1
|
Thompson EJ, Jeong A, Helfer VE, Shakhnovich V, Edginton A, Balevic SJ, James LP, Collier DN, Anand R, Gonzalez D. Physiologically-based pharmacokinetic modeling of pantoprazole to evaluate the role of CYP2C19 genetic variation and obesity in the pediatric population. CPT Pharmacometrics Syst Pharmacol 2024; 13:1394-1408. [PMID: 38837864 PMCID: PMC11330186 DOI: 10.1002/psp4.13167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/11/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Pantoprazole is a proton pump inhibitor indicated for the treatment of gastroesophageal reflux disease, a condition that disproportionately affects children with obesity. Appropriately dosing pantoprazole in children with obesity requires understanding the body size metric that best guides dosing, but pharmacokinetic (PK) trials using traditional techniques are limited by the need for larger sample sizes and frequent blood sampling. Physiologically-based PK (PBPK) models are an attractive alternative that can account for physiologic-, genetic-, and drug-specific changes without the need for extensive clinical trial data. In this study, we explored the effect of obesity on pantoprazole PK and evaluated label-suggested dosing in this population. An adult PBPK model for pantoprazole was developed using data from the literature and accounting for genetic variation in CYP2C19. The adult PBPK model was scaled to children without obesity using age-associated changes in anatomical and physiological parameters. Lastly, the pediatric PBPK model was expanded to children with obesity. Three pantoprazole dosing strategies were evaluated: 1 mg/kg total body weight, 1.2 mg/kg lean body weight, and US Food and Drug Administration-recommended weight-tiered dosing. Simulated concentration-time profiles from our model were compared with data from a prospective cohort study (PAN01; NCT02186652). Weight-tiered dosing resulted in the most (>90%) children with pantoprazole exposures in the reference range, regardless of obesity status or CYP2C19 phenotype, confirming results from previously published population PK models. PBPK models may allow for the efficient study of physiologic and developmental effects of obesity on PK in special populations where clinical trial data may be limited.
Collapse
Affiliation(s)
- Elizabeth J. Thompson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of PediatricsDuke University Medical CenterDurhamNorth CarolinaUSA
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Angela Jeong
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Victória E. Helfer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Valentina Shakhnovich
- University of Missouri‐Kansas City School of MedicineKansas CityMissouriUSA
- Divisions of Gastroenterology, Hepatology and Nutrition & Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas CityKansas CityMissouriUSA
- Ironwood PharmaceuticalsBostonMassachusettsUSA
| | - Andrea Edginton
- School of PharmacyUniversity of WaterlooWaterlooOntarioCanada
| | - Stephen J. Balevic
- Department of PediatricsDuke University Medical CenterDurhamNorth CarolinaUSA
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Laura P. James
- Department of PediatricsUniversity of Arkansas for Medical Sciences, Section of Clinical Pharmacology and Toxicology, Arkansas Children's HospitalLittle RockArkansasUSA
| | - David N. Collier
- Department of Pediatrics and Center for Health Disparities, Division of General PediatricsEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | | | - Daniel Gonzalez
- Duke Clinical Research InstituteDurhamNorth CarolinaUSA
- Division of Clinical Pharmacology, Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| |
Collapse
|
2
|
Fadel C, Łebkowska-Wieruszewskac B, Serih F, Lisowski A, Poapolathep A, Giorgi M. Comparative pharmacokinetics of intravenous and subcutaneous pantoprazole in sheep and goats. Vet J 2024; 305:106138. [PMID: 38761957 DOI: 10.1016/j.tvjl.2024.106138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Abomasal ulcers are a significant concern in intensive animal farming due to their impact on animal health and productivity. While proton pump inhibitors (PPIs) such as pantoprazole (PTZ) show promise in treating these ulcers, data on PTZ's pharmacokinetics (PK) in adult goats and sheep are limited. This study aims to fill this gap by investigating and comparing PTZ's PK in these species following single intravenous (IV) and subcutaneous (SC) administrations. Five healthy male goats and sheep were included in the study. PTZ concentrations in plasma samples were determined using a validated analytical method. Non-compartmental analysis was conducted, and statistical comparisons were made between IV and SC administrations and between species. Sheep and goats showed similar systemic exposure levels regardless of the administration route. However, sheep had a shorter t1/2 due to a higher Vd compared to goats. Cl values were comparable in both species, with low extraction ratio values. There were no significant differences in Cmax and Tmax between the two species with regards to SC administration, and complete bioavailability was observed. The MAT exceeded the t1/2 in both species, indicating a potential flip-flop phenomenon. Considering the AUC as a predictor for drug efficacy, and observing no significant differences in systemic exposure between sheep and goats for any route of administration, dosage adjustment between the two species may not be necessary. In field settings, SC administration proves more practical, providing not only complete bioavailability but also a longer half-life compared to IV. Further studies are warranted to explore the PK/PD of PTZ in small ruminants with abomasal ulcers, to fully comprehend its therapeutic efficacy in such scenarios.
