ArticlePDF Available

Pharmacokinetics of intramuscularly administered morphine in horses

Authors:

Abstract

Objective: To determine the pharmacokinetics of morphine after IM administration in a clinical population of horses. Design: Prospective clinical study. Animals: 77 horses. Procedures: Morphine sulfate (0.1 mg/kg [0.045 mg/lb], IM) was administered to horses, and blood samples were obtained at predetermined time points. Plasma morphine concentrations were measured via liquid chromatography and mass spectrometry. In preliminary investigations, samples were obtained from 2 healthy horses at 12 time points (up to 12 hours after drug administration) and analyzed via 2-stage pharmacokinetic analysis. In the clinical phase, blood samples were obtained from 75 hospitalized horses at various times (total, 2 to 3 samples/horse) up to 9 hours after drug administration, and data were analyzed via a naïve pooled pharmacokinetic model. Results: In the clinical phase, the apparent terminal half-life (t(½)) of morphine was approximately 1.5 hours, volume of distribution per bioavailability was approximately 4.5 L/kg, and clearance per bioavailability was approximately 35 mL/kg/min. Peak plasma concentration in naïve pooled analysis was 21.6 ng/mL and occurred approximately 4 minutes after administration. Morphine concentrations were below the limit of quantification ≤ 7 hours after administration in 74 horses. Adverse effects attributed to morphine administration were uncommon and considered mild. Conclusions and clinical relevance: The short t(½) of morphine in horses suggested frequent administration may be needed to maintain targeted plasma concentrations. Variations in plasma concentrations suggested optimal dosages may differ among horses. The drug was well tolerated at the described dose, but patients receiving morphine should be monitored carefully.
JAVMA, Vol 243, No. 1, July 1, 2013 Scientific Reports 105
EQUINE
The use of analgesics in horses has been evolving over
the past few decades. As veterinarians and horse
owners have become more aware of the importance of
pain management, there has been a push toward use of
multimodal analgesia. Morphine is a µ-opioid agonist
that was formerly used routinely in equine practice as
a preanesthetic and analgesic agent. However, the use
of morphine in equine practice has waned because of
the potential for adverse effects. Gastrointestinal stasis1
and CNS excitation2 are among the described adverse
effects of morphine administered at high doses. New-
er classes of drugs, such as α-2 receptor agonists and
agonist-antagonist opioids, are associated with fewer
adverse effects and have begun to take the place of mor-
phine in routine practice. As the importance of analge-
sia has become more readily accepted, administration
of NSAIDs has become common in equine practice.
Pharmacokinetics of intramuscularly
administered morphine in horses
Elizabeth P. Devine, DVM, MS, DACVS; Butch KuKanich, DVM, PhD, DACVCP; Warren L. Beard, DVM, MS, DACVS
Objective—To determine the pharmacokinetics of morphine after IM administration in a
clinical population of horses.
Design—Prospective clinical study.
Animals—77 horses.
Procedures—Morphine sulfate (0.1 mg/kg [0.045 mg/lb], IM) was administered to horses,
and blood samples were obtained at predetermined time points. Plasma morphine concen-
trations were measured via liquid chromatography and mass spectrometry. In preliminary
investigations, samples were obtained from 2 healthy horses at 12 time points (up to 12
hours after drug administration) and analyzed via 2-stage pharmacokinetic analysis. In the
clinical phase, blood samples were obtained from 75 hospitalized horses at various times
(total, 2 to 3 samples/horse) up to 9 hours after drug administration, and data were analyzed
via a naïve pooled pharmacokinetic model.
Results—In the clinical phase, the apparent terminal half-life (t1/2) of morphine was approxi-
mately 1.5 hours, volume of distribution per bioavailability was approximately 4.5 L/kg, and
clearance per bioavailability was approximately 35 mL/kg/min. Peak plasma concentration
in naïve pooled analysis was 21.6 ng/mL and occurred approximately 4 minutes after ad-
ministration. Morphine concentrations were below the limit of quantification 7 hours after
administration in 74 horses. Adverse effects attributed to morphine administration were
uncommon and considered mild.
Conclusions and Clinical Relevance—The short t1/2 of morphine in horses suggested fre-
quent administration may be needed to maintain targeted plasma concentrations. Varia-
tions in plasma concentrations suggested optimal dosages may differ among horses. The
drug was well tolerated at the described dose, but patients receiving morphine should be
monitored carefully. (J Am Vet Med Assoc 2013;243:105–112)
However, the use of these medications is not without
consequence; complications of NSAID administration
include right dorsal colitis, gastric ulceration, and re-
nal failure. Despite these effects, NSAIDs are widely
used because of their anti-inflammatory and analgesic
properties. For some conditions, including laminitis,
infected synovial structures, and fractures, the degree
of analgesia obtained with NSAIDs alone is inadequate,
and multimodal analgesia, such as the combined use of
opioids and NSAIDS, can be beneficial.
Most pharmacokinetic studies typically involve a
small number of healthy horses (eg, 4 to 8) and frequent
blood sample collection to determine the absorption,
distribution, metabolism, and excretion of the drug in
each member of a homogenous group. The mean, me-
dian, and range of pharmacokinetic parameters are re-
ported. These studies, termed standard 2-stage studies,
are useful for estimation of mean parameters in a well-
defined, homogenous population. However, a small,
From the Departments of Clinical Sciences (Devine, Beard) and Anat-
omy and Physiology (KuKanich), College of Veterinary Medicine,
Kansas State University, Manhattan, KS 66502.
This manuscript represents a portion of a thesis submitted by Dr.
Devine to the Kansas State University Department of Biomedical
Sciences as partial fulfillment of the requirements of a Master of
Science degree.
Supported in part by the Department of Clinical Sciences Research
Committee Funding and the Analytical Pharmacology Laboratory
at Kansas State University.
Address correspondence to Dr. Devine (devine@vet.k-state.edu).
ABBREVIATIONS
Cl Clearance
Cl/F Clearance per bioavailability
LOQ Limit of quantification
t1/2 Apparent terminal half-life
Vd Volume of distribution
Vd/F Volume of distribution per bioavailability
106 Scientific Reports JAVMA, Vol 243, No. 1, July 1, 2013
EQUINE
homogenous group of healthy animals may not be rep-
resentative of the clinical population of horses that may
benefit from morphine as an analgesic.
Several other methods for assessing the pharmaco-
kinetics of a drug are available, including population
pharmacokinetics and naïve pooled pharmacokinet-
ics. In contrast to standard 2-stage pharmacokinetic
studies, population and naïve pooled pharmacokinetic
studies may involve large numbers of animals, often
including the targeted clinical population, and do not
require frequent sample collection from individual pa-
tients. Population pharmacokinetics includes a larger,
more diverse group, compared with standard 2-stage
studies, and allows biological variability to be evalu-
ated. Factors affecting variability in a veterinary popu-
lation may include age, breed, body weight, concurrent
medications, and health status.3,4 Population pharma-
cokinetics allows identification of patient traits that
may alter the pharmacokinetics of a drug; however, one
of the disadvantages of this type of study is that fitting
a model to the data can be very difficult with some data
sets.
Naïve pooled pharmacokinetic studies are often
used to minimize sample collection from individual pa-
tients and to allow for evaluation of a group of patients.
They include large numbers of individuals and can be
performed in the targeted clinical population. In con-
trast to population pharmacokinetics, a single model is
fit to pooled data from all individual patients. There-
fore, factors such as age, breed, and body weight cannot
be assessed for effects on pharmacokinetic parameters.
However, naïve pooled pharmacokinetic studies typi-
cally do not require the specialized software needed for
population pharmacokinetics, and a single model is fit
to all of the data, making the modeling less complex.
Although morphine has been used for many years
in horses, pharmacokinetic parameters for the drug are
not well described in this species. The dose and fre-
quency of IM administration of morphine have been
empirical, selected on the basis of the practitioner’s
experience and on the degree of discomfort evidenced
by clinical signs in the horse. Because adverse effects
can result from excessive morphine administration, it
is imperative that a proper dose and administration fre-
quency be clarified to minimize these effects. The first
step in identifying an appropriate dose is to determine
pharmacokinetic parameters of the drug following ad-
ministration via the desired route. The purpose of the
study reported here was to determine the pharmacoki-
netics of morphine after IM administration in a clini-
cal population of horses. This population was chosen
to allow an assessment of the variability in the plasma
concentrations of morphine in patients receiving the
drug as premedication for anesthesia or as treatment
for signs of pain instead of in healthy research animals.
Materials and Methods
Animals—Two investigator-owned healthy adult
Quarter Horses, a 9-year-old gelding and a 17-year-old
mare, were used for initial evaluation of plasma mor-
phine concentrations and to determine the LOQ of the
drug prior to the clinical phase of the study. These
horses had no abnormalities detected via physical ex-
amination and were known to have been healthy for
4 and 6 years. For the clinical portion of the study, all
horses admitted to the Kansas State University Veteri-
nary Medical Teaching Hospital from December 2010
to June 2011 were eligible for enrollment. Horses were
included if morphine was administered as a periop-
erative analgesic or as treatment for signs of pain. All
horses were treated according to their clinical signs
and diagnosis as deemed necessary by the supervising
clinician. Pregnant mares and horses known to have
impaired pulmonary, hepatic, or renal function or en-
docrine disorders were not included in the study. The
preliminary investigation and the clinical study were
approved by the Kansas State University Institutional
Animal Care and Use Committee and performed in
accordance with that committee’s guidelines. Prior to
enrollment of client-owned horses in the study, own-
ers were informed of the nature of the study and pro-
vided their consent.
