Chronic oral dosing of cannabidiol and cannabidiolic acid full-spectrum hemp oil extracts has no adverse effects in horses: a pharmacokinetic and safety study

Tongxin Charlotte Wang Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY

Search for other papers by Tongxin Charlotte Wang in
Current site
Google Scholar
PubMed
Close
 MSc
,
Joseph J. Wakshlag Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY

Search for other papers by Joseph J. Wakshlag in
Current site
Google Scholar
PubMed
Close
 DVM, PhD, DACVIM, DACVSMR
,
Mason C. Jager Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY

Search for other papers by Mason C. Jager in
Current site
Google Scholar
PubMed
Close
 DVM, PhD, DACVP
,
Wayne S. Schwark Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY

Search for other papers by Wayne S. Schwark in
Current site
Google Scholar
PubMed
Close
 DVM, PhD
,
Nathalie L. Trottier Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY

Search for other papers by Nathalie L. Trottier in
Current site
Google Scholar
PubMed
Close
 PhD
,
Jacqueline M. Chevalier Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY

Search for other papers by Jacqueline M. Chevalier in
Current site
Google Scholar
PubMed
Close
 DVM, MSc https://orcid.org/0000-0002-7278-5463
,
Garett Pearson Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY

Search for other papers by Garett Pearson in
Current site
Google Scholar
PubMed
Close
 DVM
, and
Marta Cercone Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY

Search for other papers by Marta Cercone in
Current site
Google Scholar
PubMed
Close
 DVM, PhD, DACVIM https://orcid.org/0000-0002-8922-7927

Abstract

OBJECTIVE

To compare the pharmacokinetics of cannabidiol (CBD) and cannabidiolic acid (CBDA) in horses and to evaluate the safety of their chronic administration.

METHODS

CBD- and CBDA-rich oil (1 mg/kg) were administered orally twice daily to 7 adult horses over 6 weeks in a randomized, crossover design with a 2-week washout period. A 12-hour pharmacokinetic analysis was conducted on day 1 of each 6-week trial, followed by the measurement of peak and trough concentrations at weeks 1, 2, 4, and 6. The cannabinoids safety was assessed via daily physical examination, periodic bloodwork, and liver biopsy at the beginning and end of the study.

RESULTS

12-hour pharmacokinetics revealed a higher maximum serum concentration (103 vs 12 ng/mL) and greater area under the curve (259 vs 62 ng·h/mL) for CBDA when compared to CBD. Cannabidiolic acid nadir and peak serum levels over time ranged from 46 to 122 ng/mL, which was higher than CBD (12 to 38 ng/mL). Complete blood count and serum chemistry revealed no clinically relevant changes with either CBD or CBDA. No significant abnormalities were detected on liver ultrasonographic and histopathologic evaluation on day 0 and after both phases of the study.

CONCLUSIONS

A dose of either 1 mg/kg of CBD or CBDA administered long term appears safe; however, CBDA serum concentrations suggest superior absorption/retention.

CLINICAL RELEVANCE

Chronic cannabinoid supplementation in horses is safe. Considering the higher absorption of CBDA, its use is recommended to evaluate the therapeutic efficacy of this common hemp derived cannabinoid.

Abstract

OBJECTIVE

To compare the pharmacokinetics of cannabidiol (CBD) and cannabidiolic acid (CBDA) in horses and to evaluate the safety of their chronic administration.

METHODS

CBD- and CBDA-rich oil (1 mg/kg) were administered orally twice daily to 7 adult horses over 6 weeks in a randomized, crossover design with a 2-week washout period. A 12-hour pharmacokinetic analysis was conducted on day 1 of each 6-week trial, followed by the measurement of peak and trough concentrations at weeks 1, 2, 4, and 6. The cannabinoids safety was assessed via daily physical examination, periodic bloodwork, and liver biopsy at the beginning and end of the study.

RESULTS

12-hour pharmacokinetics revealed a higher maximum serum concentration (103 vs 12 ng/mL) and greater area under the curve (259 vs 62 ng·h/mL) for CBDA when compared to CBD. Cannabidiolic acid nadir and peak serum levels over time ranged from 46 to 122 ng/mL, which was higher than CBD (12 to 38 ng/mL). Complete blood count and serum chemistry revealed no clinically relevant changes with either CBD or CBDA. No significant abnormalities were detected on liver ultrasonographic and histopathologic evaluation on day 0 and after both phases of the study.

CONCLUSIONS

A dose of either 1 mg/kg of CBD or CBDA administered long term appears safe; however, CBDA serum concentrations suggest superior absorption/retention.

CLINICAL RELEVANCE

Chronic cannabinoid supplementation in horses is safe. Considering the higher absorption of CBDA, its use is recommended to evaluate the therapeutic efficacy of this common hemp derived cannabinoid.

Cannabidiol (CBD) and other minor cannabinoids deriving from Cannabis sativa chemovars have sparked considerable interest in the field of pet care and veterinary medicine.1 Two common cannabinoids, cannabidiolic acid (CBDA) and tetrahydrocannabinolic acid (THCA), are thermally unstable and decarboxylated to cannabidiol (CBD) or tetrahydrocannabinol (THC) during extraction and processing.1 While THC has psychoactive toxicity, CBD is a nonintoxicating compound with potential therapeutic applications via interaction with the homeostatic endocannabinoid system regulating, among others, neuronal development, nociception, and inflammation.2

Endocannabinoids and phytocannabinoids bind to specific receptors, like cannabinoid receptor 1 (CB1), mainly located in the brain and spinal cord, and cannabinoid receptor 2 (CB2) in the peripheral nervous system and immune cells.3 Cannabinoids have different affinity at the CB1 or CB2 receptor sites, either as an agonist or antagonist; for example, CBD functions as a CB1 negative allosteric modulator and CB2 partial agonist. Both CBD and CBDA also modulate non-CB1/CB2 receptors, like the peroxisome proliferator-activated receptor γ, 5-HT1A serotonin receptor, and multiple transient receptor potential channels, expressed in primary afferent nociceptors.49 In addition, CBDA has been reported as a selective cyclooxygenase-2 inhibitor.10

Due to its distinctive inverse cannabimimetic effect, CBD has gained significant interest in pharmaceutical and nutraceutical fields for both humans and animals. Studies1113 on dogs demonstrated that CBD or full-spectrum hemp extract CBD/CBDA can reduce seizure frequency in idiopathic epilepsy and mitigate osteoarthritis-related pain. The use of CBD in dogs and cats is supported by absorption and retention studies1416 that also confirmed its safety, although reversible rises in alkaline phosphatase have been reported. However, there is a noticeable scarcity of research on cannabinoids in horses.

In recent studies,1719 no adverse effects were found for both adult and senior horses following CBD IV and oral administration. Acute single-dose oral administration of CBD/CBDA did not produce neurologic, behavioral, or gastrointestinal effects in horses.20 In senior horses, the oral bioavailability for CBD is relatively low (7.92%), with both IV and oral administration showing no meaningful effects on hemogram or chemistry analyses.17 Other acute and long-term pharmacokinetic (PK) studies1721 in horses show low serum concentrations achieved at similar dosing to canine and feline studies, with rapid elimination of CBD, suggesting differences in absorption and retention of CBD in horses. In the study by Thomson et al,20 the absorption and retention of CBDA were superior to CBD, but there is currently no available research on chronic oral dosing of CBDA regarding its bioavailability and safety for adult horses.

