Pharmacology Flashcards
Investigation of bovine serum albumin-specific IgE expression in horses
-46 out of 65 healthy horses showed increased BSA-specific IgE at some point post-vaccination compared to baseline
-Horses with severe adverse vaccine reactions (“reactors”) had detectable BSA-specific IgE levels. However, the levels were not consistently higher than in healthy horses. This suggests that while BSA-specific IgE may play a role, it is not the sole determinant of adverse vaccine reactions.
-Plasma from donor horses used for commercial plasma production contained BSA-specific IgE levels comparable to healthy horses.
Index Case Insights:
-The index foal’s reaction illustrates the potential for mast cell degranulation and anaphylaxis when BSA-specific IgE interacts with circulating BSA.
-This aligns with anecdotal reports of fatal hypersensitivity reactions in similar scenarios. Potential for hypersensitivity reactions when commercial plasma with high BSA-specific IgE is administered to foals that have ingested powdered colostrum containing BSA.
Clinical Implications
-Routine vaccinations can elicit BSA-specific IgE, particularly in younger horses.
-Veterinarians should exercise caution when administering plasma to neonates that have consumed powdered colostrum products.
Short-term administration of flunixin meglumine or firocoxib does not alter viscoelastic coagulation profiles in healthy horses
Coagulation Profiles:
• Short-term administration of flunixin meglumine (nonselective NSAID) and firocoxib (COX-2–selective NSAID) did not result in clinically significant changes in viscoelastic coagulation parameters in healthy horses.
• Minor statistical differences were observed between the two NSAIDs for clot firmness parameters (A20 and MCF), but these were within reference intervals and not clinically significant.
• Traditional coagulation parameters, such as prothrombin time (PT) and partial thromboplastin time (PTT), were elevated in plasma stored during the flunixin meglumine phase, likely due to artifact.
Differences Between NSAIDs:
• Flunixin meglumine resulted in stronger clot formation (higher A20 and MCF values) compared to firocoxib.
• Both NSAIDs had minimal impact on fibrinogen levels, although a slight increase was noted following flunixin meglumine treatment.
Relevance to COX Pathways:
• The balance between COX-1 (procoagulant) and COX-2 (anticoagulant) activity was maintained during treatment, as evidenced by the lack of hyper- or hypocoagulability.
• The selective inhibition of COX-2 by firocoxib did not disrupt coagulation significantly, which contrasts with thrombotic risks associated with COX-2 inhibitors in humans.
Pharmacokinetics of single dose administration of three increasing doses of acetaminophen per os in 1-3-monthold foals
• The study successfully determined the pharmacokinetics of acetaminophen in foals aged 1 to 3 months at doses of 10, 20, and 40 mg/kg.
• Acetaminophen plasma concentrations varied significantly across doses, with a dose-dependent increase in peak plasma concentrations (Cmax) and area under the concentration-time curve (AUC).
• The median terminal half-life of acetaminophen (approximately 2.8 hours) was consistent across all doses, indicating predictable elimination kinetics.
• Peak plasma concentrations (Cmax) were achieved within 4.2 hours of administration for all doses, demonstrating rapid absorption.
• The median Cmax values were 4.4, 6.3, and 14 µg/mL for 10, 20, and 40 mg/kg doses, respectively.
• The AUC values also showed a clear dose-response relationship, increasing with higher doses.
• Despite the higher dose, plasma acetaminophen levels in foals at 20 mg/kg were lower than in adult horses reported in other studies.
• Plasma concentrations achieved with the 10 and 20 mg/kg doses may be insufficient for effective antipyretic or analgesic effects, which typically require levels of 10–20 µg/mL in humans.
• At 40 mg/kg, therapeutic concentrations (>10 µg/mL) were achieved in most foals, making it the most likely effective dose for clinical use.
• The lack of observed adverse effects suggests that these doses are safe for single administration, though repeated dosing requires further study.
Safety Profile:
• No adverse clinical, hematological, or biochemical effects were observed in foals at any dose.
n=6. Age 1-3 months
Single doses of acetaminophen up to 40 mg/kg are safe for 1–3-month-old foals, but only the highest dose is likely to achieve therapeutic plasma concentrations.
Pharmacokinetics and clinical efficacy of acetaminophen (paracetamol) in adult horses with mechanically induced lameness
Mechanically induced lameness model, n=9
• Acetaminophen at 30 mg/kg (A30) showed faster onset and greater improvement in lameness scores compared to 20 mg/kg (A20) and placebo (C).
• A30 significantly improved lameness at 2 and 4 hours post-treatment compared to placebo, demonstrating its potential as an effective analgesic.
• A30 was as effective as phenylbutazone (PB) in reducing lameness scores but showed a faster initial response.
