1
|
Wu X, Chen Q, Chou WC, Maunsell FP, Tell LA, Baynes RE, Davis JL, Jaberi-Douraki M, Riviere JE, Lin Z. Development of a physiologically based pharmacokinetic model for flunixin in cattle and swine following dermal exposure. Toxicol Sci 2025; 203:181-194. [PMID: 39475069 DOI: 10.1093/toxsci/kfae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
Flunixin meglumine is a nonsteroidal anti-inflammatory drug (NSAID). Banamine Transdermal is a pour-on formulation of flunixin approved for pain control in beef and dairy cattle, but not for calves and some classes of dairy cattle or swine. Violative flunixin residues in edible tissues in cattle and swine have been reported and are usually attributed to non-compliant drug use or failure to observe an appropriate withdrawal time. This project aimed to develop a physiologically based pharmacokinetic (PBPK) model for flunixin in cattle and swine to predict withdrawal intervals (WDI) after exposures to different therapeutic regimens of Banamine Transdermal. Due to the lack of comprehensive skin physiological data in cattle, the model was initially developed for swine and then adapted for cattle. Monte Carlo simulation was employed for population variability analysis. The model predicted WDIs were rounded to 1 and 2 d for liver and muscle in cattle, respectively, under FDA tolerance levels, while under EU maximum residue limits, the WDIs were rounded to 1, 3, 2, and 2 d for liver, kidney, muscle, and fat, respectively, following a labeled single transdermal 3.3 mg/kg dose in cattle. The model was converted into a user-friendly interactive PBPK (iPBPK) interface. This study reports the first transdermal absorption model for drugs in cattle. This iPBPK model provides a scientifically based tool for the prediction of WDIs in cattle and swine administered with flunixin in an extra-label manner, especially by the transdermal route.
Collapse
Affiliation(s)
- Xue Wu
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, United States
| | - Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, United States
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, United States
| | - Fiona P Maunsell
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, United States
| | - Lisa A Tell
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, United States
| | - Ronald E Baynes
- Department of Population Health and Pathobiology, Center for Chemical Toxicology Research and Pharmacokinetics, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, United States
| | - Jennifer L Davis
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24060, United States
| | - Majid Jaberi-Douraki
- Department of Mathematics, College of Arts and Sciences, Kansas State University, Manhattan, KS 66506, United States
- 1Data Consortium, Kansas State University, Olathe, KS 66061, United States
| | - Jim E Riviere
- Department of Population Health and Pathobiology, Center for Chemical Toxicology Research and Pharmacokinetics, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, United States
- 1Data Consortium, Kansas State University, Olathe, KS 66061, United States
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, United States
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
2
|
Diez R, Rodriguez JM, Lopez C, de la Puente R, Sierra M, Diez MJ, Fernandez N, Garcia JJ, Sahagun AM. Pharmacokinetic Pattern of Menbutone in Calves after Single Intravenous and Intramuscular Administration. Animals (Basel) 2024; 14:2540. [PMID: 39272325 PMCID: PMC11393952 DOI: 10.3390/ani14172540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Menbutone is a choleretic agent currently used in Europe to treat digestive disorders in livestock and dogs. Pharmacokinetic parameters were established in 4-month Holstein calves after intravenous (IV) and intramuscular (IM) administration. The drug was administered to 12 animals (10 mg/kg) for both IV and IM routes following a crossover design. Plasma samples were collected at various time points over 24 h and analyzed by reverse-phase high-performance liquid chromatography with a photodiode-array detector, following a method validated according to European Medicines Agency guidelines. Pharmacokinetic parameters were calculated using compartmental and non-compartmental methods. Menbutone followed a two-compartment open model after IV injection, with a total clearance (Cl) of 71.9 ± 13.5 mL/h/kg, an elimination half-life (t½β) of 4.53 ± 2.45 h, and a volume of distribution at steady-state (Vss) of 310.4 ± 106.4 mL/kg. Non-compartmental elimination half-life (t½λ) was 4.2 ± 1.1 h. After IM administration, drug pharmacokinetics was best described by a one-compartment open model. The peak plasma concentration (Cmax) was 15.1 ± 4.3 µg/mL; the time to reach Cmax (tmax), 1.66 ± 0.55 h; and the mean absorption time (MAT), 2.50 ± 1.42 h. Absorption was high, with a fraction of the dose absorbed (F) of 83.5 ± 22.4%. Menbutone was rapidly eliminated from plasma for both routes of administration, with a fast and high IM bioavailability.