The aim of our study was twofold: first, to measure the immediate and long-term PK of full-spectrum CBD oil versus full-spectrum, CBDA-predominant oil to determine if the absorption/retention kinetics of CBDA are superior, and second, to examine the safety of chronic administration of full-spectrum CBD and CBDA oils over a 6-week period of time recording any adverse events as observed in dogs and people.16,22,23

Methods

Study design

All experimental procedures were approved by the Cornell University IACUC (protocol #2020-0075).

Seven horses received 2 full-spectrum hemp oils (CBD and CBDA rich), each oil for a 6-week trial period, using a crossover, randomized design with a 2-week washout period. The safety of chronic administration was assessed via physical examination, periodic bloodwork, and liver biopsy at the beginning and end of the study.

A 12-hour PK analysis was conducted on day 1 of each 6-week trial period, and every 1 to 2 weeks, serum cannabinoid levels were measured prior to and at 1 and 2 hours after the morning dose to measure nadir and peak concentrations.

Animals

Seven adult female horses (3 Warmbloods, 2 Quarter Horses, and 2 mixed breeds; median age, 25 years; range, 12 to 28 years; median body weight [BW], 566 kg; range, 434 to 649 kg) of the Cornell University teaching herd were enrolled in the study. Each horse received, in computer-generated, randomized order (https://www.random.org), CBD- and CBDA-based oils (ElleVet Sciences LLC) for 6 weeks each at a dose of 1 mg/kg BW cannabinoids every 12 hours, administered orally. The oil was administered orally via a catheter tip syringe. No feeding restriction was implemented before or after the oil administration. The cannabinoid oil dose was calculated based on the horse’s BW on day 0 of each trial period. A washout period of 2 weeks separated the 2 trial periods. During the study trials, horses were housed in single-box stalls with daily turnout on a dry lot, had free access to hay and water, and were supplemented with a pelleted complete feed provided at 0.5% of their BW. During the washout period, horses were moved to pasture.

Cannabinoid products

Two cannabis extracts were tested: a CBD-rich oil and a CBDA-rich oil (ElleVet Sciences LLC). Each milliliter of CBD-rich oil contained 59.6 mg cannabinoids with the following profile: 99.2% CBD, 0.15% cannabidivarin, 0.02% Δ9-THC, and 0.01% cannabinol. Each milliliter of CBDA-rich oil contained 66.5 mg cannabinoids with the following profile: 83.5% CBDA, 12.3% CBD, 1.18% cannabigerolic acid, 0.5% cannabigerol, 0.16% cannabidivarin, 0.06% cannabinol, 0.02% D9-THC, and 0.01% THCA. The carrier ingredients consisted of sesame oil and lecithin. The components and concentration of each oil were confirmed by third-party analysis via liquid chromatography by a certified International Organization for Standardization and International Electrotechnical Commission 17025 laboratory (ProVerde Laboratory) at the beginning and end of the study, with less than 5% variability in cannabinoid results, and deemed safe for oral consumption.

Pharmacokinetics sample collection

Horses underwent a 12-hour PK assessment with blood draws at time 0, 30 minutes, and 1, 2, 4, 8, and 12 hours, with twice-a-day dosing starting immediately after the 12-hour PK sampling. Blood was drawn by direct venipuncture of the left jugular vein using an 18-gauge vacutainer needle in a red-top coagulation tube and allowed to clot. Serum was obtained after centrifugation at 2,000 X g for 10 minutes and immediately frozen at −80 °C. Serum was also obtained prior to and 1 and 2 hours after the morning dose at the end of weeks 1, 2, 4, and 6 (days 7, 14, 28, and 42) of cannabinoid oil administration to measure nadir and peak concentrations.

Serum cannabinoid analysis

Serum analysis of cannabinoids, including CBD, CBDA, THC, THCA, and their metabolite 7-COOH-CBD, was performed by LC-MS-MS (Nexera X2 and LCMS 8050; Shimadzu Corp) at the University of Illinois at Chicago Toxicology Research Laboratory as previously described.20,24 Reference standards for CBD and CBDA were obtained from Restek Corporation, whereas other reference and internal standards were obtained from Cerilliant Corporation. Concentrations of cannabinoids were calculated by LabSolutions software (version 5.65, Shimadzu Corp) using a quadratic calibration curve with 1/c2 weighing based on relative response (peak area of cannabinoids/peak area of internal standards). The calibration curve range and corresponding lowest limit of quantification and upper limit of quantification were respectively 1 ng/mL to 1,000 ng/mL for CBD, CBDA, and THC; 0.25 ng/mL and 1,000 ng/mL for THCA; and 1 ng/mL and 5,000 ng/mL for 7-COOH-CBD. Lower limits of detection for CBD, CBDA, THC, THCA, and 7-COOH-CBD were 0.5 ng/mL, 0.5 ng/mL, 0.5 ng/mL, 0.1 ng/mL, and 0.5 ng/mL, respectively.

Pharmacokinetic analysis

Serum concentrations of the various analytes versus time data for each horse were plotted on linear and semilogarithmic graphs for analysis. Data were subjected to a noncompartmental PK analysis using commercially available software (PK Solutions, version 2.0; Summit Research). Data determined for each horse included maximum serum concentration (Cmax), time of Cmax (Tmax), elimination half-life (T1/2), area under the serum concentration curve (AUC0–12h), and mean residence time (MRT). Based on data from a prior study,20 the known peak of absorption was between 1 and 2 hours of administration. Therefore, blood was collected on days 7, 14, 28, and 42 to assess serum concentrations on horses immediately before the morning dose and at 1 and 2 hours after administration to assess nadir and peak concentrations over the 42-day trial.

Safety monitoring

The health of the horses was assessed daily via physical examinations and monitoring for eventual changes in behavior and appetite. A CBC and serum chemistry panel were performed on days 0, 7, 14, 28, and 42 of each treatment trial. The CBCs were obtained from EDTA-anticoagulated blood with an automated hematology analyzer (Element HT5; Heska) and included WBC count with differential count of the individual leucocytes, RBC count and mass, hemoglobin, RBC indices and size variation, and platelet count and size. Serum chemistry measured sodium, potassium, chloride, bicarbonate, urea nitrogen, creatinine, calcium, phosphate, magnesium, total protein, albumin, globulin, glucose, AST, sorbitol dehydrogenase, glutamate dehydrogenase, GGT, total bilirubin, direct bilirubin, indirect bilirubin, creatine kinase, iron, total iron-binding capacity, and iron saturation. Biochemical analysis was performed by the Cornell Animal Health Diagnostic Center Clinical Pathology Laboratory on plasma (from heparin-anticoagulated blood) with an automated chemistry analyzer (Hitachi Modular P; Roche Diagnostics) using the manufacturer’s reagents, with the exception of sorbitol dehydrogenase (Sigma-Aldrich) and glutamate dehydrogenase (Randox Laboratories Ltd).