Preliminary investigation—Both horses were indi-
vidually housed and had access to food and water dur-
ing the preliminary investigation. The skin over the left
jugular vein was aseptically prepared, and an IV cath-
eter was placed to allow for serial blood sample collec-
tion. The 2 horses were weighed, and morphine sulfatea
(0.1 mg/kg [0.045 mg/lb], IM) was administered in the
left side of the neck at time 0. Whole blood samples
(7 mL) were collected into heparinized tubes at 5, 10,
15, 30, and 45 minutes and 1, 2, 4, 6, 8, 10, and 12
hours after morphine administration. The IV catheter
was flushed with heparinized saline (0.9% NaCl) so-
lution after each blood sample was collected and was
removed after the last sample was obtained. Paired
(duplicate) sets of blood samples were obtained from 1
horse (randomly selected via coin toss).
Physical examinations were performed at the time
of morphine administration and every 2 hours thereaf-
ter for 12 hours to monitor for any potential adverse ef-
fects. In each examination, heart rate, respiratory rate,
and rectal temperature were measured; the abdomen
was auscultated for presence of borborygmus; defeca-
tions were noted; the injection site was monitored for
heat and swelling; demeanor was assessed; and signs of
sedation (mild [lack of interest in the surroundings],
moderate [ataxic when walked], or severe [recumbent
or unresponsive]) or excitation (mild, [restlessness in
the stall], moderate [constant walking or pacing], or se-
vere [attempts to climb in the stall or other potentially
dangerous behavior]) were recorded. Horses were ad-
ditionally monitored for signs of colic or other adverse
effects.
Blood samples were centrifuged for 5 minutes at
5,000 X g, and the plasma was frozen 2 hours after
sample collection. The duplicate blood samples ob-
tained from 1 horse were stored on ice until the end of
the day and centrifuged together at that time. There-
fore, samples collected at the beginning of the day were
on ice for just over 12 hours before the serum was
separated, and the last sample obtained was centrifuged
within 1 hour after collection. This was done to deter-
mine whether sample handling would affect results.
All plasma samples were stored in a freezer at –70°C
prior to analysis. Morphine concentrations were deter-
JAVMA, Vol 243, No. 1, July 1, 2013 Scientific Reports 107
EQUINE
mined by means of liquid chromatography and mass
spectrometry.
Clinical phase—Horses were weighed, and mor-
phinea (0.1 mg/kg, IM) was administered in the neck
at time 0. The side of the neck used for morphine ad-
ministration varied among horses and was chosen to
avoid sites where other medications had been previous-
ly administered. Two or 3 blood samples (7 mL each)
were collected from each horse at different times after
morphine administration until approximately 9 hours
after injection. Samples were obtained via jugular ve-
nipuncture or from an IV catheter, when present. The
time of sample collection was recorded to the minute,
and sample collection times were evenly distributed
throughout the 9-hour collection period. After collec-
tion, samples were refrigerated at 4°C until centrifuga-
tion. Blood samples were centrifuged as previously de-
scribed 8 hours after collection, and the plasma was
separated and stored in a freezer at –70°C until analy-
sis. Physical examinations were performed at the time
of morphine administration, every 2 hours thereafter
(until the time point established in the preliminary in-
vestigation at which morphine was no longer detect-
able in samples), and at the time of each blood sample
collection. Examinations included monitoring the same
variables as described for the preliminary investigation;
evaluations were not performed when a horse was in
surgery or recovering from surgery, or if the patient was
discharged from the hospital before a scheduled exami-
nation. Concurrent medications received, including
anesthetic agents and IV fluids, were recorded for each
patient along with breed, age, body weight, sex, and di-
agnosis. Feed was withheld for 6 to 8 hours prior to
anesthesia for horses that were anesthetized for surgery.
Water was not withheld.
Analysis of plasma morphine concentrations
Liquid chromatography and mass spectrometry were
used for plasma sample analysis. Samples were thawed
on a heat block at 40°C and centrifuged at 5,000 X g
for 5 minutes. Hydromorphone D3b (internal standard;
0.1 mL; 100 ng/mL) and borate buffer (1 mL; 0.1M; pH,
9.2) were added to 1 mL of plasma, and the sample was
mixed via vortexing for 5 seconds.
The C18 solid-phase extraction cartridgesc were at-
tached to a solid-phase extraction manifold and then
conditioned with 1 mL of methanol followed by 1 mL
of deionized water. The plasma, hydromorphone, and
buffer mixtures were run through the conditioned car-
tridges attached to the solid-phase extraction manifold,
and then the cartridges were rinsed with 1 mL of deion-
ized water each. One milliliter of methanol was added
to each cartridge to elute the sample. The methanol
was evaporated to dryness at 40°C under an air stream
for 30 minutes. Each sample was then resuspended in
0.2 mL of 50% methanol in deionized water and mixed
thoroughly. The sample was centrifuged at 15,000 X g
for 5 minutes, and the supernatant was removed and
placed in injection vials.
Plasma morphine standards were processed at least
twice daily in an identical manner to that used for sam-
ples to ensure consistency of the assay. The interday ac-
curacy of the assay determined on replicates of 5 at con-
centrations of 2.5, 10.0, and 50.0 ng/mL were 102%,
98%, and 99% of the actual concentration, respectively.
The interday coefficients of variation determined on
replicates of 5 at concentrations of 2.5, 10.0, and 50.0
ng/mL were 9%, 9%, and 7%, respectively. The analytic
LOQ was 2.5 ng/mL, defined as the lowest concentra-
tion of the standard curve with measured concentra-
tions within 15% of the actual concentration.
Pharmacokinetic analysis—Only plasma samples
with morphine concentrations greater than the LOQ
were included in the pharmacokinetic analysis. A
1-compartment open model with first-order input and
output with no lag was used for analysis of samples
from the preliminary investigation and for naïve pooled
pharmacokinetic analysis in the clinical phase. Popula-
tion pharmacokinetic modeling was assessed via a soft-
ware program with a nonlinear mixed-effects model.d
One- and 2-compartment models with first-order input
and output with and without lag were assessed. The
primary pharmacokinetic parameters estimated were
the Vd, absorption rate constant, and elimination rate
constant. Secondary pharmacokinetic parameters in-
cluded the apparent absorption half-life, t1/2, area un-
der the curve, Cl, maximum plasma concentration, and
time to the maximum plasma concentration.
Statistical analysis—A Friedman repeated-mea-
sures ANOVA on ranks test was used to analyze heart
rate and respiratory rate data. Comparison between
the duplicate samples was performed by calculating
the difference between samples centrifuged 2 hours
after collection and those batched for later centrifuga-
tion. Then, a linear regression model was used to de-
termine a relationship between the samples centrifuged
2 hours after collection and those centrifuged up to
12 hours after collection. Statistical softwaree was used
for data analysis. Values of P < 0.05 were accepted as
significant for all statistical tests.
Results
Preliminary investigation—Measured variables
evaluated in the physical examinations remained with-
in respective reference ranges throughout the 12-hour
evaluation period after morphine administration in
both horses. There was no change in behavior score
observed, and no evidence of a local injection site reac-
tion was detected in either horse. Signs of gastrointes-
tinal motility were considered normal and no signs of
colic were detected. One horse defecated between the
2- and 4-hour time points and again between the 4- and
6-hour time points. The other horse defecated between
the 4- and 6-hour time points and again between the
8- and 10-hour time points. No adverse effects were ob-
served during the study period.
Peak plasma morphine concentrations in the 2
horses were estimated at 13 and 24 minutes, with
concentrations of 33.5 and 28.2 ng/mL, respectively.
Concentrations were above the LOQ (2.5 ng/mL) for
8 hours in one horse and 6 hours in the other. For du-
plicate samples obtained from 1 horse to evaluate the
effects of sample handling on drug concentrations, val-
ues were compared between the paired samples and the
percentage difference between samples collected at the
108 Scientific Reports JAVMA, Vol 243, No. 1, July 1, 2013
EQUINE
same time points was determined. On the basis of lin-
ear regression analysis with a weighting factor of 1/y2,
where y is the plasma drug concentration, the R2 value
of 0.991 (P < 0.001) indicated a strong linear relation-
ship between the samples centrifuged for plasma sepa-
ration at different times (1 to 6 hours after collection;
correlation was not evaluated at later time points be-
cause of the low morphine concentrations), suggesting
drug concentrations in samples remained stable for up
to 6 hours after collection.
