Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

C HA P T E R 3

Recognizing and Treating Pain


in Horses
Rachel C. Hector and Khursheed R. Mama

Y INTRODUCTION leukotrienes, neuropeptides, and nerve growth factors) that


sensitize peripheral nerve fibers and activate additional (silent)
Following a brief review of general concepts related to pain nociceptors at the terminal ends of Aδ and C fibers.2,3 Cen-
and its assessment in horses, this chapter will focus on avail- tral sensitization results from the cumulative effects of repeti-
able pharmacologic and to a lesser extent complementary tive and sustained nociceptive input on the dorsal horn of
therapeutic options. The intent is to provide information that the spinal cord and a subsequent release of glutamate and
veterinarians can directly apply to management of acute and neuropeptides (substance P and neurokinin A) that activate
chronic pain in horses. N-methyl-d-aspartate (NMDA) and tachykinin receptors,
resulting in a gradual “windup” of central neural pathways.2-4
Pathophysiology of Pain Visceral pain (e.g., pain from intestine, liver, spleen, kidney, or
The International Association for the Study of Pain defines bladder), unlike somatic pain, is transmitted by parasympathetic
pain as “an unpleasant sensory and emotional experience (principally vagal) and sympathetic splanchnic afferent nerve
associated with actual or potential tissue damage, or described fibers.5,6 The autonomic fibers involved in transmitting noxious
in terms of such damage” (Box 3.1).1 Nociception refers to the
activation of high threshold receptors (nociceptors) located at
the distal ends of unmyelinated (C) or poorly myelinated (Aδ)
nerve fibers. Electrical impulses conducted by these afferent   BOX 3.1    Definitions of Terms Used to Describe Pain
fibers reach the dorsal horn of the spinal cord and via ascend-
ing pathways (spinothalamic and spinoreticular thalamic) are 1. Pain: Unpleasant sensory or emotional experience
transmitted to the brain in which they may be perceived or associated with actual or potential tissue damage or
recognized as painful (Fig. 3.1). Nociception may occur in described in terms of such damage.
the absence of perception (as in the anesthetized horse) and 2. Noxious stimulus: Stimulus (mechanical, chemical, and
can trigger neuroendocrine (cortisol and catecholamines), thermal) of sufficient intensity to threaten or overtly cause
metabolic (hyperglycemia), and physiologic (heart rate and tissue damage.
respiratory rate) responses. Perception, which is influenced 3. Nociception: Process of pain perception via pain recep-
by external and animal-related factors, will trigger similar tors (nociceptors) and transmission of noxious (painful)
responses and behaviors (e.g., avoidance, lameness) intended stimuli, including transduction, transmission, modulation,
to protect the horse from further insult. Comprehensive and perception.
assessment of these responses is useful when evaluating the 4. Hyperalgesia: Increased response (hypersensitivity) to a
severity, stress, and impact of pain on the horse’s quality of life. noxious stimulus, either at the site of injury (primary) or in
Acutely painful states may resolve with discontinuation of surrounding undamaged tissue (secondary).
the stimulus and appropriate intervention. Neuronal input 5. Hyperesthesia: Increased sensitivity to nonnoxious stimuli.
may also be modulated by descending inhibitory pathways 6. Hyperpathia: Greatly exaggerated pain sensation to
originating from the brain and by use of alternate nonpain- nociceptive stimuli.
ful afferent neural input (e.g., ice at the site of injury). In the 7. Allodynia: Pain produced by nonnoxious stimuli.
absence of these, continued nociceptive input can lead to 8. Preemptive analgesia: Prevention or minimization
chronic nervous system pathology resulting from peripheral of pain by the administration of analgesics before the
and central sensitization. Clinically these conditions are asso- production of pain or before the introduction of a noxious
ciated with hypersensitivity (or a reduction in the intensity of stimulus (surgery) if pain already exists. The goal of
the stimulus required to cause pain) and allodynia (in which a preemptive analgesia is to provide a therapeutic interven-
typically nonpainful stimulus is now perceived as pain). Exag- tion in advance of pain to prevent or minimize the central
gerated responses to a noxious stimulus (hyperalgesia) and nervous system response to a noxious stimulus.
persistence of pain after removal of a noxious stimulus (hyper- 9. Multimodal therapy: Administration of multiple drugs
pathia) may also occur. that act by different mechanisms of action to produce
Peripheral sensitization is caused by the local produc- the desired (analgesic) effect.
tion, release, and accumulation of chemicals (prostaglandins,
138
CHAPTER 3  Recognizing and Treating Pain in Horses 139

Central sensitization
Dorsal ,
horn , TrkB C fiber
neuron BDNF central
NMDA axon
Ca2+
Mg2+
Signaling molecules Glutamate
Perception
Cortex
AMPA/KAI
Sub P
Projection NK1

Transmission
Modulation

Peripheral sensitization
COX2

C fiber R Prostaglandins (PGE2)


peripheral
axon RHeat
H+ Histamine
R
+
K+ Serotonin (5HT)
Signaling molecules H+
K +
H Bradykinin
R
Proteases
Cytokines (TNF )

R Nerve growth factor Transduction

FIG. 3.1  Pain nociception and perception. Painful thermal, mechanical, and chemical stimuli are trans-
duced to electric potentials (action potentials) that are transmitted to the spinal cord, in which they are
modulated and then projected to the brain (perception). The major excitatory neurotransmitter in the spine
is glutamate, which normally activates α-amino-3-hydroxy-5-isoxazole proprionic acid (AMPA) and kainate
(KAI) receptors. NMDA, N-methyl-d-aspartate; NK1, neurokinin.

inputs from visceral organs are diffuse and overlap extensively, (e.g., gastrointestinal, orthopedic, dental). Additionally, it is
resulting in difficulty in localization of the site of pain. Neuro- recognized that the pain experience is likely to be influenced
pathic pain, which is yet another form of pain, may result from by animal and environmental factors.10-12 Among physiologic
injury to the peripheral or central nervous system. For addi- assessments, stress hormones (e.g., cortisol, β-endorphins),
tional details regarding the pathophysiology of pain the reader is and catecholamines (e.g., epinephrine, norepinephrine) have
referred to McMahon et al. (2013) and Almeida et al. (2004).7,8  been measured in both clinical and laboratory settings in an
attempt to correlate these “objective data” with pain states.10,13
Pain Assessment Their usefulness has, however, been mixed because shock,
Although horses will demonstrate a variety of behavioral stress, and medications influence them like they do other
indicators of pain (Box 3.2), assessment of pain in horses physiologic measurements, such as heart rate. Further, they
has historically been considered challenging.9 This could be are not contemporaneous to the pain assessment; thus, they
because horses, as prey animals, do not wish to appear vul- only have retrospective value. Measurement of inflammatory
nerable and so do not fully exhibit signs of pain in unfamil- mediators such as prostaglandin E2 and substance P from
iar or threatening environments until pain becomes severe or joints are similarly not consistently reliable as indicators of
unrelenting. Consequently, horses may not be appropriately pain, and results are not immediately available.14,15
treated for painful conditions by the inexperienced observer. In an attempt to overcome these challenges, there has been
Although early reports suggest an emphasis on physiologic significant recent work on the development of multifaceted
or limited behavioral parameters to assess pain, today there pain assessment tools that consider physiologic (e.g., heart
is the recognition that evaluation of pain requires a multi- rate, blood pressure) and behavioral (both spontaneous and
faceted approach and should be assessed in light of the cause in response to interaction) indices developed specifically for
140 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

repeatable, and consistent categorization of pain, such that


  BOX 3.2    Potential Behavioral Indicators of Pain in Horses therapies can be both targeted and monitored. If multiple
individuals are to be involved in assessing the animal, train-
•  onsiderable restlessness, agitation, and anxiety
C ing in use of the preferred pain scale or behaviors is helpful to
• Rigid stance and reluctance to move minimize interobserver variation on parameters that are open
• Lowered head carriage to interpretation. Accurate assessment of pain will allow vet-
• Fixed stare and dilated nostrils, clenched jaw erinarians to continue to improve analgesic efforts in horses. 
• Aggression toward its own foal
• Aggression toward handlers, horses, objects, and self Development of a Treatment Plan
• Decreased interactions with handlers Given the multifaceted nature of pain, the many individual
• Vocalization (deep groaning and grunting) breeds of horses, and additional equid species (e.g., donkeys,
• Rolling mules), as well as consideration of a wide age range of individual
• Kicking at abdomen equids in which veterinarians may be faced with managing pain,
• Flank watching a systematic approach to developing and modifying a pain treat-
• Stretching ment plan should be considered. Because of the significance of
• Dullness and depression well-being, pain is considered the “5th vital sign” in human
• Weight-shifting among limbs patients. In addition to the potential emotional association
• Limb guarding and subsequent avoidance behavior horses exhibit to locations
• Abnormal weight distribution in which they may have experienced pain, physiologic conse-
• Pointing, hanging, and rotating limbs quences such as negative energy balance, poor wound healing,
• Abnormal movement and altered gastrointestinal motility may be significant.22
• Reluctance to move Management of nociceptive pain such as surgical pain or
• Arched back that associated with acute injury is a common scenario. Noci-
• Head shaking ceptive pain is considered an adaptive or protective strategy,
• Abnormal bit behavior and the associated inflammatory response is considered essen-
• Altered eating; anorexia, quidding tial to the healing process. The goal of therapy is to normalize
pain sensitivity and not allow it to progress to a maladaptive
state or one that is no longer proportional to the noxious
horses in different clinical scenarios (e.g., postarthroscopy, stimulus and healing tissue.23 Nonsteroidal antiinflammatory
colic). Each parameter on many of these scales is further cate- drugs (NSAIDs) help normalize the inflammatory response
gorized in an attempt to indicate severity of pain; for example, generated by tissue injury and are commonly used in horses.
the parameter “kicking at the abdomen” is characterized by Although there have been mixed reports of the overall advan-
frequency of the same over a fixed interval, with greater fre- tage of preemptive versus postprocedural use of NSAIDs, it
quency assigned a worse score (greater degree of pain). Hence, is most common to administer these preemptively to limit
these scales serve not only to assess the presence or absence of inflammation. Additionally, medications used in the horse for
pain but also the severity (two such scales are included in Figs. sedation (e.g., α2-adrenergic agonists) and general anesthesia
3.2 and 3.3).16-21 One of these—namely, the horse grimace (e.g., ketamine) are also analgesic and affect different sites in the
scale—presents a somewhat unique approach and is worth nociceptive pathway (e.g., α2-adrenergic agonist drugs medi-
highlighting. This scale, which was modeled after similar work ate their analgesia through descending inhibition), providing
in rodents, correlates six specific facial expressions with the a multimodal approach. Regional techniques (e.g., intraar-
degree of pain; for example, ears stiffly backward can indi- ticular, epidural, perineural) further facilitate this multimodal
cate a greater degree of pain versus none if ears face forward. approach to analgesia and minimize the adverse effects (e.g.,
Although this was developed in horses undergoing castration, gastrointestinal stasis) associated with systemic medications.
it may have applicability or provide a good adjunctive assess- Although foals are similarly deserving of a multimodal
ment tool in horses with other conditions.19 Photographic approach in treating pain, the safety of NSAIDs (e.g., gas-
examples of horses demonstrating facial expressions of pain trointestinal ulcers) and the cardiovascular side effects of
are included in Fig. 3.4. α2-adrenergic agents when used at doses similar to the adult
Note that the majority of these pain scales are targeted horse deserve consideration. Conversely, some regional tech-
toward acute or nociceptive pain in adult horses. Assessment niques (e.g., lumbosacral epidural) are more feasible, and cer-
of chronic pain may require additional and different tools (e.g., tain drugs (e.g., opioids) may not be limited by behavioral side
force plate, gait analysis), owner or trainer input, and greater effects to the same degree as in adult horses.
reliance on response to therapy. Similarly, the authors are not The goal in treating pain early and using a multimodal
aware of ethograms or pain scales for foals, but based on the approach is to minimize development of a chronic condition
author’s impressions and in talking with veterinarians who that can be more challenging to evaluate and treat. The subse-
routinely manage foals, additional behavioral signs should be quent text includes information on pain treatment modalities
considered. For example, grinding of the teeth is commonly and medications that the veterinarian may select to best man-
associated with pain from stomach ulcers in foals. Addition- age individual equine patients. 
ally, unwillingness to lie down despite buckling the front limbs,
stamping hind feet toward the belly, and lying down in lateral Y DRUGS USED TO TREAT PAIN
recumbency with one or both legs cranially toward or over the
head has been associated with pain in foals. Almost all types of drugs used to treat pain in small ani-
Although methodology may need to vary based on the cir- mals have also been used in horses, although there are
cumstance, the goal of pain assessment is to provide objective, fewer controlled trials evaluating their efficacy. This may
CHAPTER 3  Recognizing and Treating Pain in Horses 141

Physiological Data Criteria Score


/12
Heart Rate (Compared to <10% increase 0
Baseline)
>11-30% increase 1
>31-50% increase 2
>50% increase 3
Respiratory Rate <10% increase 0
(Compared to
Baseline) >11-30% increase 1
>31-50% increase 2
>50% increase 3
Digestive Sounds Normal motility 0
Decreased motility 1
No motility 2
Hypermotility 3
Rectal Temperature <0.5°C variation 0
(Compared
to Baseline) <1°C variation 1
<2°C variation 2
>2°C variation 3
Response To Interaction Criteria Score
/6
Interactive Behavior Pays attention to people 0
Exaggerated response to auditory stimuli 1
Excessive to aggressive response to auditory stimuli 2
Stupor, prostration, no response to auditory stimuli 3
Response To Palpation Of No reaction to palpation 0
Painful Area Mild reaction to palpation 1
Resistance to palpation 2
Violent reaction to palpation 3
Behavior Criteria Score
/21
Appearance (reluctance to Bright, lowered head and ears, no reluctance to move 0
move, restlessness, agitation, Bright, alert, occasional head movement, no reluctance to move 1
anxiety) Restless, pricked up ears, abnormal facial expressions, dilated 2
pupils
Excited, continuous body movements, abnormal facial expression 3
Sweating No obvious signs of sweat 0
Damp to the touch 1
Wet to the touch, beads of sweat apparent over body 2
Excessive sweating, beads of sweat running off the animal 3
Kicking At Abdomen Quietly standing, no kicking 0
Kicking at abdomen 1 - 2 times/5 mins 1
Kicking at abdomen 3 - 4 times/5 mins 2
Kicking at abdomen >5 times/5 mins, intermittent attempts to roll 3
Pawing On Floor (includes Quietly standing, no pawing 0
pointing or hanging limb) Occasionally pawing (1 - 2 times/5 mins) 1
Frequent pawing (3 - 4 times/5 mins) 2
Excessive pawing (>5 times/5 mins) 3
Posture (weight distribution Stands quietly, normal walk 0
and comfort) Occasional weight shift, slight muscle tremors 1
Non-weight bearing, abnormal weight distribution 2
Analgesic posture, attempts to urinate, prostration, muscle tremors 3
Head Movement (lateral or No evidence of discomfort, head straight ahead mostly 0
vertical head movements) Intermittent head movement, looking at flanks or lip curl 1 - 2 times 1
/5 mins
Intermittent, rapid head movement, looking at flanks or lip curl >5 2
times/5 mins
Continuous head movements, looking at flanks or lip curl >5 3
times/5 mins
Appetite Eats hay readily 0
Hesitates to eat hay 1
Little interest in eating hay, takes hay but doesn’t chew or swallow 2
Neither shows interest in nor eats hay 3
Total CPS 39