Collapse
Affiliation(s)
- Raquel Diez
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Jose M Rodriguez
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Cristina Lopez
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Raul de la Puente
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Matilde Sierra
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - M Jose Diez
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Nelida Fernandez
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Juan J Garcia
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| | - Ana M Sahagun
- Pharmacology, Department of Biomedical Sciences, Veterinary Faculty, Institute of Biomedicine (IBIOMED), University of Leon, 24071 Leon, Spain
| |
Collapse
|
3
|
Knauer WA, Barrell EA, Guedes AGP, Ventura BA. Effects of multimodal pain management strategies on acute physiological and behavioral response to cautery disbudding in neonatal goat kids. J Dairy Sci 2023; 106:2830-2845. [PMID: 36870841 DOI: 10.3168/jds.2022-22433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/02/2022] [Indexed: 03/06/2023]
Abstract
Dairy goat kids are commonly disbudded in the United States without pain relief. Our objective was to identify an efficient pain management strategy by monitoring changes in plasma biomarkers and behavior of disbudded goat kids. A total of 42 kids (5-18 d old at the time of disbudding) were randomly allocated to 1 of 7 treatments (n = 6/treatment): sham treatment; 0.05 mg/kg i.m. xylazine (X); 4 mg/kg subcutaneous buffered lidocaine (L); 1 mg/kg oral meloxicam (M); xylazine and lidocaine (XL); xylazine and meloxicam (XM); and xylazine, meloxicam, and lidocaine together (XML). Treatments were administered 20 min before disbudding. One trained individual, blinded to treatment, disbudded all kids; sham-treated kids were handled similarly except the iron was cold. Jugular blood samples (3 mL) were obtained before (-20, -10, and -1 min) and after (1, 15, and 30 min, and 1, 2, 4, 6, 12, 24, 36, 48 h) disbudding and analyzed for cortisol and prostaglandin E2 (PGE2). Mechanical nociceptive threshold (MNT) testing was performed at 4, 12, 24, and 48 h after disbudding, and kids were weighed daily until 2 d post-disbudding. Vocalizations, tail flicks, and struggle behavior during disbudding were recorded. Cameras were mounted over home pens; continuous and scan observations over 12 periods of 10 min each, in the 48 h after disbudding, captured frequency of locomotion and pain-specific behaviors. Repeated measures and linear mixed models assessed treatment effects on outcome measures during and after disbudding. Models accounted for sex, breed, and age as random effects, and Bonferroni adjustments accounted for multiple comparisons. At 15 min after disbudding, XML kids had lower plasma cortisol concentrations compared with L (50.0 ± 13.2 vs. 132.8 ± 13.6 mmol/L) and M kids (50.0 ± 13.2 vs. 145.4 ± 15.7 mmol/L). Cortisol was also lower in XML kids over the first hour after disbudding compared with L kids (43.4 ± 9 vs. 80.2 ± 9 mmol/L). Change from baseline PGE2 was not affected by treatment. Behaviors observed during disbudding did not differ by treatment group. Treatment affected MNT such that M kids were more sensitive overall compared with sham kids (0.93 ± 0.11 kgf vs. 1.35 ± 0.12 kgf). None of the recorded post-disbudding behaviors were affected by treatment, but study activities did influence behavior over time, with kid activity levels declining in the first day after disbudding but largely recovering thereafter. We conclude that none of the drug combinations investigated here appeared to fully attenuate pain indicators during or after disbudding, but triple modality seems to have offered partial relief compared with some of the single-modality treatments.