Collapse
Affiliation(s)
- Charbel Fadel
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge, 2, Pisa, PI 56124, Italy; Department of Veterinary Medicine, Lebanese University, Faculty of Agronomy Bldg., University Street, Dekwaneh, Beirut, Lebanon
| | - Beata Łebkowska-Wieruszewskac
- Department of Pharmacology, Toxicology and Environmental Protection, University of Life Sciences in Lublin, 13 Akademicka Street, Lublin 20-950, Poland
| | - Firas Serih
- Department of Veterinary Sciences, University of Sassari, Via Vienna, 2, Sassari, SS 07100, Italy
| | - Andrew Lisowski
- Institute of Animal Breeding and Biodiversity Conservation, University of Life Sciences in Lublin, 13 Akademicka Street, Lublin 20-950, Poland
| | - Amnart Poapolathep
- Faculty of Veterinary Medicine, Department of Pharmacology, Kasetsart University, 50 Paholyothin Road, Bangkok 10900, Thailand
| | - Mario Giorgi
- Department of Veterinary Sciences, University of Pisa, Viale delle Piagge, 2, Pisa, PI 56124, Italy; Department of Veterinary Sciences, University of Sassari, Via Vienna, 2, Sassari, SS 07100, Italy.
| |
Collapse
|
3
|
Poweleit EA, Vaughn SE, Desta Z, Dexheimer JW, Strawn JR, Ramsey LB. Machine Learning-Based Prediction of Escitalopram and Sertraline Side Effects With Pharmacokinetic Data in Children and Adolescents. Clin Pharmacol Ther 2024; 115:860-870. [PMID: 38297828 PMCID: PMC11046530 DOI: 10.1002/cpt.3184] [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] [Received: 09/29/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024]
Abstract
Selective serotonin reuptake inhibitors (SSRI) are the first-line pharmacologic treatment for anxiety and depressive disorders in children and adolescents. Many patients experience side effects that are difficult to predict, are associated with significant morbidity, and can lead to treatment discontinuation. Variation in SSRI pharmacokinetics could explain differences in treatment outcomes, but this is often overlooked as a contributing factor to SSRI tolerability. This study evaluated data from 288 escitalopram-treated and 255 sertraline-treated patients ≤ 18 years old to develop machine learning models to predict side effects using electronic health record data and Bayesian estimated pharmacokinetic parameters. Trained on a combined cohort of escitalopram- and sertraline-treated patients, a penalized logistic regression model achieved an area under the receiver operating characteristic curve (AUROC) of 0.77 (95% confidence interval (CI): 0.66-0.88), with 0.69 sensitivity (95% CI: 0.54-0.86), and 0.82 specificity (95% CI: 0.72-0.87). Medication exposure, clearance, and time since the last dose increase were among the top features. Individual escitalopram and sertraline models yielded an AUROC of 0.73 (95% CI: 0.65-0.81) and 0.64 (95% CI: 0.55-0.73), respectively. Post hoc analysis showed sertraline-treated patients with activation side effects had slower clearance (P = 0.01), which attenuated after accounting for age (P = 0.055). These findings raise the possibility that a machine learning approach leveraging pharmacokinetic data can predict escitalopram- and sertraline-related side effects. Clinicians may consider differences in medication pharmacokinetics, especially during dose titration and as opposed to relying on dose, when managing side effects. With further validation, application of this model to predict side effects may enhance SSRI precision dosing strategies in youth.