FIG. 3.2  Multifactorial numeric rating composite pain scale for evaluating pain in horses. (Reprinted with
permission from Elsevier. Bussières G, Jacques C, Troncy E, et al. Development of a composite orthopedic
pain scale in horses. Res Vet Sci. 2008; 85:294.)
142 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

FIG. 3.3  A multidimensional, nonvalidated pain scale that is used at the authors’ institution.
CHAPTER 3  Recognizing and Treating Pain in Horses 143

A B

FIG. 3.4  Examples of equids demonstrating facial expressions of pain. (A) Note the horse has partially
closed eyes and drawn back upper lip. (B) The donkey shows ears pointed back and strained nostrils.

be one reason why there are limited validated pharmaco- housekeeping functions in the body and play a significant role
logic options for analgesia in horses compared with those in the development of pain, inflammation, and fever.
for dogs and cats. Practically, there may also be limitations There are at least two isoforms of cyclooxygenase, and the
to the use of certain drugs in horses with respect to drug most important ones are COX-1 and COX-2. COX-1 is con-
volume or cost. As in any species, analgesic drugs also have sidered constitutively expressed in most body tissues and plays
potential adverse effects that may limit their use. Pharma- an important role in maintaining renal blood flow, protecting
cologic approaches tend to provide the greatest benefit with gastric mucosa, and facilitating platelet aggregation (among
fewer or less severe adverse effects when drugs are selected other processes). The COX-2 isoform has a greater role in the
for a specific purpose, used preemptively, or as a part of a development of inflammation and pain, and traditionally thera-
multimodal approach, and the dose is scaled to the severity peutic effects of NSAIDs are attributed to inhibition of COX-2
of pain. effects.35,36 However, COX-2 can also have housekeeping func-
Classes of drugs most commonly used in the horse include tions in the gastrointestinal tract and kidney, with greater effects
antiinflammatories, opioids, α2-adrenergic agonists, and local in diseased states,37 and the traditional absolute separation into
anesthetics. There are, however, several additional drugs that constitutive with respect to COX-1 and inducible with respect
have a place in equine pain management, and these have been to COX-2 is an oversimplification of the roles of the COX iso-
included here. Greater detail about some of these drugs may forms.38,39 The existence of a COX-3 isoform, originally dis-
be found in Chapter 2 and numerous additional resources.24-26 covered as a variant of COX-1 in dogs, is reported in canine,
Dosing information is provided in Table 3.1. rodent, and human literature.40 COX-3 is the purported target
of acetaminophen (paracetamol), although considerable debate
Antiinflammatory Drugs exists regarding this topic.41,42 Whether a COX-3 isoform exists
Antiinflammatory drugs used in horses include cortico- in horses is unknown. Regardless, important species differences
steroids and NSAIDs. They are often the first-line choice in exist in the expression of different COX isoforms in various tis-
the management of pain related to inflammatory conditions sues, and extrapolation of what works in a dog or a human is not
(e.g., synovitis and osteoarthritis)27-29 and are administered uniformly applicable to the horse.43,44
by various routes, including systemically, intraarticularly, and The most common adverse effects associated with blocking
topically.30-32 Generally, NSAIDs are the most commonly used the COX pathways are seen in the gastrointestinal and renal
drugs in horses.33 They are readily available and relatively systems. In horses gastric ulceration, right dorsal ulcerative
inexpensive and do not require precise record keeping. colitis, and renal papillary/crest necrosis are reported with
Corticosteroids act on the enzyme phospholipase A2 to NSAID use.45-47 Renal effects may be worsened by concurrent
prevent the breakdown of membrane phospholipids to arachi- dehydration.48 The reader should be aware that although the
donic acid, which is the precursor to inflammatory prostaglan- practice of combining multiple NSAIDs or corticosteroids and
dins and leukotrienes.26 NSAIDs act to block cyclooxygenase NSAIDs may result in increased analgesia, this practice is also
(COX), the enzyme that produces prostaglandins from arachi- associated with increased frequency and severity of adverse
donic acid.34 Prostaglandins and leukotrienes have important effects.49-52 In circumstances in which pain is best managed by
144 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

TABLE 3.1  Drugs Used for Analgesia in Horses

Drug Route Dose Comments


NONSTEROIDAL ANTIINFLAMMATORY DRUGS
Phenylbutazone28 IV, PO 2.2–4.4 mg/kg q12–24 h Lower doses suggested for chronic use
Flunixin meglumine230 IV, PO 1.1 mg/kg q 12–24 h IM route on label but not recommended
because of potential for local tissue
irritation
Ketoprofen28,231 IV 1.1–2.2 mg/kg q 12–24 h —
Firocoxib57 IV 0.09 mg/kg q 24 h Nonaqueous solution should not be
mixed with aqueous flush solutions in
IV lines
PO Loading dose: 0.3 mg/kg —
Then: 0.1 mg/kg q 24 h
Meloxicam55,56 PO 0.6 mg/kg q 24 h Increased dose frequency may be re-
quired for foals (q 12 h)
Diclofenac32 Topical 1% liposomal cream applied to —
bare skin over area of interest
OPIOIDS
Can be combined with acepromazine, α2-adrenergic agonists. Dilute epidural dose to desired volume.
Butorphanol62,63,179,232-235 IV, IM, SC 0.01–0.22 mg/kg Common clinically used doses are lower
than reported analgesic doses
IV CRI CRI: 0.013–0.025 mg/kg/h Loading dose recommended
Nalbuphine236 IV, IM 0.02–0.15 mg/kg —
Buprenorphine83,237-239 IV, sublingual 0.005–0.01 mg/kg —
Epidural 0.005 mg/kg —
Morphine 87-89,91,93,240,241 IV, IM 0.05–0.2 mg/kg —
IV CRI 0.1 mg/kg/h Loading dose recommended
Epidural 0.1–0.2 mg/kg —
Intraarticular 0.1–0.2 mg/kg —
Fentanyl81,201,242 IV CRI Loading dose: 0.005 mg/kg Less effective analgesic in horses com-
CRI: 0.006 mg/kg/h pared with other species
Transdermal 20- to 30-mg adhesive patch As an adjunct to other analgesics
applied to bare skin per ap-
proximately 500-kg adult horse
Remifentanil 240,243,244 IV CRI Loading dose: 0.0005 mg/kg As an adjunct to general anesthesia
CRI: 0.002–0.006 mg/kg/h
Methadone133,245,246 IV, IM, PO 0.05–0.15 mg/kg —
Epidural 0.1 mg/kg —
Meperidine (pethi- IV, IM 1 mg/kg —
dine)62,247-249 Epidural 0.3–0.8 mg/kg —
Hydromorphone 250 Epidural 0.04 mg/kg —
α2-ADRENERGIC AGONISTS
Doses provided will cause sedation. Lower doses may be effective for analgesia, especially in combination with opioids.
CRI doses are used for standing procedures or as an adjunct to general anesthesia.
Xylazine251,252-255 IV, IM 0.2–1.1 mg/kg —
IV CRI 0.016–0.69 mg/kg/h —
Epidural 0.17–0.22 mg/kg Hind limb weakness/recumbency is a
possible side effect
Detomidine 93,244,252,256-258 IV, IM 0.01–0.03 mg/kg —
IV CRI 0.005–0.02 mg/kg/h —
Epidural 0.015–0.03 mg/kg Systemic absorption is likely
Romifidine235,252,259,260 IV, IM 0.02–0.08 mg/kg —
IV CRI 0.03–0.04 mg/kg/h —
Epidural 0.03–0.06 mg/kg —
CHAPTER 3  Recognizing and Treating Pain in Horses 145

TABLE 3.1  Drugs Used for Analgesia in Horses—cont’d

Drug Route Dose Comments


Dexmedetomi- IV, IM 0.003–0.005 mg/kg —
dine240,241,261,262 IV CRI 0.001–0.008 mg/kg/h —
Medetomidine251,263,264 IV, IM 0.007–0.01 mg/kg —
IV CRI 0.003–0.005 mg/kg/h —
LOCAL ANESTHETICS
Epidural doses may be combined with opioids or α2-adrenergic agonists; recommended volume when local anesthetics are used
is not to exceed 5–7 mL in an adult horse.
Lidocaine133,247,265,266 IV CRI Loading dose: 1.3–1.5 mg/kg Loading dose given over 10–20 min
CRI: 0.05 mg/kg/min
Sacrococcygeal 0.2–0.35 mg/kg —
epidural
Mepivacaine267,268 Sacrococcygeal 0.16–0.2 mg/kg —
epidural
Ropivacaine269,270 Sacrococcygeal 0.08–0.15 mg/kg —
epidural
Bupivacaine271,272 Sacrococcygeal 0.02–0.06 mg/kg —
epidural
OTHER DRUGS
Ketamine150-152 IV CRI 0.4–0.8 mg/kg/h In conscious horses higher doses can be
used as an adjunct to general anes-
thesia
Gabapentin160,161 IV, PO 5–20 mg/kg q 8–12 h Low oral bioavailability; higher dosing
may be needed; titrate dose to effect
Methocarbamol236 PO 40–60 mg/kg q 12–24 h —
N-butylscopolamine179 IV 0.3 mg/kg Lower doses may achieve desired clinical
effect
  

IV, Intravenously; PO, orally; IM, intramuscularly; SC, subcutaneously; CRI, constant rate infusion.

a combination of drugs within these two classes, concurrent pure μ-agonists (e.g., morphine, methadone, fentanyl), partial
use of gastrointestinal protectants is suggested. μ-agonists (e.g., buprenorphine), and κ-agonist/μ-antagonists
Most NSAIDs approved for use in horses (e.g., aspirin, phen- (e.g., butorphanol). Morphine is the prototypic μ-agonist with
ylbutazone, ketoprofen, flunixin meglumine, diclofenac) work which all other opioids are compared.59,60 Although tradition-
by inhibiting both COX-1 and COX-2. However, COX-2–selec- ally pure μ-agonist opioids are considered to provide the most
tive NSAIDs (firocoxib and meloxicam) have been developed potent analgesic effects, butorphanol is reported to be equally
for horses, with the primary goal of reducing gastrointestinal as or more effective (with potentially fewer adverse effects)
side effects,53-55 especially because NSAIDs may be used chron- than μ-agonists in the treatment of gastrointestinal pain in
ically in some horses. Meloxicam specifically may offer further horses.60-63 Opioids, especially morphine, produce profound
benefit in foals, in which traditional NSAIDs have a higher risk sedative effects when combined with α2-agonists. This is com-
of adverse effects because of prolonged clearance in foals com- monly termed neuroleptanalgesia and can be useful in animals
pared with adult horses. Meloxicam is cleared more rapidly in that are refractory to analgesia or when standing interventions
foals than adults, which may result in improved tolerance to are necessary.64,65
longer treatment durations.56 Opioid use in horses is often limited by the potential for
The use of a canine chewable tablet formulation of firocoxib adverse effects. μ-Agonists in particular are known to cause
has become popular with horse owners because of low cost and excitement, agitation, and increased locomotor activity in
ease of use; however, the canine tablet has reduced bioavailabil- horses.58,66,67 This may be explained in part by the loca-
ity and reduced COX-2 inhibition in horses compared with the tion, density, and binding affinity of μ-opioid receptors in
oral equine formulation.57 Additionally, practitioners should be the equine brain.68 Although this effect is well documented,
aware that use of the canine formulation may represent extra- the clinical importance is confounded by multiple variables,
label drug use depending on their country of practice. The including potential bias in the studies because they were con-
same is true of the COX-2–selective NSAID carprofen, which is ducted using healthy, nonpainful horses. Dosages used in
licensed for dogs but has been administered to foals in place of investigational studies may also be higher than dosages used
phenylbutazone because of perceived efficacy and lower toxicity.  clinically, contributing to excitatory effects.69 As mentioned
previously, coadministration with sedative drugs ameliorates
Opioids the potential excitatory effects of opioids and augments seda-
Opioids produce their effects by activating mu (μ), kappa tion.58,68 In painful horses, low to moderate doses of opioids
(κ), or delta (δ) opioid receptors.58 Available opioids include have a calming effect and in some studies are associated with
146 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