Collapse
Affiliation(s)
- W A Knauer
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul 55108.
| | - E A Barrell
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul 55108
| | - A G P Guedes
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul 55108
| | - B A Ventura
- Department of Life Sciences, University of Lincoln, Lincoln LN6 7DL, UK
| |
Collapse
|
4
|
Jeffrey A, Gardhouse S, Kleinhenz M, Hocker SE, Weeder M, Montgomery SR, Zhang Y, Porting A, Rooney T. Examination of the pharmacokinetics and differential inhibition of cyclooxygenase isoenzymes in New Zealand white rabbits (Oryctolagus cuniculus) by the Non-Steroidal anti-inflammatory Robenacoxib. J Vet Pharmacol Ther 2023; 46:103-111. [PMID: 36478376 DOI: 10.1111/jvp.13105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/10/2022] [Accepted: 11/19/2022] [Indexed: 12/12/2022]
Abstract
Effective rabbit analgesia is challenging, and there are few studies available on the newer COX-2 selective NSAIDs, such as robenacoxib. This study aimed to establish the pharmacokinetics of oral and subcutaneous robenacoxib, describe its inhibitory actions on COX enzymes, and develop dosing, using six healthy New Zealand white rabbits. Pharmacokinetics were determined from plasma concentrations after oral administration of robenacoxib (0.83-0.96 mg/kg) and also after subcutaneous administration (2 mg/kg). The inhibitory actions of robenacoxib were evaluated by measuring plasma concentrations of thromboxane B2 (TBX2 ) and prostaglandin E2 (PGE2 ) as surrogate markers of cyclooxygenase enzyme isoform inhibition. The mean maximum concentration for oral and subcutaneous administration was 0.23 μg/ml and 5.82 μg/ml, respectively. Oral robenacoxib administration did not demonstrate a significant difference between any time point for PGE2 or TBX2 , though subcutaneous administration did for both. There was no significant difference in PGE2 or TBX2 concentrations at any time point when comparing subcutaneous versus oral routes. Although the results support that plasma robenacoxib exceeds the therapeutic levels compared to dogs and cats, there was little significance in the difference in the changes associated with COX-1 and COX-2 inhibition. Further studies are warranted to determine appropriate dosing, safety, and efficacy in rabbits.
Collapse
Affiliation(s)
- Alison Jeffrey
- Department of Clinical Science, Kansas State University, College of Veterinary Medicine, Kansas, Manhattan, USA
| | - Sara Gardhouse
- Evolution Veterinary Specialists, Colorado, Lakewood, USA
| | - Michael Kleinhenz
- Department of Clinical Science, Kansas State University, College of Veterinary Medicine, Kansas, Manhattan, USA
| | | | - Mikaela Weeder
- Department of Clinical Science, Kansas State University, College of Veterinary Medicine, Kansas, Manhattan, USA
| | - Shawnee R Montgomery
- Department of Anatomy and Physiology, Kansas State University, Kansas, Manhattan, USA
| | - Yuntao Zhang
- Veterinary Diagnostic Laboratory, Kansas State University, Kansas, Manhattan, USA
| | - Anna Porting
- Veterinary Diagnostic Laboratory, Kansas State University, Kansas, Manhattan, USA
| | - Tess Rooney
- Department of Medicine & Epidemiology, University of California, Davis, School of Veterinary Medicine, California, Davis, USA
| |
Collapse
|
5
|
Mills PC, Owens JG, Reinbold JB, McGowan M, Ellenbergner C, Woldeyohannes S, Satake N. A novel transdermal ketoprofen formulation for analgesia in cattle. J Vet Pharmacol Ther 2022; 45:530-542. [PMID: 36057922 PMCID: PMC9826033 DOI: 10.1111/jvp.13093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 07/12/2022] [Accepted: 08/14/2022] [Indexed: 01/18/2023]
Abstract
