean SD) and 5.6 3.0 years-

    old were used. They were born and housed at the

    Department of Clinical Sciences, Swedish University

    of Agricultural Sciences, Uppsala, Sweden. Nor-

    mally they lived in groups and were allowed out-

    door exercise every day.

    On the experimental days, the dogs were fed at

    0800 and were let out in the outdoor kennel pen

    according to the normal routines. At 0900, the dogs

    were weighed and prepared for the experiment. Thedogs front legs (dorsal area above carpus) were

    shaved and cleaned and one or two (when the dogs

    received intravenous drugs) catheters were inserted

    into the cephalic veins. One catheter was used for

    the IV administration and the other for blood

    sampling. A large funnel was placed around their

    neck and they were put into individual cages during

    the day. The Local Ethical Committee in Uppsala,

    Sweden approved the care of the animals and the

    experimental design.

    Drugs, study design and blood sampling

    Methadone hydrochloride (Metadon Recip, solution

    for injection 10 mg mL)1, RecipAB, Sweden) was

    used both for IV and SC administration at a dose of

    0.4 mg kg)1. In the control study, physiological

    saline solution (Natriumklorid Fresenius Kabi,

    solution for injection, 9 mg mL)1, Fresenius Kabi

    AB, Sweden) was administered IV. Methadone was

    administered IV in the cephalic vein via the catheter

    and was also administered SC in the neck region

    in a crossover design (Table 1). The order of the

    first drug administration was randomized. The wash

    out period between the different routes of adminis-tration was at least two weeks. Blood samples

    were collected before and 10, 30 and 60 minutes

    and 3, 6, 12, 22, 31 and 47 hours after adminis-

    tration of methadone. The venous catheter was used

    for blood samples up to and including 6 hours.

    The remaining blood samples were taken by direct

    cephalic vein puncture. In the control study, the

    same protocol for blood sampling as in the metha-

    done experiments was used. However, to avoid too

    much blood loss, sampling was only performed at

    10 and 60 minutes and 6 hours, while sampling at

    other time points was simulated. After 6 hours, the

    dogs returned to normal routines together with the

    other dogs.

    Blood samples, 3 mL, were collected into ice-

    chilled tubes containing EDTA (ethylenediamine-

    tetraacetic acid) as an anticoagulant. The blood was

    centrifuged at 1500 g for 20 minutes at 4 C. The

    plasma, 1.2 mL was stored at )20 C until drug

    analyses and the rest was stored at )80 C until

    hormone analyses.

    Methadone in dogs C Ingvast-Larsson et al.

    2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856 49

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    3/10

    Methadone assay

    To 0.5 mL plasma was added 100 lL internal

    standard solution [2H3]-methadone (14 ng

    100 lL)1 MeOH), 0.5 mL of MilliQ-water, 5.0 mL

    hexane/2-butanol (97:3) and 0.1 mL of sodium

    hydroxide. The mixture was extracted for 20 min-

    utes. After centrifugation, the organic phase was

    transferred to a glass tube and evaporated to dry-

    ness in a stream of nitrogen at 50 C. The residue in

    each vial was reconstituted in 60 lL of 0.1% formic

    acid in water. The reconstituted samples were

    quantified with liquid chromatographyelectrosprayionizationtandem mass spectroscopy (LCESIMS/

    MS). LC/MS/MS analyses were performed using a

    Surveyor LC system interfaced to a Finnigan TSQ

    Quantum Ultra (Thermo Electron Corporation, CA,

    USA) mass spectrometer. The separation was per-

    formed in a Zorbex Eclipse XAD-18 (2.1 50 mm,

    5 lM; Aglient Technologies, Sweden). The mobile

    phases were 0.2% formic acid in MilliQ-water (A)