Clinical phase—Seventy-five horses were in-
cluded in the clinical phase of the study. One horse, a
10-month-old Quarter Horse colt, was included twice
because it received morphine on 2 separate visits to the
clinic but was only included once in signalment-related
data. Quarter Horses (52/75 horses) were overrepre-
sented. Other breeds or breed types included warm-
bloods (n = 4), American Paint Horse (4), draft breeds
(2), Tennessee Walking Horse (2), Thoroughbred (2),
Arabian (2), mixed (2), and Morgan, Saddlebred, Mus-
tang, Rocky Mountain Spotted Horse, and Pony of the
Americas (1 each). Eighteen mares, 22 geldings, and
35 stallions were included. The mean age was 6.2 years
(range, 7 months to 31 years). Body weights ranged
from 204 to 896 kg (449 to 1,971 lb), with a mean of
415 kg (913 lb).
Seventeen of 75 horses were treated for orthopedic
abnormalities. Six of these horses did not undergo sur-
gery; these included horses with laminitis, foot abscess,
severe osteoarthritis, laceration that extended into a
joint space, and fractured sacral and caudal vertebrae.
The remaining 11 horses with orthopedic abnormali-
ties underwent surgical procedures, including tarso-
crural, middle carpal joint, and metatarsophalangeal
joint arthroscopy; right hind proximal interphalangeal
joint (pastern) arthrodesis; repair of an ulnar fracture;
removal of the apical fragment of a fractured sesamoid
bone; desmotomy of the accessory ligaments of the
deep and superficial digital flexor tendons; and seques-
trum removal from a right second metacarpal bone.
Thirty-four horses (including 4 that were cryptorchid)
underwent castration; the horse that received morphine
twice during the study had surgery postponed on the
first visit and underwent castration on the second visit.
Fifteen horses underwent soft tissue surgery other than
castration, which included surgery of the upper respi-
ratory tract, treatment of postoperative colic, repair of
a rectovaginal tear, palmar digital neurectomy, neuro-
ma removal, abdominal hernia repair, and esophageal
diverticulum repair. Three horses underwent surgical
treatment of oral or facial conditions, including incisor
removal, repair of a mandibular fracture, and frontona-
sal sinus flap surgery for ventral conchal sinusitis. Four
horses underwent ocular surgery, including removal of
a third eyelid, tumor (periocular squamous cell carci-
noma) resection, and enucleation following rupture of
a corneal ulcer. The final category included 2 horses
with soft tissue lacerations. In total, 59 horses under-
went procedures requiring general anesthesia.
Of 76 patients that received morphine during this
part of the study, 74 also received phenylbutazone or
flunixin meglumine. Because plasma morphine con-
centrations were not reliably detectable after the 8-hour
time point in the preliminary investigation, monitor-
ing of the described physical examination variables
was discontinued after 8 hours in the clinical phase of
the study. In addition, 25 horses were discharged from
the hospital before the 8-hour time point, reducing the
number of data points included in subsequent analyses.
In clinical patients, the median respiratory rate
was 20 breaths/min at time 0 and ranged from 18 to 24
breaths/min after morphine administration (Table 1).
There was no difference in respiratory rate among time
points (P = 0.742). Median heart rate was 44 beats/min
at time 0 and ranged from 40 to 44 beats/min afterward.
Median heart rate at 8 hours (40 beats/min) was signifi-
cantly (P = 0.028) lower than that at 4 hours (44 beats/
min), but no other significant differences were detected
for this variable.
Fecal output was recorded every 2 hours for each
horse for the duration of hospitalization; however,
discharge of 25 horses prior to the 8-hour time point
likely resulted in some defecations not having been re-
corded in this interval. Of 17 horses that did not un-
dergo general anesthesia, 2 defecated before the 2-hour
time point, 3 defecated between the 2- and 4-hour time
points, and 4 defecated between the 4- and 6-hour time
points. The remaining horses either did not defecate
by 8 hours (n = 2) or were discharged before 8 hours
(6). Among the 59 horses that underwent general an-
esthesia, the first defecation was within 2 hours after
morphine administration for 8 horses, between 2 and
4 hours for 4 horses, and between 4 and 6 hours for 17
horses; 9 horses defecated for the first time between the
6- and 8-hour time points. Ten horses evaluated for 8
hours did not defecate during that time, and 11 horses
were not observed to defecate but left the clinic prior to
the 8-hour time point.
Mild excitation was observed in 1 horse 4 hours af-
ter morphine administration. This was a colt admitted
for castration that had not been halter trained and had
not previously been confined in a stall. Sedation was
observed in 31 horses; however, in most (29) of these,
sedation was observed immediately after return to the
stall following anesthesia and surgery or after admin-
istration of an α-2 receptor agonist (xylazine or deto-
midine) or phenothiazine tranquilizer (acepromazine).
Two horses with signs of sedation had not received oth-
er medications. Signs of mild sedation were observed
Respiratory rate Heart rate
Time No. of horses (breaths/min) (beats/min)
0 76 20 (12–66) 44 (28–80)
2 53 24 (12–100) 44 (28–80)
4 59 18 (12–60) 44 (32–72)
6 56 18 (8–72) 43 (28–64)
8 51 20 (12–44) 40 (28–64)
Data were collected from 75 horses, with 1 horse included twice
on separate visits. Decrease in the number of horses over time re-
flects discharge of patients at various times after drug administra-
tion. The time of morphine injection was considered time 0.
Table 1—Median (range) heart and respiratory rates for 76 horses
that received morphine (0.1 mg/kg, IM, once) as a perioperative
analgesic or as treatment for signs of pain in the clinical phase of
a study to evaluate pharmacokinetics of the drug.
JAVMA, Vol 243, No. 1, July 1, 2013 Scientific Reports 109
EQUINE
in one of these horses at the 6- and 8-hour time points
and in the other horse at the 4- and 6-hour time points.
Two horses had injection site reactions; neither re-
quired treatment. One horse had a 2.5-cm, focal, cir-
cumscribed swelling at the injection site that was de-
tected at the 4-hour time point. Palpation of the region
did not elicit signs of pain. This condition persisted to
the end of the 8-hour observation period but was not
evident on the following day. The other horse had a
1.5-cm swelling detected at the 4-hour time point that
was also not associated with signs of pain on palpa-
tion. At the 8-hour time point, the size of the swell-
ing had decreased by approximately half, and other as-
pects were unchanged; the condition had completely
resolved by the next morning.
Blood samples (2 or 3/horse) were collected at
various time points throughout the day following mor-
phine administration. Twenty-six samples were collect-
ed during the first hour, 26 were collected from 1 to < 2
hours, and 22 were collected from 2 to < 3 hours. The
plasma morphine concentration was above the LOQ in
all 74 samples obtained < 3 hours after drug admin-
istration and in 49 of 51 samples obtained between 3
and 5 hours after injection. Thirteen of 48 samples had
detectable concentrations of morphine between 5 and 7
hours after injection, and only 1 of 44 samples met this
criterion after 7 hours.
Pharmacokinetic analysis—An attempt was made
to fit the clinical data to a population pharmacokinetic
model, but this could not be accomplished satisfactorily.
Therefore, a naïve pooled population analysis method
was used to determine the pharmacokinetic parameters
of morphine after IM injection in these horses (Table
2). In the preliminary experiments and in the clinical
portion of the study, morphine administered IM had a
rapid absorption phase followed by a slower elimina-
tion phase (Figure 1). Healthy horses in the prelimi-
nary investigation had higher peak plasma concentra-
tions of morphine (28.2 and 33.5 ng/mL), compared
with the value for horses of the clinical population
evaluated via naïve pooled pharmacokinetic analysis
(21.6 ng/mL). Although individual results for healthy
horses in the preliminary investigation indicated higher
maximum drug concentrations than that determined
for the clinical population, results for those 2 horses
fit within the range of values for clinical patients (11.7
to 48.7 ng/mL). The absorption rate constant for mor-
phine in clinical patients was 79.76 hours–1, compared
with a mean value of 12.95 hours–1 for the 2 horses in
the preliminary investigation. The apparent half-life of
the absorption phase had a mean value of 3.7 minutes
in horses in the preliminary investigation and was only
30 seconds in the clinical population. Maximal con-
centrations of morphine in horses in the preliminary
investigation were detected at a mean of 19 minutes,
whereas peak concentrations were detected in clinical
patients at approximately 4 minutes after administra-
tion. The t1/2 was 1.48 hours in clinical patients, with a
mean of 1.76 hours in healthy horses of the preliminary
investigation.
Because IV administration of morphine was not in-
cluded in our study, bioavailability could not be calcu-
lated. Therefore, Vd and Cl were calculated as Vd/F and
Cl/F rather than absolute values. The Vd/F in horses of
the preliminary investigation had a mean value of 2.9 L/
kg and was 4.49 L/kg for clinical patients evaluated via
naïve pooled pharmacokinetic analysis. The Cl/F had a
mean value of 19.05 mL/kg/min for horses in the pre-
liminary investigation and was 34.9 mL/kg/min for the
clinical population.