Ketoprofen is registered in many countries for injectable administration in cattle. Because it is soluble in a wide range of excipients, development of a novel transdermal (TD) ketoprofen formulation was pursued to provide a convenient and pain-free route of administration in cattle. One hundred and six excipient combinations were screened using in vitro techniques (Franz diffusion cells), with a 20%(w/v) ketoprofen formulation dissolved in a combination of 45%:45%(v/v) ethanol and isopropyl myristate (IPM) and 10%(v/v) eucalyptus oil achieving maximal penetration of ketoprofen through bovine skin. A bioavailability study was then conducted using a randomized cross-over design (n = 12), including IV, IM (both 3 mg/kg) and TD (10 mg/kg) ketoprofen formulations administered with a one-week washout period between administrations. The IV and IM formulation pharmacokinetic results were as expected. The CMAX , Tmax and AUC0-Last were significantly higher (arithmetic mean ± SD) after TD administration (20.0 ± 6.5 μg/ml, 115 ± 17 min and 3940 ± 1324 μg*min/ml, respectively), compared to IM (11.0 ± 4.0 μg/ml, 74 ± 43 min and 2376 ± 738 μg*min/ml, respectively), although there were no significant differences for T½β . However, dose corrected values CMAX and AUCinf were significantly higher for IM compared to TD. The arithmetic mean bioavailability (F) of the transdermal formulation was 50%. The plasma concentration of the TD formulation at a dose of 10 mg/kg was similar to the IM formulation at 3 mg/kg by 30 min post-dosing with an arithmetic mean ± SD of 7.97 ± 4.38 vs. 8.02 ± 3.55 μg/ml, respectively. The TD formulation was generally well tolerated by cattle, although some local irritation along the site of application was noted after 12 h of exposure during the bioavailability study. Results indicate that this novel TD formulation provides a substantial improvement in administration convenience, may improve animal welfare and end-user safety through needle-free administration, and achieves similar plasma pharmacokinetics to the IM product when administered at 10 mg/kg.
Collapse
Affiliation(s)
- Paul C. Mills
- School of Veterinary ScienceThe University of QueenslandGattonAustralia
| | | | | | - Michael McGowan
- School of Veterinary ScienceThe University of QueenslandGattonAustralia
| | | | | | - Nana Satake
- School of Veterinary ScienceThe University of QueenslandGattonAustralia
| |
Collapse
|
6
|
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; 12:3683. [PMID: 35256692 PMCID: PMC8901777 DOI: 10.1038/s41598-022-07795-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/24/2022] [Indexed: 01/17/2023] Open
Abstract
Industrial hemp (IH) is defined as Cannabis sativa containing < 0.3% delta-9 tetrahydrocannabinol (THC) and was legalized in the 2018 Farm Bill. The impact of cannabinoids in IH fed to livestock, especially after repeat exposure, has not been thoroughly investigated. Sixteen male castrated Holstein cattle weighting (± SD) 447 ± 68 kg were enrolled onto the study. Cattle were allocated into two treatment groups either receiving IH (HEMP, n = 8) or a control (CNTL, n = 8). Cattle in the HEMP group were fed 25 g IH mixed in 200 g of grain once a day for 14 days to target a daily dose of 5.5 mg/kg of cannabidiolic acid (CBDA). Behavior was continuously monitored with accelerometers and blood samples were collected at predetermined time points for plasma cannabinoid, serum cortisol, serum haptoglobin, liver enzymes, serum amyloid A, and prostaglandin E2 concentrations. The HEMP group spent a mean 14.1 h/d (95% CI 13.6–14.6 h/d) lying compared to the 13.4 h/d (95% CI 12.9–13.8 h/d) for the CNTL cattle (P = 0.03). Cortisol concentrations in the HEMP group were lower than the CNTL group (P = 0.001). Cattle in the HEMP group demonstrated an 8.8% reduction in prostaglandin E2 concentrations from baseline compared to a 10.2% increase from baseline observed in the CNTL group. No differences for haptoglobin or serum amyloid A were observed. These results suggest that feeding IH with a high CBDA content for 14 days increases lying behavior and decreases biomarkers of stress and inflammation in cattle.