    and methanol (B). The gradient program was: 0 to

    2.0 minutes, 15% B; 2.0 to 5.0 minutes, 15 to 85%

    B; 5.2 to 8.0 minutes, 15% B. The mobile phase was

    delivered at a flow rate of 200 lL minute)1. The

    mass spectrometer was run in selected reaction

    monitoring mode (SRM). The ionization technique

    was electrospray (ESI) in positive mode. The ESI

    source voltage was set at 3.5 kV and sheath gas

    flow-rate and auxiliary gas were 50 and 2 arbitrary

    units, respectively. When running collision-induced

    dissociation (CID), argon was used as the collision

    gas at a pressure of 1.5 mTorr. For SRM mode, the

    following transitions were recorded: methadone

    [M+H] m/z 310 fi 265 and [2H3]-methadone

    [M+H]+ m/z 313 fi 268 using collision energy

    20 V. The limit of quantification was 0.6 ng meth-

    adone mL)1 plasma. The standard curve was linear

    in the range 0.3 to 976 ng mL)1. The coefficients of

    variance for methadone at the 1 ng mL)1 level were

    6% (n = 6).

    Pharmacokinetic analyses

    The plasma concentrations versus time profile for

    methadone in each individual were analyzed using

    noncompartmental methods based on statisticalmoment theory (Gibaldi & Perrier 1982). A com-

    mercially available software program was used (Win

    Nonlin 5.0.1; Pharsight Corporation, CA, USA). The

    area under the curve (AUCinf) was calculated using

    the linear trapezoidal approximation. To extrapolate

    the area under the curve from time zero to infinity,

    the terminal rate constant (k) was used. The terminal

    half-lives were determined from t = ln2/k. The SC

    bioavailability (F) was calculated from the AUCsinfusing the equation:

    F% 100AUCinf;sc=AUCinf;iv

    The observed time (Tmax) to maximal plasma

    concentration (Cmax) and the Cmax were read from

    the plotted concentration-time curve for each indi-

    vidual animal after SC administration.

    Hormone assays

    Dilutions of plasma were parallel to the standard

    curve in the radioimmunoassays used. Plasma

    Table 1 Study design

    Day 1 Day 2 Day 3 Day 4 Day 5

    Dog Treatment Dog Treatment Dog Treatment Dog Treatment Dog Treatment

    First week

    A Saline D Saline G SC A IV C IV

    B* Saline E Saline H SC B SC D IV

    C Saline J SC I SC F IV E IV

    Second week

    F Saline I Saline B* IV E SC H IV

    G Saline J Saline C SC F SC I IV

    H Saline A SC D SC G IV J IV

    SC, methadone subcutaneously; IV, methadone intravenously; saline control days, saline administered IV.

    *Accidentally, dog B received another SC injection and all data were excluded.

    Wash-out period was 2 weeks between the two experimental weeks.

    Methadone in dogs C Ingvast-Larsson et al.

    50 2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    4/10

    cortisol was measured using the Coat-a-Count RIA

    (Diagnostic Products Corporation, CA, USA). The

    lower detection limit was 1.90 nmol L)1. The

    intra-assay coefficient of variance was

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    5/10

    Plasma cortisol and vasopressin concentrations

    Before IV and SC administration of methadone,

    the cortisol plasma concentrations were 86

    27 nmol L)1 and 72 25 nmol L)1 respectively

    (Fig. 2). After IV administration, the cortisol con-

    centration increased and was higher compared with

    control days at time 1 hour (p < 0.05). After SC

    administration, however, the cortisol concentration

    decreased and it was lower than the value before the

    administration at times 0.5, 1, 6 and 31 hours

    (p < 0.05), but there was no difference compared

    with the control values.

    The vasopressin concentration was 0.82 0.43pmol L)1 and 0.75 0.31 pmol L)1 before the IV

    and SC administration respectively (Fig. 3). Also the

    vasopressin concentration was higher after IV

    administration compared with the saline adminis-

    trations at times 10 minutes and 1 hour (p < 0.01).

    Behaviour

    The dogs were calm for most of the time during the

    study. In total, 13 different kinds of behaviours were

    recorded. Results are shown where the noticed

    behaviour was repeated more than three times

    during the day. The average of 90 observations

    between 1040 and 1540 hours was used in the

    statistical analyses.