Discussion
Morphine has been used as an analgesic in horses for
years but has dose-related adverse effects. Interestingly, a
Preliminary
investigation
Clinical
Parameter Mean Range phase
Ka (h–1) 12.95 8.21–17.68 79.76
Apparent absorption half-life (h) 0.0618 0.0392–0.0844 0.00869
Kel (h–1) 0.397 0.361–0.432 0.467
t1/2 (h) 1.76 1.6–1.96 1.48
Vd/F (L/kg) 2.9 2.72–3.07 4.49
AUC (h•ng/mL) 87.7 85–90.3 47.74
Cl/F (mL/min/kg) 19.05 18.5–19.6 34.9
Cmax (ng/mL) 30.85 28.2–33.5 21.6
Tmax (h) 0.31 0.22–0.4 0.065
In preliminary investigations, samples were collected at 5, 10,
15, 30, and 45 minutes and 1, 2, 4, 6, 8, 10, and 12 hours after mor-
phine administration and data were analyzed via standard 2-stage
pharmacokinetic analysis. In the clinical phase, samples were ob-
tained from hospitalized horses at various times (total, 2 to 3 sam-
ples/horse) up to 9 hours after drug administration and data were
analyzed via a naïve pooled pharmacokinetic model.
Cmax = Maximum plasma concentration. Ka = Absorption rate
constant. Kel = Elimination rate constant. Tmax = Time of maximum
plasma concentration.
See Table 1 for remainder of key.
Table 2—Pharmacokinetic parameters of morphine (0.1 mg/kg, IM, once)
following administration to 2 healthy horses during preliminary investiga-
tions and to 76 equine patients during the clinical phase of the study.
Figure 1—Plasma drug concentrations (semilogarithmic scale) of
morphine following administration in horses (0.1 mg/kg, IM). The
time of morphine injection was considered time 0. Mean values
obtained from 2 healthy horses during preliminary investigations
(triangles) and individual values obtained from 76 clinical patients
(data from 75 horses, with 1 horse included twice on separate vis-
its) that received morphine as a perioperative analgesic or as treat-
ment for signs of pain (circles) are shown. The solid line indicates the
predicted plasma kinetic profile of morphine determined via naïve
pooled pharmacokinetic analysis of samples from clinical patients.
The dashed line indicates the predicted profile determined via stan-
dard 2-stage pharmacokinetic analysis in preliminary investigations.
110 Scientific Reports JAVMA, Vol 243, No. 1, July 1, 2013
EQUINE
number of studies1,5–8 have been performed to evaluate
various doses of morphine administered via IV and IM
routes and their clinical effects in horses, but the phar-
macokinetic parameters for morphine administered IM
in horses have not been reported. In a previous study,9
morphine (0.1 mg/kg) administered IV in horses had a
t1/2 of 1.6 hours, and mean plasma concentrations of the
drug were < 10 ng/mL within 4 hours and < 5 ng/mL
within 6 hours after administration. A 3-compartment
model was used to describe pharmacokinetics of mor-
phine in that study.9 In another study in horses,10 phar-
macokinetic analysis of morphine (0.25 mg/kg [0.11 mg/
lb]) administered IV during general anesthesia with iso-
flurane revealed a t1/2 of 0.7 hours, Vd of 1.2 L/kg, and
Cl of 40 mL/min/kg. In the same study,10 the t1/2 of mor-
phine following IV administration at a dose of 2.0 mg/
kg (0.91 mg/lb) was 1 hour. Those authors10 postulated
that the t1/2 of morphine following IV administration in
horses is dose dependent. Administration of a medica-
tion into a compartment other than the central venous
compartment can alter its parameters in plasma because
of differences in absorption, and subsequently, the t1/2 of
extravascular morphine may be substantially different
following administration via different routes. Additional-
ly, poor absorption from extravascular routes may result
in low and possibly subtherapeutic drug concentrations.
Therefore, it is important to assess the pharmacokinetic
parameters of morphine following IM administration in
horses to evaluate the possibility of whether it could be a
clinically useful route of administration.
In the present study, the pharmacokinetic param-
eters of morphine administered IM as a perioperative
analgesic or as treatment for signs of pain in a clinical
population of horses included a t1/2 of approximately
1.5 hours, Vd/F of 4.49 L/kg, and Cl/F of 34.9 mL/min/
kg. The values of Vd/F and Cl/F may be useful for other
calculations involving morphine administered IM in
horses, but these do not imply 100% bioavailability of
the drug.
A 1-compartment open model with first-order in-
put and output with no lag was used for pharmacoki-
netic analysis of samples from 2 healthy horses in our
preliminary investigation and for naïve pooled pharma-
cokinetic analysis of samples from a clinical population
(75 horses, with 1 patient that received morphine on 2
visits for a total of 76). The model was chosen on the
basis of visual inspection of the data and the model that
best fit the data from the preliminary investigation. Al-
though plasma concentrations below the LOQ (2.5 ng/
mL) could not be assessed, morphine was likely to be
present in the plasma of horses after the last time point
used in the present study. We can speculate that if drug
concentrations below the LOQ in our study could be
quantified with a more sensitive assay, they would re-
sult in uniform residuals on the terminal portion of the
curve, suggesting a 1-compartment model is the most
appropriate model for the data set used in our naïve
pooled pharmacokinetic analysis. However, it is pos-
sible that a 2-compartment model could best describe
this data set if the assay used was sensitive enough to
quantify concentrations lower than the LOQ.
Peak plasma morphine concentrations following
IM administration in both healthy horses in the pre-
liminary investigation were within the range of con-
centrations detected in the clinical population, despite
numeric differences in some of the pharmacokinetic
parameters. A substantial degree of variability was de-
tected in plasma morphine concentrations after IM ad-
ministration in the clinical population of horses. It is
important to consider this variability, recognizing that
1 dose may not have the same effects in all patients,
and to perform routine clinical monitoring of equine
patients that receive morphine IM to maximize drug ef-
ficacy and minimize adverse effects.
Some apparent differences in pharmacokinetic pa-
rameters of morphine were detected between healthy
horses in the preliminary investigation and the larger
population of horses that received morphine in the
clinical phase of the study. Sample collection during the
preliminary investigation was most intensive during
the drug absorption and elimination phases, whereas
samples collected for naïve pooled analysis were pri-
marily obtained during the elimination phase. This was
done intentionally to completely capture the elimina-
tion phase because this is more clinically important for
determination of dosing intervals than is the absorption
phase. However, these data may incompletely represent
the absorption phase, thus biasing absorption rate and
time of maximal drug concentration in the naïve pooled
analysis, which was a limitation of our study. The small
number of samples during the absorption phase may
also have contributed to the inability to fit a population
pharmacokinetic model to the data.
Reported IV doses for morphine in horses range
from 0.02 to 2.4 mg/kg (0.009 to 1.09 mg/lb) in experi-
mental and clinical settings.8,11 Some doses have been
associated with adverse effects in horses, including de-
creased gastrointestinal motility and CNS excitation. At
a dose of 1.0 mg/kg (0.45 mg/lb) IV, morphine slowed
the passage of feces in the gastrointestinal tract of hors-
es.1 Other investigators reported a significant decrease
in fecal moisture content and a significant increase in
gastrointestinal transit time in horses after repeated
doses of morphine (0.5 mg/kg [0.23 mg/lb], IV, q 12 h)
in a crossover-design study.7 Horses had a 22-hour de-
lay in passing barium-impregnated spheres after mor-
phine treatment, compared with the values following
saline solution (control) treatment, and signs of CNS
stimulation were detected for 2 to 4 hours after mor-
phine administration (0.5 mg/kg, IV) in 3 of 5 horses.7
In experiments conducted to determine the morphine
dose used in that study,7 3 treatments of morphine (1.0
mg/kg, IV) caused severe signs of colic in 1 of 2 horses.
In other investigations, morphine administered IM at
0.66 mg/kg (0.30 mg/lb) caused restlessness in 7 of 8
ponies from 1 to approximately 4 hours after admin-
istration,5 and horses became ataxic and appeared un-
aware of their surroundings following a dose of 2.4 mg/
kg, IV.11 In the present study, a dose of 0.1 mg/kg, IM,
appeared to have little effect on heart rate and degree of
excitation, and no signs of colic were observed in any
horses. In a retrospective study2 to evaluate adverse ef-
fects in horses that received 0.1 to 0.17 mg of morphine/
kg (0.045 to 0.077 mg/lb), IV, the authors concluded
that morphine administration did not increase the risk
of intraoperative or postoperative complications. This
JAVMA, Vol 243, No. 1, July 1, 2013 Scientific Reports 111
EQUINE
dose range has been used clinically as an analgesic at
other institutions7 and is used for IM administration of
morphine at the hospital where the present study was
performed.
Morphine was only consistently detectable with
our assay for 3 hours after IM administration in clinical
patients. After that time, the plasma morphine concen-
tration in some horses was < 2.5 ng/mL. Because sever-
al samples lacked detectable morphine concentrations
during the later time points in the study, it is likely that
frequent administration is necessary to provide consis-
tent analgesia in some clinical patients when the drug
is administered via this route.
Determination of the efficacy of morphine at the
dose and route used was beyond the scope of the pres-
ent study. Pharmacodynamic studies evaluating asso-
ciations between pharmacokinetic data and analgesia
have not been reported in horses, and this is an area
for potential further research. Pharmacodynamic stud-
ies of morphine have been performed in humans, and
although this information cannot be extrapolated di-
rectly to horses, it does provide some general guidelines
to help equine practitioners to make educated deci-
sions about dosage recommendations. In a prospective
study12 of human cancer patients with chronic pain,
substantial individual variability was detected in the se-
rum concentrations of morphine needed for analgesia.