Liver biopsy

Ultrasonography and biopsy of the liver were performed 1 week prior to starting the oil administration and the day after the last dose (day 99). The horses were sedated with detomidine (0.01 mg/kg) and butorphanol (0.01 mg/kg), IV. A liver ultrasound (2-to-9-MHz curved array transducer; Samsung HS 60; Samsung Healthcare) evaluated the structure and echogenicity on both the left and right side of the abdomen. After the identification of a suitable window to perform the biopsy, the area was clipped and prepared for sterile procedure, followed by a 2% lidocaine injection to anesthetize the skin, SC tissue, and intercostal muscle.

Percutaneous biopsies of the liver were obtained under ultrasound guidance using a standard Tru-Cut biopsy needle (10 cm, 14 gauge; JorVet Jorgensen Laboratories). The specimens were fixed in 10% neutral-buffered formalin for at least 24 hours, then processed routinely and paraffin embedded. Sections, 6 μm in thickness, were stained with H&E, Masson trichrome, and periodic acid–Schiff for routine light microscopy. The sections were examined by a board-certified pathologist and scored for the presence of various pathologic features, including portal inflammation, lobular inflammation, individual necrotic hepatocytes, pigment accumulation, ductular reaction, cell swelling, and fibrosis, as previously described.25 Portal tract inflammation was defined as the presence of more than 5 inflammatory cells within a portal stroma. Lobular inflammation was defined as the presence of aggregates of 2 or more inflammatory cells within hepatic cords. Hepatocytes were considered necrotic if showing hypereosinophilic cytoplasm, nuclear changes (pyknosis, karyolysis, karyorrhexis), and cellular fragmentation, whereas cell swelling was defined as hepatocytes greater than 2 times the size of adjacent representative hepatocytes. Ductular reaction was characterized as proliferation of small, double-wide cords of bipotential progenitor cells breaching the limiting plate and extending into adjacent hepatic parenchyma. Fibrosis was present if immature or mature collagen bands expanded within portal stroma. For each pathologic feature, an individual score of 1 was applied if detected and 0 if absent; a total biopsy score, ranging from 0 (no detectable abnormalities) to 7 (presence of all the pathologic features), was calculated by summing the scores of each pathologic feature. The biopsy grading was performed without knowledge of the timing of collection (pre- and post-CBD/CBDA chronic administration).

Statistical analysis

Pharmacokinetic parameters, including Cmax, Tmax, T1/2, AUC0–12h, and MRT, were assessed for normality of distribution using the Shapiro-Wilk test. Only T1/2 and MRT were normally distributed (P = .241; P = .353); therefore, all PK parameters were log transformed prior to statistical analysis. Differences between treatment groups (CBD and CBDA) were determined by ANOVA using a mixed-effect model, including trial period, treatment, and treatment by trial period interaction as fixed effects and horse as a random effect. Differences between treatments were considered significant at P < .05 and tendency for difference at P < .1. Additional PK parameters for THCA were reported, and partial PK parameters for metabolite 7-COOH-CBD (Cmax, Tmax) were summarized and presented as mean ± SD.

The statistical model to determine the difference between nadir and peak serum concentration of CBD and CBDA at different time points (days 7, 14, 21, 28, and 42) included trial period, treatment, and day as fixed effects and horse nested within treatment as random effect. A covariance structure was added to ensure that the correlation between observations within the same group were constant and equal across all groups. Differences were tested using ANOVA. For 7-COOH-CBD nadir and peak concentrations, the mean ± SE was calculated.

For CBC and serum chemistry data, a Kruskal-Wallis test followed by a post hoc Dunn test with Bonferroni correction was performed to evaluate the changes in parameters over the study time. The Wilcoxon matched pairs test was used to compare the histopathology score of the liver biopsy pre- and postcannabinoid administration. The statistical analysis of PK parameters, nadir and peak serum concentrations of cannabinoids, CBC, and chemistry was performed using RStudio, version 4.3.2 (RStudio PBC). Histopathology scores are reported as mean ± SD, and analysis was conducted using GraphPad Prism, version 9 (GraphPad Software Inc).

Results

Pharmacokinetics

Pharmacokinetics data for CBD and CBDA following oral dosage are presented across the 2 trial periods as there was no treatment by trial period interaction for most parameters (Table 1). Compared to CBD, over the 2 periods, CBDA exhibited a greater Cmax (103.10 ± 87.56 ng/mL vs 12.01 ± 6.17 ng/mL; P < .001) and AUC0–12h (258.50 ± 213.80 ng/h/mL vs 61.67 ± 39.22 ng/h/mL; P < .02) and a lower Tmax (0.79 ± 0.27 hours vs 1.86 ± 0.38 hours; P < .001) and T1/2 (2.82 ± 1.48 hours vs 4.12 ± 1.37 hours; P = .018). The greater AUC0–12h value for CBDA was attributed to trial period 1. In trial period 1, the Cmax was 173.3 ± 84.33 ng/mL and 12.11 ± 8.36 ng/mL, respectively, for CBDA and CBD (P = .025). In trial period 2, the Cmax did not differ between treatments (50.47 ± 44.47 ng/mL for CBDA and 11.88 ± 3.06 ng/mL for CBD; P = .148). Mean residence time did not differ between CBDA and CBD (4.37 ± 2.14 hours vs 4.30 ± 1.19 hours; P = .122).

Table 1

Twelve-hour pharmacokinetics of cannabidiol (CBD) and cannabidiolic acid (CBDA) in horses administered 1 mg/kg of CBD- or CBDA-rich hemp oils over 2 trial periods.

Treatment
Parameter CBD CBDA Treatment P valuea Period P valuea Period X treatment P valueb
n 7 7
Cmax (ng/mL) 12.01 ± 6.17 103.10 ± 87.56 < .001c .682 .148
Tmax (h) 1.86 ± 0.38 0.79 ± 0.27 < .001c .288 .756
T1/2 (h) 4.12 ± 1.37 2.82 ± 1.48 .018c .852 .762
AUC0–12h (ng·h/mL) 61.67 ± 39.22 258.50 ± 213.80 .020c .523 .089
MRT (h) 5.20 ± 1.57 3.93 ± 1.66 .122 .153 .192

Values reported are mean ± SD.

AUC0–12h = Area under the serum concentration curve from 0 to 12 hours. Cmax = Maximum serum concentration. MRT = Mean residence time. T1/2 = Elimination half-life. Tmax = Time of Cmax.

a

Based on log-transformed data. Mixed-effects model with fixed effects for treatment and random effects for horses.

b

Based on log-transformed data. Mixed-effects model with fixed effects for treatment and period and random effects for horses.

c

Differs from CBD at P < .05.

Tetrahydrocannabinolic acid was measurable for 12-hour PK assessment following typical PK modeling, whereas 7-COOH-CBD peaked between 4 and 12 hours and plateaued afterwards, not allowing for calculations of all PK parameters (Table 2). The CBD metabolite 7-COOH-CBD showed a higher Cmax (224.6 ± 146.67 ng/mL vs 19.88 ± 9.74 ng/mL) after CBD-rich oil administration compared to CBDA-rich oil (P = .016).

Table 2

Twelve-hour pharmacokinetics of tetrahydrocannabinolic acid (THCA) and 7-COOH-CBD in horses administered 1 mg/kg of CBDA- and CBD-rich hemp oils.