improved anesthesia recovery quality,70,71 but variability exists nervous system α2-receptors in areas of the brain that are
with respect to behavioral responses to opioids in horses. In responsible for awareness, arousal, and vigilance. Activation of
other species for which extensive research exists, differences α2-receptors in the brain and spinal cord decreases the release
have been attributed to opioid receptor polymorphisms. A of excitatory neurotransmitters and interferes with sensory
μ-opioid receptor polymorphism has recently been shown to processing and transmission, producing analgesia.94
significantly affect locomotor responses to fentanyl in adult α2-Adrenoceptor agonists are profound visceral analgesics.
horses.72 Continued research in this area may help to clarify This property is crucial in the management of moderate to
varying clinical observations regarding opioid use in this severe colic. The use of α2-adrenoceptor agonists can prevent
species. injury to both practitioner and horse and allow critical diag-
A further untoward effect of opioids is that they can delay nostic and treatment procedures to be performed. These drugs,
gastric emptying and prolong intestinal transit time. Because however, may also mask signs of clinical deterioration,95,96
of this, opioids can predispose horses to constipation and especially if higher doses or drugs with longer half-lives are
colonic impaction, because water is absorbed from the slow- administered (e.g., detomidine).97 Behavioral and possibly
moving contents of the intestinal lumen.73,74 Although it has analgesic effects may be improved when α2-adrenoceptor
been the authors’ experience that horses receiving large doses agonists are administered with opioids (e.g., butorphanol or
of opioids such as morphine can show gastrointestinal sta- morphine).63
sis necessitating passage of a nasogastric tube to relieve gas, The use of α2-adrenoceptor agonists is not without sig-
studies on the risk of colic associated with morphine use in nificant adverse effects. Clinically, the administration of α2-
horses undergoing general anesthesia are conflicting.69,75-77 agonists for the purpose of analgesia is accompanied by varying
Most show no relationship between the perioperative use of levels of sedation, which may be undesirable. α2-Adrenoceptor
morphine and gastrointestinal complications. In addition, agonists also cause peripheral vasoconstriction and subse-
pain itself may predispose horses to ileus.78,79 Although the quent baroreceptor-mediated reflex bradycardia and bradyar-
potential for adverse effects should not to be ignored, it should rhythmias (e.g., second degree atrioventricular block).98,99 The
be noted that complications are most likely when opioids are increase in afterload associated with α2-adrenoceptor agonist
used systemically at large doses. administration causes a profound reduction in cardiac out-
Experimental studies on the use of opioids in horses con- put,99-101 which may be detrimental in a horse with cardio-
tinue to emerge as potential clinical benefits of this class of vascular compromise. As the vasoconstrictive effect wanes, a
drugs are increasing recognized. Several new studies describe decrease in arterial blood pressure follows, which is caused by
the pharmacology of specific opioids in horses, but the need a decrease in central nervous system sympathetic output and
for additional evidence to support specific clinical treatment continued bradycardia.94,102 Decreases in cardiac output are
protocols is great. For example, intravenous fentanyl appears relatively independent of dose.103 In horses with severe hemo-
to be a relatively ineffective analgesic with undesirable behav- dynamic compromise (e.g., strangulating colic), the benefits of
ioral effects at the doses studied in adult horses,80,81 but it has a the use of α2-adrenoceptor agonists must be weighed carefully
sedative effect in foals at relatively low doses.82 Buprenorphine with the risk of cardiovascular collapse.
shows promise as an analgesic because of its long plasma half- Other adverse effects of α2-adrenoceptor agonists include
life in horses and its efficacy via sublingual administration.83 occasional paradoxic excitement or aggression, changes in
Reports of significant buprenorphine-induced excitement or respiratory rate and tidal volume, upper airway obstruction
increases in locomotor activity exist in adult horses and foals, caused by relaxation of pharyngeal muscles, hyperglycemia,
however, even when the drug is given in combination with a increased urine production, and immediate and pronounced
sedative.84-86 The use of systemic opioids in horses will, there- decreases in gastrointestinal tone and motility that can last
fore, continue to be challenged by the need for appropriate well beyond the duration of the drug.104-110 These effects are
case selection and dose titration such that clinical benefits are dose dependent but can be related to postoperative ileus and
achieved without undesired effects. colic.111 Increased urine production could exacerbate dehy-
Systemic adverse effects can be mitigated via the use of dration in a debilitated horse if supplemental fluids are not
alternative routes of administration. Routes that have been provided. Tachypnea is sometimes observed when febrile
studied in horses with promising results include intraarticular, horses receive any of this class of drugs.
transdermal, perineural, and epidural administration. Intraar- α2-Adrenoceptor agonists can be administered as bolus
ticular morphine, for example, has a profound and lasting doses or via constant rate infusion.110 Like opioids, they
analgesic and antiinflammatory effect and produces a more can also be administered intraarticularly, perineurally,
significant reduction in lameness than intravenous morphine and epidurally. Intraarticular use of α2-adrenoceptor ago-
in horses with synovitis.87-90 Epidural morphine provides nists provides analgesia by an undetermined mechanism;
useful and prolonged analgesia for a variety of surgical pro- regional and systemic effects (from drug absorption), how-
cedures,91-93 although dose-dependent decreases in gastroin- ever, are possible.112 The potential for chondrotoxic effects
testinal transit time are reported.79,91  of α2-adrenoceptor agonists are currently under investiga-
tion, with early reports indicating the potential for chon-
α2-Adrenoreceptor Agonists drotoxicity with some but not all drugs in this class.113,114
α2-Adrenoreceptor agonists (e.g., xylazine, detomidine, romif- α2-Adrenoceptor agonists have been shown to improve dura-
idine, medetomidine, dexmedetomidine) produce sedation, tion of analgesia provided by perineural local anesthetics.115
muscle relaxation, and analgesia via activation of central and Epidural administration of α2-adrenoceptor agonists pro-
peripheral α2-receptors. Although drug effects are broadly vides beneficial analgesia for surgical procedures.93,116 Some
the same, individual differences exist with respect to α1:α2- α2-adrenoceptor agonists (e.g., detomidine) demonstrate
receptor selectivity, chemical structure, drug metabolism, systemic effects, however, because they are rapidly absorbed
and elimination. Sedation is attributed to activation of central from the epidural space.116,117
CHAPTER 3  Recognizing and Treating Pain in Horses 147

The effects of α2-adrenoceptor agonists can be reversed the local anesthetic effect has worn off. Local anesthetic drugs
with α2-adrenoceptor antagonists (e.g., atipamezole, tolazo- can also cause respiratory paralysis when administered by
line, yohimbine, idazoxan).111,118 When using these reversal epidural or spinal (subarachnoid) routes. Although unlikely
agents, all α2 effects, including analgesia, are abolished. If unless excessive drug volume is administered inadvertently,
α2-adrenoceptor antagonists are used in a situation in which the migration of local anesthetic cranially to the cervical
pain is anticipated, the practitioner should plan to provide region can paralyze the diaphragm, resulting in hypoventila-
analgesia by an alternate method. The use of α2-adrenoceptor tion and apnea. To avoid these adverse effects of epidural local
antagonists has been associated with significant adverse effects anesthetics, the authors recommend these drugs generally not
in horses, including behavioral changes, tachycardia, hypoten- be placed in epidural catheters and that the volume of local
sion, and death. Adverse effects are more often associated with anesthetic drug when using the caudal epidural space be lim-
high doses and rapid (e.g., intravenous) administration.119 ited to 5 to 7 mL for an adult horse.
Assessment of peripheral α2-adrenoceptor antagonists that Intraarticular use of local anesthetics is common in equine
retain beneficial central effects and reduce the negative cardio- practice for the diagnosis and management of lameness and
vascular effects of α2-adrenoceptor agonists is ongoing.120,121  surgery of the joints. Of concern is the potential for local anes-
thetic-induced chondrotoxicity, which has been clinically rec-
Local Anesthetics ognized in humans with the use of bupivacaine.134 An in vitro
Local anesthetics (e.g., lidocaine, mepivacaine, bupivacaine, investigation in the horse showed chondrotoxicity associated
ropivacaine) are frequently used alone or in combination with with bupivacaine, lidocaine, and mepivacaine, with mepi-
other analgesic drugs to produce loss of sensation. They may vacaine considered the least toxic to cartilage and safest for
be administered topically, regionally (perineurally and epi- clinical use.135 A subsequent in vitro human study showed that
durally), or systemically. Analgesia results from a blockade chondrotoxic effects were worse in arthritic joints.136 In con-
of sodium ion channels, which prevents the initiation and trast, a single injection of bupivacaine and lidocaine produced
conduction of electrical activity (action potentials) in small- no increases in collagen degradation biomarkers in an in vivo
diameter (C, Aδ) sensory nerve fibers. Larger (clinical) doses study in healthy horses.137 More data are necessary before the
of local anesthetics also block conduction in large fibers (Aβ), issue is clarified in horses, and practitioners should weigh
and when administered at certain sites (e.g., epidural, perineu- potential risks with benefits in individual clinical scenarios. 
ral to a mixed nerve) produce a loss of motor function and
result in temporary motor paralysis. Ketamine
The local anesthetic lidocaine when administered intrave- Ketamine is commonly used as an anesthetic induction and
nously produces mild central nervous depression and has anti- maintenance agent in horses and also possesses important
arrhythmic, antiinflammatory, free radical scavenging, and analgesic properties. NMDA receptor activation is critical
gastrointestinal promotility effects.122-127 It also provides anal- to the development of hyperalgesia and central sensitization
gesia, decreases the requirement for inhalant anesthetics, and mediated through interaction with a number of other recep-
potentiates the analgesic actions of opioids or α2-adrenoceptor tors.138,139 Ketamine’s antagonism at this receptor helps miti-
agonists.122,123,128,129 Because of variable cardiac toxicity, gate these effects. Analgesia may also be partly mediated via
administration of other local anesthetic agents by this route the monoaminergic pathway through ketamine-induced
is currently not recommended. Lidocaine produces minimal release of norepinephrine, dopamine, and serotonin.138
cardiovascular and respiratory effects in otherwise normal, At anesthetic doses, ketamine increases heart rate, cardiac
healthy horses but can decrease cardiac output, arterial blood output, and arterial blood pressure in horses.140 When used
pressure, and heart rate when administered intravenously with a goal of preventing central sensitization as an adjunct to
because of decreases in central nervous system sympathetic inhalant anesthesia, ketamine infusion both reduces inhalant
output, myocardial contractile force, and venous return.123,128 dose requirement and improves cardiac output.141 Beneficial
Lidocaine infusion does not improve cardiovascular function cardiovascular effects are attributed to increased circulating
in anesthetized horses despite decreases in inhalant anesthetic catecholamines, even though ketamine in isolation is a direct
dose, and in certain circumstances it may predispose horses myocardial depressant.142,143 Nonsystemic routes of ketamine
to a poorer quality of recovery from general anesthesia.129-131 administration have also been used with some success. For
Significant differences in metabolism, elimination, and the example, short-term local anesthesia is observed when used
potential to produce central nervous system toxicity (disori- perineurally in horses.144 Similarly when administered epi-
entation, ataxia, and seizures) are seen among different local durally, ketamine produces analgesia, but evidence of local-
anesthetics.125 Horses appear to be more sensitive to the neu- ized neuronal toxicity, especially with chronic use, limits its
rotoxic effects than some other species, with large doses pro- use in clinical patients.145,146
ducing central nervous stimulation typified by nervousness, Behavioral side effects associated with ketamine use in
agitation, sedation, ataxia, and collapse.132 Seizures have also horses must be considered. In humans, ketamine produces
been observed by the authors in clinical patients. Although hallucinations138; in horses, the potential for excitement or
less likely to occur in full-size adult horses, toxic doses may delirium during ketamine use in anesthesia is a reality mini-
also be reached when these drugs are administered locally or mized by the provision of other drugs (e.g., α2-adrenoceptor
regionally. agonists).140,147 Subanesthetic doses of ketamine used to pre-
Local anesthetics can be administered epidurally to decrease vent central sensitization do not seem to be associated with
the need for additional analgesics.133 However, as mentioned adverse behavioral effects in people, dogs, and cats.148,149 In
previously, they have the potential to produce a loss of motor horses, low-dose infusions can be used with minimal to no
function (paralysis), which can become problematic in an behavioral effects.150 Evidence of antinociceptive effect with
animal as large as a horse. The horse may injures itself while this dosing of ketamine in horses is conflicting and clinical
unable to control its limbs or may becomes recumbent until data are sparse, but subanesthetic infusions appear to be an
148 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