    The dogs were lying down at 74 21 and

    73 27 times of the 90 observation occasions after

    IV and SC administration respectively. The corre-

    sponding value after saline administration was

    69 23 [NS (not significant)] versus either admin-

    istration. While they were lying down, they were

    noticed to sleep at 32 34, 33 28 and 31 25

    times after IV and SC administration and the control

    day respectively (NS).

    The dogs sat 14 20, 14 23 and 16 23times after IV and SC administration and on the

    Table 2 Pharmacokinetic parameters in plasma

    (mean SD) after intravenous and subcutaneous admin-

    istration of methadone (0.4 mg kg)1) to dogs (n = 9)

    Parameter

    Intravenous

    administration

    Subcutaneous

    administration

    AUC (ng hour mL)1)* 257 77.8 195 57.2

    Cl (mL kg)1 minute)1) 27.9 7.6 NA

    Vd(ss) (L kg)1) 9.2 3.3 NA

    T1/2,k (hour) 3.9 1.0 10.7 4.5

    T1/2,k (hour) 3.6 1.3 9.2 5.2

    Cmax(ng mL)1) NA 19.4 10.7

    Tmax(hour) NA 1.2 1.1

    F(%) NA 79 22

    AUC, area under the concentration-time curve from time 0 to

    infinity; Cl, total body clearance; Vd(ss), volume of distribution at

    steady state; T1/2,k, terminal half-life; Cmax, maximal plasma

    concentration after intramuscular administration; Tmax, theobserved time to Cmax. F, SC bioavailability; NA, not applicable.

    *Significant difference (Students paired t-test,p< 0.02).

    Significant difference (Students paired t-test,p< 0.001).

    Harmonic mean pseudo SD.

    0 1 20

    50

    100

    150

    200

    2 8 14 20 26 32 38 44 50

    IV

    SC

    C

    **

    #

    **

    Time (hours)

    Cortisolconcentration(nmolL

    1)

    Figure 2 Plasma cortisol concentration (mean SD) after

    intravenous (IV) and subcutaneous (SC) administration of

    methadone (0.4 mg kg)1) in dogs (n = 9). Control = cor-

    tisol concentration in samples taken at time 10 minutes, 1

    and 6 hours after IV administration of isotonic saline (C).

    *p < 0.05 versus the value before the SC administration

    #p < 0.05 IV versus C.

    0 1 20

    1

    2

    3

    4

    2 8 14 20 26 32 38 44 50

    IV

    SC

    C

    #

    #

    Time (hours)

    Vasopressinconcentration(pmolL

    1)

    Figure 3 Plasma vasopressin concentration (mean SD)

    after intravenous (IV) and subcutaneous (SC) administra-

    tion of methadone (0.4 mg kg)1) in dogs (n = 9). Con-

    trol = vasopressin concentration in samples taken at time

    10 minutes, 1 and 6 hours after IV administration of

    isotonic saline (C). #p < 0.05 or less IV versus C.

    Methadone in dogs C Ingvast-Larsson et al.

    52 2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    6/10

    control day (NS). The corresponding numbers for

    eye contact were 6 11, 7 9 and 8 9 times

    (NS).

    Methadone administered SC resulted in frequent

    whining compared with control days (Fig. 4).

    Licking and swallowing were decreased aftermethadone compared with after saline administra-

    tion (Fig. 4).

    Discussion

    The pharmacokinetic parameters after IV adminis-

    tration in this study were comparable with those

    previously reported in dogs. The methadone plasma

    clearance was in accordance with previous reportedvalues (Kukanich et al. 2005; Kukanich & Borum

    2008). The clearance value is of clinical importance

    because it can be used for calculating the mainte-

    nance dose. Methadone in dogs is mainly extracted

    by the liver (Garrett et al. 1985) and the high

    clearance value corresponds to 89% of the hepatic

    blood flow in dogs (Davies & Morris 1993), which

    implies that oral bioavailability would be poor. This

    suggestion is further supported by the findings that

    after oral administration of methadone (2 mg kg)1)

    in dogs the drug could not be detected in plasma

    (Kukanich et al. 2005). In humans, methadone is a

    low-clearance drug and the oral bioavailability is

    high (Meresaar et al. 1981).