CBDA CBD
Parameter THCAa 7-COOH-CBDb 7-COOH-CBDb
n 7 7 7
Cmax (ng/mL) 17.16 ± 16.47 19.98 ± 9.74 224.6 ± 146.67c
Tmax (h) 0.86 ± 0.56 10.86 ± 1.96 9.71 ± 3.15
T1/2 (h) 6.47 ± 3.03 NA NA
AUC0–12h (ng·h/mL) 67.33 ± 51.51 NA NA
MRT (h) 8.73 ± 3.09 NA NA

Values reported are mean ± SD.

a

THCA was below the quantitation level after CBD-rich hemp oil administration.

b

Partial pharmacokinetics data.

c

7-COOH-CBD maximal concentration was higher after CBD-rich hemp oil administration compared to CBDA-rich hemp oil (P < .05).

Chronic dosing peak and nadir

Nadir and peak values for CBD, CBDA, and 7-COOH-CBD over the 42-day trial period were summarized as mean ± SE (Tables 3 and 4). For CBD, day was not significant except for a trend for higher peak serum concentration on day 7 (P = .055); peak concentrations were lower on days 14 (23.60 ± 3.57 ng/mL; P = .045) and 42 (22.30 ± 3.36 ng/mL; P = .021) compared to day 7 (38.10 ± 5.74 ng/mL). Regarding CBDA treatment, a significant effect of day was found across the 2 periods of treatment (P = .034); peak concentration was lower on day 42 (68.70 ± 10.37 ng/mL) compared to days 14 (122.30 ± 18.45 ng/mL; P = .011) and 28 (117.70 ± 17.77 ng/mL; P = .020; Figure 1). For both CBD and CBDA, nadir concentrations did not differ between days 7, 14, 28, and 42 (P = .868). There were no changes in nadir and peak concentrations for 7-COOH-CBD in both CBD and CBDA treatments.

Table 3

Nadir and peak serum concentrations (nanograms per milliliter) of horses administered 1 mg/kg of CBD- or CBDA-rich hemp oils over both 42-day trial periods.

CBD CBDA
Day Nadir Peak Nadir Peak
7 16.70 ± 3.07a 38.10 ± 5.74a 46.00 ± 8.44a 90.40 ± 13.64a,b
14 13.90 ± 2.55a 23.60 ± 3.57b 52.30 ± 9.60a 122.30 ± 18.45a
28 16.50 ± 3.03a 26.30 ± 3.97a,b 65.50 ± 12.02a 117.70 ± 17.77a
42 12.60 ± 2.31a 22.30 ± 3.36b 53.70 ± 9.86a 68.70 ± 10.37b

Values reported are mean ± SE.

a,b

Within each column, values with different superscripts are significantly different (P < .05).

Trial period was not significant except for peak serum concentration on day 7 for CBD (P = .055) and day 14 for CBDA (P = .034).

Table 4

Nadir and peak serum concentrations (nanograms per milliliter) of 7-COOH-CBD in horses administered 1 mg/kg of CBD- or CBDA-rich hemp oils over both 42-day trial periods.

CBD CBDA
Day Nadir Peak Nadir Peak
7 1,531.60 ± 263.68 1,787.61 ± 304.01 376.89 ± 32.57 386.34 ± 40.37
14 1,804.05 ± 339.62 1,961.27 ± 384.80 519.75 ± 36.87 524.36 ± 45.32
28 1,736.70 ± 311.70 1,840.38 ± 298.27 675.00 ± 130.94 678.76 ± 137.00
42 1,606.14 ± 228.97 1,700.95 ± 223.97 545.74 ± 59.15 543.63 ± 55.90

Values reported are mean ± SE.

Figure 1
Figure 1

Serum concentrations of cannabidiol (CBD) and cannabidiolic acid (CBDA) during 6 weeks of oral supplementation at 1 mg/kg every 12 hours in 7 horses. The graph illustrates the 12-hour pharmacokinetic after the first dose, followed by peak concentration at 7, 14, 28, and 42 days of chronic administration. Data are reported as mean ± SD, with the y-axis logarithmically scaled.

Citation: American Journal of Veterinary Research 2025; 10.2460/ajvr.24.08.0235

Complete blood count and serum chemistry

None of the horses demonstrated behavioral changes or complications related to the administration of the hemp extracts. One horse developed a traumatic injury on the right hind on day 28 of the first treatment trial while under CBD treatment, and a second horse developed cellulitis of the left hind on day 18 of the second treatment trial (CBDA); both horses recovered uneventfully within a few days.

The CBC at different time points (days 7, 14, 21, 28, and 42) following CBD and CBDA administration showed no clinically relevant changes (Supplementary Tables S1 and S2). One horse had a platelet count below the reference range on the automated hemogram at every time point, but smear evaluation by a board-certified pathologist confirmed platelet clumping and adequate platelet estimation.

The blood chemistry profile showed no significant changes over time for parameters reflecting renal, liver, and muscle function (Supplementary Tables S3 and S4). Electrolytes and mineral homeostasis were maintained over time except for the chloride, which had a small increase on day 28 compared to day 1 (103.30 ± 1.38 mEq/L vs 101.10 ± 1.46 mEq/L; P = .041) during CBD treatment and a mild increase during treatment with CBDA-rich oil on days 14 (103.7 ± 1.38 mEq/L; P = .0072), 28 (103.40 ± 0.53 mEq/L; P .0235), and 42 (103.10 ± 1.22 mEq/L; P = .023).

Liver biopsy

The liver could be visualized on both the right and left side only in 5 of 7 horses (71%), whereas in 2 horses it could be visualized only on the left side. No ultrasonographic abnormalities in parenchymal structure and echogenicity or within the biliary tract were detected at time 0. The biopsy was performed on the right side of 5 horses and on the left side of 2 horses. Ultrasound was repeated after the procedure to evaluate potential bleeding; no significant complications developed after the biopsies besides a small SC hematoma in 2 horses that resolved spontaneously within 48 hours. Ultrasonographic evaluation of the liver at the end of the study showed no appreciable changes in liver structure, echogenicity, and biliary tract compared to time 0.

Histopathologic evaluation of the liver biopsy revealed overall no evidence of significant inflammation or necrosis at the end of the study (Figure 2). No signs of ductular reaction or fibrosis were found in any of the biopsies. Two horses had mild sinusoidal neutrophilia at the beginning of the study and one of them also at the end. The main changes observed, already at time 0 in 6 of 7 horses, were vacuolation of hepatocytes, initially attributed to glycogen but not confirmed on periodic acid–Schiff stain. Some features scored lower in the biopsy at the end of the study. No significant difference was found between pre- and poststudy total histologic score, which was 2.57 ± 1.27 before starting cannabinoid administration and 2.14 ± 1.07 at the end of the study.

Figure 2
Figure 2

Representative photomicrographs of liver biopsy before (T0) and after (study end) chronic administration of CBD- and CBDA-rich oil in horses. Images are from 1 single horse that was representative of the series. All sections were evaluated and assigned a histologic score based on the presence of histopathologic features (0 through 7, where 0 = no detectable abnormalities and 7 = presence of all the pathologic features). At T0 (A, C, and E), pigment accumulation and cell swelling were detected (total score, 2/7); at the end of 12 weeks of hemp oil administration, no significant changes were observed except for signs of mild lobular inflammation (score, 3/7). A and B—H&E stain; scale bar = 100 µm. C and D—Masson trichrome stain; scale bar = 100 µm. E and F—Periodic acid–Schiff stain; scale bar = 100 µm.