effective adjunct for the management of laminitic pain.150-153 in horses in multiple research models and caused excitement
Prolonged infusions of ketamine lead to an increase in gastro- with increases in dose.171-173 In clinical patients, tramadol was
intestinal transit time and decreased fecal output in healthy mildly effective at reducing pain in laminitic horses in multi-
horses,154 but similar data from clinical patients are lacking.  modal approaches to pain control,151, 174 and no adverse effects
were seen when it was used as an adjunct to acepromazine,
Magnesium detomidine, ketamine, diazepam, and flunixin meglumine for
Like ketamine, magnesium owes its analgesic effects largely castration.175 Further study is necessary to determine the clin-
to its NMDA receptor blockade. Magnesium is also a calcium ical utility of tramadol in horses, but it is currently not promis-
channel blocker, and although this attribute has historically ing as a sole analgesic in this species. 
been used in the treatment of hypertension and arrhythmias,
it is also an important mechanism by which systemic admin- Soluble Epoxide Hydrolase Inhibitors
istration of magnesium is used to help treat pain.155 Although Oxidative metabolism of polyunsaturated fatty acids is a
magnesium has not been studied as an analgesic in horses, it critical step in the production of mediators important to the
has been extensively studied in human medicine. Some data development of inflammation and pain associated with tis-
are conflicting, but overall it appears that magnesium reduces sue damage. The most well-known group of drugs blocking
postoperative pain and opioid requirements after a variety of this process is the NSAIDs, which prevent arachidonic acid
surgical procedures.156,157 In an experimental setting, mag- metabolism via COX inhibition, as discussed earlier. However,
nesium also potentiates analgesia provided by morphine and the COX pathway is only part of a much more complex set of
delays the development of morphine tolerance.158 Adverse processes responsible for inflammatory and neuropathic pain.
effects appear to be minimal with appropriate dosing but Recently inhibition of the cytochrome P450–mediated
include hypotension, bradycardia, and sedation.157  transformation of polyunsaturated fatty acids into epoxides
has been explored as a potential analgesic treatment for horses
Anticonvulsants with laminitis.176 Soluble epoxide hydrolase activity measured
Gabapentin is an anticonvulsant drug that also has analgesic in the digital laminae is increased in laminitic horses, and
effects, which is likely caused by its interaction with the α2δ- experimental treatment with the soluble epoxide hydrolase
subunit on voltage-gated calcium channels, which is upregu- inhibitor t-TUCB provided moderate improvement in clini-
lated in chronic and neuropathic pain. In humans, gabapentin cal signs (e.g., limb lifting) and pain scores in a small group of
is considered a first-line drug for the treatment of neuropathic laminitic horses in a recent clinical trial.177 The novel nature
pain.159 It has low oral bioavailability in horses.160 Oral gaba- of this treatment means that further research is needed before
pentin given for 2 weeks at common clinical dosages (5–10 widespread use is adopted, but it represents a promising addi-
mg/kg every 8 hours) failed to reduce the lameness level of tion to a multimodal approach to pain control in horses with
chronically lame horses compared with placebo treatment.161 laminitis. 
However, a relatively low oral dose appeared effective for treat-
ing neuropathic pain in a horse in one case report.162 Acepromazine
When given intravenously, 20 mg/kg of gabapentin was Acepromazine is a phenothiazine tranquilizer that does not
associated only with very mild sedation,160 suggesting this have inherent analgesic properties.178,179 As such, it should not
higher dosage could be used without significant adverse be used as a sole agent with the intent to provide analgesia, nor
effects. In humans, appropriate dose titration may take several should its calming effects be interpreted as analgesic efficacy.
weeks to achieve.163 More clinical data are necessary to deter- Although not licensed for such use, acepromazine is used by
mine whether this is a factor in the efficacy of gabapentin in veterinarians in combination with α2-adrenoceptor agonists
horses. The primary dose-limiting adverse effect of gabapentin or opioids to augment sedation. The literature also suggests
in humans is somnolence.163 that acepromazine administration improves undesired loco-
Pregabalin is a similar drug with the same mechanism motor activity associated with the use of opioids in horses.58 
of action used as both an anticonvulsant and analgesic in
humans. When given via nasogastric tube to healthy horses, N-Butylscopolamine
bioavailability was high and sedation was the most common Butylscopolamine is an anticholinergic and antispasmodic
adverse effect.164 Use of pregabalin in equine clinical patients drug that has a short duration of effect on gastrointestinal pain
has not been reported.  and is used to treat spasmodic colic,179-181 and it can be given in
combination with other analgesic drugs. Because its duration
Tramadol of action is very short (20–30 minutes), a perceived benefit is
Tramadol is a synthetic weak μ-opioid receptor agonist and that it will not mask pain from a deteriorating, nonspasmodic
serotonin and norepinephrine reuptake inhibitor. Tramadol colic while still providing relief to horses with uncomplicated
is not a controlled drug in many countries. There are several colic pain. It should be noted that the anticholinergic effect of
metabolites of tramadol, but O-desmethyl-tramadol (M1) is this drug will increase the heart rate, which may complicate
considered the primary analgesic metabolite because it has physiologic assessment of pain. 
increased affinity for the μ-receptor compared with the parent
compound.165,166 Methocarbamol
In pharmacokinetic studies, horses show wide individ- Methocarbamol is a skeletal muscle relaxant that has been
ual variability in production of M1, and this metabolite has used in horses for several years to treat discomfort associated
either not been detectable or measured in very low concen- with muscle spasms, and it is moderately orally bioavailable
trations.167-170 It is possible that horses efficiently glucuroni- in horses.182 Although there is considerable anecdotal evi-
date the M1 metabolite, affecting M1 concentrations.167 dence of its use to treat equine back pain, scientific data are
Regardless, tramadol did not have an antinociceptive effect lacking. 
CHAPTER 3  Recognizing and Treating Pain in Horses 149

Bisphosphonates
Tiludronate and clodronate are bisphosphonate drugs used to
treat bone-related pain in horses. Bisphosphonates distribute
primarily to bone and inhibit bone resorption primarily via
negative effects on osteoclasts and macrophages.183,184 These
drugs are given intravenously and intramuscularly, respec-
tively, with a duration of effect of several months.
Small clinical studies with tiludronate have shown some
improvement in lameness caused by navicular disease and distal
hock arthritis,185 and a larger double-blind, placebo-controlled
trial for hock arthritis also demonstrated this drug’s efficacy.186
Tiludronate has also proven efficacious in the treatment of tho-
racolumbar vertebral pain in horses.187 Although it has been
administered during regional limb perfusion, its safety and the
efficacy of this technique are not known.188 Colic has been associ-
ated with the use of tiludronate in horses,185 and varying degrees
of renal damage have been demonstrated in humans with the use
of different bisphosphonates.189 Extensive clinical data regarding
renal toxicity in horses do not exist. However, the potential for
adverse effects should be strongly considered in horses before FIG. 3.5  An epidural catheter secured with a bandage, which prevents
treatment, because anecdotal reports of renal damage are increas- debris (such as straw or shavings) from contaminating the catheter.
ing. The practice of pretreating with flunixin meglumine to pre-
vent tiludronate-associated colic signs is discouraged because of
the concern for compounding nephrotoxicity. 

Autologous Conditioned Serum (Interleukin-1


Receptor Antagonist Protein)
Interleukin-1 receptor antagonist protein (IRAP) is an antiin-
flammatory cytokine used in the treatment of equine osteo-
arthritis. Whole blood is collected and incubated with beads
that enhance the production of IRAP.190 Serum is then sepa-
rated and injected intraarticularly. Clinically, IRAP improves
lameness scores in horses with osteoarthritis compared with
placebo.191 Because the product is autologous and includes
no drugs, it can be used in performance horses before
competition. 

Y SPECIFIC ANALGESIC TECHNIQUES


FIG. 3.6  Components of a commercially available epidural catheter kit,
Epidural Catheters including scalpel blade, catheter, connector, filter, plastic loss of resistance
Epidural catheters (Fig. 3.5) can be placed in horses to pro- syringe, and Tuohy needle for placement. (Photo courtesy Gregg Griffen-
vide a safe and efficient way to administer repeated doses of hagen, DVM, DACVAA.)
analgesic drugs (e.g., opioids, α2-adrenoceptor agonists) into
the epidural space. Epidural catheter kits that can be used in This technique involves continuous infusion (or intermittent,
horses are available commercially (Fig. 3.6). These catheters low-volume boluses) of local anesthetics in catheters placed
can be maintained successfully for several days to weeks. In adjacent to peripheral nerves. A significant advantage of this
clinical cases, patient-related complications (e.g., inflamma- technique is the ability to reduce or discontinue systemi-
tion at the catheter insertion site) appear to be very infrequent cally administered analgesics, thus avoiding the systemic side
when proper aseptic technique is used in the placement and effects of those drugs.194-197
maintenance of epidural catheters. The most common cath- In horses, this technique has been developed for use in the
eter-related complications in horses include dislodgement of distal limb, in which a perineural catheter can be placed in
the catheter or filter, loss of patency of the catheter, or catheter a standing, sedated patient and maintained under a bandage
leakage.192 Indwelling catheters do cause epidural inflamma- thereafter (Fig. 3.7). Possible complications include inadver-
tion and fibrosis, which may be responsible for loss of patency tent vascular or synovial structure puncture, local infection,
over time.192,193 These issues do not preclude replacement with nerve injury, and swelling or hematoma. Complications can
a new catheter but may make it more challenging.  be avoided by adhering to proper landmarks and using strict
aseptic technique.194,198 
Perineural Catheters
Continuous peripheral nerve block via the use of perineural Topical and Transdermal Drug Administration
catheters has been successfully used in human medicine for Topical administration of analgesic drugs offers advantages
a variety of applications, including postoperative analgesia. to systemic drug administration. Application is directed over
150 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

has not been associated with untoward side effects, and there
are promising reports of clinical efficacy in horses in pain.201
Pharmacologic data, however, show widely variable absorp-
tion in both adult horses and foals, in which effective plasma
concentrations were not reached in all individuals.202,203
Several factors may affect patch efficacy, including improper
patch placement and premature patch removal. Conditions
under which the patch is applied (e.g., whether hair is clipped
and the area cleaned) or anatomic location of the patch on the
horse might also affect absorption. Location-dependent dif-
ferences in absorption could be related to differences in the
thickness of the stratum corneum or differences in cutaneous
blood flow. In vitro work with horse skin suggests that absorp-
tion through the groin or thorax is superior to absorption
through skin on the distal limb.204 

Y COMPLEMENTARY THERAPIES
Complementary techniques may play an important role in mul-
timodal analgesic management of horses. Acupuncture, chiro-
practic manipulations, massage, and extracorporeal shock wave
therapy have been adopted and successfully used by equine vet-
erinarians.205 At their introduction, these therapies were used
in horses with primarily anecdotal evidence, but scientific data
A B on these modalities are emerging, especially as they become
increasingly popular with horse owners.

FIG. 3.7  (A) Anatomic diagram showing proper placement of a distal Acupuncture
limb perineural catheter in the horse, 5 cm distal to the accessory car- Acupuncture is purported to act by restoration of energy flow
pal bone (ACB) on the medial aspect of the limb, and 7 cm distal to ACB via stimulation of certain points on channels or “meridians”
on the lateral aspect. Dotted lines between points A and B and D and E within the body.206 Points can be stimulated with needles
represent the intended path for subcutaneous tunneling of the catheter. alone, injected substances, or electrically (“electroacupunc-
Dashed lines between points B and C and E and F represent the intended ture”).207 Modern scientific research has shown that analge-
path for the subfascial placement of the catheter, ending distal to the com- sia is produced by the stimulation of specific (acupuncture)
municating branch (cb) between the medial and lateral palmar nerves and points and not produced by similar stimulation of other points
proximal to the proximal sesamoid bones (PSB). (B) Perineural catheter on the body.208 The analgesic effect results from modulation of
secured in place on the lateral aspect of the left front limb of a horse. (Re- nociceptive input at the level of the dorsal horn of the spinal
printed with permission from Elsevier. Driessen B, Scandella M, Zarucco cord through activation of antinociceptive pathways and the
L. Development of a technique for continuous perineural blockade of the release of endogenous opioids.209 Acupuncture is an appeal-
palmar nerves in the distal equine thoracic limb. Vet Anaesth Analg. 2008; ing modality because of its relative safety and lack of systemic
35:432.) drug levels, which allows its use in performance horses during
competition.
the desired site of action such that the dose (and therefore In horses, acupuncture and electroacupuncture have posi-
side effects) may be reduced. The NSAID diclofenac is avail- tive effects on endorphin release and cutaneous analgesia.207,210
able in the United States in a liposomal formulation that has Electroacupuncture has also been used to increase pain thresh-
been shown to reduce clinical signs of lameness in horses with old in an experimental rectal distention model.211 Clinical
osteoarthritis, providing beneficial disease-modifying effects results, however, have been inconsistent. In one study, elec-
compared with systemically administered phenylbutazone.199 troacupuncture may have provided clinical improvement in
This equine formulation is not available worldwide, and the horses with chronic lameness, but differences from controls
use of topical diclofenac products formulated for humans has were not significant.212 In a more recent study of naturally
thus been adopted in many places. These formulations, how- occurring palmar heel pain, acupuncture failed to consistently
ever, do not offer similar absorption and efficacy, which is affect pain in those horses.213 Further research into the use of
likely caused by differences between human and equine skin acupuncture as a clinical modality is needed to define which
for which these products were not designed.200 conditions might benefit the most from this treatment. 
Transdermal drug administration offers a method for
continuously delivering systemic drugs over several hours or Extracorporeal Shock Wave Therapy
days. This is especially beneficial for drugs with a short dura- Extracorporeal shock wave therapy is used to treat a variety
tion of effect, such that multiple bolus doses can be avoided. of musculoskeletal injuries in horses, including osteoarthri-
The opioid fentanyl, for example, is widely available as a trans- tis, back pain, stress fractures, tendonitis, and desmitis.214
dermal patch designed to provide controlled fentanyl release Although the exact mechanism of action is not known, the
for 72 hours. Intravenous administration of fentanyl has been basic principle is that when focused, pulsed, high-energy
associated with adverse behavioral effects and questionable waves are applied to the injured area, they lead to neovascu-
analgesic efficacy in horses. In contrast, transdermal fentanyl larization and tissue remodeling.214,215 In some studies, shock
CHAPTER 3  Recognizing and Treating Pain in Horses 151