• Heart rate was consistently lower in the A30 group compared to other treatments at various time points, indicating effective pain relief and reduced physiological stress.
Pharmacokinetics:
• Both doses of acetaminophen were rapidly absorbed, with peak plasma concentrations achieved within 0.66 hours (A20) and 0.43 hours (A30).
• A30 achieved higher peak concentrations (Cmax) and greater overall exposure (AUC) than A20, consistent with dose-proportional pharmacokinetics.
• The mean elimination half-life was longer for A30 (5.3 hours) than A20 (3.5 hours), likely reflecting slower clearance at the higher dose.
• Therapeutic plasma concentrations (>10 μg/mL) were maintained for longer durations in the A30 group, contributing to its superior clinical efficacy.
• Acetaminophen at 30 mg/kg provides significant pain relief and is suitable as a monotherapy for managing lameness in horses.
• The faster onset of action with A30 makes it advantageous for acute pain management compared to phenylbutazone.
• A therapeutic plasma concentration of 8 μg/mL was exceeded for an average of 5.8 hours (A20) and 7.3 hours (A30), supporting the efficacy of these doses.
• Higher concentrations achieved with A30 align with better clinical outcomes, suggesting the need for dose optimization in future studies.
• A30 reduced heart rate more consistently than other treatments, supporting its analgesic effect.
• Heart rate variability (HRV) data indicated a shift toward vagal tone in the A30 group, correlating with reduced pain and stress.
Pharmacokinetics, clinical efficacy, and safety of acetaminophen (paracetamol) in adult horses with naturally occurring chronic lameness
• 30 mg/kg administered orally twice daily for 21 days was safe and resulted in transient improvements in lameness in horses with naturally occurring chronic lameness.
• Plasma pharmacokinetics of APAP demonstrated rapid absorption and no drug accumulation over the 21-day dosing period.
• Tmax (time to maximum concentration) increased from day 7 to day 21, potentially due to variability in gastric emptying.
• Mean plasma protein binding of APAP was approximately 50%, higher than reported in humans.
• No detection of the hepatotoxic metabolite NAPQI, suggesting the administered dose did not overwhelm primary glucuronidation and sulfation pathways.
• Elimination half-life extended slightly by day 21 but remained consistent with previous equine studies.
Lameness Improvements:
• Subjective Evaluations: Whole-body lameness scores showed significant improvement at 2 and 4 hours post-treatment compared to untreated controls. Lamest limb scores also improved significantly, particularly in hindlimb lameness cases.
• Objective Evaluations (BMIS): Improvements in hindlimb push-off lameness were significant at 1, 2, and 8 hours post-treatment. No significant effects were observed for forelimb lameness or hindlimb impact lameness parameters, indicating variability in response depending on lameness type.
Safety Profile:
• No significant changes in liver biopsy scores, clinicopathological markers, or gastric ulcer scores were observed after 21 days of dosing.
• Mild elevations in GGT and calcium were noted but remained within reference ranges and were not clinically significant.
• Total bilirubin levels normalized, likely reflecting the resolution of fasting-related elevations at baseline.
Clinical Implications:
• Acetaminophen at 30 mg/kg twice daily offers a viable option for managing mild chronic lameness in horses.
• Effective as a short-term monotherapy but may not suffice for moderate to severe cases or long-term management.
• Higher doses may be required to achieve comparable pharmacological effects to other species due to increased protein binding and interspecies differences in metabolism.
Pharmacokinetics and safety of repeated oral dosing of acetaminophen in adult horses
PK:
• Acetaminophen was rapidly absorbed in horses, with a mean Tmax =1.35 hours after a single dose and 0.99 hours after multiple doses.
• The mean t1/2 increased from 2.78 hours (single dose) to 3.99 hours (multiple doses), suggesting a nonlinear elimination pattern over time.
• No significant drug accumulation occurred with twice-daily dosing over 14 days, as demonstrated by consistent maximum plasma concentrations (Cmax) and lack of significant increases in trough levels.
Efficacy and Therapeutic Plasma Concentrations:
• Plasma acetaminophen concentrations exceeded the therapeutic threshold for analgesia in humans (10 µg/mL) for only two hours post-administration.
• Previous studies showed that a single 20 mg/kg dose could reduce lameness for up to five hours, suggesting that horses may require lower therapeutic plasma concentrations than humans.
Safety Profile:
• The 14-day dosing regimen was generally well-tolerated, with no clinically significant changes in gastric ulcer scores or hepatic enzyme levels.
• Statistically significant but clinically insignificant changes were noted in albumin, alkaline phosphatase (ALP), total protein, calcium, and creatine kinase levels.
• Sorbitol dehydrogenase (SDH) levels decreased, and total bilirubin increased during the study but were attributed to factors such as grazing or fasting rather than acetaminophen toxicity.