Citation: American Journal of Veterinary Research 2025; 10.2460/ajvr.24.08.0235

Discussion

The objective of this study was to assess the PK of CBD and CBDA and their safety over a 6-week oral administration period. A recent study suggested that CBDA, when present with full hemp extract, provides a natural vehicle to enhance the plasma absorption of cannabinoids, and many species appear to preferentially absorb CBDA when compared to CBD in full-spectrum hemp extracts.11,26,27 We confirmed a higher absorption of CBDA compared to CBD in horses, maintained during chronic administration.

Compared to a previous PK study19 in horses administered the same dosage of CBD at 1 mg/kg BW, CBD had a greater Cmax (12.01 ± 6.17 ng/mL vs 4.3 ± 2.1 ng/mL); lower Tmax (1.86 ± 0.38 hours vs 4.1 ± 4.1 hours), T1/2 (4.12 ± 1.37 hours vs 14.8 ± 4.6 hours), and MRT (5.20 ± 1.57 hours vs 13.5 ± 2.3 hours); and a similar AUC0–12h (61.67 ± 39.22 ng/h/mL vs 51.6 ± 21.1 ng/h/mL). These differences in Cmax, Tmax, and MRT might be caused by the variations in the CBD oil profile and the presence of minor cannabinoid compounds between studies or differences in feeding regimens between the 2 studies. In contrast, CBDA consistently exhibited a higher Cmax (103.10 ± 87.56 ng/mL vs 12.01 ± 6.17 ng/mL) and AUC0–12h (258.50 ± 213.80 ng/h/mL vs 61.67 ± 39.22 ng/h/mL), suggesting increased bioavailability and potentially altered uptake or retention compared to CBD. Comparable findings were reported with CBD/CBDA-rich, full-spectrum hemp oil at a total dose of 2 mg/kg.20 Additionally, a shorter Tmax, T1/2, and MRT were observed for CBDA, indicating its potential as a compound with more rapid onset and clearance compared to CBD.

The predicted average steady-state serum concentration based on 12-hour PK is not presented in this study because actual serum concentrations were measured periodically over the 42-day study. Cannabidiolic acid serum concentrations were greater than CBD over the 42-day study (Figure 1; Table 3), with nadir serum concentrations of both cannabinoids remaining relatively constant throughout the 6-week period. These data suggest that there is not a significant bioaccumulation of the more lipophilic CBD over time, although there is a recent study28 suggesting mild accumulation. Eichler et al28 calculated a terminal half-life of 161.29 ± 43.65 hours, with an accumulation ratio of 2.63, after a CBD oral paste trial at 3 mg/kg twice daily for 15 days. Data regarding CBDA accumulation is not currently available in horses, and in our study CBD and CBDA peak concentrations behaved differently over time. While peak serum concentration for CBD decreased after the first week, CBDA peak concentration initially increased on day 14 and later decreased to reach a minimum on day 42. The cause of the decrease in CBDA peak concentration after a few weeks is unknown but could reflect acquired changes in its absorption, distribution, or metabolism, potentially involving the induction of specific metabolizing enzymes. Cannabinoids can interact with the cytochrome P450 (CYP) enzyme system, which is present in the small intestine epithelium and responsible for drug metabolism. Indeed, CYPs are major metabolic pathways for phytocannabinoids, especially for CYP3A4, which is more abundant in the small intestine and liver.29 Several species of CYPs are inducible by increased synthesis of mRNAs coding for specific enzymes, and the induction of drug-metabolizing enzymes has been described and considered one of the factors involved in development of drug tolerance.30 This suggests that CBDA post dosing serum concentration might not remain steady by week 6, and further study is required to understand long-term dosing of CBDA and its metabolism when considering the potential for therapeutic dosing.

Compared to dogs and humans, horses exhibit lower plasma concentrations of CBD.12,16,19,31,32 It is possible that horses have greater hepatic carboxylation of CBD to 7-COOH-CBD. However, small intestinal absorption of cannabinoids via the transepithelial portal circulation was reported to be limited to 2.1% to 9.3% compared via lymphatic chylomicrons absorption in a rat study; lymphatic CBD concentrations can be up to 1,000 times higher than serum concentrations.33 If there is greater hepatic carboxylation in horses, such a process would be limited to second-pass metabolism rather than first pass. In addition, dietary fat consumption in horses is relatively low, and chylomicron transport may be limited by low dietary fat intake. Given the large abundance of squamous epithelium and a resident microbiota, CBD carboxylation via microbial metabolism in the horse stomach should be considered as a possible mechanism in addition to the intestinal CYP enzyme system. Regarding the matrix and absorption of neutral (CBD) and acidic (CBDA) cannabinoids, little work has been performed to assess protein binding or differences in delivery vehicle between the two. However, it has been shown that protein emulsification in conjunction with various forms of lipid may enhance the absorption and retention of CBD in rats, whereas CBDA appears to be found in biological fluids in humans at higher concentrations when delivered in an aqueous versus a lipid matrix.34,35 These limited studies show that there may be ways to improve the absorption of CBD and CBDA by utilizing ideal vehicles for delivery and that the study of optimal delivery is in its infancy across species.

In horses, only a few longer-term PK studies17,19,28,36 were conducted with doses of 0.5 to 3 mg CBD/kg BW. Turner et al36 measured PK over 264 hours after CBD administration of a single dose at 2 mg/kg BW in senior horses. Compared to our study, CBD appeared to have a similar Cmax (18.54 ± 9.80 ng/mL vs 12.01 ± 6.17 ng/mL) and Tmax (2.46 ± 1.62 hours vs 1.86 ± 0.38 hours), and by 72 hours after oral administration, the plasma CBD concentration was either below the limit of quantitation of the assay or nondetectable. In a consecutive study,36 the chronic use of a once-a-day oral administration of CBD oil (2 mg/kg) for 90 days was found to maintain a much lower mean CBD plasma concentration, around 3 ng/mL. In Yocom et al,19 during chronic administration of 1.5 mg/kg twice daily of oil-based CBD, only nadir steady-state plasma CBD concentration at weeks 2 and 4 were presented and are comparable to our current study. In Eichler et al,28 after CBD paste (3 mg/kg BW) oral administration twice daily for 15 days, CBD and 7-COOH-CBD serum concentrations on days 7 and 14 were comparable to our study.28

As shown in previous studies,17,20,28 7-COOH CBD is the predominant CBD metabolite in horses, with accumulation within the first week of oil administration. This finding is not surprising considering the long terminal half-life (43 ± 16 hours) reported after a single dose of CBD oil.17 From a metabolic standpoint, an interesting finding in this study was a relatively high 7-COOH-CBD serum concentration in the CBDA group during the 7-, 14-, 28-, and 42-day time points that were not evident during the acute 12-hour PK analysis. The reason for this is unclear as one would have expected some 7-COOH-CBD within the first 12 hours also in the CBDA group since the CBDA oil had approximately 12% CBD as a constituent, meaning that horses would have received approximately a 0.1-mg/kg dose of CBD from that oil. It is possible that the horses cleared the 7-COOH-CBD quickly after the acute 12-hour PK, making it undetectable while accumulation could be happening over time, or there is a decarboxylation and rapid carboxylation event occurring in the intestinal epithelium or liver that turns CBDA into 7-COOH-CBD as a metabolite. The mechanism is unclear, and further studies utilizing pure CBDA are needed.