    The volume of distribution (Vd(ss)), calculated in

    the present study, was large and was of the same

    magnitude as shown in both humans and

    dogs (Meresaar et al. 1981; Kukanich et al. 2005;

    Kukanich & Borum 2008). This parameter is

    required to calculate an appropriate loading dose.

    Terminal half-life post-IV administration has

    previously been reported to be between 24 hours

    when measuring methadone plasma concentration

    until 8 hours after administration (Garrett et al.

    1985; Kukanich et al. 2005; Kukanich & Borum2008) and approximately 4 hours, measuring the

    methadone plasma concentrations up to 15 hours

    (Schmidt et al. 1994). The longer the plasma

    concentrations are measured the less is the possi-

    bility that the half-life is underestimated. In the

    present study, we measured the concentrations of

    methadone in plasma up to between 24 and

    30 hours and the terminal half-life was 3.9 hours

    after IV administration. As the terminal half-life

    after IV administration is dependent both on volume

    of distribution and clearance the half-life in humans

    is considerably longer than in dogs. The published

    kinetic studies on methadone in humans indicated a

    terminal half-life of between 1555 hours (Meres-

    aar et al. 1981; Wolff et al. 1993, 1997). The

    terminal half-life is of clinical importance when

    choosing the dose interval.

    The half-life increased significantly after SC

    administration but also the individual variation

    compared with IV administration. The longer half-

    life is probably because of absorption rate-limited

    IV SC C0

    200

    400

    600

    800

    1000

    1200*

    Whining

    (no/30min)

    IV SC C

    0

    10

    20

    30

    40

    **

    Licking(no/30min)

    IV SC C

    0

    2

    4

    6

    8

    *

    *

    Swallowing(no/30min)

    (a)

    (b)

    (c)

    Figure 4 Whining, licking and swallowing frequencies

    during 30 minutes (mean SD of 90 observations in

    six 5-minute sessions) recorded after intravenous (IV) and

    subcutaneous (SC) administration of methadone

    (0.4 mg kg)1) in dogs (n = 9) and during days when

    saline was administered (C). *p < 0.05 versus C.

    Methadone in dogs C Ingvast-Larsson et al.

    2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856 53

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    7/10

    elimination (flip-flop phenomenon). The variability

    in absorption kinetics is dependent on the site of

    administration and also of massaging of the admin-

    istration site (Rowland & Tozer 1995). The

    increased half-life is an advantage if the drug will

    be used for a longer period as the dose interval canbe longer. With a half-life of 11 hours once to twice

    daily dosing will be adequate. The SC bioavailability

    was high, but the Cmaxwas low compared with the

    initial methadone plasma concentrations received

    after IV administration. SC administration will

    produce less plasma fluctuations but also a need

    for a loading dose as a steady state will not occur

    until about approximately 2 days (4 half-lives). The

    most suitable approach if immediate analgesia is

    required is to begin with IV administration of the

    drug and after approximately 3 hours start the SC

    dosage regimen.

    After IV administration of methadone, the plasma

    concentrations of both cortisol and vasopressin

    increased. These effects correlated with the drug

    plasma concentrations, were short-lasting and the

    hormone concentrations were back to control

    values within 3 hours post-drug administration.

    Increased cortisol plasma concentration after acute

    administration of opioids has been reported in many

    other species including rat (Guaza et al. 1979),

    mouse, guinea pig, (Pechnick 1993), cat (Borrell

    et al. 1975; Guaza et al. 1979) and goat (Ingvast-

    Larsson et al. 2007). Vasopressin concentrations

    after opioid administration have been reported toincrease in the dog (Hellebrekers et al. 1989) and in