Collapse
|
7
|
De Koster J, Boucher JF, Tena JK, Gehring R, Stegemann MR. Co-formulation of ketoprofen with tulathromycin alters pharmacokinetic and pharmacodynamic profile of ketoprofen in cattle. J Vet Pharmacol Ther 2021; 45:69-82. [PMID: 34387365 DOI: 10.1111/jvp.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/25/2021] [Indexed: 12/01/2022]
Abstract
The current studies aimed to evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) profile and to establish a PK-PD model for ketoprofen in a new fixed combination product containing tulathromycin (2.5 mg/kg) and ketoprofen (3 mg/kg) to treat bovine respiratory disease associated with pyrexia in cattle. Firstly, the effect of different ketoprofen doses as mono-substance (1, 3, and 6 mg/kg subcutaneous) on lipopolysaccharide-induced fever was evaluated which indicated that rectal temperature reduction lasted longer in the calves receiving 3 and 6 mg/kg ketoprofen. Secondly, the PK profile of the combination product was compared with mono-substance products (3 mg/kg subcutaneous and intramuscular). The PK profile of ketoprofen in the combination product was characterized by longer t1/2 , lower Cmax and increased AUC in comparison with mono-substance products. Due to prolonged ketoprofen exposure in the combination product, the pyrexia reducing effect of the combination product lasted longer in a second lipopolysaccharide challenge study in comparison with mono-substance products. Finally, a PK-PD model for the anti-pyretic effect of ketoprofen was developed based on the data from the different studies. The PK-PD model eliminated the need for additional animal experiments and indicated that a 3 mg/kg ketoprofen dose in the combination product provided optimal efficacy.
Collapse
Affiliation(s)
- Jenne De Koster
- Zoetis, Veterinary Medicine Research and Development, Zaventem, Belgium
| | - Joseph F Boucher
- Zoetis, Veterinary Medicine Research and Development, Kalamazoo, MI, USA
| | - Jezaniah-Kira Tena
- Zoetis, Veterinary Medicine Research and Development, Kalamazoo, MI, USA
| | - Ronette Gehring
- Division of Veterinary and Comparative Pharmacology, Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | | |
Collapse
|
8
|
Graves MT, Schneider L, Cox S, Caldwell M, Krawczel P, Lee A, Lear A. Evaluation of the pharmacokinetics and efficacy of transdermal flunixin for pain mitigation following castration in goats. Transl Anim Sci 2020; 4:txaa198. [PMID: 33324963 PMCID: PMC7724925 DOI: 10.1093/tas/txaa198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/27/2020] [Indexed: 11/14/2022] Open
Abstract
The mitigation of pain associated with common management procedures is a rising concern among veterinarians, producers and consumers. Nonsteroidal anti-inflammatory drugs are vital compounds for this purpose due to their cost, convenience, and efficacy. A transdermal formulation of flunixin meglumine (FM) was approved for the treatment of pain in cattle; however, the efficacy has yet to be determined for small ruminants. The current study had two aims: 1) to determine the pharmacokinetics of transdermal flunixin meglumine (TD FM) in bucklings and 2) to evaluate pain mitigation by TD FM following castration. To evaluate pharmacokinetics, 12 male goats (mean age = 6 mo) received 2.2 mg/kg of FM IV (n = 6) or 3.3 mg/kg TD FM (n = 6). Plasma FM concentrations were measured. The mean Cmax, Tmax, and harmonic mean half-life for TD FM were 1.09 ± 0.65 μg/mL, 5.50 ± 2.95 h, and 7.16 ± 2.06 h, respectively. To evaluate the efficacy of pain mitigation, 18 goats were randomly assigned to three treatment groups: 1) TD FM and castration (FM CAST) (n = 6); 2) transdermal placebo and castration (PL CAST) (n = 6); and 3) TD FM and sham castration (SHAM) (n = 6). Plasma samples were collected at 0, 12, 24, 36, 48, 72, and 96 h to assess cortisol and prostaglandin E2 (PGE2). Daily dry matter intake (DMI) was recorded and body weight was measured at the beginning and end of the study. Thermography (IRT) images of the scrotum, as well as heart rate (HR), respiratory rate (RR), and rectal temperature, were taken twice daily. Separate mixed analysis of variance models were used to test the effects of treatment, time, and their interaction on mean body temperature, IRT, HR, and RR. Autoregressive covariance structure was utilized to account for repeated measures and individual goat DMI prior to the study was added as a covariate. There were no differences in vital parameters, IRT measurements, cortisol, or PGE2 in animals receiving either TD FM or placebo following castration (P > 0.05). DMI had a treatment by hour interaction and was significantly higher in FM CAST and SHAM groups than the PL CAST group (P = 0.04). Goats in the SHAM group gained weight throughout the study, whereas goats in all other groups lost weight (P = 0.02). Results indicate that TD FM may mitigate pain as demonstrated by increased DMI; however, a single dose may not be sufficient to reduce physiological indicators of pain associated with castration in goats.