We confirmed the safety of chronic CBD and CBDA administration in horses, ruling out adverse effects via clinical assessment and bloodwork. The CBC and chemistry profile remained stable over the 6-week study without significant effects on albumin or calcium as described in other studies.19,36,37 We detected relative increases in sodium and chloride over time without clinically relevant consequence. Considering that this electrolyte variation was not accompanied by changes in renal values or bicarbonate, the relative increase in chloride was attributed to the change in horse management. During each trial period, the horses were moved from 24 hours’ continuous access to grass at pasture to stall housing with turnout in a dry lot; removing grass from the diet potentially reduced the overall body free water with relative increases in sodium and chloride.

More importantly, in our study we detected no increase in the serum liver enzymes that have been reported as potential issues in 3 studies of horses.19,31,37 Our dosing was relatively low at 1 mg/kg for both cannabinoids, providing longer-term data that these modest doses appear safe. In view of hepatic adverse events noted in the human literature and recent work in dogs and horses reporting rises in liver enzymes, it was important to examine hepatic biopsies.19,38,39 Considering the horses’ welfare, we only performed hepatic biopsies immediately prior to and after the entire 14-week study period, including both CBD at 1 mg/kg and CBDA at 1 mg/kg twice daily. Cannabidiol use in humans at relatively high doses (above 5 mg/kg) has been associated with an increase in liver enzymes, in some cases consistent with drug-induced liver injury, and CBD-induced cellular damage of human hepatocytes has been reported in vitro.38,40,41 A dose-dependent CBD toxicity has been described in rodents with cytoplasmic swelling and centrilobular hypertrophy.37 The most common drug-induced liver injury histopathologic lesions also include cholestatic hepatitis, fibrosis, ductular reaction, portal inflammation, and necrosis.42 The liver histopathology in our study detected some cell swelling in samples collected prior to the treatment period and confirmed no significant hepatocellular changes associated with the CBD and CBDA use. Additionally, special stains for fibrosis and glycogen also confirmed that there were no negative effects on the liver with CBD and CBDA at our dosing regimen.

Products that contain CBD or hemp extract (other cannabinoids) are commonly seen in the equine market for potential alternative treatments for arthritis, pain relief, or anxiety,37 and both CBDA and CBD have been reported to be useful in the treatment of epilepsy and other neurological disorders in humans and dogs.13,23 In cows, CBDA has been shown to have positive effects on behavior and biomarkers of inflammation, piquing interest in CBDA therapeutic use in ruminants.43 In addition, CBDA at high doses has the potential to work through the cyclooxygenase-2 pathway, potentially regulating pain and modulating the inflammatory process.4,44 The pharmaceutical use of hemp extract or cannabinoids remains unexplored in horses as the efficacy and therapeutic concentrations of these compounds have not been established for any common condition in this species except for 1 case report on horse cribbing and a recent study by Interlandi et al46 examining the treatment of horses in conjunction with phenylbutazone.45,46 From very few efficacy studies it could be extrapolated, for CBD only, that a serum concentration of 102 ng/mL would be an appropriate therapeutic level for arthritis pain treatment in dogs and 54.8 to 78.9 ng/mL to control seizures in humans.12,47 The 12-hour PK and long-term administration of this study suggest that CBDA bioavailability is superior to CBD and should be further evaluated as a more physiologically pertinent cannabinoid supplement for horses. Alternatively, potentially far higher doses of CBD should be used to obtain similar responses that have been observed in other species.

Despite the significant findings, our dataset is characterized by high variance in the observed cannabinoid serum concentrations. This wide variability in the oil absorption between horses could be attributed to different factors, starting from the administration protocol. The oil was administered orally via a catheter tip syringe, without any feeding restriction, about 1 to 2 hours after pelleted feed, and while the horses had free access to hay. We cannot rule out potential partial loss of oil during administration or interference in the oil absorption due to the stomach content. The amount of forage in the gastrointestinal tract, pH variations in the gastrointestinal tract, protein binding, and enzymes specific to the equine digestive tract have already been considered factors affecting cannabinoid oral bioavailability in the horses.17 Moreover, even if the horses were healthy on physical examination and bloodwork, the group included several senior horses, whose intestinal absorption and metabolism could be affected by age-related changes in gastric secretion and small intestinal absorption as described in humans.48 In fact, a similar high variance in cannabinoid serum concentration with low bioavailability after oral administration has been already observed in senior horses.17

This study represents an initial assessment of long-term oral administration of CBD and CBDA in adult horses. The results indicate that there were no clinically significant adverse effects observed with multidose administration. Horses appeared to tolerate full-spectrum CBD or CBDA hemp oil extract well when dosed at 1 mg/kg BW. Notably, the nadir and peak serum concentrations over 6 weeks for CBD and CBDA comparison suggest a better absorption for CBDA, highlighting the need for further evaluations before providing a comprehensive guide for clinical use. Additional studies are necessary to elucidate the therapeutic benefits and determine the optimal dosage for adult horses as well as to evaluate the PK and potential adverse effects associated with longer-term dosing.

Supplementary Materials

Supplementary materials are posted online at the journal website: avmajournals.avma.org.

Acknowledgments

The authors would like to thank Stephen Parry from the Cornell Statistical Consulting Center for guidance in selecting statistical models.

Disclosures

Dr. Wakshlag and Dr. Schwark are paid consultants of ElleVet Sciences. Dr. Chevalier was an Associate Editor for AJVR. The current AJVR Editor-in-Chief, Dr Lisa Fortier, was a coprincipal investigator of this project. The other authors have nothing to disclose.

Funding

This study was funded by ElleVet Sciences.

References

  • 1.

    Aizpurua-Olaizola O, Soydaner U, Öztürk E, et al. Evolution of the cannabinoid and terpene content during the growth of Cannabis sativa plants from different chemotypes. J Nat Prod. 2016;79(2):324331. doi:10.1021/acs.jnatprod.5b00949

    • Search Google Scholar
    • Export Citation
  • 2.

    Matei D, Trofin D, Iordan DA, et al. The endocannabinoid system and physical exercise. Int J Mol Sci. 2023;24(3):1989. doi:10.3390/ijms24031989

    • Search Google Scholar
    • Export Citation
  • 3.

    Kendall DA, Yudowski GA. Cannabinoid receptors in the central nervous system: their signaling and roles in disease. Front Cell Neurosci. 2017;10:294. doi:10.3389/fncel.2016.00294

    • Search Google Scholar
    • Export Citation
  • 4.

    Ruhaak LR, Felth J, Karlsson PC, Rafter JJ, Verpoorte R, Bohlin L. Evaluation of the cyclooxygenase inhibiting effects of six major cannabinoids isolated from Cannabis sativa. Biol Pharm Bull. 2011;34(5):774778. doi:10.1248/bpb.34.774

    • Search Google Scholar
    • Export Citation
  • 5.