    the goat (Ingvast-Larsson et al. 2007), but the

    majority of studies have suggested an inhibition of

    vasopressin release of opioids in vivo(Pfeiffer & Herz

    1984). Increased cortisol plasma concentrations are

    often used as a marker of pain and stress in animals

    and increased vasopressin concentrations in con-

    nection with stress and pain have also been

    suggested (Hydbring et al. 1999; Hydbring-Sand-

    berg et al. 2004). After SC administration, no

    increases of hormone concentrations were observed;

    most likely because of the low plasma concentra-

    tions of methadone. Instead, the plasma concentra-

    tions of cortisol were lower when compared to the

    concentrations before drug administration probably

    reflecting the dogs becoming stressed by the han-

    dling before the drug was administered. The results

    show that increased hormone concentrations are

    not useful parameters when assessing analgesic

    efficacy of methadone in dogs as the drug per se

    induced changes in hormone concentrations in

    plasma. The plasma concentrations of methadone

    were unlikely to have exceeded clinically relevant

    concentrations as the dose used was in the lower

    range of the dose interval that often is recom-

    mended in dogs (Plumb 2005). In humans, meth-

    adone plasma concentrations eliciting 50% ofmaximal pain relief are reported to be between

    53604 ng mL)1 (Inturrisi et al.1990; Garrido &

    Troconiz 2000). The individual variation in hu-

    mans is dependent on previous exposure to opioids

    (cross-tolerance) and degree of pain. A mean

    estimate for methadone plasma concentrations

    eliciting 50% of maximal pain relief in nontolerant

    subjects was 60 ng mL)1 (Gourlay et al.1984). In

    this study, the dogs at the plasma concentrations of

    between 5282 ng methadone mL)1 were calm and

    the hormone concentrations were increased, but the

    plasma concentration able to provide analgesia in

    dogs is not yet established.

    The individual variations in plasma vasopressin

    concentration were large and thus the values were

    high in some dogs. This vasopressin concentration

    will cause a maximal antidiuretic effect in the

    kidney, which should be taken into account in the

    evaluation of fluid therapy if methadone is admin-

    istered.

    We observed drug-induced effects on behaviour.

    For technical reasons, the same person had to be

    responsible for both blood sampling and the obser-

    vations of behaviour and hence the administration

    order could not be blinded. This was unfortunate,but not considered as a major obstacle to perform

    the study, since the observations were done using

    standardized scoring. Methadone increased the

    whining and this phenomenon was most obvious

    after SC administration. It is important to appreciate

    that methadone may cause whining and this should

    not be misinterpreted as the dog being in pain. A

    similar result has been observed with butorphanol

    where dogs vocalized both after butorphanol alone

    and after surgery and butorphanol (Fox et al.

    2000). Behaviours such as licking and swallowing

    known to be typical for dogs experiencing discom-

    fort (Beerda et al. 1997, 1998) decreased after

    methadone administration indicating whining was

    not because of discomfort. None of the dogs vomited

    and no signs of agitation and excitement were

    noticed in any dog. Morphine, another opioid acting

    on the l-receptor, has been shown to induce

    vomiting in dogs (Kukanich et al. 2005).

    In conclusion, when methadone was adminis-

    tered by the SC route of administration the half-life

    Methadone in dogs C Ingvast-Larsson et al.

    54 2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    8/10

    was longer, but the inter-individual variation in

    plasma concentrations was greater compared with

    IV administration. Increased frequency of whining

    occurred after administration of methadone and

    may be a drug effect and not a sign of pain. Cortisol

    and vasopressin concentrations in plasma are notsuitable for evaluating analgesia after IV methadone

    treatment. Future studies are required to establish

    the methadone plasma concentration needed for

    analgesia in the dog.

    Acknowledgements

    This study was supported by the Swedish Animal

    Welfare Agency and by the Swedish Research

    Council Medicine. We thank Gunilla Drugge,

    Department of Anatomy, Physiology and Biochem-

    istry and Emma Hornebro, Department of Clinical

    Sciences, Swedish University of Agricultural Sci-

    ences, Uppsala, Sweden for excellent assistance.

    References

    Beerda B, Schilder MB, vanHooff JA et al. (1997) Mani-

    festations of chronic and acute stress in dogs. Appl

    Animal Behav 52, 307319.