Collapse
Affiliation(s)
- Meggan T Graves
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - Liesel Schneider
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN
| | - Sherry Cox
- Biomedical and Diagnostic Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - Marc Caldwell
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| | - Peter Krawczel
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN.,Department of Agricultural Sciences and Animal Science, University of Helsinki, Helsinki, Finland.,Department of Production Animal Medicine at the Research Centre for Animal Welfare, University of Helsinki, Helsinki, Finland.,Helsinki One Health (HOH), University of Helsinki, Helsinki, Finland
| | - Amanda Lee
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN
| | - Andrea Lear
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN
| |
Collapse
|
9
|
Gu Y, Gu Q, Yang Q, Yang M, Wang S, Liu J. Finite Element Analysis for Predicting Skin Pharmacokinetics of Nano Transdermal Drug Delivery System Based on the Multilayer Geometry Model. Int J Nanomedicine 2020; 15:6007-6018. [PMID: 32884260 PMCID: PMC7439786 DOI: 10.2147/ijn.s261386] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/30/2020] [Indexed: 11/23/2022] Open
Abstract
Background Skin pharmacokinetics is an indispensable indication for studying the drug fate after administration of transdermal drug delivery systems (TDDS). However, the heterogeneity and complex skin structured with stratum corneum, viable epidermis, dermis, and subcutaneous tissue inevitably leads the drug diffusion coefficient (Kp) to vary depending on the skin depth, which seriously limits the development of TDDS pharmacokinetics in full thickness skin. Methods A multilayer geometry skin model was established and the Kp of drug in SC, viable epidermis, and dermis was obtained using the technologies of molecular dynamics simulation, in vitro permeation experiments, and in vivo microdialysis, respectively. Besides, finite element analysis (FEA) based on drug Kps in different skin layers was applied to simulate the paeonol nanoemulsion (PAE-NEs) percutaneous dynamic penetration process in two and three dimensions. In addition, PAE-NEs skin pharmacokinetics profile obtained by the simulation was verified by in vivo experiment. Results Coarse-grained modeling of molecular dynamic simulation was successfully established and the Kp of PAE in SC was 2.00×10−6 cm2/h. The Kp of PAE-NE in viable epidermis and in dermis detected using penetration test and microdialysis probe technology, was 1.58×10−5 cm2/h and 3.20×10−5 cm2/h, respectively. In addition, the results of verification indicated that PAE-NEs skin pharmacokinetics profile obtained by the simulation was consistent with that by in vivo experiment. Discussion This study demonstrated that the FEA combined with the established multilayer geometry skin model could accurately predict the skin pharmacokinetics of TDDS.