Potential Causes of Observed Changes:
• The variability in absorption (Cmax and Tmax) was attributed to differences in gastric emptying and intestinal absorption among individual horses, particularly under fed conditions.
• The decrease in bioavailability (AUC) and increase in volume of distribution (Vd/F) with multiple doses could reflect changes in protein binding or drug metabolism, though further research is needed to confirm this.
Clinical Implications:
• Efficacy:
-The rapid absorption and short duration above therapeutic concentrations suggest that acetaminophen may require higher or more frequent dosing for sustained analgesic effects in horses.
-Despite the short plasma half-life, its clinical effects on lameness were reported to last longer, potentially indicating local tissue effects or a lower therapeutic requirement in horses.
• Safety:
-Repeated administration of acetaminophen at 20 mg/kg appears safe over two weeks in healthy adult horses, with no evidence of cumulative toxicity.
-Combining acetaminophen with nonsteroidal anti-inflammatory drugs (NSAIDs) may increase the risk of gastrointestinal side effects and warrants careful monitoring.
Pharmacokinetics and efficacy of orally administered acetaminophen in adult horses with experimentally induced endotoxemia
PK:
• APA) administered at 30 mg/kg PO in horses with endotoxemia exhibited reduced plasma concentrations compared to healthy horses, likely due to altered drug absorption.
• Cmax in endotoxemic horses was 13.97 µg/mL (30.02 µg/mL in healthy horses)
• Tmax remained similar between endotoxemic and healthy horses, suggesting that delayed gastric emptying was not the primary cause of reduced absorption.
• Reduced AUC in endotoxemic horses points to compromised drug absorption, potentially due to reduced gastric and intestinal perfusion.
Antipyretic Efficacy:
• APAP reduced rectal temperature significantly at 4 and 6 hours post-administration compared to placebo.
• There was no statistical difference in antipyretic efficacy between APAP and flunixin meglumine (FLU), suggesting APAP is a viable alternative for fever management in horses.
• Despite efficacy in reducing fever, APAP showed no significant effect on systemic inflammatory cytokines (IL-1β, IL-10, TNF-α).
Heart Rate Reduction:
• FLU demonstrated superior reduction in heart rate compared to APAP at 4 and 6 hours post-treatment.
• The greater effect of FLU on heart rate was attributed to its more potent COX inhibition and subsequent suppression of thromboxane A2 (TXA2), which has direct chronotropic effects.
Mechanistic Insights:
• Altered Drug Absorption in Endotoxemia:
-Decreased Cmax and AUC in endotoxemic horses likely result from reduced gastrointestinal perfusion and potential venous congestion rather than delayed gastric emptying.
-Findings align with studies in other species where endotoxemia similarly decreased absorption of orally administered drugs.
• COX Inhibition:
-FLU’s ability to reduce heart rate more effectively than APAP highlights its stronger impact on prostanoid-mediated systemic effects during endotoxemia.
• Safety Profile:
-APAP administration did not result in significant changes in hematological or biochemical parameters, indicating good tolerability.
-No adverse effects related to hepatotoxicity or gastrointestinal ulceration were observed, supporting its safety in acute administration for endotoxemic horses.
Ex vivo COX-1 and COX-2 inhibition in equine blood by phenylbutazone, flunixin meglumine, meloxicam and firocoxib
COX-1 and COX-2 Selectivity:
• Firocoxib and meloxicam demonstrated significantly less COX-1 inhibition compared to phenylbutazone and flunixin meglumine.
• All four NSAIDs exhibited comparable levels of COX-2 inhibition after multiple doses, indicating similar anti-inflammatory efficacy.
• The high COX-2 selectivity of firocoxib aligns with its reported COX-1/COX-2 inhibitory ratio of 263–643, compared to meloxicam’s lower ratio of 3.8.
Pharmacokinetics:
• Firocoxib had the longest half-life (48.7 hours) and largest volume of distribution among the studied NSAIDs.
• Phenylbutazone and flunixin meglumine displayed shorter half-lives (~6 and ~4.8 hours, respectively), reflecting their need for more frequent dosing.
• Despite short plasma half-lives, phenylbutazone and flunixin maintained COX-1 inhibition beyond expected therapeutic windows, suggesting prolonged physiological effects.
Efficacy of COX-2 Inhibition:
• Firocoxib achieved COX-2 inhibition comparable to traditional NSAIDs, supporting its use as an effective anti-inflammatory agent.
• The addition of a loading dose of firocoxib (0.3 mg/kg) is recommended to achieve therapeutic concentrations more quickly, as it does not reach steady-state until the fifth dose.
Clinical Implications:
NSAID Selection:
• COX-2 selective NSAIDs, such as firocoxib, are preferable for long-term pain management, particularly in patients at risk for gastrointestinal or renal complications.