    Beerda B, Schilder MB, van Hooff JA et al. (1998)

    Behavioural, saliva cortisol and heart rate responses to

    different types of stimuli in dogs. Appl Animal Behav 58,

    365381.

    Borrell J, Llorens I, Borrell S (1975) Adrenal, plasma and

    urinary corticosteroids during single or repeated

    administration of morphine in cats. Eur J Pharmacol 31,

    237242.

    Dale O, Hoffer C, Sheffels P et al. (2002) Disposition of

    nasal, intravenous, and oral methadone in healthy

    volunteers. Clin Pharmacol Ther 72, 536545.

    Dale O, Sheffels P, Kharasch ED (2004) Bioavailabilities of

    rectal and oral methadone in healthy subjects. Br J Clin

    Pharmacol 58, 156162.

    Davies B, Morris T (1993) Physiological parameters in

    laboratory animals and humans. Pharm Res 10, 1093

    1095.

    Devitt CM, Cox RE, Hailey JJ (2005) Duration, complica-

    tions, stress, and pain of open ovariohysterectomy versusa simple method of laparoscopic-assisted ovariohysterec-

    tomy in dogs. J Am Vet Med Assoc 227, 921927.

    Fox SM, Mellor DJ, Lawoko CR et al. (1998) Changes in

    plasma cortisol concentrations in bitches in response to

    different combinations of halothane and butorphanol,

    with or without ovariohysterectomy. Res Vet Sci 65,

    125133.

    Fox SM, Mellor DJ, Stafford KJ et al. (2000) The

    effects of ovariohysterectomy plus different combina-

    tions of halothane anaesthesia and butorphanol

    analgesia on behaviour in the bitch. Res Vet Sci 68,

    265274.

    Garrett ER, Derendorf H, Mattha AG (1985) Pharmacoki-

    netics of morphine and its surrogates. VII: High-perfor-

    mance liquid chromatographic analyses and

    pharmacokinetics of methadone and its derived metab-

    olites in dogs. J Pharm Sci 74, 12031214.

    Garrido MJ, Troconiz IF (2000) Methadone: a review of its

    pharmacokinetic/pharmacodynamic properties. J Phar-

    macol Toxicol Methods 42, 6166.

    Gibaldi M, Perrier D (1982) Noncompartmental analysis

    based on statistical moment theory. In: Pharmacoki-

    netics (2nd edn). Swarbrick J (ed). Marcel Dekker, New

    York, NY, USA. pp. 409417.

    Gourlay GK, Willis RJ, Wilson PR (1984) Postoperative

    pain control with methadone: influence of supplemen-

    tary methadone doses and blood concentration-response

    relationships. Anesthesiology 61, 1926.

    Guaza C, Torellas A, Borell J et al. (1979) Effects of mor-phine upon the pituitary-adrenal system and adrenal

    catecholamines: a comparative study in cats and rats.

    Pharmacol Biochem Behav 11, 5763.

    Hellebrekers LJ, van den Brom WE, Mol JA (1989) Plasma

    arginine vasopressin response to intravenous metha-

    done and naloxone in conscious dogs. J Pharmacol Exp

    Ther 248, 329333.

    Hydbring E, Madej A, MacDonald E et al. (1999) Hor-

    monal changes during parturition in heifers and goats

    are related to the phases and severity of labour.

    J Endocrinol 160, 7585.

    Hydbring-Sandberg E, von Walter LW, Hoglund K et al.

    (2004) Physiological reactions to fear provocation in

    dogs. J Endocrinol 180, 439448.

    Ingvast-Larsson C, Svartberg K, Hydbring-Sandberg E

    et al. (2007) Clinical pharmacology of buprenorphine in

    healthy, lactating goats. J Vet Pharmacol Ther 30, 249

    256.

    Inturrisi CE, Portenoy RK, Max MB et al. (1990) Phar-

    macokinetic-pharmacodynamic relationships of metha-

    done infusions in patients with cancer pain. Clin

    Pharmacol Ther 47, 565577.

