+ All Categories
Home > Documents > Comparison of phenobarbital with bromide as a first-choice antiepileptic drug for treatment of...

Comparison of phenobarbital with bromide as a first-choice antiepileptic drug for treatment of...

Date post: 14-Dec-2016
Category:
Upload: david-mark
View: 213 times
Download: 0 times
Share this document with a friend
11
JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1073 SMALL ANIMALS S ince the early 1990s, bromide (most commonly ad- ministered as a potassium salt) has been used as an AED for the long-term management of epilepsy in dogs. 1–4 Increasingly, it is considered as an alternative to phenobarbital as the first-choice sole AED used for long-term control of epilepsy in dogs. 5–8 Successful use of both phenobarbital and bromide is facilitated by a long elimination half-life, which minimizes fluctuation in drug concentrations during a 12-hour dosing interval. 7–11 Therapeutic drug monitoring is widely available and is supported by canine (rather than human) therapeutic ranges as follows: serum phenobarbital concentrations of 15 to 45 µg/mL 7,12–14 and serum bromide concentrations of 1 to 3 mg/mL (when used as a sole treatment). 7,12–17 Comparison of phenobarbital with bromide as a first-choice antiepileptic drug for treatment of epilepsy in dogs Dawn Merton Boothe, DVM, PhD, DACVIM, DACVCP; Curtis Dewey, DVM, MS, DACVS, DACVIM; David Mark Carpenter, PhD Objective—To compare efficacy and safety of treatment with phenobarbital or bromide as the first-choice antiepileptic drug (AED) in dogs. Design—Double-blinded, randomized, parallel, clinical trial. Animals—46 AED-naïve dogs with naturally occurring epilepsy. Procedures—Study inclusion was based on age, history, findings on physical and neuro- logic examinations, and clinicopathologic test results. For either phenobarbital treatment (21 dogs) or bromide treatment (25), a 7-day loading dose period was initiated along with a maintenance dose, which was adjusted on the basis of monthly monitoring. Efficacy and safety outcomes were compared between times (baseline and study end [generally 6 months]) and between drugs. Results—Phenobarbital treatment resulted in eradication of seizures (17/20 [85%]) signifi- cantly more often than did bromide (12/23 [52%]); phenobarbital treatment also resulted in a greater percentage decrease in seizure duration (88 ± 34%), compared with bromide (49 ± 75%). Seizure activity worsened in 3 bromide-treated dogs only. In dogs with seizure eradication, mean ± SD serum phenobarbital concentration was 25 ± 6 µg/mL (phenobar- bital dosage, 4.1 ± 1.1 mg/kg [1.9 ± 0.5 mg/lb], PO, q 12 h) and mean serum bromide con- centration was 1.8 ± 0.6 mg/mL (bromide dosage, 31 ± 11 mg/kg [14 ± 5 mg/lb], PO, q 12 h). Ataxia, lethargy, and polydipsia were greater at 1 month for phenobarbital-treated dogs; vomiting was greater for bromide-treated dogs at 1 month and study end. Conclusions and Clinical Relevance—Both phenobarbital and bromide were reasonable first-choice AEDs for dogs, but phenobarbital was more effective and better tolerated during the first 6 months of treatment. (J Am Vet Med Assoc 2012;240:1073–1083) Each drug has its disadvantages. Both are associated with polyuria, polydipsia, and polyphagia and the sequelae as- sociated with general sedation (eg, lethargy and ataxia), adverse effects to which an animal may develop toler- ance. 1,2,5–7,17 In addition, each has unique characteristics that complicate successful treatment. Long-term pheno- barbital treatment has been associated with hepatotox- icity, although a cause-and-effect relationship has not been proven. 18–20 Phenobarbital also has been associated with unpredictable, idiosyncratic adverse drug reactions, including pancytopenia, 21,22 and drug interactions that reflect induction of selected hepatic drug-metabolizing enzymes that target xenobiotics 23–30 as well as endoge- nous compounds. 31,32 Induction of its own metabolism might result in a decrease in plasma drug concentrations and therapeutic failure. In contrast, although renal ex- cretion of bromide limits hepatotoxicity or induction of hepatic enzymes, its elimination half-life in dogs is so long that several months of treatment must occur with each change in dosage before steady-state con- centrations (and thus maximum effect) are reached. 33,34 Although this delay can be overcome by administration From the Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine (Boothe) and the Department of Mathematics and Statistics, College of Sciences and Mathematics (Carpenter), Auburn University, Auburn, AL 36849; and the De- partment of Clinical Sciences, College of Veterinary Medicine, Cor- nell University, Ithaca, NY 14850 (Dewey). This study was implemented at Texas A&M University’s Texas Vet- erinary Medical Center but included the participation of referral practitioners throughout the United States. Supported by the Canine Health Foundation and the American Ken- nel Club. Presented as a poster at the 20th American College of Veterinary In- ternal Medicine Annual Forum, Dallas, May 2002. Address correspondence to Dr. Boothe ([email protected]). ABBREVIATIONS AED Antiepileptic drug CI Confidence interval
Transcript

JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1073

SM

ALL A

NIM

ALS

Since the early 1990s, bromide (most commonly ad-ministered as a potassium salt) has been used as

an AED for the long-term management of epilepsy in dogs.1–4 Increasingly, it is considered as an alternative to phenobarbital as the first-choice sole AED used for long-term control of epilepsy in dogs.5–8 Successful use of both phenobarbital and bromide is facilitated by a long elimination half-life, which minimizes fluctuation in drug concentrations during a 12-hour dosing interval.7–11 Therapeutic drug monitoring is widely available and is supported by canine (rather than human) therapeutic ranges as follows: serum phenobarbital concentrations of 15 to 45 µg/mL7,12–14 and serum bromide concentrations of 1 to 3 mg/mL (when used as a sole treatment).7,12–17

Comparison of phenobarbital with bromide as a first-choice antiepileptic drug for treatment of epilepsy in dogs

Dawn Merton Boothe, dvm, phd, dacvim, dacvcp; Curtis Dewey, dvm, ms, dacvs, dacvim; David Mark Carpenter, phd

Objective—To compare efficacy and safety of treatment with phenobarbital or bromide as the first-choice antiepileptic drug (AED) in dogs.Design—Double-blinded, randomized, parallel, clinical trial.Animals—46 AED-naïve dogs with naturally occurring epilepsy.Procedures—Study inclusion was based on age, history, findings on physical and neuro-logic examinations, and clinicopathologic test results. For either phenobarbital treatment (21 dogs) or bromide treatment (25), a 7-day loading dose period was initiated along with a maintenance dose, which was adjusted on the basis of monthly monitoring. Efficacy and safety outcomes were compared between times (baseline and study end [generally 6 months]) and between drugs.Results—Phenobarbital treatment resulted in eradication of seizures (17/20 [85%]) signifi-cantly more often than did bromide (12/23 [52%]); phenobarbital treatment also resulted in a greater percentage decrease in seizure duration (88 ± 34%), compared with bromide (49 ± 75%). Seizure activity worsened in 3 bromide-treated dogs only. In dogs with seizure eradication, mean ± SD serum phenobarbital concentration was 25 ± 6 µg/mL (phenobar-bital dosage, 4.1 ± 1.1 mg/kg [1.9 ± 0.5 mg/lb], PO, q 12 h) and mean serum bromide con-centration was 1.8 ± 0.6 mg/mL (bromide dosage, 31 ± 11 mg/kg [14 ± 5 mg/lb], PO, q 12 h). Ataxia, lethargy, and polydipsia were greater at 1 month for phenobarbital-treated dogs; vomiting was greater for bromide-treated dogs at 1 month and study end.Conclusions and Clinical Relevance—Both phenobarbital and bromide were reasonable first-choice AEDs for dogs, but phenobarbital was more effective and better tolerated during the first 6 months of treatment. (J Am Vet Med Assoc 2012;240:1073–1083)

Each drug has its disadvantages. Both are associated with polyuria, polydipsia, and polyphagia and the sequelae as-sociated with general sedation (eg, lethargy and ataxia), adverse effects to which an animal may develop toler-ance.1,2,5–7,17 In addition, each has unique characteristics that complicate successful treatment. Long-term pheno-barbital treatment has been associated with hepatotox-icity, although a cause-and-effect relationship has not been proven.18–20 Phenobarbital also has been associated with unpredictable, idiosyncratic adverse drug reactions, including pancytopenia,21,22 and drug interactions that reflect induction of selected hepatic drug-metabolizing enzymes that target xenobiotics23–30 as well as endoge-nous compounds.31,32 Induction of its own metabolism might result in a decrease in plasma drug concentrations and therapeutic failure. In contrast, although renal ex-cretion of bromide limits hepatotoxicity or induction of hepatic enzymes, its elimination half-life in dogs is so long that several months of treatment must occur with each change in dosage before steady-state con-centrations (and thus maximum effect) are reached.33,34 Although this delay can be overcome by administration

From the Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine (Boothe) and the Department of Mathematics and Statistics, College of Sciences and Mathematics (Carpenter), Auburn University, Auburn, AL 36849; and the De-partment of Clinical Sciences, College of Veterinary Medicine, Cor-nell University, Ithaca, NY 14850 (Dewey).