The range of effective plasma concentrations was 30 to
120 nmol/L (8.58 to 34 ng/mL) for the 25th to 75th
percentiles. When patient-controlled analgesia was as-
sessed in the immediate postoperative period, there was
also a large degree of individual variability, with a mean
± SD minimum effective plasma concentration of 16 ±
9 ng/mL.13 If 16 ng of morphine/mL is used as an ap-
proximate value for the mean minimum analgesic con-
centration in horses, only 18 of 26 (69%) samples col-
lected between 0 to 1 hours and 9 of 26 (35%) samples
collected between 1 and 2 hours had morphine concen-
trations that exceeded this value.
Although the model used in the present study can-
not predict the safety of other doses of morphine in
horses, it can be used to predict the plasma concentra-
tions of morphine after the IM administration at other
doses. According to our model, if this population of
horses received morphine IM at a dose of 0.2 mg/kg
(0.09 mg/lb) and the plasma concentrations are pro-
portional to the dose, all 26 samples collected between
0 and 1 hours after injection and 20 of 26 samples
obtained between 1 and 2 hours after injection would
contain > 16 ng of morphine/mL. Two to 3 hours after
drug administration, 8 of 22 samples collected would
have morphine concentrations above this value. Al-
though dose-related changes in circulating morphine
concentrations can be extrapolated with this model,
other studies1,7 have shown that increasing the dose of
morphine may increase the number of adverse effects.
Therefore, studies should be conducted assessing the
pharmacokinetic parameters and adverse effects of
morphine at higher doses than that used in the pres-
ent study. Marked variability was detected in plasma
morphine concentrations in clinical patients in our
study. This model does suggest that higher doses
would be needed to achieve targeted plasma morphine
concentrations in a larger percentage of horses and
that higher doses would likely extend the duration for
which concentrations of morphine exceed 16 ng/mL.
Clinical judgment and careful patient monitoring is
required in determining the appropriate dose to maxi-
mize analgesia and minimize adverse effects.
Morphine use for patient-controlled analgesia was
measured in a human study13 with morphine as the
only analgesic administered. Although administration
of 1 type of analgesic can be effective, the use of > 1
class of analgesics can also be beneficial. Multimodal
analgesia combines analgesics with different mecha-
nisms of action in an attempt to enhance analgesia
and minimize the adverse effects of a single medica-
tion.14,15 A systematic review16 of 60 human studies
concluded that the amount of morphine required for
analgesia was reduced by the concurrent administra-
tion of an NSAID. There was a substantial decrease in
postoperative nausea and vomiting, which are among
the adverse effects of morphine in humans, in patients
that received an NSAID as well as morphine.16 There
is clear evidence that multimodal analgesia is effec-
tive in human medicine, and at least 1 clinical report14
suggests the same is true in horses. In the present
study, almost every horse in the clinical population
that received morphine (74/76) also received either
phenylbutazone or flunixin meglumine. The effects
of concurrent administration of an NSAID on mor-
phine analgesic effects have not been evaluated in a
population of horses. It is likely that combined drug
treatments in horses will decrease the plasma concen-
tration of morphine needed for analgesia, but further
study is needed to determine effective concentrations
of morphine when used alone or in combination with
other medications.
One limitation of our study was that the observer
assessing sedation and excitation was not blinded,
and this could have biased these evaluations. Anoth-
er limitation is that it is difficult to draw any mean-
ingful conclusions about fecal output because most
(59/76) horses underwent general anesthesia and had
feed withheld for 6 to 8 hours before surgery. Even
without opioid administration, general anesthesia
has been reported to inhibit gastrointestinal motility
in horses.17 Most (47/76) horses in the present study
defecated within 8 hours after morphine administra-
tion and, of the 29 horses that were not observed to
defecate during this time, 17 were discharged before
the end of the 8-hour observation period. Therefore,
it seems reasonable to conclude that if morphine
administered at this dose inhibited gastrointestinal
motility, the effect was transient. However, it is im-
portant to note that associations between higher or
multiple doses of morphine and gastrointestinal mo-
tility were not evaluated.
Most horses (59/76) underwent general anesthesia
and surgery on the same day that morphine was admin-
istered. To our knowledge, a comparison of the phar-
macokinetic parameters of morphine with and without
concurrent general anesthesia has not been reported.
Similarly, the effects of health status, breed, and sex on
these values have not been reported in horses. There-
fore, numerous factors could have influenced the re-
112 Scientific Reports JAVMA, Vol 243, No. 1, July 1, 2013
EQUINE
sults of pharmacokinetic analysis of morphine in this
study. These data were obtained in clinical patients and
therefore should be more representative of the phar-
macokinetics of the targeted population than data ob-
tained in a study of healthy horses.
Injection site reactions were detected in only 2
horses in the present study. Both horses had a localized
swelling at the injection site that did not seem pain-
ful on palpation and resolved without treatment. These
swellings may have represented hematomas, seromas,
or allergic reactions. However, because the reactions
were self-limiting and transient, no other tests were
performed to determine the nature of the swelling. We
are unaware of any other reports describing injection
site reactions after IV or IM administration of morphine
in horses. Although median heart rate in clinical pa-
tients was significantly lower at 8 hours (40 beats/min)
than at 4 hours (44 beats/min) after drug administra-
tion, both values were within the reference range for
horses (28 to 44 beats/min). Therefore, the difference
did not appear to have any clinical relevance.
Time from sample collection to centrifugation and
freezing varied during the clinical phase of our study,
but data collected during the preliminary investigation
indicated that time from collection to centrifugation of
the sample under similar storage conditions resulted
in minimal differences in the measured plasma con-
centration of morphine. This was important because
it can be challenging to obtain multiple blood samples
from a large number of clinical patients and perform
plasma separation immediately after collection. Our
preliminary data showed that separation of plasma up
to 6 hours after collection had minimal effects on the
amount of morphine that was detected in the plasma.
Although further testing is needed, results of the
present study indicate that plasma concentrations of
morphine equivalent to concentrations considered ef-
ficacious in humans are transient following administra-
tion of a single 0.1 mg/kg dose IM in horses. Because
of the potential for adverse effects at higher or more
frequent doses, care should be taken to monitor equine
patients receiving morphine for evidence of efficacy or
adverse effects and to use the lowest dose that has ther-
apeutic benefit for the patient.
a. Morphine sulfate, Baxter Healthcare Corp, Deerfield, Ill.
b. Cerilliant Inc, Round Rock, Tex.
c. Bond Elut, Varian Inc, Palo Alto, Calif.
d. WinNonMix, version 2.0.1, Pharsight Corp, Cary, NC.
e. SigmaStat, version 3.1, Systat Software Inc, Chicago, Ill.
References
1. Alexander F. The effect of some anti-diarrhoeal drugs on intes-
tinal transit and faecal excretion of water and electrolytes in the
horse. Equine Vet J 1978;10:229–234.
2. Mircica E, Clutton RE, Kyles KW, et al. Problems associated
with perioperative morphine in horses: a retrospective case
analysis. Vet Anaesth Analg 2003;30:147–155.
3. Cox SR, Liao S, Payne-Johnson M, et al. Population pharma-
cokinetics of mavacoxib in osteoarthritic dogs. J Vet Pharmacol
Ther 2011;34:1–11.
4. Peyrou M, Doucet MY, Vrins A, et al. Population pharmacoki-
netics of marbofloxacin in horses: preliminary analysis. J Vet
Pharmacol Ther 2004;27:283–288.
5. Kalpravidh M, Lumb WV, Wright M, et al. Effects of butorpha-
nol, flunixin, levorphanol, morphine, and xylazine in ponies.
Am J Vet Res 1984;45:217–223.
6. Phaneuf LP, Grivel ML, Ruckebusch Y. Electromyoenterography
during normal gastro-intestinal activity, painful or non-painful
colic and morphine analgesia, in the horse. Can J Comp Med
1972;36:138–144.
7. Boscan P, Van Hoogmoed LM, Farver TB, et al. Evaluation of the
effects of the opioid agonist morphine on gastrointestinal tract
function in horses. Am J Vet Res 2006;67:992–997.
8. Clark L, Clutton RE, Blissitt KJ, et al. The effects of morphine
on the recovery of horses from halothane anaesthesia. Vet An-
aesth Analg 2008;35:22–29.
9. Combie JD, Nugent TE, Tobin T. Pharmacokinetics and protein
binding of morphine in horses. Am J Vet Res 1983;44:870–874.
10. Steffey EP, Eisele JH, Baggot JD. Interactions of morphine and
isoflurane in horses. Am J Vet Res 2003;64:166–175.
11. Combie J, Dougherty J, Nugent E, et al. Pharmacology of nar-
cotic analgesics in the horse IV. Dose and time response rela-
tionships for behavioral-responses to morphine, meperidine,
pentazocine, anileridine, methadone, and hydromorphone. J
Equine Med Surg 1979;3:377–385.