Collapse
Affiliation(s)
- Ethan A. Poweleit
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Biomedical Informatics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Division of Research in Patient Services, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Samuel E. Vaughn
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Zeruesenay Desta
- Division of Clinical Pharmacology, Indiana University, School of Medicine, Indianapolis, IN
| | - Judith W. Dexheimer
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH
- Division of Emergency Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jeffrey R. Strawn
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Laura B. Ramsey
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, MO, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
4
|
Sintusek P, Mutalib M, Thapar N. Gastroesophageal reflux disease in children: What’s new right now? World J Gastrointest Endosc 2023; 15:84-102. [PMID: 37034973 PMCID: PMC10080553 DOI: 10.4253/wjge.v15.i3.84] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/15/2023] [Accepted: 02/08/2023] [Indexed: 03/16/2023] Open
Abstract
Gastroesophageal reflux (GER) in children is very common and refers to the involuntary passage of gastric contents into the esophagus. This is often physiological and managed conservatively. In contrast, GER disease (GERD) is a less common pathologic process causing troublesome symptoms, which may need medical management. Apart from abnormal transient relaxations of the lower esophageal sphincter, other factors that play a role in the pathogenesis of GERD include defects in esophageal mucosal defense, impaired esophageal and gastric motility and clearance, as well as anatomical defects of the lower esophageal reflux barrier such as hiatal hernia. The clinical manifestations of GERD in young children are varied and nonspecific prompting the necessity for careful diagnostic evaluation. Management should be targeted to the underlying aetiopathogenesis and to limit complications of GERD. The following review focuses on up-to-date information regarding of the pathogenesis, diagnostic evaluation and management of GERD in children.
Collapse
Affiliation(s)
- Palittiya Sintusek
- Thai Pediatric Gastroenterology, Hepatology and Immunology Research Unit (TPGHAI), Division of Gastroenterology, Department of Pediatrics, King Chulalongkorn Memorial Hospital and Thai Red Cross, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Bangkok, Thailand
| | - Mohamed Mutalib
- Department of Paediatric Gastroenterology, Pediatric and Gastroenterology Services, Evelina London Children’s Hospital, London SE1 7EH, United Kingdom
| | - Nikhil Thapar
- Department of Gastroenterology, Hepatology and Liver Transplant, Queensland Children’s Hospital, Brisbane, Queensland 4101, Australia
- School of Medicine, University of Queensland, Brisbane, Queensland 4006, Australia
- Woolworths Centre for Child Nutrition Research, Queensland University of Technology, Brisbane, Queensland 4101, Australia
| |
Collapse
|
5
|
Pharmacogenetic Aspects of Drug Metabolizing Enzymes and Transporters in Pediatric Medicine: Study Progress, Clinical Practice and Future Perspectives. Paediatr Drugs 2023; 25:301-319. [PMID: 36707496 DOI: 10.1007/s40272-023-00560-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 01/28/2023]
Abstract
As the activity of certain drug metabolizing enzymes or transporter proteins can vary with age, the effect of ontogenetic and genetic variation on the activity of these enzymes is critical for the accurate prediction of treatment outcomes and toxicity in children. This makes pharmacogenetic research in pediatrics particularly important and urgently needed, but also challenging. This review summarizes pharmacogenetic studies on the effects of genetic polymorphisms on pharmacokinetic parameters and clinical outcomes in pediatric populations for certain drugs, which are commonly prescribed by clinicians across multiple therapeutic areas in a general hospital, organized from those with the most to the least pediatric evidence among each drug category. We also further discuss the research status of the gene-guided dosing regimens and clinical implementation of pediatric pharmacogenetics. More and more drug-gene interactions are demonstrated to have clinical validity for children, and pharmacogenomics in pediatrics have shown evidence-based benefits to enhance the efficacy and precision of existing drug dosing regimens in several therapeutic areas. However, the most important limitation to the implementation is the lack of high-quality, rigorous pediatric prospective clinical studies, so adequately powered interventional clinical trials that support incorporation of pharmacogenetics into the care of children are still needed.