wave therapy produced acute local analgesia for 48 to 72 hours showing benefits of many different drugs and nontraditional
after treatment,216,217 which has raised concern about poten- analgesic techniques. In addition, the importance of treating
tial catastrophic injury in performance horses subjected to pain and the use of multimodal techniques is more and more
work after minor injury and subsequent treatment. However, widely accepted. Although pain management in the horse
in further studies with specific lesions, shock wave therapy did continues to be challenging, the increasing availability of
not reduce lameness in the immediate posttreatment period, pharmacologic and complementary options provides a reason
and its acute analgesic effect in these cases has been ques- for optimism.
tioned.15,218 Nevertheless, shock wave remains a viable poten-
tial adjunct therapy in a multimodal approach to analgesia.  REFERENCES
Chiropractic Manipulations 1. Merskey HR, Bogduk N. Classification of chronic pain, descrip-
tions of chronic pain syndromes and definitions of pain terms.
Chiropractic manipulations rely on “high-velocity, low-ampli- 2nd ed. Seattle, WA: IASP Press; 1994.
tude” manual thrusts that attempt to restore normal joint 2. Muir WW, Woolf CJ. Mechanisms of pain and their therapeutic
motion and improve tissue function of the equine spine.219 implications. J Am Vet Med Assoc. 2001;219:1346.
These factors might be negatively altered by an underlying 3. Woolf CJ, Salter MW. Neuronal plasticity: increasing the gain in
pathologic condition, such as limb lameness.220 Addressing pain. Science. 2000;288:1765.
this additional potential source of pain could theoretically 4. Woolf CJ, Decosted I. Implications of recent advances in the
improve the overall clinical condition, especially if the under- understanding of pain pathophysiology for the assessment of
pain in patients. Pain Suppl. 1999;6:S141.
lying condition is also managed. Chiropractic manipulations 5. Blackshaw LA, Gabhart GF. The pharmacology of gastrointesti-
change the kinematics of horses with back pain, increase nal nociceptive pathways. Curr Opin Pharmacol. 2002;2:642.
mechanical nociceptive thresholds in healthy horses compared 6. Bueno L, Fioramonti J, Delvaux M, et al. Mediators and phar-
with controls, and reduce cervical muscle activity.219,221,222 The macology of visceral sensitivity: from basic to clinical investiga-
significance of these findings with respect to acute or chronic tions. Gastroenterology. 1997;112:1714.
pain control is unknown.  7. McMahon S, Koltzenburg M, Tracey I, et al. Wall and Melzack’s
textbook of pain. 6th ed. Philadelphia: Saunders; 2013.
Low-Level Laser Therapy 8. Almeida TF, Roizenblatt S, Tufik S. Afferent pain pathways: a
Low-level lasers (“cold lasers”) have become a popular thera- neuroanatomical review. Brain Res. 2004;1000:40.
peutic adjunct in the equine veterinary world. Cold lasers are 9. Love EJ, Taylor PM, Clark C, et al. Analgesic effect of butorpha-
nol in ponies following castration. Equine Vet J. 2009;41:552.
typically relatively low power and nonthermal, such that tis- 10. Bussières G, Jacques C, Troncy E, et al. Development of a com-
sue damage is avoided while beneficial effects are achieved. posite orthopaedic pain scale in horses. Res Vet Sci. 2008;85:294.
There is anecdotal support from performance horse owners 11. Ashley FH, Waterman-Pearson AE, Whay HR. Behavioural as-
regarding the use of laser therapy in the treatment of acute sessment of pain in horses and donkeys: application to clinical
and chronic musculoskeletal pain. Despite enthusiasm for this practice and future studies. Equine Vet J. 2005;37:565.
modality, strong scientific data supporting the use of lasers in 12. Van Loon JPAM, Back W, Hellebrekers LJ, et al. Application of
the horse are sparse.223 Evidence of laser therapy as an analge- a composite pain scale to objectively monitor horses with so-
sic, antiinflammatory, and aid to wound healing is emerging matic and visceral pain under hospital conditions. J Equine Vet
in human medicine and research models.224-226 Studies sug- Sci. 2010;30:641.
gest that possible mechanisms of action include modulation 13. Raekallio M, Taylor PM, Bennett RC. Preliminary investiga-
tions of pain and analgesia assessment in horses administered
of local inflammatory mediators (e.g., prostaglandins, tumor phenylbutazone or placebo after arthroscopic surgery. Vet Surg.
necrosis factor-α [TNF-α]) and reduction or alteration in 1997;26:150.
inflammatory cell infiltration or distribution.227 14. deGraue JC, van de Lest CH, van Weeren R, et al. Arthrogenic
A wide variety of cold lasers are marketed that can be sold lameness of the fetlock: synovial fluid markers or inflammation
directly to horse owners.228 Lasers are classified on a 1 to 4 and cartilage turnover in relation to clinical joint pain. Equine
scale by the U.S. Food and Drug Administration or by the Vet J. 2006;38:305.
International Electrotechnical Commission based on the level 15. Kirker-Head CA, Chandna VK, Agarwal RK, et  al. Con-
of potential operating hazard, with higher classes being more centrations of substance P and prostaglandin E2 in synovial
powerful and also more likely to cause injury if improperly fluid of normal and abnormal joints of horses. Am J Vet Res.
operated.229 Lasers available for equine therapy come from 2000;61:714.
16. Taylor D, Hood DM, Wagner IP. Short-term effect of therapeu-
one of the two highest classes: class 3B or class 4. Class 4 lasers tic shoeing on severity of lameness in horses with chronic lami-
are typically more expensive, more powerful and more effi- nitis. Am J Vet Res. 2002;63:1629.
cient and can treat larger or deeper areas. Protocols and laser 17. Price J, Catriona S, Welsh EM, et al. Preliminary evaluation of
types used in human medicine vary across studies, and there is a behaviour-based system for assessment of post-operative pain
no scientific consensus as to the most appropriate laser or pro- in horses following arthroscopic surgery. Vet Anaesth Analg.
tocol for use in equine therapy. Positive results from human 2003;30:124.
studies, however, should encourage further research on laser 18. Graubner C, Gerber V, Doherr M, et al. Clinical application and
therapy in horses, which will help to clarify the place of cold reliability of a post abdominal surgery pain assessment scale
laser therapy in management of equine pain.  (PASPAS) in horses. Vet J. 2011;188:178.
19. Dalla Costa E, Minero M, Lebelt D, et al. Development of the
Horse Grimace Scale (HGS) as a pain assessment tool in horses
Y SUMMARY undergoing routine castration. PLoS One. 2014;9:e92281.
20. van Loon JP, Jonckheer-Sheehy VSM, Back W, et al. Monitor-
Equine practitioners have more tools now than ever before to ing equine visceral pain with a composite pain scale score and
assess and manage pain. Detailed pain scales improve clinical correlation with survival after emergency gastrointestinal sur-
pain assessment, and promising clinical studies have emerged gery. Vet J. 2014;200:109.
152 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

21. Sutton GA, Dahan R, Turner D, et  al. A behavior based pain 44. Marshall JF, Blikslager AT. The effect of nonsteroidal anti-
scale for horses with acute colic: scale construction. Vet J. inflammatory drugs on the equine intestine. Equine Vet J Suppl.
2013;196:394. 2011;39:140.
22. Taylor PM, Pascoe PJ, Mama KR. Diagnosing and treating pain 45. McConnico RS, Morgan TW, Williams CC, et al. Pathophysi-
in the horse. Where are we today? Vet Clin North Am Equine ologic effects of phenylbutazone on the right dorsal colon in
Pract. 2002;18:1. horses. Am J Vet Res. 2008;69:1496.
23. Woolf CJ. Pain: moving from symptom control toward mecha- 46. MacAllister CG, Morgan SJ, Borne AT, et  al. Comparison of
nism-specific pharmacologic management. Ann Internal Med. adverse effects of phenylbutazone, flunixin meglumine, and ke-
2004;140:441. toprofen in horses. J Am Vet Med Assoc. 1993;202:71.
24. Riviere JE, Papich MG. Veterinary pharmacology and therapeu- 47. Read WK. Renal medullary crest necrosis associated with phe-
tics. 9th ed. Somerset: John Wiley & Sons; 2013. nylbutazone therapy in horses. Vet Pathol. 1983;20:662.
25. Cunningham FM. Comparative and veterinary pharmacology. 48. Gunson DE, Soma LR. Renal papillary necrosis in horses after
1st ed. Heidelberg: Springer; 2010. phenylbutazone and water deprivation. Vet Pathol. 1983;20:603.
26. Papich MG. Saunders handbook of veterinary drugs: small and 49. Keegan KG, Messer NT, Reed SK, et al. Effectiveness of admin-
large animal. 3rd ed. Philadelphia: Elsevier/Saunders; 2011. istration of phenylbutazone alone or concurrent administration
27. Owens JG, Kamerling SG, Stanton SR, et al. Effects of pretreat- of phenylbutazone and flunixin meglumine to alleviate lame-
ment with ketoprofen and phenylbutazone on experimentally ness in horses. Am J Vet Res. 2008;69:167.
induced synovitis in horses. Am J Vet Res. 1996;57:866. 50. Boston SE, Moens NMM, Kruth SA, et al. Endoscopic evalua-
28. Soma LR, Uboh CE, Maylin GM. The use of phenylbutazone in tion of the gastroduodenal mucosa to determine the safety of
the horse. J Vet Pharmacol Ther. 2012;35:1. short-term concurrent administration of meloxicam and dexa-
29. Hu HH, MacAllister CG, Payton ME, et  al. Evaluation of the methasone in healthy dogs. Am J Vet Res. 2003;64:1369.
analgesic effects of phenylbutazone administered at a high or 51. Kivett L, Taintor J, Wright J. Evaluation of the safety of a combi-
low dosage in horses with chronic lameness. J Am Vet Med As- nation of oral administration of phenylbutazone and firocoxib
soc. 2005;226:414. in horses. J Vet Pharmacol Ther. 2014;37:413.
30. Soma LR, Uboh CE, Liu Y, et  al. Pharmacokinetics of 52. Reed SK, Messer NT, Tessman RK, et  al. Effects of phenyl­
dexamethasone following intra-articular, intravenous, in- butazone alone or in combination with flunixin meglumine
tramuscular, and oral administration in horses and its ef- on blood protein concentrations in horses. Am J Vet Res.
fects on endogenous hydrocortisone. J Vet Pharmacol Ther. 2006;67:398.
2013;36:181. 53. Doucet MY, Bertone AL, Hendrickson D, et al. Comparison of
31. de Grauw JC, Visser-Meijer MC, Lashley F, et al. Intra-articular efficacy and safety of paste formulations of firocoxib and phe-
treatment with triamcinolone compared with triamcinolone nylbutazone in horses with naturally occurring osteoarthritis. J
with hyaluronate: a randomised open-label multicentre clinical Am Vet Med Assoc. 2008;232:91.
trial in 80 lame horses. Equine Vet J. 2016;48:152. 54. Orsini JA, Ryan WG, Carithers DS, et al. Evaluation of oral ad-
32. Levine DG, Epstein KL, Neelis DA, et al. Effect of topical ap- ministration of firocoxib for the management of musculoskel-
plication of 1% diclofenac sodium liposomal cream on inflam- etal pain and lameness associated with osteoarthritis in horses.
mation in healthy horses undergoing intravenous regional limb Am J Vet Res. 2012;73:664.
perfusion with amikacin sulfate. Am J Vet Res. 2009;70:1323. 55. D’Arcy-Moskwa E, Noble G, Weston L, et al. Effects of meloxi-
33. Goodrich LR, Nixon AJ. Medical treatment of osteoarthritis in cam and phenylbutazone on equine gastric mucosal permeabil-
the horse–a review. Vet J. 2006;171:51. ity. J Vet Intern Med. 2012;26:1494.
34. Lees P, May SA, McKellar QA. Pharmacology and therapeutics 56. Raidal SL, Edwards S, Pippia J, et al. Pharmacokinetics and safe-
of non-steroidal anti-inflammatory drugs in the dog and cat: 1. ty of oral administration of meloxicam to foals. J Vet Intern Med.
General pharmacology. J Small Anim Pract. 1991;32:183. 2013;27:300.
35. Brune K, Patrignani P. New insights into the use of currently 57. Holland B, Fogle C, Blikslager AT, et  al. Pharmacokinetics
available non-steroidal anti-inflammatory drugs. J Pain Res. and pharmacodynamics of three formulations of firocoxib in
2015;8:105. healthy horses. J Vet Pharmacol Ther. 2015;38:249.
36. Lees P, Landoni MF, Giraudel J, et  al. Pharmacodynamics 58. Combie J, Shults T, Nugent EC, et al. Pharmacology of narcotic
and pharmacokinetics of nonsteroidal anti-inflammatory analgesics in the horse: selective blockade of narcotic-induced
drugs in species of veterinary interest. J Vet Pharmacol Ther. locomotor activity. Am J Vet Res. 1981;42:716.
2004;27:479. 59. Combie J, Blake JW, Ramey BE, et al. Pharmacology of narcotic
37. DeMaria AN, Weir MR. Coxibs—beyond the GI tract: re- analgesics in the horse: quantitative detection of morphine in
nal and cardiovascular issues. J Pain Symptom Manage Suppl. equine blood and urine and logit-log transformations of this
2003;25:41. data. Am J Vet Res. 1981;42:1523.
38. Simmons DL, Botting RM, Hla T. Cyclooxygenase isozymes: 60. Combie J, Nugent TE, Tobin T. Pharmacokinetics and protein
the biology of prostaglandin synthesis and inhibition. Pharma- binding of morphine in horses. Am J Vet Res. 1983;44:870.
col Rev. 2004;56:387. 61. Kalpravidh M, Lumb WV, Wright M, et al. Effects of butorpha-
39. KuKanich B, Bidgood T, Knesl O. Clinical pharmacology of nol, flunixin, levorphanol, morphine, and xylazine in ponies.
nonsteroidal anti-inflammatory drugs in dogs. Vet Anaesth An- Am J Vet Res. 1984;45:217.
alg. 2012;39:69. 62. Muir WW, Robertson JT. Visceral analgesia: effects of xylazine,
40. Kis B, Snipes JA, Busija DW. Acetaminophen and the cyclooxy- butorphanol, meperidine, and pentazocine in horses. Am J Vet
genase-3 puzzle: sorting out facts, fictions, and uncertainties. J Res. 1985;46:2081.
Pharmacol Exp Ther. 2005;315:1. 63. Sellon DC, Roberts MC, Blikslager AT, et al. Effects of continu-
41. Botting R, Ayoub SS. COX-3 and the mechanism of action of ous rate intravenous infusion of butorphanol on physiologic
paracetamol/acetaminophen. Prostaglandins Leukot Essent Fat- and outcome variables in horses after celiotomy. J Vet Intern
ty Acids. 2005;72:85. Med. 2004;18:555.
42. Davies NM, Good RL, Roupe KA, et al. Cyclooxygenase-3: ax- 64. Robertson JT, Muir WW. A new analgesic drug combination in
iom, dogma, anomaly, enigma or splice error? Not as easy as 1, the horse. Am J Vet Res. 1983;44:1667.
2, 3. J Pharm Pharm Sci. 2004;7:217. 65. Muir WW, Skarda RT, Sheehan W. Hemodynamic and respira-
43. Radi ZA. Pathophysiology of cyclooxygenase inhibition in ani- tory effects of xylazine-morphine sulfate in horses. Am J Vet
mal models. Toxicol Pathol. 2009;37:34. Res. 1979;40:1417.
CHAPTER 3  Recognizing and Treating Pain in Horses 153