This study was implemented at Texas A&M University’s Texas Vet-erinary Medical Center but included the participation of referral practitioners throughout the United States.

Supported by the Canine Health Foundation and the American Ken-nel Club.

Presented as a poster at the 20th American College of Veterinary In-ternal Medicine Annual Forum, Dallas, May 2002.

Address correspondence to Dr. Boothe ([email protected]).

Abbreviations

AED Antiepileptic drugCI Confidence interval

1074 Scientific Reports JAVMA, Vol 240, No. 9, May 1, 2012

SM

ALL

AN

IMA

LS

of a loading dose that targets steady-state serum bromide concentrations, such an approach also increases the risk of adverse effects, the most common of which is vomit-ing.3,4,6,11,35,36 Although drug interactions are limited with bromide, dietary chloride content predictably and pro-portionately impacts bromide clearance and thus plasma drug concentrations.33,34 Both phenobarbital and bro-mide continue to be used as first-choice AEDs,5–7 yet no studies have compared their use as drugs for control of epilepsy in dogs. The purpose of the study reported here was to compare efficacy and safety of bromide with those of phenobarbital in dogs with spontaneously occurring epilepsy.

Materials and Methods

This study was implemented in client-owned AED-naïve dogs with spontaneous epilepsy. Informed con-sent was obtained and the protocol was approved by the Clinical Research Committee of the Texas Veteri-nary Teaching Hospital. A sample size of 25 dogs/group was targeted to allow detection of a 40% difference in number of seizures per month between groups with a power of 80%. Patients were solicited for participation through advertisements in selected veterinary journals and through the therapeutic drug monitoring service at the Texas Veterinary Medical Center. Evaluation at Texas A&M University was not necessary for a patient to participate in the study.

Patient selection—Diagnosis of epilepsy was made on the basis of clinicopathologic test results, history, and findings on physical and neurologic ex-aminations performed by the referring veterinarian at the time of evaluation because of seizures. Clinico-pathologic tests consisted of a CBC, serum biochemi-cal analysis, and measurements in serum of prepran-dial and postprandial bile acids concentrations and amylase and lipase activities. A standardized neuro-logic examination form and questionnaire were com-pleted by the referring veterinarian, and a standard-ized questionnaire focusing on baseline seizure data was completed by the client. A criterion for candi-date inclusion in the study included age of onset of disease between 0.5 and 6.5 years; age criterion for enrollment was selected on the basis of the currently described range that encompasses the emergence of idiopathic epilepsy. Inclusion criteria related to sei-zure activity included a seizure interval ≤ 1 month (no stipulation was made as to duration or number other than that each patient must have endured at least 2 seizures so that a baseline interval could be determined), no abnormal neurologic examination findings or abnormal neurologic behavior between seizure episodes, no prior AED treatment (other than diazepam necessary to control status epilepticus), and clinicopathologic test results within reference limits. When identifiable from baseline data, patients with secondary epilepsy, particularly that caused by infection, neoplasia, or metabolic diseases, were ex-cluded. However, invasive (eg, CSF analysis) or ex-pensive diagnostic tests (eg, MRI) necessary to rule out some causes of secondary epilepsy were not re-quired for participation in the study.

Drug administration—Dogs enrolled into the study were assigned by random blocking (blocks of 10) to receive either phenobarbital or bromide. All study participants except the principal investigator (DMB) were blinded to AED administration. No study inves-tigator served as the primary veterinarian for any study subject. However, the principal investigator (DMB), in consultation with the referring veterinarian and client, made all recommendations regarding changes in drug administration regimens for all patients. Recommen-dations subsequently were implemented by the client through the referring veterinarian.

To individualize dosage on the basis of response to treatment, drugs were prepared each month for each patient on the basis of therapeutic drug monitoring results. Phenobarbitala was purchased as the finished product for administration. Bromide is not available as an approved drug. A bromide product is commercially available, but this product has not been subjected to pre-market approval or assessment by any regulatory agen-cy; rather, it is a manufactured, compounded product. Thus, bromide was compounded by use of a pure crystal substrate.b Both drugs were prepared in opaque gelatin capsules,c with the capsule size selected on the basis of the smallest size in which the calculated dose would fit. The calculated daily dose for each patient was adminis-tered in a maximum of 2 capsules. Filler was not used for either drug. All compounding was performed by a registered pharmacist with adherence to stipulations set forth in American Medicinal Drug Use Clarification Act and appropriate compliance guidelines.37,38 Drugs were identified on their labels by numeric codes. Clients were instructed to administer each drug at 12-hour intervals; the drug could be administered in food with the excep-tion of the day before monitoring, for which food was withheld from dogs after midnight. Diets that dogs were receiving at the time of enrollment into the study were continued throughout the study period; treats were al-lowed as prior to enrollment.

The amount of AED administered to each patient was designed to target the low end of the therapeutic range at steady state, with dosages individualized for each patient on the basis of therapeutic drug monitor-ing.7,12–17 The elimination half-life for phenobarbital (53 to 96 hours)9,10 is considerably shorter than that for bro-mide (range, 15 ± 9 days to 69 ± 22 days, depending in part on chloride intake).11,33,34 To circumvent the differ-ences in time to steady state and thus maximum efficacy for comparison between the 2 drugs, a loading dose was administered orally over a 7-day period (14 equal doses administered every 12 hours) such that steady-state drug concentrations that targeted the low end of the therapeu-tic range for each drug would be achieved for either drug by day 8. For bromide, a total loading dose of 450 mg/kg (204.5 mg/lb) was administered orally to achieve serum bromide concentrations of approximately 1.25 mg/mL.17 For phenobarbital, a loading dose was calculated by use of the following equation39,40:

DL = Target•Vd/F

where DL is loading dose, Vd is volume of distribution,

and F is oral bioavailability. To target serum phenobar-

JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1075

SM

ALL A

NIM

ALS

bital concentrations of approximately 17 µg/mL, as-suming 85% oral bioavailability and a volume of dis-tribution of 0.65 L/kg, a total loading dose of 12 mg/kg (5.45 mg/lb) was calculated for oral administration.

The total loading dose for each drug was divided into 14 equal doses and administered every 12 hours over a 7-day period along with the maintenance dose. Administration of maintenance doses began on day 1 for bromide (15 mg/kg [6.8 mg/lb], PO, q 12 h) or phe-nobarbital (3.5 mg/kg [1.6 mg/lb], PO, q 12 h).7,11 On day 8 of treatment for either group, blood samples were obtained to monitor the serum concentration (peak at 5 hours for phenobarbital and trough for both phenobar-bital and bromide) of each drug achieved with the load-ing dose. Also on day 8, the dosage for each drug was decreased to a maintenance dose. Serum drug concen-trations were monitored again 3 weeks later (month 1) after the loading dose was completed to make sure se-rum drug concentrations were being maintained. If the 8-day and 3-week serum drug concentrations were not similar, the dosage was adjusted on the basis of patient response and the difference in serum drug concentra-tions. Thus, in general, by month 1 of treatment, a tar-get serum drug concentration and dosage had been es-tablished for each patient. Blood samples were obtained from dogs for monitoring monthly thereafter, and ad-justments were made by the investigator as described.

The target serum drug concentration for each pa-tient was the lowest possible concentration associated with eradication of seizures (≥ 50% reduction in sei-zures being an acceptable second goal) and absence of intolerable adverse effects. The maintenance dose was decreased if seizures were well controlled and ad-verse effects emerged. The dosage also was decreased for some patients for which seizures were well con-trolled (ie, the patient remained seizure free through 2 prestudy seizure intervals), even if adverse effects were not present, if the serum concentration of the drug was considered higher than necessary to control seizures (ie, concentrations were in the mid to high population therapeutic range). All decreases in dosage were made in 25% decrements. The targeted concentration for a patient was increased in the event that seizures were not controlled and adverse effects were either absent or tolerable. For such patients, serum drug concentrations were increased by approximately 25% by administra-tion of a reduced loading dose and an increase in the maintenance dose. A decreased total loading dose for bromide was 200 to 250 mg/kg (91 to 114 mg/lb) and 3 to 3.5 mg/kg (1.36 to 1.6 mg/lb) for phenobarbital. The new total loading dose was divided into 6 equal doses and administered every 12 hours over a 3-day period along with the new maintenance dose. This stepwise approach to increasing serum drug concentrations was implemented repetitively in any patient that continued to have seizures until either seizures were acceptably controlled or intolerable adverse effects emerged and persisted. Because of the concern of phenobarbital- induced hepatotoxicity, the dosage of phenobarbital was not increased any further in patients for which se-rum phenobarbital concentration was approximately 32 µg/mL (an additional 25% increase in phenobarbi-tal concentrations would result in serum phenobarbi-

tal concentration ≥ 40 µg/mL). Cases in which patients continued to have seizures despite serum phenobar-bital concentration ≥ 32 µg/mL were thus considered failures, and those patients were withdrawn from the study at that point. Serum bromide concentrations could reach or exceed 3 mg/mL as long as unacceptable adverse effects did not emerge. For either drug, cases in which patients were having seizures and adverse effects were unacceptable and were thus considered therapeu-tic failures, regardless of serum drug concentrations, and those patients were withdrawn from the study at that point.