Collapse
|
6
|
Jafri F, Taylor ZL, Gonzalez D, Shakhnovich V. Effects of obesity on the pharmacology of proton pump inhibitors: current understanding and future implications for patient care and research. Expert Opin Drug Metab Toxicol 2023; 19:1-11. [PMID: 36800927 PMCID: PMC10065909 DOI: 10.1080/17425255.2023.2178897] [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] [Received: 11/29/2022] [Accepted: 02/07/2023] [Indexed: 02/20/2023]
Abstract
INTRODUCTION In the United States, obesity affects approximately ⅖ adults and ⅕ children, leading to increased risk for comorbidities, like gastroesophageal reflux disease (GERD), treated increasingly with proton pump inhibitors (PPIs). Currently, there are no clinical guidelines to inform PPI dose selection for obesity, with sparse data regarding whether dose augmentation is necessary. AREAS COVERED We provide a review of available literature regarding the pharmacokinetics (PK), pharmacodynamics (PD), and/or metabolism of PPIs in children and adults with obesity, as a step toward informing PPI dose selection. EXPERT OPINION Published PK data in adults and children are limited to first-generation PPIs and point toward reduced apparent oral drug clearance in obesity, with equipoise regarding obesity impact on drug absorption. Available PD data are sparse, conflicting, and limited to adults. No studies are available to inform the PPI PK→PD relationship in obesity and if/how it differs compared to individuals without obesity. In the absence of data, best practice may be to dose PPIs based on CYP2C19 genotype and lean body weight, so as to avoid systemic overexposure and potential toxicities, while monitoring closely for efficacy.
Collapse
Affiliation(s)
- Farwa Jafri
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO
| | - Zachary L. Taylor
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valentina Shakhnovich
- University of Missouri-Kansas City School of Medicine, Kansas City, MO
- Children’s Mercy Kansas City, Kansas City, MO
- Center for Children’s Healthy Lifestyles and Nutrition, Kansas City, MO
| |
Collapse
|
7
|
Feldman K, Kearns GL, Pearce RE, Abdel-Rahman SM, Leeder JS, Friesen A, Staggs VS, Gaedigk A, Weigel J, Shakhnovich V. Utility of the 13 C-pantoprazole breath test as a CYP2C19 phenotyping probe for children. Clin Transl Sci 2022; 15:1155-1166. [PMID: 35099109 PMCID: PMC9099127 DOI: 10.1111/cts.13232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/29/2022] Open
Abstract
The 13 C-pantoprazole breath test (PAN-BT) is a safe, non-invasive, in-vivo CYP2C19 phenotyping probe for adults. Our objective was to evaluate PAN-BT performance in children, with a focus on discriminating individuals who, according to guidelines from the Clinical Pharmacology Implementation Consortium (CPIC), would benefit from starting dose escalation vs. reduction for proton pump inhibitors (PPIs). Children (n=65; 6-17 years) genotyped for CYP2C19 variants *2, *3, *4, and *17 received a single oral dose of 13 C-pantoprazole. Plasma concentrations of pantoprazole and its metabolites, and changes in exhaled 13 CO2 (termed delta-over-baseline or DOB), were measured 10 times over 8 hours using HPLC-UV and spectrophotometry, respectively. Pharmacokinetic parameters of interest were generated and DOB features derived using feature engineering for the first 180 minutes post-administration. DOB features, age, sex, and obesity status were used to run bootstrap analysis at each timepoint (Ti) independently. For each iteration, stratified samples were drawn based on genotype prevalence in the original cohort. A random forest was trained, and predictive performance of PAN-BT evaluated. Strong discriminating ability for CYP2C19 intermediate vs. normal/rapid metabolizer phenotype was noted at DOBT30min (mean sensitivity: 0.522, specificity: 0.784), with consistent model outperformance over a random or a stratified classifier approach at each time-point (p<0.001). With additional refinement and investigation, the test could become a useful and convenient dosing tool in clinic to help identify children who would benefit most from PPI dose escalation vs. dose reduction, in accordance with CPIC guidelines.