12. Klepstad P, Dale O, Kaasa S, et al. Influences on serum con-
centrations of morphine, M6G and M3G during routine clini-
cal drug monitoring: a prospective survey in 300 adult cancer
patients. Acta Anaesthesiol Scand 2003;47:725–731.
13. Dahlström B, Tamsen A, Paalzow L, et al. Patient-controlled an-
algesic therapy, part IV: pharmacokinetics and analgesic plas-
ma concentrations of morphine. Clin Pharmacokinet 1982;7:
266–279.
14. Dutton DW, Lashnits KJ, Wegner K. Managing severe hoof pain
in a horse using multimodal analgesia and a modified composite
pain score. Equine Vet Educ 2009;21:37–43.
15. Wideman GL, Keffer M, Morris E, et al. Analgesic efficacy of a
combination of hydrocodone with ibuprofen in postoperative
pain. Clin Pharmacol Ther 1999;65:66–76.
16. Maund E, McDaid C, Rice S, et al. Paracetamol and selective
and non-selective non-steroidal anti-inflammatory drugs for the
reduction in morphine-related side-effects after major surgery:
a systematic review. Br J Anaesth 2011;106:292–297.
17. Singh S, McDonell WN, Young SS, et al. Cardiopulmonary and
gastrointestinal motility effects of xylazine/ketamine-induced
anesthesia in horses previously treated with glycopyrrolate. Am
J Vet Res 1996;57:1762–1770.
... Recent changes in attitudes regarding animal welfare have increased the frequency of use of pain-relieving agents in production animal species (George et al., 2003). Non-steroidal antiinflammatories (NSAIDs) are the most commonly used medications by most practitioners, but repeated dosing leads to concerns with prostaglandin inhibition related side effects such as gastrointestinal ulcers and nephrotoxicity (Devine et al., 2013). In addition, meat and milk residues as well as withdrawal times are of concern with NSAID use and NSAID's alone are often insufficient for controlling severe unrelenting pain, such as fracture repair, and obstructive urolithiasis. ...
... In addition, meat and milk residues as well as withdrawal times are of concern with NSAID use and NSAID's alone are often insufficient for controlling severe unrelenting pain, such as fracture repair, and obstructive urolithiasis. Therefore, alternate modalities of analgesia must be investigated (Devine et al., 2013;George et al., 2003;Uhrig et al., 2007). ...
... It is commonly used in dogs both IV and IM and provides rapid and effective analgesia and sedation (Barnhart et al., 2000). In large animal species, morphine use has been evaluated in llamas (Uhrig et al., 2007), sheep (Sloan et al., 1991), and horses (Devine et al., 2013). These studies found morphine to be well tolerated; adverse effects were uncommon and mild in horses and appeared to be dose related. ...
Article
This study aimed to evaluate pharmacokinetic profiles of morphine in goats following a single dose administered intravenously, intramuscularly, or subcutaneously at 0.1 mg/kg, 0.25 mg/kg, and 0.4 mg/kg. Study population included eight healthy adult goats in a randomized cross-over study. Serial plasma samples were collected and morphine was quantified using high-performance liquid chromatography/mass spectrometry. Data fit a two-compartment model following intravenous administration and a non-compartmental model following both intramuscular and subcutaneous administration. Plasma elimination half-life was 2.88 ± 1.13 h (0.1 mg/kg), 2.30 ± 0.49 h (0.25 mg/kg), and 2.67 ± 0.82 h (0.4 mg/kg) following IV morphine. Intramuscular Cmax values were 13.4 ± 2.77 ng/ml (0.1 mg/kg), 34 ± 11.50 ng/ml (0.25 mg/kg), and 68.9 ± 24.5 ng/ml (0.4 mg/kg). Intramuscular Tmax f(h) or IM dosing (in hrs) was 0.19 ± 0.14 (0.1 mg/kg), 0.24 ± 0.24 (0.25 mg/kg), and 0.21 ± 0.24 (0.4 mg/kg). Subcutaneous Cmax values were 9.88 ± 3.31 ng/ml (0.1 mg/kg), 28.5 ± 11.6 ng/ml (0.25 mg/kg), and 39.4 ± 14.3 ng/ml (0.4 mg/kg). Subcutaneous Tmax (h) values for SC dosing were 0.36 ± 0.21 (0.1 mg/kg), 0.31 ± 0.17 (0.25 mg/kg), and 0.4 ± 0.13 (0.4 mg/kg). Intramuscular bioavailability values were 153.77 ± 12.60% (0.4 mg/kg), 104.8 ± 25.12% (0.25 mg/kg), and 100.7 ± 29.57% (0.1 mg/kg). Subcutaneous bioavailability values were 130.58 ± 19.07% (0.4 mg/kg), 116.6 ± 27.03% (0.25 mg/kg), and 111.6 ± 23.24% (0.1 mg/kg). No adverse effects were observed. Assuming plasma concentration required to induce analgesia is 16 ± 9 ng/ml in goats, as demonstrated in humans, it is suggested to administer morphine intramuscularly at 0.4 mg/kg every 3–4 h or SC every 2–3 h. This is a speculative conclusion therefore further studies evaluating pharmacodynamics and plasma analgesic threshold in goats is recommended.
... Immunohistochemical staining with c-Fos protein was performed 120 min after the final CTA or CPP test ( Figure 2). Notably, some previous studies have demonstrated that morphine injections could induce different half-lives based on different methods of administering morphine in different species (Hartvig et al., 1984;Hasselstrom et al., 1990;Taylor et al., 2001;Devine et al., 2013). For example, the elimination half-life of morphine is 4.2 h for humans (Hasselstrom et al., 1990), 30-45 min for Rhesus monkeys (Hartvig et al., 1984), 1.5 h for horses (Devine et al., 2013), and 76.3 min (intravenous injections) or 93.4 min (intramuscular injections) for cats (Taylor et al., 2001). ...
... Notably, some previous studies have demonstrated that morphine injections could induce different half-lives based on different methods of administering morphine in different species (Hartvig et al., 1984;Hasselstrom et al., 1990;Taylor et al., 2001;Devine et al., 2013). For example, the elimination half-life of morphine is 4.2 h for humans (Hasselstrom et al., 1990), 30-45 min for Rhesus monkeys (Hartvig et al., 1984), 1.5 h for horses (Devine et al., 2013), and 76.3 min (intravenous injections) or 93.4 min (intramuscular injections) for cats (Taylor et al., 2001). Therefore, the experimental procedure of morphine injections was staggered across Days 12,14,16,18,and 20. ...
Article
Full-text available
To re-examine the paradoxical effect hypothesis of abused drugs, the present study concerned whether different doses of morphine disparately affect neuronal activity and associations among the subareas of the medial prefrontal cortex (mPFC: cingulate cortex 1-Cg1, prelimbic cortex-PrL, infralimbic cortex-IL), the subregions of the nucleus accumbens (NAc; both core and shell), and the basolateral amygdala (BLA) following conditioned taste aversion (CTA) and conditioned place preference (CPP). All rats were given a 0.1% saccharin solution for 15-min, and they were intraperitoneally injected with saline or 20, 30, or 40 mg/kg morphine to form the aversive CTA learning. Later, half of the rats were tested for CPP (including the CTA and then CPP tests) for 30-min. Finally, the immunohistochemical staining with c-Fos was conducted after the behavioral test. After the CTA test, c-Fos (%) in the Cg1 and PrL (but not the IL) was more in 20–40 mg/kg of the morphine groups; c-Fos (%) in the NAc core, NAc shell, and BLA was more in the 30–40 mg/kg morphine group. After the CPP test, the Cg1, PrL, IL, and BLA showed more c-Fos (%) in 20 mg/kg morphine; the NAc core showed fewer in c-Fos (%) in the 30–40 mg/kg morphine groups. The mPFC subregions (e.g., Cg1, PrL, and IL), NAc subareas (e.g., NAc core and NAc shell), and BLA were involved in the different doses of morphine injections. The correlation analysis showed that a positive correlation was observed between PrL and IL with NAc core with low doses of morphine and with NAc shell with increasing doses of morphine after the CTA test. After the CPP, an association between PrL and NAc core and NAc shell at low doses and between IL and BLA and NAc shell with increasing doses of morphine. Therefore, different neural substrates and the neural connectivity are observed following different doses of morphine and after the CTA and CPP tests. The present data extend the paradoxical effect hypothesis of abused drugs.
... One notable finding from this study was that morphine and M6G concentrations following oral administration of 0.6 mg/kg codeine were comparable to that observed following administration of the reported analgesic dose of morphine (0.2 mg/kg, intravenous) [15]. While morphine administration at clinically effective doses has been shown to cause neuroexcitation in horses, increases in heart rate and adverse gastrointestinal effects [16][17][18], oral administration of 0.2 mg/kg of codeine was well tolerated [14]. ...