    Cui M, Honore P, Zhong C, et al. TRPV1 receptors in the CNS play a key role in broad-spectrum analgesia of TRPV1 antagonists. J Neurosci. 2006;26(37):93859393. doi:10.1523/JNEUROSCI.1246-06.2006

    • Search Google Scholar
    • Export Citation
  • 6.

    Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):47904805. doi:10.1111/bph.13250

    • Search Google Scholar
    • Export Citation
  • 7.

    Lu HC, Mackie K. Review of the endocannabinoid system. Biol Psychiatry Cogn Neurosci Neuroimaging. 2021;6(6):607615. doi:10.1016/j.bpsc.2020.07.016

    • Search Google Scholar
    • Export Citation
  • 8.

    Muller C, Morales P, Reggio PH. Cannabinoid ligands targeting TRP channels. Front Mol Neurosci. 2019;11:487. doi:10.3389/fnmol.2018.00487

    • Search Google Scholar
    • Export Citation
  • 9.

    Landucci E, Pellegrini-Giampietro DE, Gianoncelli A, Ribaudo G. Cannabidiol preferentially binds TRPV2: a novel mechanism of action. Neural Regen Res. 2022;17(12):26932694. doi:10.4103/1673-5374.335821

    • Search Google Scholar
    • Export Citation
  • 10.

    Takeda S, Misawa K, Yamamoto I, Watanabe K. Cannabidiolic acid as a selective cyclooxygenase-2 inhibitory component in cannabis. Drug Metab. Dispos. 2008;36(9):19171921. doi:10.1124/dmd.108.020909

    • Search Google Scholar
    • Export Citation
  • 11.

    Vega-García A, Feria-Romero I, García-Juárez A, et al. Cannabinoids: a new perspective on epileptogenesis and seizure treatment in early life in basic and clinical studies. Front Behav Neurosci. 2021;14:610484. doi:10.3389/fnbeh.2020.610484

    • Search Google Scholar
    • Export Citation
  • 12.

    Gamble LJ, Boesch JM, Frye CW, et al. Pharmacokinetics, safety, and clinical efficacy of cannabidiol treatment in osteoarthritic dogs. Front Vet Sci. 2018;5:165. doi:10.3389/fvets.2018.00165

    • Search Google Scholar
    • Export Citation
  • 13.

    Rozental AJ, Weisbeck BG, Corsato Alvarenga I, et al. The efficacy and safety of cannabidiol as adjunct treatment for drug-resistant idiopathic epilepsy in 51 dogs: a double-blinded crossover study. J Vet Intern Med. 2023;37(6):22912300. doi:10.1111/jvim.16912

    • Search Google Scholar
    • Export Citation
  • 14.

    Hunt ABG, Flint HE, Logan DW, King T. A single dose of cannabidiol (CBD) positively influences measures of stress in dogs during separation and car travel. Front Vet Sci. 2023;10:1112604. doi:10.3389/fvets.2023.1112604

    • Search Google Scholar
    • Export Citation
  • 15.

    Bradley S, Young S, Bakke AM, et al. Long-term daily feeding of cannabidiol is well-tolerated by healthy dogs. Front Vet Sci. 2022;9:977457. doi:10.3389/fvets.2022.977457

    • Search Google Scholar
    • Export Citation
  • 16.

    Deabold KA, Schwark WS, Wolf L, Wakshlag JJ. Single-dose pharmacokinetics and preliminary safety assessment with use of CBD-rich hemp nutraceutical in healthy dogs and cats. Animals (Basel). 2019;9(10):832. doi:10.3390/ani9100832

    • Search Google Scholar
    • Export Citation
  • 17.

    Turner SE, Knych HK, Adams AA. Pharmacokinetics of cannabidiol in a randomized crossover trial in senior horses. Am J Vet Res. 2022;83(9):ajvr.22.02.0028. doi:10.2460/ajvr.22.02.0028

    • Search Google Scholar
    • Export Citation
  • 18.

    Sánchez D, Medina A, Serrano-Rodríguez JM, et al. Pharmacokinetics and oral bioavailability of cannabidiol in horses after intravenous and oral administration with oil and micellar formulations. Equine Vet J. 2023;55(6):10941103. doi:10.1111/evj.13923

    • Search Google Scholar
    • Export Citation
  • 19.

    Yocom AF, O’Fallon ES, Gustafson DL, Contino EK. Pharmacokinetics, safety, and synovial fluid concentrations of single- and multiple-dose oral administration of 1 and 3 mg/kg cannabidiol in horses. J Equine Vet Sci. 2022;113:103933. doi:10.1016/j.jevs.2022.103933

    • Search Google Scholar
    • Export Citation
  • 20.

    Thomson ACS, McCarrel TM, Zakharov A, et al. Pharmacokinetics and tolerability of single-dose enteral cannabidiol and cannabidiolic acid rich hemp in horses (Equus caballus). Front Vet Sci. 2024;11:1356463. doi:10.3389/fvets.2024.1356463

    • Search Google Scholar
    • Export Citation
  • 21.

    Williams MR, Holbrook TC, Maxwell L, Croft CH, Ientile MM, Cliburn K. Pharmacokinetic evaluation of a cannabidiol supplement in horses. J Equine Vet Sci. 2022;110:103842. doi:10.1016/j.jevs.2021.103842

    • Search Google Scholar
    • Export Citation
  • 22.

    McGrath S, Bartner LR, Rao S, Packer RA, Gustafson DL. Randomized blinded controlled clinical trial to assess the effect of oral cannabidiol administration in addition to conventional antiepileptic treatment on seizure frequency in dogs with intractable idiopathic epilepsy. J Am Vet Med Assoc. 2019;254(11):13011308. doi:10.2460/javma.254.11.1301

    • Search Google Scholar
    • Export Citation
  • 23.

    Georgieva D, Langley J, Hartkopf K, et al. Real-world, long-term evaluation of the tolerability and therapy retention of Epidiolex (cannabidiol) in patients with refractory epilepsy. Epilepsy Behav. 2023;141:109159. doi:10.1016/j.yebeh.2023.109159

    • Search Google Scholar
    • Export Citation
  • 24.

    Wakshlag JJ, Schwark WS, Deabold KA, et al. Pharmacokinetics of cannabidiol, cannabidiolic acid, Δ9-tetrahydrocannabinol, tetrahydrocannabinolic acid and related metabolites in canine serum after dosing with three oral forms of hemp extract. Front Vet Sci. 2020;7:505. doi:10.3389/fvets.2020.00505

    • Search Google Scholar
    • Export Citation
  • 25.

    Jager MC, Choi E, Tomlinson JE, Van de Walle G. Naturally acquired equine parvovirus-hepatitis is associated with a wide range of hepatic lesions in horses. Vet Pathol. 2024;61(3):442452. doi:10.1177/03009858231214024

    • Search Google Scholar
    • Export Citation
  • 26.

    Rooney TA, Carpenter JW, KuKanich B, Gardhouse SM, Magnin GC, Tully TN. Feeding decreases the oral bioavailability of cannabidiol and cannabidiolic acid in hemp oil in New Zealand white rabbits (Oryctolagus cuniculus). Am J Vet Res. 2022;83(10):ajvr.22.01.0006. doi:10.2460/ajvr.22.01.0006

    • Search Google Scholar
    • Export Citation
  • 27.