Collapse
Affiliation(s)
- Keith Feldman
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA
| | - Gregory L Kearns
- Texas Christian University and UNTHSC School of Medicine, Fort Worth, TX, USA
| | - Robin E Pearce
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA
| | - Susan M Abdel-Rahman
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA
| | - J Steven Leeder
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA
| | - Alec Friesen
- Oklahoma School of Community Medicine, Tulsa, OK, USA
| | - Vincent S Staggs
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA
| | - Andrea Gaedigk
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA
| | - Jaylene Weigel
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Valentina Shakhnovich
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA.,Children's Mercy Kansas City, Kansas City, MO, USA.,Center for Children's Healthy Lifestyles & Nutrition, Kansas City, MO, USA
| |
Collapse
|
8
|
Smith JS, Mochel JP, Soto-Gonzalez WM, Rahn RR, Fayne BN, Escher OG, Geletka AM, Harvill LE, Bergman JB, Cox S. Pharmacokinetics of Pantoprazole and Pantoprazole Sulfone in Goats After Intravenous Administration: A Preliminary Report. Front Vet Sci 2021; 8:744813. [PMID: 34631865 PMCID: PMC8492921 DOI: 10.3389/fvets.2021.744813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Ruminant species are at risk of developing abomasal ulceration, but there is a lack of pharmacokinetic data for anti-ulcer therapies, such as the proton pump inhibitor pantoprazole, in goats. Objective: The primary study objective was to estimate the plasma pharmacokinetic parameters for pantoprazole in adult goats after intravenous administration. A secondary objective was to describe the pharmacokinetic parameters for the metabolite, pantoprazole sulfone, in goats. Methods: Pantoprazole was administered intravenously to six adult goats at a dose of 1 mg/kg. Plasma samples were collected over 36h and analyzed via reverse phase high performance liquid chromatography for determination of pantoprazole and pantoprazole sulfone concentrations. Pharmacokinetic parameters were determined by non-compartmental analysis. Results: Plasma clearance, elimination half-life, and volume of distribution of pantoprazole were estimated at 0.345 mL/kg/min, 0.7 h, and 0.9 L/kg, respectively following IV administration. The maximum concentration, elimination half-life and area under the curve of pantoprazole sulfone were estimated at 0.1 μg/mL, 0.8 h, and 0.2 hr*μg/mL, respectively. The global extraction ratio was estimated 0.00795 ± 0.00138. All animals had normal physical examinations after conclusion of the study. Conclusion: The reported plasma clearance for pantoprazole is lower than reported for foals, calves, and alpacas. The elimination half-life appears to be < that reported for foals and calves. Future pharmacodynamic studies are necessary for determination of the efficacy of pantoprazole on acid suppression in goats.