• Traditional NSAIDs, like flunixin meglumine, may be more effective for immediate, short-term use due to their stronger initial COX-1 and COX-2 inhibition.
Safety Considerations:
• Prolonged COX-1 inhibition by phenylbutazone and flunixin poses a risk of gastrointestinal and renal side effects, especially in dehydrated or systemically compromised horses.
• Firocoxib and meloxicam are safer options for horses with pre-existing renal or gastrointestinal issues, due to reduced COX-1 inhibition.
Meloxicam ameliorates the systemic inflammatory response syndrome associated with experimentally induced endotoxemia in adult donkeys
Systemic Inflammatory Response:
• Meloxicam significantly ameliorated SIRS associated with LPS-induced endotoxemia in donkeys.
• Key clinical improvements included reduced tachycardia, faster recovery from leukopenia and neutropenia, and attenuated increases in plasma lactate levels.
• Plasma tumor necrosis factor-alpha (TNFα) and interleukin-1β (IL-1β) concentrations were significantly lower in the meloxicam-treated group compared to controls.
Ex vivo Gene Expression:
• Pro-inflammatory cytokines (TNFα, IL-1β, IL-6, and IL-8) were upregulated in both groups following LPS infusion. NSD between groups
–> Attenuation of TNFα and IL-1β release may reflect post-transcriptional modulation rather than direct suppression of gene expression.
• Anti-inflammatory IL-10 expression peaked earlier in the meloxicam group, suggesting enhanced resolution of inflammation.
Clinical Efficacy:
• Heart Rate: Meloxicam delayed the onset of tachycardia and significantly reduced mean heart rates during the acute phase of endotoxemia.
• Temperature Regulation: While fever was observed in both groups, meloxicam-treated donkeys exhibited a slower rise and earlier stabilization of body temperature.
• Gut Motility: Meloxicam preserved gut motility, which was significantly impaired in the control group.
• Biochemical Changes: Plasma lactate levels normalized faster in meloxicam-treated donkeys.
• Total solids and fibrinogen concentrations were not significantly different between groups, indicating minimal acute phase protein response during the study period
Effects of phenylbutazone, firocoxib, and dipyrone on the diuretic response to furosemide in horses
NSAID Impact on Furosemide-Induced Diuresis:
• Pretreatment with phenylbutazone (PB), firocoxib (FX), and dipyrone (DP) reduced furosemide-induced diuresis by approximately 25%, compared to furosemide alone (FU).
• The magnitude of diuretic inhibition was similar across the three NSAIDs, indicating no significant difference in their effects on renal response to furosemide.
Electrolyte Excretion:
• All NSAID treatments attenuated furosemide-induced increases in fractional sodium (FClNa) and chloride (FClCl) clearances during the first hour post-administration.
• Potassium excretion (FClK) increased after furosemide administration but was not significantly affected by NSAID pretreatment.
Interindividual Variability:
• Considerable variability in the magnitude of diuresis was observed among individual mares, both with and without NSAID pretreatment.
• Differences in urine production were less pronounced in horses with lower diuretic response to furosemide.
Clinical Implications:
Renal Function Monitoring:
• The similar inhibitory effects of all three NSAIDs suggest that even COX-2 selective (firocoxib) or atypical NSAIDs (dipyrone) may not provide “renoprotective” benefits compared to nonselective NSAIDs like phenylbutazone.
• Routine monitoring of renal function is advised when using NSAIDs in horses receiving furosemide, especially in cases with underlying dehydration or compromised renal function.
NSAID Selection:
• While dipyrone and firocoxib may offer a reduced risk of gastrointestinal toxicity, their lack of advantage in maintaining diuretic response limits their potential benefit in renal protection.
• Phenylbutazone remains effective but may pose greater risks in horses with pre-existing renal concerns.
Mechanistic Insights:
• Furosemide-induced diuresis depends on prostaglandin E2 (PGE2) production, which enhances solute delivery to the distal nephron and reduces water reabsorption.
• NSAIDs inhibit cyclooxygenase (COX) enzymes, reducing PGE2 production and thereby attenuating the diuretic and natriuretic effects of furosemide.
Effects of phenylbutazone alone or in combination with a nutritional therapeutic on gastric ulcers, intestinal permeability, and fecal microbiota in horses
9 days at 4.4mg/kg PBZ:
• increased gastric ulcer scores (1), intestinal permeability, and altered fecal microbiota.
• Increased permeability, as measured by circulating 16S rDNA, was transient, peaking on Day 54 (five days into treatment) and resolving by Day 59.
• Significant loss of beneficial genera, such as Pseudobutyrivibrio, was observed, potentially contributing to intestinal inflammation.
Nutritional Therapeutic Mitigation:
• Co-administration of a nutritional therapeutic attenuated phenylbutazone-induced increases in intestinal permeability and gastric ulcer scores.