    Kukanich B, Borum SL (2008) The disposition and

    behavioral effects of methadone in Greyhounds. Vet

    Anaesth Analg 35, 242248.

    Kukanich B, Lascelles BD, Aman AM et al. (2005) The

    effects of inhibiting cytochrome P450 3A, p-glycopro-tein, and gastric acid secretion on the oral bioavailability

    of methadone in dogs. J Vet Pharmacol Ther 28, 461

    466.

    Meresaar U, Nilsson MI, Holmstrand J et al. (1981) Single

    dose pharmacokinetics and bioavailability of methadone

    in man studied with a stable isotope method. Eur J Clin

    Pharmacol 20, 473478.

    Minton JE (1994) Function of the hypothalamic-pituitary-

    adrenal axis and the sympathetic nervous system in

    Methadone in dogs C Ingvast-Larsson et al.

    2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856 55

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    9/10

    models of acute stress in domestic farm animals. J Anim

    Sci 72, 18911898.

    Molony V, Kent JE (1997) Assessment of acute pain in

    farm animals using behavioural and physiological

    measurements. J Anim Sci 75, 266272.

    Nanda AS, Dobson H, Ward WR (1992) Opioid

    modulation of the hypothalamo-pituitary-adrenal

    axis in dairy cows. Domest Anim Endocrinol 9, 181

    186.

    Pechnick RN (1993) Effects of opiods on the hypothalamo-

    pituitary-adrenal axis. Annu Rev Pharmacol Toxicol 32,

    353382.

    Pfeiffer A, Herz A (1984) Endocrine actions of opioids.

    Horm Metab Res 16, 386397.

    Plumb DC (2005) Veterinary Drug Handbook. (5th edn),

    Pharma vet Inc, Stockholm, WI, USA. pp 718719.

    Rowland M, Tozer NT (1995) Clinical Pharmacokinetics,

    Concepts and Applications. (3rd edn), Lippincott

    Williams & Wilkins, Philadelphia, PA, USA. p. 127.

    Schlitt SC, Schroeter LM, Wilson JE et al. (1978) Metha-done-induced respiratory depression in the dog: com-

    parison of steady-state and rebreathing techniques and

    correlation with serum drug concentration. J Pharmacol

    Exp Ther 207, 109122.

    Schmidt N, Brune K, Williams KM et al. (1994) Stereose-

    lective pharmacokinetics of methadone in beagle dogs.

    Chirality 6, 492495.

    Selley DE, Cao CC, Sexton T et al. (2001) l-Opioid recep-

    tor-mediated G-protein activation by heroin metabolites:

    evidence for greater efficacy of 6-monoacetylmorphine

    compared with morphine. Biochem Pharmacol 62,

    447455.

    Sibanda S, Hughes JM, Pawson PE et al. (2006) The effects

    of preoperative extradural bupivacaine and morphine on

    the stress response in dogs undergoing femoro-tibial

    joint surgery. Vet Anaesth Analg 33, 246257.

    Vinuela-Fernandez I, Jones E, Welsh EM et al. (2007) Pain

    mechanismsand their implication for the management of

    pain in farm and companion animals. Vet J 174, 227

    239.

    Wolff K, Hay AW, Raistrick D et al. (1993) Steady-state

    pharmacokinetics of methadone in opioid addicts. Eur J

    Clin Pharmacol 44, 189194.

    Wolff K, Rostami-Hodjegan A, Shires S et al. (1997) Thepharmacokinetics of methadone in healthy subjects and

    opiate users. Br J Clin Pharmacol 44, 325334.

    Received 8 July 2008; accepted 22 September 2008.

    Methadone in dogs C Ingvast-Larsson et al.

    56 2010 The Authors. Journal compilation 2010 Association of Veterinary Anaesthetists, 37, 4856

  • 7/27/2019 Clinical Pharmacology of Methadone in Dogs.

    10/10

    Copyright of Veterinary Anaesthesia & Analgesia is the property of Blackwell Publishing Limited and its

    content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's

    express written permission. However, users may print, download, or email articles for individual use.