Collapse
Affiliation(s)
- Joe S Smith
- Systems Modeling and Reverse Translational Pharmacology, Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States.,Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Jonathan P Mochel
- Systems Modeling and Reverse Translational Pharmacology, Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Windy M Soto-Gonzalez
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Rebecca R Rahn
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Bryanna N Fayne
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Olivia G Escher
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Anastasia M Geletka
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Lainey E Harvill
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Joan B Bergman
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Sherry Cox
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
9
|
Lima JJ, Thomas CD, Barbarino J, Desta Z, Van Driest SL, El Rouby N, Johnson JA, Cavallari LH, Shakhnovich V, Thacker DL, Scott SA, Schwab M, Uppugunduri CRS, Formea CM, Franciosi JP, Sangkuhl K, Gaedigk A, Klein TE, Gammal RS, Furuta T. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C19 and Proton Pump Inhibitor Dosing. Clin Pharmacol Ther 2021; 109:1417-1423. [PMID: 32770672 PMCID: PMC7868475 DOI: 10.1002/cpt.2015] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/15/2020] [Indexed: 12/27/2022]
Abstract
Proton pump inhibitors (PPIs) are widely used for acid suppression in the treatment and prevention of many conditions, including gastroesophageal reflux disease, gastric and duodenal ulcers, erosive esophagitis, Helicobacter pylori infection, and pathological hypersecretory conditions. Most PPIs are metabolized primarily by cytochrome P450 2C19 (CYP2C19) into inactive metabolites, and CYP2C19 genotype has been linked to PPI exposure, efficacy, and adverse effects. We summarize the evidence from the literature and provide therapeutic recommendations for PPI prescribing based on CYP2C19 genotype (updates at www.cpicpgx.org). The potential benefits of using CYP2C19 genotype data to guide PPI therapy include (i) identifying patients with genotypes predictive of lower plasma exposure and prescribing them a higher dose that will increase the likelihood of efficacy, and (ii) identifying patients on chronic therapy with genotypes predictive of higher plasma exposure and prescribing them a decreased dose to minimize the risk of toxicity that is associated with long-term PPI use, particularly at higher plasma concentrations.
Collapse
Affiliation(s)
- John J. Lima
- Center for Pharmacogenomics and Translational Research, Nemours Children’s Health, Jacksonville, FL, USA
| | - Cameron D. Thomas
- Department of Pharmacotherapy and Translational Research, and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julia Barbarino
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Zeruesenay Desta
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sara L. Van Driest
- Departments of Pediatrics and Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nihal El Rouby
- Department of Pharmacotherapy and Translational Research, and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
- Division of Pharmacy Practice & Administrative Sciences, University of Cincinnati James Winkle College of Pharmacy, Cincinnati, OH, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research, and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Larisa H. Cavallari
- Department of Pharmacotherapy and Translational Research, and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Valentina Shakhnovich
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children’s Mercy Kansas City and University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Mercy Kansas City, Kansas City, MO, USA
- Center for Children’s Healthy Lifestyles & Nutrition, Kansas City, MO, USA
| | - David L. Thacker
- Department of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA
- Translational Software, Bellevue, WA, USA
| | - Stuart A. Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Sema4, Stamford, CT, USA
| | - Matthias Schwab
- Dr Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- Department of Clinical Pharmacology, University Hospital, Tuebingen, Germany
- Department of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
| | - Chakradhara Rao S Uppugunduri
- CANSEARCH Research Laboratory, Department of Paediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Oncology-Hematology Unit, Department of Paediatrics, Gynaecology and Obstetrics, Geneva University Hospital, Geneva, Switzerland
| | - Christine M. Formea
- Department of Pharmacy Services and Intermountain Precision Genomics, Intermountain Healthcare, Salt Lake City, UT, USA
| | - James P. Franciosi
- Division of Gastroenterology, Hepatology and Nutrition, Nemours Children’s Hospital, Orlando, FL
- Department of Pediatrics, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Katrin Sangkuhl
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children’s Mercy Kansas City and University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Teri E. Klein
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Roseann S. Gammal
- Department of Pharmacy Practice, MCPHS University School of Pharmacy, Boston, MA, USA
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Takahisa Furuta