• 6 of the 10 horses developed glandular ulceration with a mean increase of 1.1 grades. Only 1 horse in the nutritional therapeutic group developed phenylbutazone-induced glandular ulceration. The difference in glandular ulcer scores was significant in the
phenylbutazone-treated horses (P = .02) but not in the control and phenylbutazone plus nutritional therapeutic groups
• Horses receiving the nutritional therapeutic exhibited stabilization in fecal microbiota, maintaining beneficial bacteria like Pseudobutyrivibrio.
Phenylbutazone-induced GI effects likely resulted from:
• Topical effects causing intestinal epithelial cell death.
• Reduced prostaglandin (PGE2) production leading to diminished mucus secretion and mucosal protection.
• Disruption of tight junction proteins (e.g., ZO-1) via oxidative stress from reactive oxygen species (ROS), increasing permeability.
• Altered antimicrobial peptide secretion by Paneth cells.
• Loss of beneficial bacteria, such as Pseudobutyrivibrio, may exacerbate intestinal inflammation due to their role in butyrate production, a short-chain fatty acid crucial for maintaining epithelial barrier integrity.
Clinical Implications:
• Healthy adult horses showed only transient increases in GI permeability, but more severe effects could occur in compromised or young horses.
• Clinical signs of GI injury were absent, indicating potential subclinical injury in this population.
• NSAIDs like phenylbutazone pose risks for right dorsal colitis and other GI complications, emphasizing the need for protective strategies.
• The nutritional therapeutic showed potential in mitigating NSAID-induced GI effects, possibly through its components (e.g., omega-3 fatty acids, antioxidants, glutamine, prebiotics).
Pharmacokinetics and anti-inflammatory effects of flunixin meglumine as a sole agent and in combination with phenylbutazone in exercised Thoroughbred horses
• Administration of phenylbutazone (PBZ) 24 hours after flunixin meglumine (FM) significantly increased FM clearance compared to FM administered alone.
• No significant differences were noted in the elimination half-life or other pharmacokinetic parameters, except clearance.
Anti-inflammatory Effects:
• FM alone effectively inhibited both constitutively expressed (COX-1) and inducible (COX-2) cyclooxygenase enzymes, as shown by the suppression of thromboxane B2 (TXB2), prostaglandin E2 (PGE2), and prostaglandin F2 alpha (PGF2α).
• Co-administration of PBZ prolonged the suppression of TXB2 and PGE2, indicating additive or synergistic inhibition of COX enzymes.
• Leukotriene B4 (LTB4) and 5-HETE concentrations increased transiently post-FM administration
Metabolism Interaction:
• The slight increase in FM concentrations shortly after PBZ administration may result from competitive displacement from plasma protein binding sites due to PBZ’s high binding affinity.
Clinical Implications:
• The prolonged anti-inflammatory effects observed with combined FM and PBZ administration suggest potential benefits in managing severe inflammatory conditions.
• However, the enhanced COX inhibition may increase the risk of adverse effects, such as gastrointestinal or renal complications, particularly with prolonged use.
Additive Effects:
• The suppression of COX-1 (TXB2) and COX-2 (PGE2) by both FM and PBZ suggests an additive effect when the drugs are used in combination.
• Prolonged inhibition of COX-2–related mediators, such as PGE2, highlights the cumulative pharmacodynamic impact of sequential NSAID administration.
Arachidonic Acid Pathway:
• NSAID-induced COX inhibition leads to arachidonic acid being redirected to the lipoxygenase pathway, explaining the transient increase in LTB4 and 5-HETE concentrations.
Localisation of cannabinoid and cannabinoid-related receptors in the equine dorsal root ganglia
Expression of Cannabinoid and Related Receptors:
• CB1R: Observed in all sensory neurons and satellite glial cells (SGCs)
• CB2R: Expressed in 80% of neurons and SGCs
• PPARα: Detected in sensory neurons, SGCs, and vascular endothelial cells, aligning with its known roles in anti-inflammatory and neuroprotective effects.
• TRPA1: Found in sensory neurons and SGCs, with higher expression in unmyelinated nerve fibers, supporting its involvement in nociception and pain signaling.
• 5-HT1aR: Localized in sensory neurons, SGCs, and Schwann cells, indicating its participation in serotonergic modulation of nociception.
The distribution of these receptors in equine dorsal root ganglia (DRG) aligns partially with observations in humans, rodents, and dogs, though notable differences exist in receptor localization (e.g., CB1R and CB2R in SGCs).
Mechanistic Insights:
• CB1R influences nociception by modulating neurotransmitter release at sensory neuron synapses.
• CB2R, through its expression in both neurons and SGCs, is implicated in peripheral pain modulation and anti-inflammatory pathways. Activation of CB2R has demonstrated efficacy in reducing nociceptive signaling in human and rodent studies, suggesting similar therapeutic potential in equine medicine.