After each monthly follow-up evaluation and 1 week prior to the next follow-up evaluation, a 6-week supply of the appropriate AED at the previous or new dosage was mailed to the referring veterinarian. Dur-ing each monthly visit to the referring veterinarian, any AED not administered to the dog by its owner dur-ing the previous month was returned by the owner to the veterinarian who, in turn, returned the AED to the principal investigators. Client compliance was verified by random checks (counting) of remaining drug and by monthly therapeutic drug monitoring.

Data collection—Clients maintained a calendar and log that required daily entries regarding their dog’s response to treatment. These were sent to the client through the veterinarian at least 6 weeks prior to the start of the study, when such a delay in the implemen-tation of treatment was acceptable to owner and veteri-narian; otherwise, clients provided baseline data for the 30 days prior to study enrollment on the basis of records or the history as provided to the referring veterinarian. The calendar directed clients to mark each day either yes or no if they observed abnormal activity, includ-ing seizures, in their dog. A yes designation required a follow-up description on the log of the abnormal activ-ity, including the time of day the activity occurred and, for seizures, the number, duration, and severity. Clients also were specifically directed to document daily any activity that might be indicative of adverse effects, in-cluding a yes or no designation for lethargy (defined as sleepiness or apathy), ataxia (described as wobbliness), appetite increase or decrease, vomiting or diarrhea, in-creased urination or water intake, or hyperactivity.

Patients were reevaluated by their referring vet-erinarian at the end of each month of treatment. Vet-erinarians were sent the new month’s packet contain-ing forms and tubes for collection of blood and a new month’s supply of the AED (at the previous month’s dosage) at least 1 week prior to the monthly follow-up evaluation. Evaluation was accomplished by use of in-vestigator-supplied standardized forms that addressed questions specifically relating to response (including evidence of adverse drug events or adverse effects) to AED treatment. Additionally, blood samples were col-lected for clinicopathologic tests (as collected at base-line) and therapeutic drug monitoring. Food was with-held from dogs the night before the scheduled visit. Clients were instructed not to give the morning dose of AED and to deliver their dog to the referring veterinar-ian by the time the morning dose was due to be given. At the time of arrival at the clinic, a preprandial blood

1076 Scientific Reports JAVMA, Vol 240, No. 9, May 1, 2012

SM

ALL

AN

IMA

LS

sample was collected for clinicopathologic testing and determination of the trough serum drug concentration. The morning dose of AED was then administered, and 5 hours later, a peak serum drug concentration was measured. The dog was then fed, and a postprandial serum bile acids concentration was determined 2 hours later. During each reevaluation, clients submitted to the veterinarian completed monthly logs and calendars and any unused drug. Blood samples, unused drug, patient calendar and logs, and the referring veterinarian’s assess-ment were mailed to the principal investigator by use of a prepaid, overnight courier service. After analysis and evaluation by the study investigator, if a new drug dosage was indicated, a 6-week supply of the drug at the new dosage was immediately mailed to the referring veteri-narian such that it was received within 1 week after the last follow-up evaluation and at least 3 weeks before the next follow-up evaluation. At the time of each dog’s com-pletion of the study, the code for each drug was revealed to the referring veterinarian and client. Clinicopatho-logic testing and therapeutic drug monitoring results for the last month of the study and a 6-week supply of drug were provided. As an additional incentive for participa-tion, therapeutic drug monitoring services through the investigator’s laboratory were offered at a 50% discount for the remainder of the dog’s life.

Criteria for withdrawal from the study—With-drawal from the study could be either dog owner or investigator driven. Clients could withdraw their dog from the study at any time for any reason. Investigator-driven withdrawals occurred if clients or veterinarians failed to return daily logs, calendars, and unused drug or failed to submit the dog for monthly blood sample collection. Emergence of unacceptable adverse drug events also was a condition of withdrawal or study end as determined by communication between the owner, referring veterinarian, and principal investigator. Inclu-sion of data for withdrawn patients depended on the reasons for withdrawal and the time of the withdraw-al.41 If withdrawal occurred ≥ 2 months into the study, given that sufficient time had elapsed to allow stabili-zation of serum drug concentrations and response of seizure activity to the AED, all data were included and the patient was considered to have completed the study, with the ending month being the month of withdrawal rather than month 6. If an unacceptable adverse event necessitating withdrawal occurred prior to 2 months into the study, safety data for that patient were retained in the analysis and the case was considered a therapeu-tic failure rather than a success, even if seizures were controlled. Efficacy data for such patients were retained only if seizures had been eradicated in the patient by month 2.

Data analysis—Phenobarbital was assayed in serum with an automated polarized immunofluorescence as-say.42 Bromide was assayed in serum by UV spectroscopy with a previously described gold chloride method; this assay has been validated by the US Pharmacopeia.43,44 Both assays were validated in canine serum. Intra-assay and interassay variability for both assays was < 10% for control samples that spanned the lower and up-per limits of quantitation for bromide (0.5 and 4.5 mg/mL,

respectively) and phenobarbital (5 and 60 µg/mL, respec-tively). Disappearance half-life was calculated monthly for phenobarbital on the basis of peak and trough concentra-tions and the following equation:

t1/2

= 0.693/kel

where t1/2

is disappearance half-life and kel is the dis-

appearance rate constant, calculated as (ln [peak/trough]/12 h).39,40 The timing of blood sample collec-tion to measure peak serum phenobarbital concen-tration was chosen on the basis of an oral absorption half-life of 1.3 hours, indicating that approximately 93% of the orally administered drug should have been absorbed from the gastrointestinal tract.9,10

Efficacy outcome measures—At the end of the study for each dog, efficacy of each drug in controlling seizures was evaluated via several outcome measures. Seizure activity was measured as the duration (minutes) of each seizure episode (by use of a mean duration for all seizures that occurred in the month of report), num-ber of seizures in 1 month, and duration of time (days) that lapsed between seizure episodes (seizure interval) at baseline and for the last month of the study. For dogs in which seizures were eradicated (ie, seizure interval > 180 days at study end), the number of days since the last seizure was recorded at study end (eg, 180 days if the patient did have a seizure throughout the 6-month period). Additionally, for each dog at baseline and at study end, severity of seizure activity was scored such that 0 indicated no seizure activity; 1 indicated partial motor seizures with or without altered consciousness; 2 indicated generalized tonic clonic seizures ≤ 3 minutes in duration, with or without loss of consciousness, and with only a single seizure during a 24-hour period; 3 indicated generalized tonic clonic seizures > 3 minutes, with or without loss of consciousness, defecation, uri-nation, and vocalization, and with seizures as frequent as 2 in a 30-minute period; and 4 indicated general-ized tonic clonic seizures with loss of consciousness and clustering. Clustering was defined as ≥ 3 seizures with < 30 minutes elapsing between seizures. Clients in conjunction with referring veterinarians and the prin-cipal investigator (DMB) determined the score for each patient. Finally, at study end, response to each AED was designated for each dog as eradication (0 seizures at month 6), > 50% reduction in seizure number, no re-sponse (≤ 50% decrease in seizure number), or worsen-ing of seizure activity (an increase of any magnitude in seizure numbers at study end vs baseline). These data, along with safety data, were used to designate treatment in each dog as either a success or failure in regard to AED treatment.

Safety and adverse effects outcome measures—Clinicopathologic test data were evaluated monthly for evidence of covert illness. Hepatotoxicity, rather than in-duction of hepatic enzymes, was considered to be pres-ent if serum alkaline phosphatase activity and serum ala-nine aminotransaminase activity were ≥ 2 and 3 times, respectively, the reference range activities.20,45 In con-trast, evidence of hepatic dysfunction was determined on the basis of changes in hepatic function tests (albumin,

JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1077

SM

ALL A

NIM

ALS

cholesterol, and BUN concentrations) outside the refer-ence range or, for serum bile acids, concentrations > 13 µmol/L (preprandial) or 30 µmol/L (postprandial).20,45 Hepatic dysfunction was considered present if results of at least 3 of the 4 hepatic function tests were outside of the reference range at study end. Adverse effects (eg, vomiting or diarrhea; lethargy; ataxia; polyuria, polydip-sia, or polyphagia; or hyperactivity) were quantitated at baseline and month 6 by counting the number of days that the adverse effect was documented by the client as occurring in the patient during that month. In addition, the percentage of dogs that developed adverse effects (yes or no) was determined for each treatment group at months 1 and 6 (or study end) and throughout the study. For percentages, a dog was considered to have had an adverse effect regardless of the number of episodes dur-ing that month (eg, a dog that vomited once during the month was treated the same as a dog that vomited 10 times during the month).