Article
Full-text available
Background In humans, codeine is a commonly prescribed analgesic that produces its therapeutic effect largely through metabolism to morphine. In some species, analgesic effects of morphine have also been attributed to the morphine-6-glucuronide (M6G) metabolite. Although an effective analgesic, administration of morphine to horses produces dose-dependent neuroexcitation at therapeutic doses. Oral administration of codeine at a dose of 0.6 mg/kg has been shown to generate morphine and M6G concentrations comparable to that observed following administration of clinically effective doses of morphine, without the concomitant adverse effects observed with morphine administration. Based on these results, it was hypothesized that codeine administration would provide effective analgesia with decreased adverse excitatory effects compared to morphine. Seven horses received a single oral dose of saline or 0.3, 0.6 or 1.2 mg/kg codeine or 0.2 mg/kg morphine IV (positive control) in a randomized balanced 5-way cross-over design. Blood samples were collected up to 72 hours post administration, codeine, codeine 6-glucuronide, norcodeine morphine, morphine 3-glucuronide and M6G concentrations determined by liquid chromatography- mass spectrometry and pharmacokinetic analysis performed. Pre- and post-drug related behavior, locomotor activity, heart rate and gastrointestinal borborygmi were recorded. Response to noxious stimuli was evaluated by determining thermal threshold latency. Results Morphine concentrations were highest in the morphine dose group at all times post administration, however, M6G concentrations were significantly higher in all the codeine dose groups compared to the morphine group starting at 1 hour post drug administration and up to 72-hours in the 1.2 mg/kg group. With the exception of one horse that exhibited signs of colic following administration of 0.3 and 0.6 mg/kg, codeine administration was well tolerated. Morphine administration, led to signs of agitation, tremors and excitation. There was not a significant effect on thermal nociception in any of the dose groups studied. Conclusions The current study describes the metabolic profile and pharmacokinetics of codeine in horses and provides information that can be utilized in the design of future studies to understand the anti-nociceptive and analgesic effects of opioids in this species with the goal of promoting judicious and safe use of this important class of drugs.
... The quantification of pain is relevant to identify various degrees of suffering and aid decision-making for qualitative selection and dosage of analgesic intervention. Although it was expected that horses castrated with prior analgesia would express an intermediate degree of pain between those castrated without previous analgesia and those only anesthetized, this was not observed even 8 h after the anesthesia, when the analgesic effect of the morphine (2 h) [49], dipyrone (5 h) [50], and flunixin meglumine (8 h) would be fading [51]. It can be inferred that the analgesic protocol produced preventive analgesia. ...
Article
Full-text available
This prospective and longitudinal study aimed to identify spontaneous post-orchiectomy pain behaviors in horses regardless of the effects of anesthesia, analgesia, and recording time of day. Twenty-four horses divided into four groups were submitted to: inhalation anesthesia only (GA), or combined with previous analgesia (GAA), or orchiectomy under pre (GCA), or postoperative (GC) analgesia. The data obtained from the subtraction of frequency and/or duration of 34 behaviors recorded during seven 60-min time-points in the 24 h after the anesthesia from those recorded in the mirrored time-points in the 24 h before the anesthesia (delta) were compared over time and among groups by Friedman and Kruskal–Wallis tests, respectively (p < 0.05). Time of day influenced the behaviors of walk, look out the window, rest the pelvic limb, and rest standing still. The only pain-related behaviors were decreased mirrored proportional differences in time spent drinking, and eating, and increased mirrored proportional differences in the frequency or duration of look at the wound, retract the pelvic limb, expose the penis, and look at the back of the stall. In conclusion, confounding factors rather than pain may influence several suggestive pain-related behaviors documented in the literature.
Article
Although morphine has demonstrated antinociceptive effects in horses, its administration has been associated with dose-dependent adverse effects. In humans and rats, part of the analgesic effect of morphine has been attributed to the active metabolite, morphine-6-glucuronide (M6G). Although morphine can cause several undesirable effects, M6G has a more favorable safety profile. The objective of this study was to characterize the pharmacokinetics, tissue distribution, and behavioral and select physiological effects of M6G following intravenous administration to a small group of horses. In Part 1 of the study, 3 horses received a single intravenous administration of saline, 0.5 mg/kg body weight (BW) M6G, or 0.5 mg/kg BW morphine in a 3-way crossover design. Blood samples were collected up to 96 hours post-administration, concentrations of drug and metabolites measured, and pharmacokinetics determined. Behavioral and physiological effects were then recorded. In Part 2 of the study, 2 horses scheduled to be euthanized for other reasons, were administered 0.5 mg/kg BW M6G. Blood, cerebrospinal fluid (CSF), and various tissue samples were collected post-administration and concentrations of drug were determined. The clearance of M6G was more rapid and the volume of distribution at steady state was smaller for M6G compared to morphine. A reaction characterized by head shaking, pawing, and slight ataxia was observed immediately following administration of both morphine and M6G to horses. After M6G administration, these behaviors subsided rapidly and were followed by a longer period of sedation. Following administration, M6G was detected in the kidney, liver, CSF, and regions of the brain. Results of this study encourage further investigation of M6G in order to assess its clinical feasibility as an analgesic in horses.
Chapter
Due to benefits such as decreased pain, reduced tissue damage, and quicker return to performance, laparoscopic and thoracoscopic procedures are increasing in popularity. In the horse, these may be done under standing sedation or general anesthesia. The chapter summarizes the effects on cardiovascular parameters, oxygenation, ventilation, and intracranial pressure. It briefly reviews sedation and locoregional anesthetic techniques used for standing surgery. In some cases, the nature of the procedure or temperament of the horse preclude performing standing surgery. The chapter also reviews anesthetic techniques as well as the unique challenges faced during laparoscopic and thoracoscopic procedures performed under general anesthesia. Recommendations for monitoring and support and considerations of the same are similar to those for any horse undergoing procedures under general anesthesia and may include non-invasive and invasive measures.
Article
Full-text available
This study aimed to elucidate the sedative effect and physiological changes associated with the intramuscular injection of detomidine combined with morphine in horses. Six healthy crossbred horses, aged 2 to 10 years, were included. A crossover experimental design was used to compare the effects of intramuscular injection of 30 µg/kg of detomidine alone (IMD) and intramuscular administration of 30 µg/kg of detomidine and 0.1 mg/kg of morphine (IMDM). The degree of sedation, height of head above ground, were assessed at the time points before and 5, 10, 20, 30, 40, 50, 60, 75, 90, 105, and 120 minutes after drug administration, and heart rate, respiratory rate, systolic blood pressure, rectum temperature and intestinal motility were assessed at the time points before and 10, 20, 30, 40, 50, 60, 75, 90, 105, and 120 minutes after drug administration. The physiological parameters were analyzed using the Kruskal-Wallis test with Dunn’s post-hoc test and analysis of variance with t-test for independent samples and the sedation scores using the Friedman test and Mann Whitney U-test. P-values <0.05 indicated a statistically significant difference. IMDM promoted a higher sedative effect as compared to IMD, but the sedation occurred inconsistently. Additionally, a reduction in intestinal motility was observed with IMDM at 60, 75, 90, and 105 minutes after administration. IMDM promoted more variable sedation and prolonged reduction in the intestinal motility in the horses as compared to IMD.
Article
Full-text available
Non-opioid analgesics, paracetamol, non-steroidal anti-inflammatory drugs (NSAIDs), or cyclo-oxygenase 2 (COX-2) inhibitors are often given along with morphine as part of multimodal analgesia after major surgery. We have undertaken a systematic review and a mixed treatment comparison (MTC) analysis in order to determine explicitly which class of non-opioid analgesic, paracetamol, NSAIDs, or COX-2 inhibitors is the most effective in reducing morphine consumption and morphine-related adverse effects. Sixty relevant studies were identified. The MTC found that when paracetamol, NSAIDs, or COX-2 inhibitors were added to patient-controlled analgesia (PCA) morphine, there was a statistically significant reduction in morphine consumption: paracetamol [mean difference (MD) -6.34 mg; 95% credibility interval (CrI) -9.02, -3.65], NSAIDs (MD -10.18; 95% CrI -11.65, -8.72), and COX-2 inhibitors (MD -10.92; 95% CrI -12.77, -9.08). There was a significant reduction in nausea and postoperative nausea and vomiting with NSAIDs compared with placebo (odds ratio 0.70; 95% CrI 0.53, 0.88) but not for paracetamol or COX-2 inhibitors, nor for NSAIDs compared with paracetamol or COX-2 inhibitors. There was no statistically significant difference in sedation between any intervention and comparator. On the basis of six trials (n=695), 2.4% of participants receiving an NSAID experienced surgical-related bleeding compared with 0.4% with placebo. The MTC found that there is a decrease in 24 h morphine consumption when paracetamol, NSAID, or COX-2 inhibitors are given in addition to PCA morphine after surgery, with no clear difference between them. Similarly, the benefits in terms of reduction in morphine-related adverse effects do not strongly favour one of the three non-opioid analgesics.
Article
This report describes a clinically useful modified composite pain score (MCPS) for horses with hoof pain. The horse in this report initially suffered from acute pain from a subsolar seroma as well as suspected pain from chronic laminitis. Following surgical debridement, corium prolapsed through the wound and it became infected. During the course of conventional hoof and wound management, the pain experienced by the patient was refractory to nonsteroidal anti-inflammatory drugs. Using the MCPS as a guide, inflammatory and neuropathic pain states were identified and multimodal analgesia was employed in response to the varying pain states. Drugs used for the anti-inflammatory pain management protocol included firocoxib, butorphanol, phenylbutazone, aspirin and fish oil. Neuropathic pain modulators included parenteral and local anaesthetics, pentoxifylline, ketamine and gabapentin. Composite pain scoring in horses, which includes observational, physiological and interactive components, may have greater sensitivity for demonstrating response to therapy when multiple types and stages of pain exist.