• PPARα: Acts as a transcription factor with rapid non-genomic effects, contributing to the anti-inflammatory and neuroprotective properties of cannabinoids. Its vascular localization suggests a role in regulating blood flow and homeostasis within the DRG.
• TRPA1: Known for mediating somatic and visceral pain responses, particularly to mechanical, chemical, and thermal stimuli.
• 5-HT1aR: Plays a critical role in serotonergic analgesia by inhibiting glutamate release and reducing pain signal transmission.
Pharmacokinetics and oral bioavailability of cannabidiol in horses
• Cannabidiol (CBD) displayed a tri-compartmental distribution profile, indicative of widespread tissue penetration.
• The volume of distribution (Vss) was large (36 L/kg), suggesting significant sequestration in tissues like fat, central nervous system, and joints.
• Clearance (Cl) was high at 1.46 L/h/kg, pointing to efficient hepatic elimination.
• The oral bioavailability of CBD was low (~14%), consistent across both sesame oil and micellar formulations.
• Micellar formulations provided faster absorption and higher peak plasma concentrations (Cmax) compared to oil formulations, but bioavailability remained similar.
Absorption and Bioavailability:
• Low oral bioavailability is attributed to CBD’s high lipophilicity, limited intestinal absorption, and extensive hepatic first-pass metabolism.
Half-life and Steady-State:
• The terminal half-life (t1/2) was approximately 24-34 hours, longer than prior studies, reflecting slow drug release from peripheral compartments.
• Simulations indicated steady-state concentrations were reached after 6-7 days with twice-daily oral administration.
Therapeutic Applications:
• Both micellar and oil-based formulations can be used for oral CBD administration, with the choice depending on desired pharmacokinetic profiles:
-Micellar formulations: Faster peak levels, suitable for acute conditions.
-Oil formulations: Sustained levels, possibly better for chronic conditions.
Mechanistic Insights:
• First-Pass Effect: Extensive first-pass metabolism by the liver contributes significantly to CBD’s low systemic availability after oral administration. The presence of major metabolites (e.g., 7-COOH CBD) in other studies supports this finding.
• Lipophilic Drug Challenges: The high lipophilicity (log P ~6.3) and low water solubility of CBD necessitate lipid-based or micellar delivery systems for improved absorption.
The pharmacokinetics of a fentanyl matrix patch applied at three different anatomical locations in horses
• Fentanyl matrix patches provided measurable plasma concentrations across all anatomical locations: inguinal abdomen (TXA), dorsal metacarpus (TXM), and ventral tail base (TXT).
• The maximum plasma concentration (Cmax) ranged from 1.55 to 2.07 ng/mL, comparable to prior studies using reservoir patches.
• Tmax was location-dependent, with TXA and TXT showing shorter times (10 hours) compared to TXM (14.3 hours).
• The area under the curve (AUC) values were smaller than those reported in studies with reservoir patches, likely due to differences in patch formulations and methodologies.
Site-Specific Absorption:
• TXA and TXT demonstrated higher plasma concentrations and greater AUC values than TXM, aligning with previous findings showing superior absorption in areas with thinner, more vascularized skin.
• Variability in absorption across locations may reflect differences in skin thickness, vascularity, and local drug penetration.
Comparison with Reservoir Patches:
• Matrix patches had lower AUC values than reservoir patches, likely due to:
-Differences in patch design and drug delivery mechanisms.
-Variability in application methodology, including preparation of the application site (e.g., shaving, cleaning).
-Potential residual drug in patches after removal, which was not quantified in this study.
Analgesic Potential:
• Plasma fentanyl levels achieved in this study (1.55–2.07 ng/mL) are consistent with concentrations reported to provide analgesia in other species.
• However, additional studies are needed to determine whether these levels provide effective pain relief in horses without adjunct NSAIDs or other therapies.
• Pain types (e.g., visceral vs. somatic) may respond differently to fentanyl, requiring tailored clinical applications.
Safety and Tolerability:
• No significant effects on heart rate, respiratory rate, or rectal temperature were observed during the study, indicating the patches were well-tolerated.
• No local adverse effects, such as erythema or irritation, were noted at any application sites, contrasting with previous studies reporting mild irritation with reservoir patches.
Plasma disposition of gabapentin after escalating intragastric doses in adult horses
PK:
• Gabapentin plasma concentrations increased with escalating doses from 10 to 160 mg/kg, but not in a proportional manner.
• AUC and Cmax increased less than dose-proportionally, indicating saturation of intestinal absorption pathways.
• Terminal half-life varied across doses, with a range of 2 to 15.7 hours, longer than previously reported in lower-dose studies (5–20 mg/kg).