Overall success versus failure—Each patient was designated as either a success or a failure in regard to treatment. Success was defined as > 50% reduction in the number of seizures, a severity score ≤ 2, and the lack of unacceptable adverse effects as defined by the client or referring veterinarian. Adverse effects designated as unacceptable must have been sufficiently severe to lead to patient withdrawal from the study prior to the end of the 6-month study period (including during the load-ing period) or cause the client to choose an alternative anticonvulsant at study end.

Statistical analysis—Descriptive data analysis in-cluded generation of mean ± SD for continuous data (clinicopathologic test results, body weight, drug dos-age, and serum drug concentrations) for each time (baseline and study end) for each treatment group. An exception was for disappearance half-life, which was reported as the harmonic mean and pseudo-SD.46 Medi-an and range were determined for nonparametric data, including categorical data (seizure severity scores) or data that were not normally distributed (seizure effi-cacy and adverse effects).

Data analysis established the efficacy and safety of each drug by comparing responses at study end to base-line seizure activity within groups. Nonparametric vari-ables were comparedd between times within each treat-ment group via the Wilcoxon signed rank test, whereas continuous variables were compared between times via a paired Student t test. For phenobarbital, comparisons were also made for disappearance half-life after initial loading period (7 days) and at study end by use of a paired t test. Values of P ≤ 0.05 were considered signifi-cant for all comparisons.

Data analysis also compared the efficacy at study end and safety at study beginning (month 1) and end between the 2 drugs. Analysis of variance could not be performed on nonparametic data, which included most of the efficacy data and adverse effects safety data. Thus, for efficacy comparisons between drugs, percent-age change of study-end data from baseline data was compared via the Wilcoxon rank sum test. For continu-ous, normally distributed data, comparisons were made at study end between drugs via a Student t test for ei-

ther equal or unequal variance as appropriate for each parameter. The proportion of dogs with each degree of seizure severity was compared between groups at base-line and study end by means of χ2 analysis. Additional-ly, the proportion of dogs for which seizure activity was eradicated and the proportion of dogs for which AED treatment was considered a success were compared be-tween treatment groups by means of χ2 analysis. For adverse effects, χ2 analysis was also used to compare the number of dogs with a specific adverse effect at month 1, throughout the study (total), and at study end.

Results

Study animals—Forty-six patients were enrolled in the study: 21 in the phenobarbital group and 25 in the bromide group. The study included 7 mixed-breed dogs, 5 Labrador Retrievers, 3 Golden Retrievers, 3 Miniature Schnauzers, 3 Standard Poodles, 2 Welsh Springer Spaniels, 2 Siberian Huskies, 2 Greyhounds, 2 German Shepherd Dogs, 2 Dalmatians, 2 Collies, and 1 each of the following dogs: Basset Hound, Beagle, Bi-chon Frise, Border Collie, Cocker Spaniel, Dachshund, English Springer Spaniel, German Shorthaired Pointer, Italian Greyhound, Miniature Dachshund, Pug, Rott-weiler, and Schnauzer. Male dogs were disproportion-ately represented; there were 11 sexually intact males, 4 sexually intact females, 21 castrated males, and 10 spayed females. Seizures in the majority (33/46 [72%]) of patients were scored at baseline as either 4 (17/46 [37%]) or 3 (16/46 [35%]) in severity. At baseline, se-verity of seizures for bromide-treated dogs was less than that of phenobarbital-treated dogs. The proportion of dogs with a seizure severity score of 2 at baseline was significantly (P = 0.005) lower in phenobarbital-treated dogs (1/21 [4.8%]), compared with bromide-treated dogs (7/25 [28%]). Although the proportion of patients at baseline with scores of 3 or 4 was 86% (18/21) for phenobarbital-treated dogs and 60% (15/25) for bro-mide-treated dogs, these proportions did not differ sig-nificantly. Groups did not differ significantly in terms of sex (phenobarbital-treated dogs, 14/21 [67%] males and 7/21 [33%] females; bromide-treated dogs, 18/25 [72%] males and 7/25 [28%] females), body weight (phenobarbital-treated dogs, mean ± SD of 22.7 ± 13 kg [49.9 ± 13.0 lb] and median of 22 kg [48.4 lb]; bro-mide-treated dogs, mean ± SD of 22.8 ± 11.5 kg [50.2 ± 25.3 lb] and median of 23 kg [50.6 lb]), or age (pheno-barbital-treated dogs, mean ± SD of 3.3 ± 1.7 years and median of 3 years; bromide-treated dogs, mean ± SD of 3.1 ± 1.5 years and median of 3 years).

Twenty of 21 phenobarbital-treated dogs and 23 of 25 bromide-treated dogs completed the study (indi-cating both safety and efficacy could be evaluated); the power to detect a 40% change in seizures per month was decreased from the target of 80% to 70%. One dog in the phenobarbital group ingested 1 month’s supply of phe-nobarbital on day 7. This patient was treated supportive-ly and was discharged 3 days after ingestion with no ad-verse events; the owner chose to withdraw the dog from the study. All data on this patient other than baseline and safety (prior to accidental ingestion) were excluded from data analysis, and this patient was considered neither a

1078 Scientific Reports JAVMA, Vol 240, No. 9, May 1, 2012

SM

ALL

AN

IMA

LS

failure nor a success. Treatment of this patient was con-tinued by administration of bromide with no adverse effects. For the bromide group, 2 dogs were withdrawn from the study at day 7 (during the loading period) be-cause of an unacceptable adverse effect (ie, vomiting). Because duration of treatment was < 2 months, efficacy data were not available for these dogs. However, safety data were included; furthermore, both dogs were con-sidered failures. Month 6 was not the final month (study end) for all dogs. One dog in the phenobarbital group was withdrawn from the study at month 2 because of the development of neutropenia (not present at month 1); month 2 data were considered as study-end data for this dog. All data were included for this patient; despite the fact that the dog had responded to treatment (sei-zures eradicated), it was considered a failure because of the unacceptable adverse effect. Noncompliance (failure to bring dogs to monthly follow-up examinations) led to investigator-directed completion at month 3 for 1 phe-nobarbital-treated patient and at month 2 for 1 bromide-treated patient. The failure to bring these dogs to follow-up examinations was unrelated to either drug safety or efficacy for these dogs. Data collected at the respective month of withdrawal for each dog were considered as study-end data for these 2 dogs, and their data were in-cluded. In all, study withdrawal occurred in 1 phenobar-bital-treated dog and 2 bromide-treated dogs (at day 7) and study end occurred prior to 6 months in 1 bromide-treated dog and 2 phenobarbital-treated dogs.

Efficacy data—Several efficacy measures improved at study end, compared with baseline, for both drugs (Table 1). For phenobarbital-treated dogs, seizure num-ber (P = 0.001), duration (P = 0.001), and severity (P = 0.001) were significantly decreased and seizure interval was significantly (P = 0.001) increased at study end, compared with baseline. For bromide-treated dogs, sei-zure number (P = 0.005) and severity (P = 0.001) were significantly decreased and seizure interval was signifi-cantly (P = 0.001) increased at study end, compared with baseline; seizure duration decreased over time, although not significantly (P = 0.08). The percentage

of dogs in which seizure activity was eradicated was sig-nificantly (P < 0.001) greater for phenobarbital-treated dogs (17/20 [85%]), compared with bromide-treated dogs (12/23 [52%]). For those dogs that still had sei-zures at study end, the percentage decrease in seizure duration also was greater (P = 0.04) for phenobarbital-treated dogs, compared with bromide-treated dogs. The percentage of dogs in which seizures were successfully controlled was greater for phenobarbital-treated dogs (15/20 [75%]), compared with bromide-treated dogs (15/23 [65%]), albeit not significantly (P = 0.059).

Insufficient numbers of dogs had seizures at study end; therefore, severity scores could not be compared be-tween treatment groups. A seizure severity score of 0 was achieved by study end in 17 of 20 phenobarbital-treated dogs and in 12 of 23 bromide-treated dogs. Seizure activ-ity worsened at study end, compared with baseline, in 3 of 23 (13%) bromide-treated dogs (2 dogs were 2.5 years old and 1 was 5 years old), but not in any phenobarbital-treated dogs. However, the risk of an increase in seizures with bromide treatment, compared with phenobarbital treatment, was not significant (P = 0.14).