    Sosa-Higareda M, Guzman DSM, Knych H, et al. Twice-daily oral administration of a cannabidiol and cannabidiolic acid–rich hemp extract was well tolerated in orange-winged Amazon parrots (Amazona amazonica) and has a favorable pharmacokinetic profile. Am J Vet Res. 2023;84(4):ajvr.22.11.0197.

    • Search Google Scholar
    • Export Citation
  • 28.

    Eichler F, Poźniak B, Machnik M, et al. Pharmacokinetic modelling of orally administered cannabidiol and implications for medication control in horses. Front Vet Sci. 2023;10:1234551. doi:10.3389/fvets.2023.1234551

    • Search Google Scholar
    • Export Citation
  • 29.

    Smith RT, Gruber SA. Contemplating cannabis? The complex relationship between cannabinoids and hepatic metabolism resulting in the potential for drug-drug interactions. Front Psychiatry. 2023;13:1055481. doi:10.3389/fpsyt.2022.1055481

    • Search Google Scholar
    • Export Citation
  • 30.

    Okey AB, Roberts EA, Harper PA, Denison MS. Induction of drug-metabolizing enzymes: mechanisms and consequences. Clin Biochem. 1986;19(2):132141. doi:10.1016/S0009-9120(86)80060-1

    • Search Google Scholar
    • Export Citation
  • 31.

    Ryan D, McKemie DS, Kass PH, Puschner B, Knych HK. Pharmacokinetics and effects on arachidonic acid metabolism of low doses of cannabidiol following oral administration to horses. Drug Test Anal. 2021;13(7):13051317. doi:10.1002/dta.3028

    • Search Google Scholar
    • Export Citation
  • 32.

    Bartner LR, McGrath S, Rao S, Hyatt LK, Wittenburg LA. Pharmacokinetics of cannabidiol administered by 3 delivery methods at 2 different dosages to healthy dogs. Can J Vet Res. 2018;82(3):178183.

    • Search Google Scholar
    • Export Citation
  • 33.

    Sitovs A, Logviss K, Lauberte L, Mohylyuk V. Oral delivery of cannabidiol: revealing the formulation and absorption challenges. J Drug Deliv Sci Technol. 2024;92:105316. doi:10.1016/j.jddst.2023.105316

    • Search Google Scholar
    • Export Citation
  • 34.

    Wang C, Dong C, Lu Y, Freeman K, Wang C, Guo M. Digestion behavior, in vitro and in vivo bioavailability of cannabidiol in emulsions stabilized by whey protein-maltodextrin conjugate: impact of carrier oil. Colloids Surf B Biointerfaces. 2023;223:113154. doi:10.1016/j.colsurfb.2023.113154

    • Search Google Scholar
    • Export Citation
  • 35.

    Perez-Acevedo A, Pacifici R, Mannocchi G, et al. Disposition of cannabinoids and their metabolites in serum, oral fluid, sweat patch and urine from healthy individuals treated with pharmaceutical preparations of medical cannabis. Phytother Res. 2021;35(3):16461657. doi:10.1002/ptr.6931

    • Search Google Scholar
    • Export Citation
  • 36.

    Turner S, Knych HK, Adams AA. The effects of cannabidiol on immune function and health parameters in senior horses. Vet Immunol Immunopathol. 2023;257:110549. doi:10.1016/j.vetimm.2023.110549

    • Search Google Scholar
    • Export Citation
  • 37.

    Leise JM, Leatherwood JL, Paris BL, et al. Evaluation of an oral supplemental cannabidiol product for acceptability and performance in mature horses. Animals (Basel). 2023;13(2):245. doi:10.3390/ani13020245

    • Search Google Scholar
    • Export Citation
  • 38.

    Lo LA, Christiansen A, Eadie L, et al. Cannabidiol-associated hepatotoxicity: a systematic review and meta-analysis. J Intern Med. 2023;293(6):724752. doi:10.1111/joim.13627

    • Search Google Scholar
    • Export Citation
  • 39.

    Corsato Alvarenga I, Wilson KM, McGrath S. Tolerability of long-term cannabidiol supplementation to healthy adult dogs. J Vet Intern Med. 2024;38(1):326335. doi:10.1111/jvim.16949

    • Search Google Scholar
    • Export Citation
  • 40.

    Gingrich J, Choudhuri S, Cournoyer P, Downey J, Jacobs KM. Review of the oral toxicity of cannabidiol (CBD). Food Chem Toxicol. 2023;176:113799. doi:10.1016/j.fct.2023.113799

    • Search Google Scholar
    • Export Citation
  • 41.

    Chen S, Li X, Wu Q, et al. Investigation of cannabidiol-induced cytotoxicity in human hepatic cells. Toxicology. 2024;506:153884. doi:10.1016/j.tox.2024.153884

    • Search Google Scholar
    • Export Citation
  • 42.

    Venkataswamy C, Thomas E, Kartikayan RK. Drug induced liver injury: a descriptive analyses of histopathological patterns and clinical outcomes. Int J Adv Med Health Res. 2024;11(1):1923.

    • Search Google Scholar
    • Export Citation
  • 43.

    Kleinhenz MD, Weeder M, Montgomery S, et al. Short term feeding of industrial hemp with a high cannabidiolic acid (CBDA) content increases lying behavior and reduces biomarkers of stress and inflammation in Holstein steers. Sci Rep. 2022;12(1):3683. doi:10.1038/s41598-022-07795-z

    • Search Google Scholar
    • Export Citation
  • 44.

    Simon LS. Role and regulation of cyclooxygenase-2 during inflammation. Am J Med. 1999;106(5B):37S42S. doi:10.1016/S0002-9343(99)00115-1

    • Search Google Scholar
    • Export Citation
  • 45.

    Cunha RZ, Felisardo LL, Salamanca G, Marchioni GG, Neto OI, Chiocchetti R. The use of cannabidiol as a novel treatment for oral stereotypic behaviour (crib-biting) in a horse. Vet Anim Sci. 2023;19:100289. doi:10.1016/j.vas.2023.100289

    • Search Google Scholar
    • Export Citation
  • 46.

    Interlandi C, Tabbì M, Di Pietro S, et al. Improved quality of life and pain relief in mature horses with osteoarthritis after oral transmucosal cannabidiol oil administration as part of an analgesic regimen. Front Vet Sci. 2024;11:1341396. doi:10.3389/fvets.2024.1341396

    • Search Google Scholar
    • Export Citation
  • 47.

    Huntsman RJ, Tang-Wai R, Alcorn J, et al. Dosage related efficacy and tolerability of cannabidiol in children with treatment-resistant epileptic encephalopathy: preliminary results of the CARE-E study. Front Neurol. 2019;10:716. doi:10.3389/fneur.2019.00716

    • Search Google Scholar
    • Export Citation
  • 48.

    Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2003;57(1):614. doi:10.1046/j.1365-2125.2003.02007.x

    • Search Google Scholar
    • Export Citation
All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 604 604 604
PDF Downloads 221 221 221
Advertisement