Dose Proportionality:
• Plasma gabapentin concentrations did not increase linearly with higher doses, a phenomenon attributed to the saturation of intestinal transporters, similar to findings in humans and dogs.
• At doses ≥120 mg/kg, further increases in gabapentin exposure were minimal, suggesting limited additional benefit at higher doses.
Efficacy Insights:
• Simulated plasma concentrations indicated that dosing regimens of 10 mg/kg every 8 hours, 20 mg/kg every 12 hours, or 80 mg/kg every 24 hours could maintain plasma levels above the estimated effective concentration (EC50) for neuropathic pain in other species.
• Despite these pharmacokinetic insights, the lack of validated neuropathic pain models in horses limits the ability to correlate plasma levels with clinical efficacy.
Safety:
• Gabapentin was well-tolerated at all doses, with mild sedation observed in one horse at higher doses (120 and 160 mg/kg). No significant changes in physical examination parameters were noted.
• Long-term safety of gabapentin, particularly concerning hepatic and renal function, remains unverified.
Pharmacokinetics and pharmacodynamics of repeat dosing of gabapentin in adult horses
PK:
• Gabapentin administered at 40 mg/kg and 120 mg/kg every 12 hours resulted in predictable plasma drug accumulation over 14 days.
• Higher doses (120 mg/kg) produced significantly greater plasma concentrations and AUC compared to 40 mg/kg, suggesting dose-dependent absorption and accumulation.
• Median trough concentrations after repeated administration were 6.4 µg/mL (40 mg/kg) and 17 µg/mL (120 mg/kg), with the latter achieving theoretical effective analgesic concentrations (EC50 = 16.7 µg/mL).
• shows dose-dependent accumulation, consistent with saturation of renal elimination or absorption mechanisms.
Safety:
• No clinical signs of sedation or ataxia were observed in any horses at either dose during the study.
• Biochemistry profiles remained within normal limits, suggesting no adverse effects on liver or kidney function.
• healthy adult mares for 14 days
Efficacy Potential:
• The 120 mg/kg dose is more likely to achieve plasma concentrations associated with analgesia in humans and rats, making it suitable for treating equine pain.
• The 40 mg/kg dose, while safe, may require adjustment in dosing intervals to maintain therapeutic levels in some horses.
• Variability in plasma concentrations suggests the need for individualized dosing regimens based on the horse’s pharmacokinetic profile.
Effect of the p38 MAPK inhibitor doramapimod on the systemic inflammatory response to intravenous lipopolysaccharide in horses
• Doramapimod significantly reduced the systemic inflammatory response to LPS infusion in horses.
• The drug decreased heart rate, rectal temperature, blood pressure changes, leukocyte depletion, and cytokine production (TNF-α and IL-1β) following LPS administration.
• Peak cytokine levels were reduced, and the total inflammatory burden, as measured by the AUC for TNF-α and IL-1β, was significantly lower in the doramapimod-treated group.
Mechanism of Action:
• Doramapimod inhibits the p38 MAPK pathway, a key regulator of inflammatory cytokine transcription and post-transcriptional stabilization.
• This mechanism directly targets the upstream activation of pro-inflammatory mediators, offering broader anti-inflammatory potential compared to traditional NSAIDs, which act further downstream.
Safety:
• Doramapimod was well tolerated in healthy horses, with no observed adverse effects on clinical parameters or hematological and biochemical profiles.
• The drug’s safety profile supports its potential for short-term use in systemic inflammatory conditions, such as SIRS in equine patients.
• Short-term Use: Doramapimod’s effectiveness in reducing inflammation within hours of administration suggests its utility for acute inflammatory crises. The transient nature of cytokine suppression aligns with the critical time frame for intervention in acute SIRS cases.
• Doramapimod could complement existing treatments such as NSAIDs or polymyxin B in managing endotoxemia or other inflammatory conditions.
Safety and efficacy of subcutaneous alpha-tocopherol in healthy adult horses
• Subcutaneous administration of the 500 IU/mL alpha-tocopherol formulation significantly increased serum and cerebrospinal fluid (CSF) concentrations compared to the 600 IU/mL formulation and oral administration.
• Serum alpha-tocopherol levels exceeded normal reference ranges within 24–48 hours of subcutaneous administration in Phase 2, suggesting effective systemic absorption.
• All horses developed notable tissue swelling at the injection site, regardless of the product concentration. One horse experienced persistent swelling that required surgical drainage, and histology confirmed sterile granulomatous inflammation.
• The extent of tissue reaction limits the clinical applicability of the subcutaneous formulation, despite its efficacy in raising vitamin E levels.
• Muscle concentrations were highly variable and showed no statistically significant increase post-administration, indicating preferential distribution to other tissues.