Drug concentrations—Mean serum concentra-tions and dosages in phenobarbital-treated dogs and bromide-treated dogs at study end were summarized (Table 2). After the loading period (day 8), the mean trough serum phenobarbital concentration was 29 ± 13 µg/mL (95% CI, 23 to 34 µg/mL), whereas at month 1, the concentration was 22 ± 7.7 µg/mL (95% CI, 19 to 26 g/mL). For those patients in which seizures were eradicated, mean phenobarbital concentration at study end was 25.4 ± 5.4 µg/mL (range, 12 to 34 µg/mL; 95% CI, 23 to 28 µg/mL). Seizures in 1 patient were > 50% controlled but not eradicated despite a serum drug con-centration of 30.7 µg/mL. Seizures in 2 patients did not improve at all despite drug concentrations of 35 and 36 µg/mL. The correlation between phenobarbital dosage and either treatment response or serum drug concen-trations (R2 = 0.242) was poor.

Because changes in serum phenobarbital concen-trations over time might reflect changes in drug dosage

PB treatment KBr treatment

Variable Mean 6 SD 95% CI Mean 6 SD 95% CI

Seizure duration (min) Baseline 5.7 6 5.3a 3.5–7.9 5.1 6 7.9 3.2–8.3 Study end 1.2 6 3.6b 0.0–2.7 1.5 6 2.6 1.1–2.6 Change (%) 88 6 34 74–102 49 6 75 31–80Seizure interval (d) Baseline (d) 12.7 6 8.1a 9–16 15.1 6 11a 4.5–20 Study end (d) 93 6 76b 62–124 96 6 71b 29–125 Change (% X 100) 16 6 39c 0.1–32 20 6 40d 16–36No. of seizures/mo Baseline 4.4 6 6.3a 1.8–7.0 5.4 6 9.7a 4.0–9.4 Study end 0.4 6 0.9b 0.0–0.8 1.2 6 2.4b 1.0–2.2 Change (%) 87 6 32 13–74 67 6 61 42–92

*Study withdrawal occurred in 1 phenobarbital-treated dog and 2 bromide-treated dogs (at day 7), and study end occurred prior to 6 months in 1 bromide-treated dog and 2 phenobarbital-treated dogs.

a,bMeans with different letters were significantly (P , 0.05) different within treatment groups over time; for bromide-treated dogs, seizure duration decreased over time, although not significantly (P = 0.08). c,dMeans with different letters were significantly (P # 0.05) different between treatment groups at that time.

KBr = Potassium salt of bromide. PB = Phenobarbital.

Table 1—Comparison of seizure activity between phenobarbital-treated (n = 21) and bromide-treated (25) epileptic dogs* at baseline and study end.

JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1079

SM

ALL A

NIM

ALS

or changes in disappearance half-life (ie, resulting from autoinduction), peak and trough serum phenobarbi-tal concentrations were measured to calculate half-life. Blood samples to measure both peak and trough serum phenobarbital concentrations were obtained on both day 8 and month 6 for 15 of the 21 patients. Mean disappearance half-life at study end of 46 ± 31 hours (range, 14 to 120 hours; 95% CI, 29 to 56 hours) was significantly (P = 0.05) less than that at baseline (68 ± 31 hours; 95% CI, 56 to 88 hours). The great-est decrease in half-life in a single dog was from 120 hours on day 8 to 21 hours at study end (month 6). Half-life decreased by ≥ 25% in 9 of 15 patients; an increase in seizure activity necessitated an increase in the phenobarbital dosage in 5 of these 9 dogs. An in-crease in serum phenobarbital concentration by ≥ 25% at study end, compared with month 1, occurred in 6 of 20 (30%) patients.

For bromide, mean serum bromide concentration after the loading period (day 8) was 1.3 ± 0.4 mg/mL (95% CI, 1.2 to 1.5 mg/mL). At month 1, the mean se-rum bromide concentration was 1.3 ± 0.5 mg/mL (95% CI, 1.1 to 1.5 mg/mL), indicating the maintenance dose was successful in maintaining serum concentrations achieved by loading. An increase in serum bromide concentrations at study end, compared with month 1, occurred in 52% (12/23) of patients. For those patients in which seizures were eradicated, mean serum bro-mide concentration was 1.8 ± 0.6 mg/mL (range, 0.9 to 3.3 mg/mL). In the 5 patients in which seizures were decreased by ≥ 50% but not eradicated, mean serum bromide concentration was 2.1 ± 0.6 mg/mL. In the pa-tient in which seizures were < 50% controlled, serum bromide concentration was 1.39 mg/mL; this patient became lethargic, and a further increase in the dosage of bromide was not pursued (Table 2). For the 3 dogs receiving bromide for which seizure activity worsened (as determined on the basis of number of seizures per month), serum bromide concentrations were 1.8, 2.5, and 2.9 mg/mL; the dosage of bromide could not be in-creased without causing unacceptable lethargy in these dogs. As with phenobarbital, the correlation between bromide dosage and either treatment response or serum drug concentrations (R2 = 0.0018) was poor.

Safety and adverse-effects data—Mean ± SD body weight was significantly higher at study end, compared with month 1, for dogs in the phenobarbital group (23 ± 13 kg vs 25 ± 14 kg [50.6 ± 28.6 lb vs 55.0 ± 30.8 lb],

respectively). Of 21 phenobarbital-treated dogs, body weight increased in 12 and decreased in 2. Mean ± SD body weight was significantly higher at study end, com-pared with month 1, for dogs in the bromide group (23 ± 11 kg vs 24 ± 13 kg [50.6 ± 24.2 lb vs 52.8 ± 28.6 lb], respectively). Of 23 bromide-treated dogs, body weight increased in 10 and decreased in 8. Whereas percentage change in body weight did not differ between groups at study end, the percentage of dogs that lost weight was significantly (P = 0.02) greater in the bromide group, compared with that for the phenobarbital group.

The percentage of phenobarbital-treated dogs with clinical signs considered to be adverse effects at month 6 was significantly (P < 0.001) less than that for month 1 for polyuria, polydipsia, polyphagia, lethargy, and atax-ia; most clinical signs were absent by month 6 (Table 3). For clinicopathologic test results of phenobarbital-treated dogs, significant differences over time included serum albumin concentration (decreased; P = 0.009) and serum alkaline phosphatase activity (increased; P < 0.001). However, despite these significant differenc-es, mean clinicopathologic test data were within refer-ence limits at study end for dogs in the phenobarbital group (Table 4). One dog in the phenobarbital group was withdrawn from the study at month 2 because of neutropenia with fever. The total WBC and neutrophil counts in this patient were 2,700 X 103 WBCs/µL and 900 X 103 neutrophils/µL, respectively; the PCV in this patient at month 2 was 44%, compared with 54% at baseline. Because of this adverse event, the patient was withdrawn from the study and treatment was switched to bromide. The WBC count was normalizing in this patient at 2 weeks following discontinuation of pheno-barbital treatment.

Two patients in the bromide group were withdrawn from the study within the first 2 weeks after drug ad-ministration because of vomiting (Table 3). One dog was almost withdrawn because of hyperactivity. The percentage of dogs with clinical signs indicative of ad-verse effects at month 6 was significantly (P < 0.001) less than that for month 1. Mean clinicopathologic data did not differ between times and was within reference limits at all times for dogs in the bromide group.

There were no significant differences between treatment groups in clinicopathologic test results at baseline (Table 4). Significantly different percentage change in selected clinicopathologic data at study end included decreased serum albumin concentration (P = 0.02) and increased serum alkaline phosphatase activ-

Serum PB (µg/mL) Serum bromide (mg/mL)

Variable No. of dogs (%) Mean 6 SD Range No. of dogs (%) Mean 6 SD Range

All dogs 20 27 6 6 12.4–36 23 1.9 6 0.6 0.9–3.3100% controlled 17 (85) 25.4 6 5.4 12.4–34 12 (52) 1.8 6 0.6 0.9–3.350% to , 100% controlled 1 (5) 30.7 NA 5 (22) 2.1 6 0.6 1.5–2.7. 0% to , 50% controlled 2 (10) 36 NA 1 (4) 1.39 NAWorsened seizure activity 0 (0) NA NA 3 (13) 2.3 6 0.5 1.8–2.9

†Dogs received phenobarbital (mean 6 SD, 4.11 6 1.1 mg/kg [1.87 6 0.5 mg/lb]; range, 3.9 to 4.9 mg/kg [1.77 to 2.23 mg/lb]) or bromide (mean 6 SD, 30.6 6 10.9 mg/kg [13.9 6 4.95 mg/lb]; range, 26 to 35 mg/kg [11.8 to 15.9 mg/lb]) orally twice a day.

See Table 1 for remainder of key.