Article
Objective To identify the incidence of adverse effects caused by morphine 100–170 µg kg−1 administration during surgery in horses. Design Retrospective case record analysis (1996–2000). Animals Eighty-four healthy (ASA 1 or 2) horses, mean age 5.5 ± 3.1 (SD) years (2 months to 16 years), mean weight 524 ± 14 kg (100–950). Methods Physiological data and evidence of complications were collected from the anaesthetic records of all animals anaesthetized with romifidine, ketamine, diazepam and halothane and undergoing laryngeal surgery or orchiectomy at the Royal (Dick) School of Veterinary Studies. Cases were divided into those receiving (group M+; n = 18) and those not receiving morphine (M−; n = 29), and the data compared. Values for heart and respiratory rate and mean arterial pressure were compared at 15-minute intervals between 30 and 120 minutes after induction using anova for repeated measures. The incidence of intraoperative problems was compared using Fisher's exact test. Recovery scores were compared using Student's unpaired t-test. The records of a further 37 horses undergoing umbilical herniorrhaphy (n = 5), arthroscopy (n = 29) or tarsal arthrodesis (n = 3) were also studied but not analysed statistically due to disparate treatment distribution. Results There were no significant differences between the M+ and M− groups. The incidence of post-operative complications such as box-walking and colic were similar in each group. Conclusions Morphine doses of 100–170 µg kg−1 do not increase the risk of problems when used to provide perioperative analgesia in horses anaesthetized with romifidine, ketamine, diazepam and halothane. Clinical relevance Morphine provides an acceptable and relatively inexpensive way to provide perioperative analgesia in horses.
Article
Mavacoxib (Trocoxil™) is an oral long-acting COX-2 inhibitor approved for the treatment of osteoarthritis in dogs. Two field trials were conducted in client-owned dogs suffering from osteoarthritis, with dosages of 4 mg/kg body weight (BW) (Study 1) or 2 mg/kg BW (Study 2). Mavacoxib plasma concentrations were determined from trough blood samples and from blood samples collected at 4-10 months after the last dose. A one-compartment linear model was fitted to the concentration data (1317 concentration records from 286 patients), and parameters for oral clearance (Cl/F), apparent volume of distribution (V d/F) and their between-subject variabilities (BSV) were estimated. Covariates were included in the model based on the outcomes of stepwise regression procedures. In the final model, the typical value of Cl/F was a function of BW, age and breed. German shepherds and Labrador retrievers were found to have 31% higher values of Cl/F than patients from different breeds with similar ages and BWs. The typical value of V d/F was found to be dependent only on BW. The two field studies appeared to differ similarly with respect to Cl/F and V d/F. The explanation for this difference is not known, but the difference was accounted for in the final model as a 23.9% lower bioavailability in Study 2. Mavacoxib exhibited relatively broad BSV in Cl/F and V d/F, with coefficients of variation of 47% and 19%, respectively. The typical value for mavacoxib's terminal elimination plasma half-life (t 1/2) was 44 days, but a minority of patients (approximately 5%) had empirical Bayes estimates of t 1/2 exceeding 80 days. Simulations with the model indicated that the majority of patients treated with mavacoxib 2 mg/kg will maintain trough plasma mavacoxib concentrations associated with efficacy. Results of the population pharmacokinetic analysis helped to reduce the dose from 4 to 2 mg/kg and thus increased the therapeutic index for this molecule.
Article
The effect of morphine, Tinct. opii , loperamide, pethidine and atropine on intestinal transit and the faecal and urinary excretion of water and electrolytes was studied in ponies. The rate of passage of a particulate marker was slowed by morphine, hastened then slowed by loperamide and Tinct. opii , and hastened by atropine. The liquid marker was slowed by Tinct. opii and hastened then slowed by the other drugs. Only loperamide decreased the faecal sodium excretion. This drug also decreased faecal water and weight; it appeared worthy of clinical trial in diarrhoea. Tinct. opii decreased but morphine, pethidine and atropine increased faecal water. RÉSUMÉ Les effets de la morphine, de la teinture d'Opium, du loperamide, de la pethidine et de l'atropine sur le transit intestinal et sur l'excrétion urinaire et fecale de l'eau et des électrolytes ont étéétudié sur des poneys. La vitesse de passage d'un marqueur a été ralentie par la morphine accélérée puis ralentie par le loperamide et par la teinture d'Opium et accélérée par l'atropine. Un marqueur liquide a été freiné par la teinture d'Opium et accéléré puis ralenti par les autres médicaments. Seul la loperamide a diminué l'excrétion fecale de sodium. Ce médicament a également diminué le poids et la quantité d'eau des feces. Il est apparu interessant d'emploi dans le traitement des diarrhées. La teinture d'Opium a diminué l'eau fecale au contraire de la morphine de la pethidine et de l'atropine qui l'augmentaient. ZUSAMMENFASSUNG Die Wirkung von Morphium, Tinctura opii, Loperamid, Pethidin und Atropin auf die Intestinalpassage und die Exkretion von Wasser und Elektrolyten in Faeces und Urin wurde an Ponies untersucht. Die Passagezeit eines festen Markers wurde durch Morphium verlangsamt, zuerst beschleunigt dann verlangsamt durch Loperamid und Tinct. opii, beschleunigt durch Atropin. Der flüssige Marker wurde durch Tinct. opii verlangsamt, beschleunigt und nachher verlangsamt durch die anderen Medikamente. Nur Loperamid setzte die Na‐Exkretion in den Faeces herab. Diese Substanz verminderte auch das faekale Wasser und das Gewicht; sie scheint einer klinischen Prüfung bei Durchfällen würdig. Tinc. opii setzte die faekale Wasserausscheidung herab, Morphium, Pethidin und Atropin vermehrten sie.
Article
The electrical potentials were recorded from the antrum, the duodenum, the ileum and the first part of the colon of ponies under (a) normal resting conditions, (b) during nonpainful colic and (c) after intravenous morphine administration. The normal pony, at rest, had five contractions of the antrum per minute. On the small intestine, the basal electrical activity decreased from the duodenum (14-15/min) to the ileum (10-11/min). The small bowel also had three types of motility: peristaltic waves, rhythmic segmentations and random contractions. On the colon, bursts of potentials indicating intense motor activity occurred at the rate of 20 to 30 per hour. Morphine given intravenously (IV) greatly increased the frequency of the electrical potentials of the antrum and the longitudinal bands of the colon. During non-painful colic, hyperactivity of the cranial small intestine was continuous. Spasms of the jejunum occurred every minute and could not be relieved by morphine (IV). When colic was painful, jejunal spasms announced the crisis of intense abdominal pain. After morphine (IV) the spasms and pain disappeared; the jejunum remained hyperactive, the motility of the colon was increased while the antrum became quiet.
Article
The analgesic and behavioral effects of butorphanol (0.22 mg/kg), flunixin (2.2 mg/kg), levorphanol (0.033 mg/kg), morphine (0.66 mg/kg), and xylazine (2.2 mg/kg), given IM were observed in 8 ponies. These ponies were instrumented to measure response objectively to painful superficial and visceral stimuli. Effects on the cardiopulmonary system and rectal temperature also were evaluated in 6 of these ponies. Observations were conducted before drug injection (base-line values) and after injection at 30, 60, 120, 180, and 240 minutes. Xylazine provided the highest pain threshold for the first 60 minutes and a sedative effect for 105 minutes. The effects for superficial pain and visceral pain persisted 3 hours and 4 hours, respectively. Morphine produced good analgesia for superficial pain (30 minutes), whereas butorphanol provided good effect for visceral pain (4 hours). A slight degree of analgesia for visceral pain was obtained after morphine (1 hour) and levorphanol (4 hours); flunixin did not induce analgesia. Butorphanol, levorphanol, and morphine stimulated motor activity. Behavioral effects did not occur after flunixin was given. Xylazine decreased systolic, diastolic, and mean blood pressures. Marked increases in these pressures, heart rate, and respiratory rate were observed after morphine was given. Changes of central venous pressure, rectal temperature, and blood gas values remained within base-line limits after both drugs were given. Butorphanol increased heart rates for 1 hour; flunixin and levorphanol did not alter any of the above values.
Article
Morphine could be detected in horses dosed with 0.1 mg of drug/kg of body weight for up to 48 hours in blood and 144 hours in urine. This dose of morphine elicited no observable effects and is a suggested analgesic dose. Computer analysis revealed that a 3-compartment open system was the best fitting model with a serum half life (t1/2(beta)) of 87.9 minutes and a urine t1/2(beta) of 101.1 minutes. Binding to equine serum proteins was linear over a drug concentration range of 3.88 X 10(-5)M to 3.50 X 10(-8)M and averaged 31.6%. In RBC-partitioning experiments, 78.1% of the drug was found in the plasma fraction. The data indicated that a horse should not be given morphine closer than 1 week before a race.