66. Mama KR, Pascoe PJ, Steffey EP. Evaluation of the interaction 88. Lindegaard C, Thomsen MH, Larsen S, et al. Analgesic efficacy
of mu and kappa opioid agonists on locomotor behavior in the of intra-articular morphine in experimentally induced radio-
horse. Can J Vet Res. 1993;57:106. carpal synovitis in horses. Vet Anaesth Analg. 2010;37:171.
67. Nugent TE, Combie JD, Weld JM, et al. Effects of enkephalins 89. van Loon JP, De Grauw JC, van Dierendonck M, et  al. Intra-
versus opiates on locomotor activity of the horse. Res Commun articular opioid analgesia is effective in reducing pain and in-
Chem Pathol Pharmacol. 1982;35:405. flammation in an equine LPS induced synovitis model. Equine
68. Hellyer PW, Bai L, Supon J, et al. Comparison of opioid and al- Vet J. 2010;42:412.
pha-2 adrenergic receptor binding in horse and dog brain using 90. Lindegaard C, Frost AB, Thomsen MH, et al. Pharmacokinetics
radioligand autoradiography. Vet Anaesth Analg. 2003;30:172. of intra-articular morphine in horses with lipopolysaccharide-
69. Andersen MS, Clark L, Dyson SJ, et al. Risk factors for colic in induced synovitis. Vet Anaesth Analg. 2010;37:186.
horses after general anaesthesia for MRI or nonabdominal sur- 91. Martin-Flores M, Campoy L, Kinsley MA, et al. Analgesic and
gery: absence of evidence of effect from perianaesthetic mor- gastrointestinal effects of epidural morphine in horses after
phine. Equine Vet J. 2006;38:368. laparoscopic cryptorchidectomy under general anesthesia. Vet
70. Taylor PM. Effect of postoperative pethidine on the anaesthetic Anaesth Analg. 2014;41:430.
recovery period in the horse. Equine Vet J. 1986;18:70. 92. Van Hoogmoed LM, Galuppo LD. Laparoscopic ovariectomy
71. Clark L, Clutton RE, Blissitt KJ, et al. The effects of morphine on using the endo-GIA stapling device and endo-catch pouches
the recovery of horses from halothane anaesthesia. Vet Anaesth and evaluation of analgesic efficacy of epidural morphine sul-
Analg. 2005;35:22. fate in 10 mares. Vet Surg. 2005;34:646.
72. Wetmore L, Pascoe P, Shilo-Benjamini Y, et al. Effects of fentanyl 93. Goodrich LR, Nixon AJ, Fubini SL, et  al. Epidural morphine
administration on locomotor response in horses with the G57C and detomidine decreases postoperative hindlimb lameness in
μ-opioid receptor polymorphism. Am J Vet Res. 2016;77:828. horses after bilateral stifle arthroscopy. Vet Surg. 2002;31:232.
73. Boscan P, Van Hoogmoed LM, Farver TB, et al. Evaluation of 94. England GCW, Clarke KW. Alpha2 adrenoceptor agonists in
the effects of the opioid agonist morphine on gastrointestinal the horse: a review. Br Vet J. 1996;152:641.
tract function in horses. Am J Vet Res. 2006;67:992. 95. Jochle W, Moore JN, Brown J, et al. Comparison of detomidine,
74. Sojka JE, Adams SB, Lamar CH, et  al. Effect of butorphanol, butorphanol, flunixin meglumine and xylazine in clinical cases
pentazocine, meperidine, or metoclopramide on intestinal mo- of equine colic. Equine Vet J Suppl. 1989;7:111.
tility in female ponies. Am J Vet Res. 1988;49(4):527. 96. Lowe JE, Hilfiger J. Analgesic and sedative effects of detomidine
75. Senior JM, Pinchbeck GL, Dugdale AHA, et  al. Retrospective compared to xylazine in a colic model using IV and IM routes
study of the risk factors and prevalence of colic in horses after of administration. Acta Vet Scand. 1986;82:85.
orthopaedic surgery. Vet Rec. 2004;155:321. 97. Lowe JE, Hilfiger J. Analgesic and sedative effects of detomidine
76. Nelson BB, Lordan EE, Hassel DM. Risk factors associated in a colic model: blind studies on efficacy and duration of ef-
with gastrointestinal dysfunction in horses undergoing elec- fects. Proc Annu Conv Am Equin. 1984;30:225.
tive procedures under general anaesthesia. Equine Vet J Suppl. 98. Muir WW, Skarda RT, Sheehan W. Hemodynamic and respira-
2013;45:8. tory effects of a xylazine-acetylpromazine drug combination in
77. Mircica E, Clutton RE, Kyles KW, et  al. Problems associated horses. Am J Vet Res. 1979;40:1518.
with perioperative morphine in horses: a retrospective case 99. Wagner AE, Muir WW, Hinchcliff KW. Cardiovascular effects
analysis. Vet Anaesth Analg. 2003;30:147. of xylazine and detomidine in horses. Am J Vet Res. 1991;52:651.
78. Koenig J, Cote N. Equine gastrointestinal motility—ileus and 100. Still J, Serteyn D, Van der Merwe CA. Cardiovascular and respir-
pharmacological modification. Can Vet J. 2006;47:551. atory effects of detomidine in isoflurane-anaesthetised horses.
79. Sano H, Martin-Flores M, Santos LCP, et al. Effects of epidural J S Afr Vet Assoc. 1996;67:199.
morphine on gastrointestinal transit in unmedicated horses. Vet 101. Bettschart-Wolfensberger R, Freeman SL, Bowen I, et al. Car-
Anaesth Analg. 2011;38:121. diopulmonary effects and pharmacokinetics of i.v. dexmedeto-
80. Sanchez L, Robertson S, Maxwell L, et al. Effect of fentanyl on midine in ponies. Equine Vet J. 2005;37:60.
visceral and somatic nociception in conscious horses. J Vet In- 102. Kamerling SG, Cravens WMT, Bagwell CA. Dose-related ef-
tern Med. 2007;21:1067. fects of detomidine on autonomic responses in the horse. J Au-
81. Ohta M, Wakuno A, Okada J, et al. Effects of intravenous fen- ton Pharmacol. 1988;8:241.
tanyl administration on end-tidal sevoflurane concentrations in 103. Pypendop BH, Verstegen JP. Hemodynamic effects of medeto-
thoroughbred racehorses undergoing orthopedic surgery. J Vet midine in the dog: a dose titration study. Vet Surg. 1998;27:612.
Med Sci. 2010;72:1107. 104. Lester GD, Merritt AM, Neuwirty L, et  al. Effect of α2-
82. Knych HK, Steffey EP, Casbeer HC, et al. Disposition, behav- adrenergic, cholinergic, and nonsteroidal anti-inflammatory
ioural and physiological effects of escalating doses of intra- drugs on myoelectric activity of ileum, cecum, and right ventral
venously administered fentanyl to young foals. Equine Vet J. colon and on cecal emptying of radiolabeled markers in clini-
2015;47:592. cally normal ponies. Am J Vet Res. 1998;58:320.
83. Messenger KM, Davis JL, LaFevers DH, et al. Intravenous and 105. Merritt AM, Burrows JA, Hartless CS. Effect of xylazine, deto-
sublingual buprenorphine in horses: pharmacokinetics and in- midine, and a combination of xylazine and butorphanol on
fluence of sampling site. Vet Anaesth Analg. 2011;38:374. equine duodenal motility. Am J Vet Res. 1998;59:619.
84. Davis JL, Messenger KM, LaFevers DH, et al. Pharmacokinetics 106. Sutton DGM, Preston T, Christley RM, et al. The effects of xy-
of intravenous and intramuscular buprenorphine in the horse. J lazine, detomidine, acepromazine and butorphanol on equine
Vet Pharmacol Ther. 2012;35:52. solid phase gastric emptying rate. Equine Vet J. 2002;34:486.
85. Potter JJ, MacFarlane PD, Love EJ, et  al. Preliminary investi- 107. Nuñez E, Steffey EP, Ocampo L, et al. Effects of alpha2-adrener-
gation comparing a detomidine continuous rate infusion com- gic receptor agonists on urine production in horses deprived of
bined with either morphine or buprenorphine for standing food and water. Am J Vet Res. 2004;65:1342.
sedation in horses. Vet Anaesth Analg. 2016;43:189. 108. Valdes-Vazquez MA, Aguilera-Tejero E, Mayer-Valor R. Effect
86. Risberg ÅI, Spadavecchia C, Ranheim B, et al. Antinociceptive of xylazine during endoscopic evaluation of functional upper
effect of buprenorphine and evaluation of the nociceptive with- respiratory disorders in horses. J Equine Vet Sci. 1993;13:84.
drawal reflex in foals. Vet Anaesth Analg. 2015;42:329. 109. Ducharme NG, Hackett RP, Fubini SL, et al. The reliability of
87. Santos LCP, De Moraes AN, Saito ME. Effects of intraarticu- endoscopic examination in assessment of arytenoid cartilage
lar ropivacaine and morphine on lipopolysaccharide-induced movement in horses. Part II. Influence of side of examination,
synovitis in horses. Vet Anaesth Analg. 2009;36:280. reexamination, and sedation. Vet Surg. 1991;20:180.
154 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

110. Ringer SK, Schwarzwald CC, Portier KG, et  al. Effects on 130. Valverde A, Gunkel C, Doherty TJ, et al. Effect of a constant rate
cardiopulmonary function and oxygen delivery of doses of infusion of lidocaine on the quality of recovery from sevoflu-
romifidine and xylazine followed by constant rate infusions in rane or isoflurane general anaesthesia in horses. Equine Vet J.
standing horses. Vet J. 2013;195:228. 2005;37:559.
111. Grubb TL, Muir WW, Bertone AL, et al. Use of yohimbine to re- 131. Schuhbeck MM, Kuhn M, Spadavecchia C, et  al. Continuous
verse prolonged effects of xylazine hydrochloride in a horse being intravenous lidocaine infusion during isoflurane anaesthesia
treated with chloramphenicol. J Am Vet Med Assoc. 1997;210:1771. in horses undergoing surgical procedures. Pferdeheilkunde.
112. Di Salvo A, Della Rocca G, Bazzica C, et al. A pharmacokinetic/ 2012;28:252.
clinical approach to postulate a local action of intra-articular 132. Meyer GA, Lin HC, Hanson RR, et  al. Effects of intravenous
xylazine administration in the horse: a preliminary study. J Vet lidocaine overdose on cardiac electrical activity and blood pres-
Pharmacol Ther. 2014;37:464. sure in the horse. Equine Vet J. 2001;33:431.
113. Ullmer J, Mama KR, Lee C, et al. The effects of xylazine on nor- 133. Olbrich VH, Mosing M. A comparison of the analgesic effects
mal and interleukin-1 conditioned equine articular cartilage ex- of caudal epidural methadone and lidocaine in the horse. Vet
plants. CA: Proc 38th Am College Vet Anesth San Diego; 2013. Anaesth Analg. 2003;30:156.
114. Mama KR, Ullmer J, King M, et  al. The effects of xylazine, 134. Rapley JH, Beavis RC, Barber FA, et  al. Glenohumeral chon-
dexmedetomidine and clonidine on normal and interleukin-1 drolysis after shoulder arthroscopy associated with continuous
conditioned equine cartilage explants. Japan: Proc 12th World bupivacaine infusion. Arthroscopy. 2009;25:1367.
Congress of Vet Anaesth Kyoto; 2015. 135. Park J, Sutradhar BC, Hong G, et al. Comparison of the cytotox-
115. Abdallah FW, Brull R. Facilitatory effects of perineural dexme- ic effects of bupivacaine, lidocaine, and mepivacaine in equine
detomidine on neuraxial and peripheral nerve block: a system- articular chondrocytes. Vet Anaesth Analg. 2011;38:127.
atic review and meta-analysis. Br J Anaesth. 2013;110(915):2013. 136. Breu A, Rosenmeier K, Kujat R, et al. The cytotoxicity of bupiv-
116. Skarda RT, Muir WW. Comparison of antinociceptive, cardiovas- acaine, ropivacaine, and mepivacaine on human chondrocytes
cular, and respiratory effects, head ptosis, and position of pelvic and cartilage. Anesth Analg. 2013;117:514.
limbs in mares after caudal epidural administration of xylazine and 137. Piat P, Richard H, Beauchamp G, et al. In vivo effects of a single
detomidine hydrochloride solution. Am J Vet Res. 1996;57:1338. intra-articular injection of 2% lidocaine or 0.5% bupivacaine on
117. Skarda RT, Muir WW. Caudal analgesia induced by epidural articular cartilage of normal horses. Vet Surg. 2012;41:1002.
or subarachnoid administration of detomidine hydrochloride 138. Mion G, Villevieille T. Ketamine pharmacology: an update
solution in mares. Am J Vet Res. 1994;55:670. (pharmacodynamics and molecular aspects, recent findings).
118. Hubbell JAE, Muir WW. Antagonism of detomidine sedation CNS Neurosci Ther. 2013;19:370.
in the horse using intravenous tolazoline or atipamezole. Equine 139. De Kock MF, Lavand’homme PM. The clinical role of NMDA
Vet J. 2006;38:238. receptor antagonists for the treatment of postoperative pain.
119. Carroll GL, Matthews NS, Hartsfield SM, et  al. The effect of Best Pract Res Clin Anaesthesiol. 2007;21:85.
detomidine and its antagonism with tolazoline on stress-related 140. Muir WW, Skarda RT, Milne DW. Evaluation of xylazine and
hormones, metabolites, physiologic responses, and behavior in ketamine hydrochloride for anesthesia in horses. Am J Vet Res.
awake ponies. Vet Surg. 1997;26:69. 1977;38:195.
120. Pakkanen SAE, Raekallio MR, Mykkänen AK, et al. Detomidine 141. Muir WW, Sams R. Effects of ketamine infusion on halo-
and the combination of detomidine and MK-467, a peripheral thane minimal alveolar concentration in horses. Am J Vet Res.
alpha-2 adrenoceptor antagonist, as premedication in horses 1802;53:1992.
anaesthetized with isoflurane. Vet Anaesth Analg. 2015;42:527. 142. Lin HC, Passler T, Wilborn RR, et al. A review of the general
121. Vainionpää MH, Raekallio MR, Pakkanen SA, et  al. Plasma pharmacology of ketamine and its clinical use for injectable an-
drug concentrations and clinical effects of a peripheral alpha- aesthesia in horses. Equine Vet Educ. 2015;27:146.
2-adrenoceptor antagonist, MK-467, in horses sedated with 143. Takki S, Nikki P, Jäättelä A, et al. Ketamine and plasma catecho-
detomidine. Vet Anaesth Analg. 2013;40:257. lamines. Br J Anaesth. 1972;44:1318.
122. Doherty TJ, Frazier DL. Effect of intravenous lidocaine on halo- 144. López-Sanromán FJ, Cruz JM, Santos M, et  al. Evaluation of
thane minimum alveolar concentration in ponies. Equine Vet J. the local analgesic effect of ketamine in the palmar digital nerve
1998;30:300. block at the base of the proximal sesamoid (abaxial sesamoid
123. Robertson SA, Sanchez LC, Merritt AM, et al. Effect of systemic block) in horses. Am J Vet Res. 2003;64:475.
lidocaine on visceral and somatic nociception in conscious 145. Gómez de Segura IA, De Rossi R, Santos M, et al. Epidural in-
horses. Equine Vet J. 2005;37:122. jection of ketamine for perineal analgesia in the horse. Vet Surg.
124. Harkins JD, Mundy GD, Woods WE, et  al. Lidocaine in the 1998;27:384.
horse: its pharmacological effects and their relationship to ana- 146. Braun S, Gaza N, Werdehausen R, et al. Ketamine induces apop-
lytical findings. J Vet Pharmacol Ther. 1998;21:462. tosis via the mitochondrial pathway in human lymphocytes and
125. Harkins JD, Stanley S, Mundy GD, et al. A review of the phar- neuronal cells. Br J Anaesth. 2010;105:347.
macology, pharmacokinetics, and regulatory control in the US of 147. Taylor PM, Bennett RC, Brearley JC, et al. Comparison of deto-
local anaesthetics in the horse. J Vet Pharmacol Ther. 1995;18:397. midine and romifidine as premedicants before ketamine and
126. Brianceau P, Chevalier H, Karas A, et al. Intravenous lidocaine halothane anesthesia in horses undergoing elective surgery. Am
and small-intestinal size, abdominal fluid, and outcome after J Vet Res. 2001;62:359.
colic surgery in horses. J Vet Intern Med. 2002;16:736. 148. Wagner AE, Walton JA, Hellyer P, et  al. Use of low doses of
127. Guschlbauer M, Feige K, Geburek F, et al. Effects of in vivo li- ketamine administered by constant rate infusion as an ad-
docaine administration at the time of ischemia and reperfusion junct for postoperative analgesia in dogs. J Am Vet Med Assoc.
on in vitro contractility of equine jejunal smooth muscle. Am J 2002;221:72.
Vet Res. 2011;72:1449. 149. Carstensen M, Møller AM. Adding ketamine to morphine for
128. Rezende ML, Wagner AE, Mama KR, et al. Effects of intrave- intravenous patient-controlled analgesia for acute postopera-
nous administration of lidocaine on the minimum alveolar con- tive pain: a qualitative review of randomized trials. Br J Anaesth.
centration of sevoflurane in horses. Am J Vet Res. 2011;72:446. 2010;104:401.
129. Wagner AE, Mama KR, Steffey EP, et al. Comparison of the car- 150. Fielding CL, Brumbaugh GW, Matthews NS, et  al. Pharma-
diovascular effects of equipotent anesthetic doses of sevoflurane cokinetics and clinical effects of a subanesthetic continu-
alone and sevoflurane plus an intravenous infusion of lidocaine ous rate infusion of ketamine in awake horses. Am J Vet Res.
in horses. Am J Vet Res. 2011;72:452. 2006;67:1484.
CHAPTER 3  Recognizing and Treating Pain in Horses 155