Collapse
Affiliation(s)
- Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Qing Gu
- Department of Pharmacy, Jingan District Zhabei Central Hospital, Shanghai 200070, People's Republic of China
| | - Qing Yang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Meng Yang
- Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Shengzhang Wang
- Institute of Biomechanics, Department of Aeronautics and Astronautics, Fudan University, Shanghai 200433, People's Republic of China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Pharmacy, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| |
Collapse
|
10
|
Smith JS, Marmulak TL, Angelos JA, Lin Z, Rowe JD, Carlson JL, Shelver WL, Lee EA, Tell LA. Pharmacokinetic Parameters and Estimated Milk Withdrawal Intervals for Domestic Goats ( Capra Aegagrus Hircus) After Administration of Single and Multiple Intravenous and Subcutaneous Doses of Flunixin Meglumine. Front Vet Sci 2020; 7:213. [PMID: 32509803 PMCID: PMC7248982 DOI: 10.3389/fvets.2020.00213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction: The study objectives were to estimate plasma flunixin (FLU) pharmacokinetic parameters and milk depletion profiles for FLU and its metabolite (5-hydroxy flunixin; 5-OH) after subcutaneous (SC) and intravenous (IV) administration of single and multiple flunixin meglumine (FM) doses to non-lactating (nulliparous and pregnant does) and lactating dairy goats. Analytical methods (ELISA and UPLC-MS/MS) for quantifying plasma FLU concentrations were compared. The final objective was to use regulatory (FDA and EMA) methods to estimate milk withdrawal intervals following extra-label drug use in goats. Methods: FM was administered IV and SC to commercial dairy goats at 1.1 mg/kg for single and multiple doses. Plasma and milk samples were analyzed for FLU and 5-OH via UPLC-MS/MS. Plasma samples were also analyzed for FLU concentrations via ELISA. Using statistical approaches recommended by regulatory agencies, milk withdrawal intervals were estimated following FM extra-label use. Results: Following IV administration of a single FM dose, clearances were 127, 199, and 365 ml/kg/h for non-lactating (NL) pregnant does, NL nulliparous does, and lactating dairy does, respectively. Following multiple SC doses, clearance/F was 199 ml/kg/h for lactating does. After IV administration of a single FM dose, terminal elimination half-lives were 4.08, 2.87, and 3.77 h for NL pregnant does, NL nulliparous does, and lactating dairy does, respectively. After multiple SC doses, the terminal elimination half-life was 3.03 h for lactating dairy does. No significant differences were noted for samples analyzed by UPLC-MS/MS or ELISA. Milk withdrawal intervals ranged from 36 to 60 h depending on the regulatory statistical method and dosage regimen. Conclusions: Subcutaneous administration of FM to goats results in similar plasma pharmacokinetic parameters as IV administration. ELISA analysis is an alternative method to UPLC-MS/MS for quantifying FLU concentrations in caprine plasma samples. Following FM extra-label administration to dairy goats, clinicians could consider 36–60 h milk withdrawal intervals.
Collapse
Affiliation(s)
- Joe S Smith
- School of Veterinary Medicine, William R. Pritchard Veterinary Medical Teaching Hospital (VMTH), University of California, Davis, Davis, CA, United States
| | - Tara L Marmulak
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - John A Angelos
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Zhoumeng Lin
- Department of Anatomy and Physiology, Institute of Computational Comparative Medicine (ICCM), College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Joan D Rowe
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Jan L Carlson
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
| | - Weilin L Shelver
- USDA-ARS Biosciences Research Laboratory, Fargo, ND, United States
| | - Elizabeth A Lee
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Lisa A Tell
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
11
|
Reppert EJ, Kleinhenz MD, Montgomery SR, Heiman J, Sura A, Bornheim HN, Magnin G, Sidhu PK, Zhang Y, Joo H, Coetzee JF. Pharmacokinetics and pharmacodynamics of intravenous and transdermal flunixin meglumine in alpacas. J Vet Pharmacol Ther 2019; 42:572-579. [PMID: 31353535 DOI: 10.1111/jvp.12800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/18/2019] [Accepted: 07/01/2019] [Indexed: 11/28/2022]
Abstract
The aim of this study was to determine the pharmacokinetics and prostaglandin E2 (PGE2 ) synthesis inhibiting effects of intravenous (IV) and transdermal (TD) flunixin meglumine in eight, adult, female, Huacaya alpacas. A dose of 2.2 mg/kg administered IV and 3.3 mg/kg administered TD using a cross-over design. Plasma flunixin concentrations were measured by LC-MS/MS. Prostaglandin E2 concentrations were determined using a commercially available ELISA. Pharmacokinetic (PK) analysis was performed using noncompartmental methods. Plasma PGE2 concentrations decreased after IV flunixin meglumine administration but there was minimal change after TD application. Mean t1/2 λz after IV administration was 4.531 hr (range 3.355 to 5.571 hr) resulting from a mean Vz of 570.6 ml/kg (range, 387.3 to 1,142 ml/kg) and plasma clearance of 87.26 ml kg-1 hr-1 (range, 55.45-179.3 ml kg-1 hr-1 ). The mean Cmax, Tmax and t1/2 λz for flunixin following TD administration were 106.4 ng/ml (range, 56.98 to 168.6 ng/ml), 13.57 hr (range, 6.000-34.00 hr) and 24.06 hr (18.63 to 39.5 hr), respectively. The mean bioavailability for TD flunixin was calculated as 25.05%. The mean 80% inhibitory concentration (IC80 ) of PGE2 by flunixin meglumine was 0.23 µg/ml (range, 0.01 to 1.38 µg/ml). Poor bioavailability and poor suppression of PGE2 identified in this study indicate that TD flunixin meglumine administered at 3.3 mg/kg is not recommended for use in alpacas.