- Center for Clinical Research, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
10
|
Duan Y, Chen J, Cui H, Zhao C. The effect of pantoprazole and somatostatin combined with thrombin in the treatment of non-esophagogastric varicosity upper gastrointestinal bleeding. Am J Transl Res 2021; 13:5484-5490. [PMID: 34150147 PMCID: PMC8205661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/10/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To explore the effect of pantoprazole and somatostatin combined with thrombin in the treatment of non-esophagogastric varicosity upper gastrointestinal bleeding (UGB) as well as its influence on serum hs-CRP and coagulation function. METHODS From June 2016 to May 2018, patients with upper gastrointestinal hemorrhage due to non-esophagogastric varices in our hospital were selected as research subjects. After screening, they were randomly divided into the combined group (57 cases) and the control group (57 cases). After the two groups are treated, the therapeutic effect was observed. The two groups of patients were followed up for 6 consecutive months, and the data were statistically analyzed. RESULTS It was found that there wass no significant difference between the two groups in gender, age, amount of bleeding, and etiology (P > 0.05). It was found that the immediate hemostasis rate and the hemostasis rate within 24 hours in the combined group were distinctly higher compared to the control group. The difference has statistical significance (P < 0.05). The total effective rate of the combined group was distinctly higher compared to the control group (P < 0.05). By comparing the expression levels of hs-CRP and IL-6 protein in the serum of the two groups before and after treatment, it was found that there was no significant difference in the expression levels of hs-CRP and IL-6 protein before treatment. However, after treatment, it was found that the levels of hs-CRP and IL-6 protein in the combined group were distinctly lower compared to the control group (P < 0.05). By analyzing adverse reactions, it was found that the combined group had distinctly lower adverse reactions compared to the control group (P < 0.05). CONCLUSION This work provides an experimental basis for the diagnosis and treatment of non-esophagogastric varicose UGB in the clinic.
Collapse
Affiliation(s)
- Yu Duan
- Department of Gastroenterology, The Third Hospital of Inner Mongolia Medical University (Inner Mongolia Baogang Hospital) Baotou 014010, Inner Mongolia Autonomous Region, China
| | - Ji Chen
- Department of Gastroenterology, The Third Hospital of Inner Mongolia Medical University (Inner Mongolia Baogang Hospital) Baotou 014010, Inner Mongolia Autonomous Region, China
| | - Hong Cui
- Department of Gastroenterology, The Third Hospital of Inner Mongolia Medical University (Inner Mongolia Baogang Hospital) Baotou 014010, Inner Mongolia Autonomous Region, China
| | - Cuijuan Zhao
- Department of Gastroenterology, The Third Hospital of Inner Mongolia Medical University (Inner Mongolia Baogang Hospital) Baotou 014010, Inner Mongolia Autonomous Region, China
| |
Collapse
|
11
|
Roberts TA, Wagner JA, Sandritter T, Black BT, Gaedigk A, Stancil SL. Retrospective Review of Pharmacogenetic Testing at an Academic Children's Hospital. Clin Transl Sci 2021; 14:412-421. [PMID: 33048453 PMCID: PMC7877836 DOI: 10.1111/cts.12895] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/02/2020] [Indexed: 12/28/2022] Open
Abstract
There is limited evidence to support pharmacogenetic (PGx) testing in children. We conducted a retrospective review of PGx testing among 452 patients at an academic children's hospital to determine the potential utility of PGx in diseases of childhood and to identify targets for future pediatric pharmacogenetic research. An actionable gene-drug pair associated with the 28 genes tested (Clinical Pharmacogenetics Implementation Consortium (CPIC) level A or B, Pharmacogenomics Knowledge Base (PharmGKB) level 1A or B, or US Food and Drug Administration (FDA) recommendation and a PharmGKB level) was present in 98.7% of patients. We identified 203 actionable gene-drug-diagnosis groups based on the indications for each actionable drug listed in Lexicomp. Among patients with an actionable gene-drug-diagnosis group, 49.3% had a diagnosis where the drug was a therapeutic option and PGx could be used to guide treatment selection. Among patients with an associated diagnosis, 30.9% had a prescription for the actionable drug allowing PGx guided dosing. Three genes (CYP2C19, CYP2D6, and CYP3A5) accounted for all the gene-drug-diagnosis groups with matching diagnoses and prescriptions. The most common gene-drug-diagnosis groups with matching diagnoses and prescriptions were CYP2C19-citalopram-escitalopram-depression 3.3% of patients tested; CYP2C19-dexlansoprazole-gastritis-esophagitis 3.1%; CYP2C19-omeprazole-gastritis-esophagitis 2.4%; CYP2D6-atomoxetine-attention deficit hyperactivity disorder 2.2%; and CYP2C19-citalopram-escitalopram-obsessive-compulsive disorder 1.5%. PGx could be used to guide selection of current treatment options or medication dosing in almost half (48.7%) of pediatric patients tested. Mood disorders and gastritis/esophagitis are promising targets for future study of PGx testing because of the high prevalence of these diagnoses and associated actionable gene-drug pairs in the pediatric population.