151. Guedes AGP, Matthews NS, Hood DM. Effect of ketamine hy- 172. Carregaro AB, Freitas GC, Ribeiro MH, et  al. Physiological
drochloride on the analgesic effects of tramadol hydrochloride and analgesic effects of continuous-rate infusion of morphine,
in horses with signs of chronic laminitis-associated pain. Am J butorphanol, tramadol or methadone in horses with lipopol-
Vet Res. 2012;73:610. ysaccharide (LPS)-induced carpal synovitis. BMC Vet Res.
152. Peterbauer C, Larenza PM, Knobloch M, et  al. Effects of a 2014;10:1.
low dose infusion of racemic and S-ketamine on the nocicep- 173. Milaré AS, De Oliveira FA, Luna SPL, et al. Intravenous trama-
tive withdrawal reflex in standing ponies. Vet Anaesth Analg. dol injection has no antinociceptive effect in horses undergoing
2008;35:414. electrical and thermal stimuli. J Equine Vet Sci. 2013;33:823.
153. Wagner AE, Mama KR, Contino EK, et al. Evaluation of seda- 174. Guedes A, Knych H, Hood D. Plasma concentrations, anal-
tion and analgesia in standing horses after administration of xy- gesic and physiological assessments in horses with chronic
lazine, butorphanol, and subanesthetic doses of ketamine. J Am laminitis treated with two doses of oral tramadol. Equine Vet J.
Vet Med Assoc. 2011;238:1629. 2016;48:528.
154. Elfenbein JR, Robertson SA, Corser AA, et al. Systemic effects of 175. Cagnardi P, Ferraresi C, Zonca A, et al. Clinical pharmacoki-
a prolonged continuous infusion of ketamine in healthy horses. netics of tramadol and main metabolites in horses undergoing
J Vet Intern Med. 2011;25:1134. orchiectomy. Vet Q. 2014;34:143.
155. Tramer MR, Schneider J, Marti RA, et al. Role of magnesium 176. Guedes A, Galuppo L, Hood D, et al. Soluble epoxide hydrolase
sulfate in postoperative analgesia. Anesthesiology. 1997;84:340. activity and pharmacologic inhibition in horses with chronic
156. Stomatology FM, Yan Q. Effects of systemic magnesium on severe laminitis. Equine Vet J. 2016.
post-operative analgesia: is the current evidence strong enough? 177. Guedes AG, Morrisseau C, Sole A, et al. Use of a soluble epoxide
Pain Physician. 2015;18:405. hydrolase inhibitor as an adjunctive analgesic in a horse with
157. Albrecht E, Kirkham KR, Liu SS, et al. Peri-operative intrave- laminitis. Vet Anaesth Analg. 2013;40:440.
nous administration of magnesium sulphate and postoperative 178. Love EJ, Taylor PM, Murrell J, et  al. Effects of acepromazine,
pain: a meta-analysis. Anaesthesia. 2013;68:79. butorphanol and buprenorphine on thermal and mechanical
158. McCarthy RJ, Kroin JS, Tuman KJ, et  al. Antinociceptive po- nociceptive thresholds in horses. Equine Vet J. 2012;44:221.
tentiation and attenuation of tolerance by intrathecal co-infu- 179. Sanchez LC, Elfenbein JR, Robertson SA. Effect of aceproma-
sion of magnesium sulfate and morphine in rats. Anesth Analg. zine, butorphanol, or N-butylscopolammonium bromide on
1998;86(830):1998. visceral and somatic nociception and duodenal motility in con-
159. Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy scious horses. Am J Vet Res. 2008;69:579.
for neuropathic pain in adults: a systematic review and meta- 180. Roelvink ME, Goossens L, Kalsbeek HC, et  al. Analgesic and
analysis. Lancet Neurol. 2015;14:162. spasmolytic effects of dipyrone, hyoscine-N-butylbromide and
160. Terry RL, McDonnell SM, Van Eps AW, et al. Pharmacokinetic a combination of the two in ponies. Vet Rec. 1991;129:378.
profile and behavioral effects of gabapentin in the horse. J Vet 181. Boatwright CE, Fubini SL, Grohn YT, et al. A comparison of N-
Pharmacol Ther. 2010;33:485. butylscopolammonium bromide and butorphanol tartrate for
161. Caldwell FJ, Taintor J, Waguespack RW, et al. Effect of PO ad- analgesia using a balloon model of abdominal pain in ponies.
ministered gabapentin on chronic lameness in horses. J Equine Can J Vet Res. 1996;60:65.
Vet Sci. 2015;35:536. 182. Rumpler MJ, Colahan P, Sams RA. The pharmacokinetics of
162. Davis JL, Posner LP, Elce Y. Gabapentin for the treatment of methocarbamol and guaifenesin after single intravenous and
neuropathic pain in a pregnant horse. J Am Vet Med Assoc. multiple-dose oral administration of methocarbamol in the
2007;231:755. horse. J Vet Pharmacol Ther. 2014;37:25.
163. Dworkin RH, O’Connor AB, Backonja M, et al. Pharmacologic 183. Cremers S, Papapoulos S. Pharmacology of bisphosphonates.
management of neuropathic pain: evidence-based recommen- Bone. 2011;49:42.
dations. Pain. 2007;132:237. 184. Catterall JB, Cawston TE. Drugs in development: bisphos-
164. Mullen KR, Schwark W, Divers TJ. Pharmacokinetics of single- phonates and metalloproteinase inhibitors. Arthritis Res Ther.
dose intragastric and intravenous pregabalin administration in 2003;5:12.
clinically normal horses. Am J Vet Res. 2013;74:1043. 185. Kamm L, McIlwraith W, Kawcak C. A review of the efficacy of
165. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin tiludronate in the horse. J Equine Vet Sci. 2008;28:209.
Pharmacokinet. 2004;43:879. 186. Gough MR, Thibaud D, Smith RKW. Tiludronate infusion in
166. Vettorato E, Zonca A, Cagnardi P, et al. Pharmacokinetics and the treatment of bone spavin: a double blind placebo-controlled
efficacy of intravenous and extradural tramadol in dogs. Vet J. trial. Equine Vet J. 2010;42:381.
2010;183:310. 187. Coudry V, Thibaud D, Riccio B, et al. Efficacy of tiludronate in
167. Knych HK, Corado CR, McKemie DS, et  al. Pharmacokinet- the treatment of horses with signs of pain associated with osteo-
ics and selected pharmacodynamic effects of tramadol fol- arthritic lesions of the thoracolumbar vertebral column. Am J
lowing intravenous administration to the horse. Equine Vet J. Vet Res. 2007;68:329.
2013;45:490. 188. Hunter BG, Duesterdieck-Zellmer KF, Larson MK. Tiludronate
168. Stewart AJ, Boothe DM, Cruz-Espindola C, et  al. Pharmaco­ concentrations and cytologic findings in synovial fluid after in-
kinetics of tramadol and metabolites O-desmethyltramadol travenous regional limb perfusion with tiludronate in horses.
and N-desmethyltramadol in adult horses. Am J Vet Res. Peer J. 2015;3:e889.
2011;72:967. 189. Body JJ, Pfister T, Bauss F, et al. Preclinical perspectives on bis-
169. Shilo Y, Britzi M, Eytan B, et al. Pharmacokinetics of tramadol phosphonate renal safety. Oncologist. 2005;10(suppl 1):3.
in horses after intravenous, intramuscular and oral administra- 190. Meijer H, Reinecke J, Becker C, et al. The production of anti-
tion. J Vet Pharmacol Ther. 2008;31:60. inflammatory cytokines in whole blood by physico-chemical
170. Giorgi M, Soldani G, Manera C, et  al. Pharmacokinetics of induction. Inflamm Res. 2003;52:404.
tramadol and its metabolites M1, M2 and M5 in horses fol- 191. Frisbie DD, Kawcak CE, Werpy NM, et al. Clinical, biochemi-
lowing intravenous, immediate release (fasted/fed) and sus- cal, and histologic effects of intra-articular administration of
tained release single dose administration. J Equine Vet Sci. autologous conditioned serum in horses with experimentally
2007;27:481. induced osteoarthritis. Am J Vet Res. 2007;68:290.
171. Dhanjal JK, Wilson DV, Robinson E, et al. Intravenous trama- 192. Martin CA, Kerr CL, Pearce SG, et  al. Outcome of epidural
dol: effects, nociceptive properties, and pharmacokinetics in catheterization for delivery of analgesics in horses: 43 cases
horses. Vet Anaesth Analg. 2009;36:581. (1998–2001). J Am Vet Med Assoc. 2003;222:1394.
156 PART 1  MECHANISMS OF DISEASE AND PRINCIPLES OF TREATMENT