Table 2—Serum drug concentrations in phenobarbital-treated (n = 20) and bromide-treated (23) epileptic dogs*† with controlled or uncontrolled seizure activity at study end.

1080 Scientific Reports JAVMA, Vol 240, No. 9, May 1, 2012

SM

ALL

AN

IMA

LS

ity (P < 0.001) for the phenobarbital group, compared with the bromide group. Adverse effects for which the proportion of affected dogs was greater for the pheno-barbital group, compared with the bromide group, at month 1 were polyuria, lethargy, and ataxia; this differ-ence resolved by study end. Adverse effects that were greater for the bromide group, compared with the phe-nobarbital group, at 1 month were vomiting and diar-rhea; the proportion of dogs with vomiting remained significantly (P < 0.001) greater at study end for the bromide group. By study end, the proportion of dogs with lethargy also was significantly (P = 0.04) greater for the bromide group, compared with the phenobar-bital group.

Discussion

This study supports preferential use of phenobar-bital, compared with bromide, for sole treatment of epi-lepsy in dogs. Efficacy of phenobarbital was better as determined on the basis of a higher proportion of dogs for which seizures were eradicated and the shorter du-ration of seizures; overall success was better, albeit not significantly (P = 0.06), for phenobarbital. The poten-tial role of differences in drug concentrations (relative to the therapeutic range) for either drug as a reason for

better efficacy comparison between treatment groups was minimized in the present study by our approach of using monitoring to guide treatment in each pa-tient. For patients that continued to have seizures once steady-state concentrations were reached, the drug dosage (and thus serum concentration) for either drug was increased. Drug dosages continued to be increased until either seizures were controlled or the dog devel-oped adverse effects that were considered unacceptable by the client (or the risk was considered unacceptable by the study investigator), in which case the dog was considered a failure in terms of treatment. From a safe-ty standpoint, polyuria, lethargy, and ataxia were more common in the phenobarbital group as treatment was begun (month 1). However, after the loading period, mean serum phenobarbital concentrations (29 µg/mL; in the mid to high therapeutic range) exceeded the tar-get (17 µg/mL; low end of the therapeutic range) by 70%. This is likely to have increased the incidence of adverse effects related to sedation as treatment was started. In contrast, mean serum bromide concentra-tion (1.3 mg/mL) achieved after the loading period (low end of the therapeutic range) did not substan-tially exceed the target (1.25 mg/mL). Despite better improvement, by study end, mean serum phenobarbi-tal concentrations had decreased, compared with base-

Month 1 Study end Total

Variable PB (n = 20) KBr (n = 23) P value* PB (n = 20) KBr (n = 23) P value* PB (n = 20) KBr (n = 23) P value*

Polyuria (%) 30 9 0.020 0 0 NS 35 13 0.01Polydipsia (%) 40 26 NS 0 4.3 NS 40 39 NSPolyphagia (%) 30 39 NS 0 4.3 NS 30 43 NSLethargy (%) 50 22 , 0.001 5 13 0.040 50 35 NSAtaxia (%) 55 9 , 0.001 5 8.7 NS 55 22 , 0.001Vomiting (%) 15 48 , 0.001 0 21 , 0.001 20 57 , 0.001Diarrhea (%) 5 13 0.040 5 0 NS 15 22 NSHyperactivity (%) 30 35 NS 10 4.3 NS 35 43 NS

*P value for those proportions that differed significantly (P , 0.05) between groups at each time point.n = Number of dogs included in each treatment group at each time point. NS = No significant difference.See Table 1 for remainder of key.

Table 3—Proportion of epileptic dogs with adverse effects that received either phenobarbital or bromide at 1 month, study end, and throughout the study (total).

PB KBr

Variables RR Baseline Study end Change (%) Baseline Study end Change (%)

Albumin (g/dL) 2.4–3.6 4.1 6 0.6* 3.8 6 0.8* 9.2 6 15† 3.9 6 0.5 3.9 6 0.5 –2.2 6 15†ALT (U/L) 10–130 45 6 22 47 6 23 24 6 74 44 6 18 57 6 51 27 6 87ALP (U/L) 24–147 48 6 30* 125 6 95* 205 6 204† 41 6 21 47 6 40 29 6 116†Total bilirubin (mg/dL) 0–0.8 0.3 6 0.2 0.3 6 0.4 19 6 105 0.4 6 0.2 0.3 6 0.4 11 6 169BUN (mg/dL) 5.0–29 16 6 4.3 18.1 6 7.7 –32 6 89 18 6 7 18 6 7.2 –0.6 6 31Cholesterol (mg/dL) 120–247 205 6 64 211 6 53 –8.8 6 28 256 6 89 248 6 82 2 6 22GGT (U/L) 0–25 6.5 6 3 5.9 6 2.6 6.4 6 49 7 6 3.4 5.8 6 3 –3.7 6 51PreBA (µmol/L) , 13 14 6 13.2 6.3 6 2.9 53 6 361 11.5 6 16 7.5 6 7 30.3 6 145PostBA (µmol/L) , 30 20 6 18.3 8.3 6 12 41 6 334 17.5 6 18 13 6 11 –3.8 6 82PCV (%) 37–55 53 6 6.8 50 6 6.6 4.7 6 12 49 6 6 49 6 4.9 –1.6 6 15WBC count (X 103/mL) 6,000–17,000 9,190 6 3,639 8,923 6 3,943 –2.9 6 41 8,896 6 2,263 10,727 6 3,236 –27 6 50

*Significantly different between times within phenobarbital group (P = 0.009 for serum albumin concentration and , 0.001 for serum ALP ac-tivity). †Percentage change is significantly different between treatment groups (P = 0.02 for serum albumin concentration and , 0.001 for serum ALP activity).

ALP = Alkaline phosphatase. ALT = Alanine aminotransferase. BA = Bile acids. GGT = γ-Glutamyl transferase. PreBA = Preprandial bile acids. PostBA = Postprandial bile acids. RR = Reference range.

See Table 1 for remainder of key.

Table 4—Mean ± SD changes in clinicopathologic data over time in dogs with spontaneous epilepsy that were treated with either phe-nobarbital (n = 21) or bromide (25).

JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1081

SM

ALL A

NIM

ALS

line, but serum bromide concentrations were increased relative to baseline. Vomiting and lethargy persisted in the bromide group, compared with the phenobarbital group, at study end. Seizure activity worsened in 13% of bromide-treated dogs but no phenobarbital-treated dogs. Worsening seizures are a recognized, albeit rare, reaction to selected anticonvulsants.47 Although not a statistically significant change, the increase might none-theless be clinically relevant. When considered together with safety data, successful control of seizures, which in-cluded both efficacy and safety data, was greater but not significantly (P = 0.06) so with phenobarbital. Nonethe-less, this study does not exclude bromide as a reasonable first-choice AED for control of epilepsy in dogs; seizures were eradicated in > 50% of bromide-treated patients.

For phenobarbital, mean serum concentrations in-creased approximately 20% between months 1 and 6; for bromide, mean serum concentrations increased ap-proximately 50%; these changes reflected dose increases. Yet, for both drugs, the percentage of dogs that devel-oped adverse effects markedly decreased. As such, this study does support accommodation to the common ad-verse effects caused by either phenobarbital or bromide in dogs. These clinical signs were documented by the owner, and as such, it is possible that it was the own-er, rather than the dog, that adapted to the adverse ef-fects. However, veterinarians assessed patients monthly, thereby decreasing the risk of owner bias in assessment. One adverse effect for which accommodation was less common was vomiting, which occurred more frequently in the bromide group. Hypertonicity and direct gastric irritation are likely to have contributed to these clinical signs. The risk of vomiting was likely increased by the loading approach to drug treatment; vomiting was suf-ficiently unacceptable in 2 dogs during the loading pe-riod that their owners requested withdrawal from the study. Both dogs were subsequently started on pheno-barbital with no adverse effects. However, close to 50% of bromide-treated dogs still were vomiting at day 30 (3 weeks after the loading period) and 20% were still vom-iting at study end. It is possible that vomiting may have been decreased if bromide had been administered as a sodium salt, rather than potassium salt, or as a liquid, rather than capsule. Bromide and phenobarbital have been associated with increased serum triglyceride con-centration48 or pancreatitis49 in dogs, suggesting this as a possible cause of vomiting in the present study. Serum pancreatic lipase immunoreactivity has proven useful for assessing the likelihood of pancreatitis.50 However, at the time that the present study was implemented, this test was not routinely offered. Serum amylase and lipase activity remained normal, supporting but not proving the lack of pancreatitis as a cause of vomiting in the bromide-treated patients. The incidence of vom-iting in dogs of the present study suggests that a load-ing dose with bromide be avoided if there is no need to rapidly achieve therapeutic concentrations.