Collapse
Affiliation(s)
- Emily J Reppert
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Michael D Kleinhenz
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Shawnee R Montgomery
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Jared Heiman
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Amanda Sura
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Heather N Bornheim
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Geraldine Magnin
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Pritam K Sidhu
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas.,Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| | - Yuntao Zhang
- Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| | - Hyun Joo
- Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| | - Johann F Coetzee
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas.,Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| |
Collapse
|
12
|
Reppert EJ, Kleinhenz MD, Montgomery SR, Bornheim HN, Magnin G, Sidhu PK, Zhang Y, Joo H, Coetzee JF. Pharmacokinetics and pharmacodynamics of intravenous and transdermal flunixin meglumine in meat goats. J Vet Pharmacol Ther 2019; 42:309-317. [PMID: 30802981 DOI: 10.1111/jvp.12756] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/19/2019] [Accepted: 02/03/2019] [Indexed: 11/27/2022]
Abstract
The aim of this study was to determine the pharmacokinetics and prostaglandin E2 (PGE2 ) synthesis inhibiting effects of intravenous (IV) and transdermal (TD) flunixin meglumine in eight adult female Boer goats. A dose of 2.2 mg/kg was administered intravenously (IV) and 3.3 mg/kg administered TD using a cross-over design. Plasma flunixin concentrations were measured by LC-MS/MS. Prostaglandin E2 concentrations were determined using a commercially available ELISA. Pharmacokinetic (PK) analysis was performed using noncompartmental methods. Plasma PGE2 concentrations decreased after flunixin meglumine for both routes of administration. Mean λz -HL after IV administration was 6.032 hr (range 4.735-9.244 hr) resulting from a mean Vz of 584.1 ml/kg (range, 357.1-1,092 ml/kg) and plasma clearance of 67.11 ml kg-1 hr-1 (range, 45.57-82.35 ml kg-1 hr-1 ). The mean Cmax , Tmax, and λz -HL for flunixin following TD administration was 0.134 μg/ml (range, 0.050-0.188 μg/ml), 11.41 hr (range, 6.00-36.00 hr), and 43.12 hr (15.98-62.49 hr), respectively. The mean bioavailability for TD flunixin was calculated as 24.76%. The mean 80% inhibitory concentration (IC80 ) of PGE2 by flunixin meglumine was 0.28 μg/ml (range, 0.08-0.69 μg/ml) and was only achieved with IV formulation of flunixin in this study. The PK results support clinical studies to examine the efficacy of TD flunixin in goats. Determining the systemic effects of flunixin-mediated PGE2 suppression in goats is also warranted.
Collapse
Affiliation(s)
- Emily J Reppert
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Michael D Kleinhenz
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Shawnee R Montgomery
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Heather N Bornheim
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Geraldine Magnin
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Pritam K Sidhu
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas.,Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| | - Yuntao Zhang
- Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| | - Hyun Joo
- Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| | - Johann F Coetzee
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas.,Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, Kansas
| |
Collapse
|