193. Sysel AM, Pleasant RS, Jacobson JD, et  al. Systemic and lo- 214. Waguespack RW, Burba DJ, Hubert JD, et al. Effects of extra-
cal effects associated with long-term epidural catheterization corporeal shock wave therapy on desmitis of the accessory liga-
and morphine-detomidine administration in horses. Vet Surg. ment of the deep digital flexor tendon in the horse. Vet Surg.
1997;26:141. 2011;40:450.
194. Zarucco L, Driessen B, Scandella M, et al. Continuous perineu- 215. Imboden I, Waldern NM, Wiestner T, et al. Short term analge-
ral block of the palmar nerves: a new technique for pain relief in sic effect of extracorporeal shock wave therapy in horses with
the distal equine forelimb. Clin Tech Equine Pract. 2007;6:154. proximal palmar metacarpal/plantar metatarsal pain. Vet J.
195. Chelly JE, Greger J, Casati A, et  al. Continuous lateral sciatic 2009;179:50.
blocks for acute postoperative pain management after major 216. McClure SR, Sonea IM, Evans RB, et al. Evaluation of analgesia
ankle and foot surgery. Foot Ankle. 2002;23:749. resulting from extracorporeal shock wave therapy and radial
196. Ganesh A, Rose JB, Wells T, et al. Continuous peripheral nerve pressure wave therapy in the limbs of horses and sheep. Am J
blockade for inpatient and outpatient postoperative analgesia in Vet Res. 2005;66:1702.
children. Anesth Analg. 2007;105:1234. 217. Dahlberg JA, McClure SR, Evans RB, et  al. Force platform
197. Boezaart AP. Perineural infusion of local anesthetics. Anesthesi- evaluation of lameness severity following extracorporeal shock
ology. 2006;104:872. wave therapy in horses with unilateral forelimb lameness. J Am
198. Driessen B, Scandella M, Zarucco L. Development of a tech- Vet Med Assoc. 2006;229:100.
nique for continuous perineural blockade of the palmar 218. Brown KE, Nickels FA, Caron JP, et  al. Investigation of the
nerves in the distal equine thoracic limb. Vet Anaesth Analg. immediate analgesic effects of extracorporeal shock wave
2008;35:432. therapy for treatment of navicular disease in horses. Vet Surg.
199. Frisbie DD, McIlwraith CW, Kawcak CE, et  al. Evaluation of 2005;34:554.
topically administered diclofenac liposomal cream for treat- 219. Gomez Alvarez CBG, L’ami JJ, Moffat D, et al. Effect of chiro-
ment of horses with experimentally induced osteoarthritis. Am practic manipulations on the kinematics of back and limbs in
J Vet Res. 2009;70:210. horses with clinically diagnosed back problems. Equine Vet J.
200. Andreeta A, Verde C, Babusci M, et  al. Comparison of di- 2008;40:153.
clofenac diethylamine permeation across horse skin from five 220. Landman M, De Blaauw JA, Van Weeren PR, et al. Field study
commercial medical human formulations. J Equine Vet Sci. of the prevalence of lameness in horses with back problems. Vet
2011;31:502. Rec. 2004;155:165.
201. Thomasy SM, Slovis N, Maxwell LK, et al. Transdermal fentanyl 221. Langstone J, Ellis J, Cunliffe C. A preliminary study of the ef-
combined with nonsteroidal anti-inflammatory drugs for anal- fect of manual chiropractic treatment on the splenius muscle in
gesia in horses. J Vet Intern Med. 2004;18:550. horses when measured by surface electromyography. Equine Vet
202. Eberspächer E, Stanley SD, Rezende M, et al. Pharmacokinetics J. 2015;47(18):2015.
and tolerance of transdermal fentanyl administration in foals. 222. Sullivan KA, Hill AE, Haussler KK. The effects of chiroprac-
Vet Anaesth Analg. 2008;35:249. tic, massage and phenylbutazone on spinal mechanical noci­
203. Orsini JA, Moate PJ, Kuersten K, et al. Pharmacokinetics of fen- ceptive thresholds in horses without clinical signs. Equine Vet J.
tanyl delivered transdermally in healthy adult horses–variabil- 2008;40:14.
ity among horses and its clinical implications. J Vet Pharmacol 223. Klide AM. Treatment of chronic back pain in horses stimulation
Ther. 2006;29:539. of acupuncture points with a low powered infrared laser. Vet
204. Mills PC, Cross SE. Regional differences in transdermal pene- Surg. 1987;16:106.
tration of fentanyl through equine skin. Res Vet Sci. 2007;82:252. 224. Hagiwara S, Iwasaka H, Okuda K, et al. GaAlAs (830 nm) low-
205. Hubbell JAE, Saville WJA, Bednarski RM, et al. The use of seda- level laser enhances peripheral endogenous opioid analgesia in
tives, analgesic and anaesthetic drugs in the horse: an electronic rats. Lasers Surg Med. 2007;39:797.
survey of members of the American Association of Equine 225. Ginani F, Soares DM, Barboza CAG. Effect of low-level laser
Practitioners (AAEP). Equine Vet J. 2010;42:487. therapy on mesenchymal stem cell proliferation: a systematic
206. Vincent CA, Richardson PH. The evaluation of therapeutic acu- review. Lasers Med Sci. 2015;30:2189.
puncture: concepts and methods. Pain. 1986;24:1. 226. Smoot B, Chiavola-Larson L, Lee J, et al. Effect of low-level la-
207. Skarda RT, Tejwani GA, Muir WW. Cutaneous analgesia, ser therapy on pain and swelling in women with breast cancer-
hemodynamic and respiratory effects, and beta-endorphin con- related lymphedema: a systematic review and meta-analysis. J
centration in spinal fluid and plasma of horses after acupunc- Cancer Surviv. 2014;9:287.
ture and electroacupuncture. Am J Vet Res. 2002;63:1435. 227. Bjordal JM, Johnson MI, Iversen V, et al. Low-level laser ther-
208. Takeshige C, Oka K, Mizuno T, et  al. The acupuncture point apy in acute pain: a systematic review of possible mechanisms
and its connecting central pathway for producing acupuncture of action and clinical effects in randomized placebo-controlled
analgesia. Brain Res Bull. 1993;30:53. trials. Photomed Laser Surg. 2006;24:158.
209. Tobaldini G, Aisengart B, Lima MMS, et al. Ascending nocicep- 228. ColdLasers.org: A guide to equine lasers. http://www.coldlasers
tive control contributes to the antinociceptive effect of acupunc- .org/guide-to-equine-lasers; 2016.
ture in a rat model of acute pain. J Pain. 2014;15:422. 229. U.S. Food and Drug Administration. Laser products and instru-
210. Bossut DFB, Leshin LS, Stromberg MW, et al. Plasma cortisol ments. http://www.fda.gov/radiation-emittingproducts/radiati
and beta-endorphin in horses subjected to electro-acupuncture onemittingproductsandprocedures/homebusinessandentertain
for cutaneous analgesia. Peptides. 1983;4:501. ment/laserproductsandinstruments/default.htm; 2016.
211. Skarda RT, Muir WW. Comparison of electroacupuncture and 230. Lee CD, Maxwell LK. Effect of body weight on the pharmacoki-
butorphanol on respiratory and cardiovascular effects and rec- netics of flunixin meglumine in miniature horses and quarter
tal pain threshold after controlled rectal distention in mares. horses. J Vet Pharmacol Ther. 2014;37:35.
Am J Vet Res. 2003;64:137. 231. Landoni MF, Lees P. Pharmacokinetics and pharmacodynam-
212. Steiss JE, White NA, Bowen JM. Electroacupuncture in the ics of ketoprofen enantiomers in the horse. J Vet Pharmacol
treatment of chronic lameness in horses and ponies: a con- Ther. 1996;19:466.
trolled clinical trial. Can J Vet Res. 1989;53:239. 232. Spadavecchia C, Arendt-Nielsen L, Spadavecchia L, et  al. Ef-
213. Robinson KA, Manning ST. Efficacy of a single-formula acu- fects of butorphanol on the withdrawal reflex using threshold,
puncture treatment for horses with palmar heel pain. Can Vet J. suprathreshold and repeated subthreshold electrical stimuli in
2015;56:1257. conscious horses. Vet Anaesth Analg. 2007;34:48.
CHAPTER 3  Recognizing and Treating Pain in Horses 157

233. Dias BP, Araújo MA, Deschk M, et al. Effects of a continuous 253. Neto PIN, Luna SP, Queiroz-Williams P, et al. Cardiorespiratory
rate infusion of butorphanol in isoflurane-anesthetized horses and antinociceptive effects of two different doses of lidocaine
on cardiorespiratory parameters, recovery quality, gastrointes- administered to horses during a constant intravenous infusion
tinal motility and serum cortisol concentrations. Acta Cir Bras. of xylazine and ketamine. BMC Vet Res. 2013;9:1.
2014;29:801. 254. Moens Y, Lanz F, Doherr MG, et  al. A comparison of the
234. Chiavaccini L, Claude AK, Lee JH, et al. Pharmacokinetics and antinociceptive effects of xylazine, detomidine and romi-
pharmacodynamics comparison between subcutaneous and in- fidine on experimental pain in horses. Vet Anaesth Analg.
travenous butorphanol administration in horses. J Vet Pharma- 2003;30:183.
col Ther. 2015;38:365. 255. LeBlanc PH, Caron JP, Patterson JS, et al. Epidural injection of
235. Marly C, Bettschart-Wolfensberger R, Nussbaumer P, et  al. xylazine for perineal analgesia in horses. J Am Vet Med Assoc.
Evaluation of a romifidine constant rate infusion protocol with 1988;193:1405.
or without butorphanol for dentistry and ophthalmologic pro- 256. Wilson DV, Bohart GV, Evans AT, et al. Retrospective analy-
cedures in standing horses. Vet Anaesth Analg. 2014;41:491. sis of detomidine infusion for standing chemical restraint in 51
236. Orsini JA. Equine emergencies: treatment and procedures. 4th ed. horses. Vet Anaesth Analg. 2002;29:54.
St. Louis: Elsevier/Saunders; 2014. 257. da Silva Serpa PB, Natalini CC, Cavalcanti RL, et al. Effects of
237. Cruz FSF, Carregaro AB, Machado M, et al. Sedative and car- detomidine constant rate infusion on blood glucose and lactate
diopulmonary effects of buprenorphine and xylazine in horses. in sevoflurane anesthetized horses. Acta Sci Vet. 2012;40:1051.
Can J Vet Res. 2011;75:35. 258. Serpa PB, Garbade P, Natalini CC, et al. Energy imbalance and
238. Taylor P, Coumbe K, Henson F, et al. Evaluation of sedation for physiological changes during detomidine hydrochloride constant
standing clinical procedures in horses using detomidine com- rate infusion in standing horses. J Equine Vet Sci. 2015;35:232.
bined with buprenorphine. Vet Anaesth Analg. 2014;41:14. 259. Ringer SK, Portier K, Torgerson PR, et al. The effects of a loading
239. Fischer BL, Ludders JW, Asakawa M, et  al. A comparison of dose followed by constant rate infusion of xylazine compared
epidural buprenorphine plus detomidine with morphine plus with romifidine on sedation, ataxia and response to stimuli in
detomidine in horses undergoing bilateral stifle arthroscopy. horses. Vet Anaesth Analg. 2013;40:157.
Vet Anaesth Analg. 2009;36:67. 260. Devisscher L, Schauvliege S, Dewulf J, et  al. Romifidine as a
240. Benmansour P, Husulak ML, Bracamonte JL, et al. Cardiopul- constant rate infusion in isoflurane anaesthetized horses: a clin-
monary effects of an infusion of remifentanil or morphine in ical study. Vet Anaesth Analg. 2010;37:425.
horses anesthetized with isoflurane and dexmedetomidine. Vet 261. Rezende ML, Grimsrud KN, Stanley SD, et al. Pharmacokinet-
Anaesth Analg. 2014;41:346. ics and pharmacodynamics of intravenous dexmedetomidine in
241. Gozalo-Marcilla M, Steblaj B, Schauvliege S, et al. Comparison the horse. J Vet Pharmacol Ther. 2015;38:15.
of the influence of two different constant-rate infusions (dex- 262. Marcilla MG, Schauvliege S, Segaert S, et al. Influence of a con-
medetomidine versus morphine) on anaesthetic requirements, stant rate infusion of dexmedetomidine on cardiopulmonary
cardiopulmonary function and recovery quality in isoflurane function and recovery quality in isoflurane anaesthetized horses.
anaesthetized horses. Res Vet Sci. 2013;95:1186. Vet Anaesth Analg. 2012;39:49.
242. Thomasy SM, Steffey EP, Mama KR, et al. The effects of iv fen- 263. Creighton CM, Lemke KA, Lamont LA, et  al. Comparison of
tanyl administration on the minimum alveolar concentration of the effects of xylazine bolus versus medetomidine constant rate
isoflurane in horses. Br J Anaesth. 2006;97:232. infusion on cardiopulmonary function and depth of anesthe-
243. Benmansour P, Duke-Novakovski T. Prolonged anesthesia us- sia in horses anesthetized with isoflurane. J Am Vet Med Assoc.
ing sevoflurane, remifentanil and dexmedetomidine in a horse. 2012;240:991.
Vet Anaesth Analg. 2013;40:521. 264. Kempchen S, Kuhn M, Spadavecchia C, et  al. Medetomidine
244. Lamuraglia R, Kirkby P, Funcia JP. Cardiopulmonary effects continuous rate intravenous infusion in horses in which sur-
and recovery quality of remifentanil–isoflurane anesthesia in gical anaesthesia is maintained with isoflurane and intrave-
horses. J Equine Vet Sci. 2015;35:271. nous infusions of lidocaine and ketamine. Vet Anaesth Analg.
245. Nilsfors L, Kvart C, Kallings P, et  al. Cardiorespiratory and 2012;39:245.
sedative effects of a combination of acepromazine, xylazine and 265. Nannarone S, Cenani A, Gialletti R, et al. Clinical comparison
methadone in the horse. Equine Vet J. 1988;20:364. of two regimens of lidocaine infusion in horses undergoing
246. Linardi RL, Stokes AM, Keowen ML, et al. Bioavailability and laparotomy for colic. Vet Anaesth Analg. 2015;42:150.
pharmacokinetics of oral and injectable formulations of metha- 266. Peiró JR, Barnabe PA, Cadioli FA, et al. Effects of lidocaine in-
done after intravenous, oral, and intragastric administration in fusion during experimental endotoxemia in horses. J Vet Intern
horses. Am J Vet Res. 2012;73:290. Med. 2010;24:940.
247. DeRossi R, Medeiros U, de Almeida RG, et al. Meperidine pro- 267. Skarda RT, Muir WW. Segmental epidural and subarachnoid
longs lidocaine caudal epidural anaesthesia in the horse. Vet J. analgesia in conscious horses: a comparative study. Am J Vet
2008;178:294. Res. 1870;44:1983.
248. Foreman JH, Ruemmler R. Efficacy of intramuscular meperi- 268. Hendrickson DA, Wilson DG. Laparoscopic cryptorchid cas-
dine hydrochloride versus placebo in experimental foot lame- tration in standing horses. Vet Surg. 1997;26:335.
ness in horses. Equine Vet J Suppl. 2013;45:48. 269. Skarda RT, Muir WW. Analgesic, hemodynamic and respira-
249. Skarda RT, Muir WW. Analgesic, hemodynamic, and respira- tory effects of caudal epidurally administered ropivacaine hy-
tory effects induced by caudal epidural administration of me- drochloride in mares. Vet Anaesth Analg. 2001;28:61.
peridine hydrochloride in mares. Am J Vet Res. 2001;62:1001. 270. van Loon JP, Menke ES, Doornenbal A, et al. Antinociceptive
250. Natalini CC, Linardi RL. Analgesic effects of epidural adminis- effects of low dose lumbosacral epidural ropivacaine in healthy
tration of hydromorphone in horses. Am J Vet Res. 2006;67:11. ponies. Vet J. 2012;193:240.
251. Yamashita K, Muir WW, Tsubakishita S, et al. Clinical compari- 271. DeRossi R, Módolo TJ, Pagliosa RC, et al. Comparison of anal-
son of xylazine and medetomidine for premedication of horses. gesic effects of caudal epidural 0.25% bupivacaine with bupiv-
J Am Vet Med Assoc. 2002;221:1144. acaine plus morphine or bupivacaine plus ketamine for analgesia
252. López-Sanromán FJ, Holmbak-Petersen R, Varela M, et al. Ac- in conscious horses. J Equine Vet Sci. 2012;32:190.
celerometric comparison of the locomotor pattern of horses se- 272. DeRossi R, Miguel GLS, Frazílio FO, et al. L-Bupivacaine 0.5%
dated with xylazine hydrochloride, detomidine hydrochloride, vs. racemic 0.5% bupivacaine for caudal epidural analgesia in
or romifidine hydrochloride. Am J Vet Res. 2013;74:828. horses. J Vet Pharmacol Ther. 2005;28:293.

You might also like