• Oral supplementation achieved a modest increase in serum alpha-tocopherol levels, but CSF concentrations remained suboptimal.
• Subcutaneous administration resulted in faster and more substantial increases in both serum and CSF alpha-tocopherol levels, making it potentially beneficial for urgent correction of deficiency.
• Subcutaneous alpha-tocopherol could serve as a rapid intervention for severe vitamin E deficiency, particularly in horses with conditions affecting oral absorption.
• Its application may be considered for equine neurological diseases where increasing CSF concentrations quickly is critical.
• The 500 IU/mL formulation is preferable to the 600 IU/mL formulation due to its higher efficacy and potentially reduced local irritation.
• Oral administration remains the safest option for long-term supplementation but may not suffice for rapid correction in acute cases.
Antimicrobial susceptibility of bacterial isolates from ambulatory and referral practices
• Resistance rates were significantly higher in isolates from referral hospitals compared to ambulatory practices.
• Penicillin-Gentamicin (P-G) sensitivity: 91% of ambulatory isolates were sensitive, compared to only 64% of referral hospital isolates.
• Trimethoprim-Sulfamethoxazole (TMPS) sensitivity: 82% of ambulatory isolates were sensitive, compared to 56% of referral hospital isolates.
• No second-line antimicrobial showed consistently high efficacy for isolates resistant to first-line antimicrobials.
• Referral hospital isolates had the lowest sensitivity rates to second-line antimicrobials:
-Tetracycline: 8%
-Ceftiofur: 29%
-Enrofloxacin: 40%
The higher resistance rates in referral hospitals are likely due to:
• Greater antimicrobial selection pressure.
• Increased transmission of resistant strains in hospital settings.
• Higher stress levels in hospitalized horses, leading to increased shedding of resistant bacteria.
Effects of corn oil on ponazuril serum and CSF concentrations in horses
Co-administration of ponazuril with corn oil (PONOIL) resulted in significantly higher serum and cerebrospinal fluid (CSF) concentrations compared to ponazuril alone (PON).
• Steady-state serum concentrations for PONOIL (6.2 ± 0.9 mg/L) were higher than PON (4.5 ± 1.0 mg/L; P = .004).
• Steady-state CSF concentrations for PONOIL (0.213 ± 0.04 mg/L) exceeded those for PON (0.162 ± 0.04 mg/L; P = .03).
• CSF ponazuril concentrations were not significantly higher on Day 7, despite higher serum concentrations, suggesting delayed equilibration between serum and CSF.
• Higher serum concentrations on Days 14 and 21 directly correlated with increased CSF levels, indicating that serum concentrations drive CSF accumulation.
• Substantial inter-horse variability in both serum and CSF concentrations was observed, especially in the PON group.
• Some horses achieved CSF concentrations close to or below the effective inhibitory concentration for Sarcocystis neurona, highlighting variability in pharmacokinetics.
Enhanced Absorption:
• Corn oil enhances ponazuril absorption due to the drug’s high lipophilicity, improving bioavailability and systemic concentrations.
• Higher serum concentrations translate into greater CSF penetration, driven by passive diffusion across the blood-brain barrier (BBB).
Delayed CSF Equilibration:
• The time to achieve maximal CSF concentrations may reflect slower BBB crossing and compartmental distribution dynamics of ponazuril.
Evaluation of safety, humoral immune response and faecal shedding in horses inoculated with a modified-live bovine coronavirus vaccination
• The modified-live bovine coronavirus (BCoV) vaccine was safe to administer via oral, intranasal, and intrarectal routes.
• No clinical signs of enteric coronavirus infection (e.g., lethargy, fever, diarrhea, or colic) were observed in vaccinated horses.
• No evidence of vaccine virus shedding in feces supports the hypothesis that the vaccine virus did not replicate in the equine intestinal tract.
• Viral shedding was detected in nasal secretions of two intranasally vaccinated horses within 24 hours post-vaccination, attributed to residual vaccine virus rather than active replication.
• Absence of viral shedding in feces across all groups indicates minimal risk of environmental contamination.
• Seroconversion occurred in only 27% of vaccinated horses, with one seroconversion from each vaccination route.
-The limited humoral response may result from inadequate replication of the vaccine virus in equine cells, the low antigenic mass in the vaccine, or pre-existing cross-reactive antibodies to equine coronavirus (ECoV).
Immune Activation:
• The study aimed to bypass gastric degradation by testing alternative administration routes (e.g., intrarectal), hypothesizing improved mucosal immunity. However, the serological response remained limited.
• Prior evidence from foal studies indicates that intrarectal vaccination may enhance cell-mediated immunity, but this was not assessed here.
• BCoV’s close antigenic relationship to ECoV makes it a suitable model for vaccine testing, but the limited replication of the modified-live BCoV strain in equine tissues may have reduced its immunogenicity.