For phenobarbital, the concern for liver disease caused us to not increase drug dosage further in dogs in which trough concentrations were approximately 32 µg/mL. Among the difficulties associated with phenobarbi-tal use is discriminating phenobarbital-associated hepa-totoxicity from phenobarbital-associated induction of

liver enzymes.19,20 Phenobarbital, but not bromide, was associated with a mean increase in serum alkaline phos-phatase activity at study end, compared with baseline. This increase may be due to induction of serum alkaline phosphatase activity (rather than liver disease) as was in-dicated by the lack of changes indicative of hepatic dis-ease (ie, alanine aminotransaminase activity and BUN, serum albumin, or bile acid concentrations). However, the results of the present study can only be applied to a 6-month period and only if trough concentrations are maintained at < 36 µg/mL (the highest trough measured at month 6 in the present study). The lack of adverse effects at month 6 for phenobarbital, compared with bro-mide, in this study must be balanced with the potential need to monitor hepatic function tests.

Another sequela of phenobarbital-associated in-duction of metabolizing enzymes is the risk of drug interactions, which may have occurred in this study. Be-cause we monitored both peak and trough serum phe-nobarbital each month, we were able to demonstrate a decrease in disappearance half-life in most patients re-ceiving phenobarbital. Changes in disappearance half-life likely contributed to the poor correlation between phenobarbital dosage and either treatment response or serum phenobarbital concentration. The shortest half-life measured in any patient was 14 hours; the greatest change measured in any 1 patient was a decrease from 120 to 21 hours. For such patients, serum phenobarbi-tal concentrations will decrease over time, increasing the risk of therapeutic failure. For the present study, a decrease in serum phenobarbital in association with a shortened half-life often required a dosage increase. Another sequela of shortened phenobarbital half-life is the potential for significant changes in peak and trough concentrations during a single dosing interval, as Levitski and Trepanier14 previously demonstrated in 9% of canine patients (n = 33) receiving phenobarbital for control of epilepsy. The greatest difference between peak and trough phenobarbital concentration occurred in the patient with a 14-hour half-life; concentrations declined from a peak of 17 to a trough of 12 µg/mL. Because a lower serum phenobarbital concentration in-creases the risk of therapeutic failure, we recommend that serum phenobarbital concentration monitoring be performed on the basis of collection of a trough sample such that the lowest concentration that occurs during a dosing interval can be determined. In patients in which control is difficult, collection of both a peak and trough sample might be prudent to identify the potential role of short disappearance half-life in causing therapeutic failure.

Although previously reported therapeutic ranges for both bromide and phenobarbital are supported by the present study, we also demonstrate that the concen-trations (and drug dosages) required to control seizures in dogs are quite variable. For phenobarbital, the low-est (12 µg/mL) and highest (34 µg/mL) effective serum concentrations differed by almost 3-fold. Likewise, for bromide, the lowest (0.9 mg/mL) and highest (3.3 mg/mL) serum concentrations associated with control dif-fered almost 4-fold. This variability exemplifies the im-portance of therapeutic drug monitoring as a tool for guiding treatment. Monitoring allowed us to maintain

1082 Scientific Reports JAVMA, Vol 240, No. 9, May 1, 2012

SM

ALL

AN

IMA

LS

low concentrations in patients that otherwise would have been exposed unnecessarily to higher concentra-tions. We also did not consider a therapeutic failure to have occurred unless serum drug concentrations were high enough to be associated with a risk of liver disease (phenobarbital) or the patient had signs of overdose (phenobarbital or bromide). Although a population therapeutic range for either phenobarbital or bromide is a reasonable target as treatment is begun, monitoring of drug concentrations and clinical signs should deter-mine the range necessary to control seizures in indi-vidual patients. Failure should be ascribed to subthera-peutic concentrations for the patient until the patient continues to have unacceptable seizure activity despite concentrations at or above the population therapeutic range or if unacceptable adverse effects emerge.

One of the weaknesses of this study is the lack of diagnostic procedures that would be necessary to rule in idiopathic epilepsy (a diagnosis by exclusion). Al-though the inclusion criteria were designed to increase the likelihood that idiopathic epilepsy was the cause of seizures in study participants, it cannot be confirmed in any patient. At the time the study was funded, MRI was not a routine option in practice, and including this as a requirement for participation would have caused lo-gistic and financial burdens that would have precluded obtaining an adequate number of study participants. Our inclusion criteria for age ranged from 1 to 6 years (with the most likely age of onset of epilepsy being 1 to 5 years). Four study participants that met all crite-ria for inclusion (2 in each treatment group) were > 5 but < 6.5 years of age. For 3 of these dogs, seizures were eradicated; in the fourth (bromide), seizures were decreased in duration and number by 50% and the se-verity score was decreased from 4 to 3. Thus, although the seizures in these patients may be less likely to be associated with idiopathic epilepsy than were the sei-zures in the other patients, the inclusion of these older patients does not appear to have biased the data toward therapeutic failure.

This study demonstrates that both phenobarbital and bromide are reasonable first-choice AEDs, but phe-nobarbital may be more efficacious. Regarding adverse effects, phenobarbital may be more difficult to start if a loading dose is used; however, once steady-state serum concentrations are reached, adverse effects are more likely to persist for bromide. This study also suggests a poor relationship between drug dosage and serum drug concentrations, suggesting that serum drug concentra-tions should be monitored for guidance in adjustment of drug dosage as control is sought in epileptic dogs.

a. Phenobarbital, Henry Schein Inc, Port Washington, NY.b. Potassium bromide, product No. JT2998-5, VWR Scientific, Suwanee,

Ga.c. Capsuline Inc, Pompano Beach, Fla.d. SAS/STAT Statistical Software, SAS Institute Inc, Cary, NC.

References1. Parent JM. Clinical management of canine seizures. Vet Clin

North Am Small Anim Pract 1988;18:947–965.2. Lane SB, Bunch SE. Medical management of recurrent seizures

in dogs and cats. J Vet Intern Med 1990;4:26–39.3. Pearce LK. Potassium bromide as an adjunct to phenobarbital

for the management of uncontrolled seizures in dogs. Prog Vet Neurol 1990;1:195–101.

4. Podell M, Fenner WR. Bromide therapy in refractory canine id-iopathic epilepsy. J Vet Intern Med 1993;7:318–327.

5. Podell M. Seizures in dogs. Vet Clin North Am Small Anim Pract 1996;26:779–809.

6. Trepanier LA. Use of bromide as an anticonvulsant for dogs with epilepsy. J Am Vet Med Assoc 1995;207:163–166.

7. Boothe DM. Anticonvulsant and other neurologic therapies. In: Boothe DM, ed. Small animal clinical pharmacology and thera-peutics. Philadelphia: WB Saunders Co, 2001;431–456.

8. Dewey CW. Anticonvulsant therapy in dogs and cats. Vet Clin North Am Small Anim Pract 2006;36:1107–1127.

9. Pedersoli WM, Wike JS, Ravis WR. Pharmacokinetics of single doses of phenobarbital given intravenously and orally to dogs. Am J Vet Res 1987;48:679–683.

10. Ravis WR, Pedersoli WM, Wike JS. Pharmacokinetics of phenobarbital in dogs given multiple doses. Am J Vet Res 1989;50:1343–1347.

11. Trepanier LA, Babish JG. Pharmacokinetic properties of bro-mide in dogs after the intravenous and oral administration of single doses. Res Vet Sci 1995;58:248–251.

12. Cunningham JG, Haidukewych D, Jensen HA. Therapeutic se-rum concentrations of primidone and its metabolites, pheno-barbital and pheynlethylmalonamide in epileptic dogs. J Am Vet Med Assoc 1983;182:1091–1094.

13. Morton DJ, Honhold N. Effectiveness of a therapeutic drug monitoring service as an aid to the control of canine seizures. Vet Rec 1988;122:346–349.

14. Levitski RE, Trepanier LA. Effect of timing of blood collection on serum phenobarbital concentrations in dogs with epilepsy. J Am Vet Med Assoc 2000;217:200–204.

15. Grewal MS, Swinyard EA, Jensen HV, et al. Correlation between anticonvulsant activity and plasma concentration of bromide. J Pharmacol Exp Ther 1954;112:109–115.

16. Trepanier LA, Van Schoick A, Schwark WS, et al. Therapeutic se-rum drug concentrations in epileptic dogs treated with potassium bromide alone or in combination with other anticonvulsants: 122 cases (1992–1996). J Vet Intern Med 1998;213:1449–1453.

17. US Pharmacopeia. Bromide compounding monograph. In: Unit-ed States pharmacists’ pharmacopeia 2008–2009. Rockville, Md: US Pharmacopeial Convention, 2009.

18. Dayrell-Hart B, Steinberg SA, VanWinkle TJ, et al. Hepatotoxic-ity of phenobarbital in dogs: 18 cases (1985–1989). J Am Vet Med Assoc 1991;199:1060–1066.

19. Gaskill CL, Miller LM, Mattoon JS, et al. Liver histopatholo-gy and liver and serum alanine aminotransferase and alkaline phosphatase activities in epileptic dogs receiving phenobarbital. Vet Pathol 2005;42:147–160.

20. Muller PB, Taboada J, Hosgood G, et al. Effects of long-term phenobarbital treatment on the liver in dogs. J Vet Intern Med 2000;15:165–171.

21. Jacobs G, Calvert C, Kaufman A. Neutropenia and thrombocy-topenia in three dogs treated with anticonvulsants. J Am Vet Med Assoc 1998;212:681–684.

22. Stokol T, Blue JT, French TW. Idiopathic pure red cell aplasia and nonregenerative immune-mediated anemia in dogs: 43 cas-es (1988–1999). J Am Vet Med Assoc 2000;216:1429–1436.

23. Aldridge A, Neims AH. The effects of phenobarbital and β-naphthoflavone on the elimination kinetics and metabolite pat-tern of caffeine in the Beagle dog. Drug Metab Dis 1979;7:378–382.

24. Bekersky I, Maggio AC, Mattaliano V Jr, et al. Influence of phe-nobarbital on the disposition of clonazepam and antipyrine in the dog. J Pharmacokinet Biopharm 1977;5:507–512.

25. Sams RA, Muir WW. Effects of phenobarbital on thiopental phar-macokinetics in Greyhounds. Am J Vet Res 1988;49:245–249.

26. Ciaccio PJ, Halpert JR. Characterization of a phenobarbital-inducible dog liver cytochrome P450 structurally related to rat and human enzymes of the P450IIIA (steroid-inducible) gene subfamily. Arch Biochem Biophys 1989;271:284–299.

27. Nossaman BC, Amouzadeh HR, Sangiah S. Effects of chloramphen-icol, cimetidine and phenobarbital on and tolerance to xylazine-ketamine anesthesia in dogs. Vet Hum Toxicol 1990;32:216–219.

28. Kantrowitz LB, Peterson ME, Trepanier LA, et al. Serum total

JAVMA, Vol 240, No. 9, May 1, 2012 Scientific Reports 1083

SM

ALL A

NIM

ALS

thyroxine, total triiodothyronine, free thyroxine, and thyrotro-pin concentrations in epileptic dogs treated with anticonvul-sants. J Am Vet Med Assoc 1999;214:1804–1808.

29. Hojo T, Ohno R, Shimoda M, et al. Enzyme and plasma protein induc-tion by multiple oral administrations of phenobarbital at a therapeutic dosage regimen in dogs. J Vet Pharmacol Ther 2002;25:121–127.

30. Orito K, Saito M, Fukunaga K, et al. Pharmacokinetics of zonisamide and drug interaction with phenobarbital in dogs. J Vet Pharmacol Ther 2008;31:259–264.

31. Gieger TL, Hosgood G, Taboada J, et al. Thyroid function and serum hepatic enzyme activity in dogs after phenobarbital ad-ministration. J Vet Intern Med 2000;14:277–281.

32. Muller PB, Wolfsheimer KJ, Taboada J, et al. Effects of long-term phenobarbital treatment on the thyroid and adrenal axis and ad-renal function tests in dogs. J Vet Intern Med 2000;15:157–164.

33. Trepanier LA, Babish JG. Effect of dietary chloride content on the elimination of bromide by dogs. Res Vet Sci 1995;58:252–255.

34. Shaw N, Trepanier LA, Center SA, et al. High dietary chloride con-tent associated with loss of therapeutic serum bromide concentra-tions in an epileptic dog. J Am Vet Med Assoc 1996;208:234–236.

35. Nichols ES, Trepanier LA, Linn K. Bromide toxicosis secondary to renal insufficiency in an epileptic dog. J Am Vet Med Assoc 1996;208:231–233.

36. Rossmeisl JH, Inzana KD. Clinical signs, risk factors, and out-comes associated with bromide toxicosis (bromism) in dogs with idiopathic epilepsy. J Am Vet Med Assoc 2009;234:1425–1431.

37. Extralabel drug use in animals; final rule, 21 CFR Part 530. Fed Regist 1996;61:57731–57746.

38. US FDA Center for Veterinary Medicine. Chapter 6, Subchapter 600, Sec. 608.400—compounding of drugs for use in animals, July 2003. In: Compliance policy guide: compliance policy guidance for FDA staff and in-dustry. Rockville, Md: US FDA, 2003. Available at: www.fda.gov/ICECI/ComplianceManuals/CompliancePolicyGuidanceManual/ucm074656.htm. Accessed Mar 27, 2012.

39. Boothe DM. Therapeutic drug monitoring. In: Boothe DM, ed. Small animal clinical pharmacology and therapeutics. Philadel-phia: WB Saunders Co, 2001;60–72.

40. Bauer LH. Handbook of clinical pharmacokinetics. Columbus, Ohio: McGraw-Hill, 2005.

41. US FDA Good Clinical Practice Program Office of Science & Health Coordination, Office of the Commissioner. Guidance for sponsors, clinical investigators, and IRBs: data retention when sub-jects withdraw from FDA-regulated clinical trials. HF-34. Available at: www.fda.gov/oc/gcp/draft.html. Accessed Jun 22, 2010.

42. Loomis KF, Frye RM. Evaluation of the Abbott TDx for the stat measurement of phenobarbital, phenytoin, carbamazepine, and theophylline. Am J Clin Pathol 1983;80:686–691.

43. Sunshine I. Bromide. Type A. Procedure. In: Sunshine I, ed. CRC meth-odology for analytical toxicology. Cleveland: CRC Press, 1975;54–55.

44. US Pharmacopeia (USP-NF). USP33–NF28. Potassium bromide oral solution monograph. In: The United States pharmacopeia. Rockville, Md: US Pharmacopeial Convention, 2010;3:4375.

45. Navarro VJ, Senior JR. Drug-related hepatotoxicity. N Engl J Med 2006;354:731–739.

46. Lam FC, Hung CT, Perrier DG. Estimation of variance for har-monic mean half-lives. J Pharm Sci 1985;74:229–231.

47. Sazgar M, Bourgeois BF. Aggravation of epilepsy by antiepileptic drugs. Pediatr Neurol 2005;33:227–234.

48. Kluger EK, Malik R, Ilkin WJ, et al. Serum triglyceride concentra-tion in dogs with epilepsy treated with phenobarbital or with phe-nobarbital and bromide. J Am Vet Med Assoc 2008;233:1270–1277.

49. Gaskill CL, Cribb AE. Pancreatitis associated with potassium bromide/phenobarbital combination therapy in epileptic dogs. Can Vet J 2000;41:555–558.

50. Steiner JM, Xenoulis PG, Anderson JA, et al. Serum pancreatic lipase immunoreactivity concentrations in dogs treated with po-tassium bromide and/or phenobarbital. Vet Ther 2008;9:37–44.

From this month’s AJVR

Effects of acepromazine maleate on platelet function assessed by use of adenosine diphosphate–activated and arachidonic acid–activated modified thromboelastography in healthy dogsBobbi J. Conner et al

Objective—To evaluate the effect of acepromazine maleate administered IV on platelet function assessed in healthy dogs by use of a modified thromboelastography assay.Animals—6 healthy adult mixed-breed dogs.Procedures—Dogs received each of 3 treatments (saline [0.9% NaCl] solution [1 to 2 mL, IV] and acepromazine maleate [0.05 and 0.1 mg/kg, IV]) in a randomized crossover study with a minimum 3-day washout period between treatments. From each dog, blood samples were collected via jugular venipuncture immediately before and 30 and 240 minutes after administration of each treatment. A modified thromboelastography assay, consisting of citrated kaolin–activated (baseline assessment), reptilase-ADP–activated (ADP-activated), and reptilase-arachidonic acid (AA)–activated (AA-activat-ed) thromboelastography was performed for each sample. Platelet inhibition was evaluated by as-sessing the percentage change in maximum amplitude for ADP-activated or AA-activated samples, compared with baseline values. Percentage change in maximum amplitude data were analyzed by use of Skillings-Mack tests with significance accepted at a family-wise error rate of P < 0.05 by use of Bonferroni corrections for multiple comparisons.Results—No significant differences were found in the percentage change of maximum amplitude from baseline for ADP-activated or AA-activated samples among treatments at any time.Conclusions and Clinical Relevance—Platelet function in dogs, as assessed by use of a modi-fied thromboelastography assay, was not inhibited by acepromazine at doses of 0.05 or 0.1 mg/kg, IV. This was in contrast to previous reports in which it was suggested that acepromazine may alter platelet function via inhibition of ADP and AA. (Am J Vet Res 2012;73:595–601)

See the midmonth issues of JAVMA

for the expanded table of contents

for the AJVR or log on to

avmajournals.avma.org for access

to all the abstracts.

May 2012


Recommended