Collapse
Affiliation(s)
- Timothy A. Roberts
- Division of Adolescent MedicineChildren’s Mercy Kansas CityKansas CityMissouriUSA
- Department of PediatricsUMKC School of MedicineKansas CityMissouriUSA
| | - Jennifer A. Wagner
- Department of PediatricsUMKC School of MedicineKansas CityMissouriUSA
- Division of Clinical PharmacologyToxicology, and Therapeutic InnovationChildren’s Mercy Kansas CityKansas CityMissouriUSA
| | - Tracy Sandritter
- Division of Clinical PharmacologyToxicology, and Therapeutic InnovationChildren’s Mercy Kansas CityKansas CityMissouriUSA
| | - Benjamin T. Black
- Department of PediatricsUMKC School of MedicineKansas CityMissouriUSA
- Division of Developmental and Behavioral HealthChildren’s Mercy Kansas CityKansas CityMissouriUSA
| | - Andrea Gaedigk
- Department of PediatricsUMKC School of MedicineKansas CityMissouriUSA
- Division of Clinical PharmacologyToxicology, and Therapeutic InnovationChildren’s Mercy Kansas CityKansas CityMissouriUSA
| | - Stephani L. Stancil
- Division of Adolescent MedicineChildren’s Mercy Kansas CityKansas CityMissouriUSA
- Division of Clinical PharmacologyToxicology, and Therapeutic InnovationChildren’s Mercy Kansas CityKansas CityMissouriUSA
| |
Collapse
|
12
|
Rossow KM, Aka IT, Maxwell-Horn AC, Roden DM, Van Driest SL. Pharmacogenetics to Predict Adverse Events Associated With Antidepressants. Pediatrics 2020; 146:peds.2020-0957. [PMID: 33234666 PMCID: PMC7786826 DOI: 10.1542/peds.2020-0957] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES To determine the association between cytochrome P450 2C19 (CYP2C19) metabolizer status and risk for escitalopram and citalopram, collectively termed (es)citalopram, and sertraline adverse events (AEs) in children. METHODS In this retrospective cohort study, we used deidentified electronic health records linked to DNA. The cohort included children ≤18 years with ≥2 days of (es)citalopram or ≥7 days of sertraline exposure. The primary outcome was AEs assessed by manual chart review. CYP2C19 was genotyped for functional variants (*2, *3, *4, *6, *8, and *17), and individuals were assigned metabolizer status. Association between AEs and metabolizer status was determined by using Cox regression adjusting for age, race, ethnicity, dose, and concomitant CYP2C19-inhibiting medications. RESULTS The cohort included 249 sertraline-exposed and 458 (es)citalopram-exposed children, with a median age of 14.2 years (interquartile range 11.2-16.2) and 13.4 years (interquartile range 10.1-15.9), respectively. Sertraline AEs were more common in normal metabolizers (NMs) compared to poor metabolizers (PMs) or intermediate metabolizers (IMs) (hazard ratio [HR] 1.8; 95% confidence interval [CI] 1.01-3.2; P = .047) in unadjusted analysis and after adjustment (HR 1.9; CI 1.04-3.4; P = .04). For (es)citalopram, more AEs were observed in NMs than PMs and IMs without statistically significant differences (unadjusted HR 1.6; CI 0.95-2.6; P = .08; adjusted HR 1.6; CI 0.95-2.6; P = .08). CONCLUSIONS In contrast to adults, in our pediatric cohort, CYP2C19 NMs experienced increased sertraline AEs than PMs and IMs. (Es)citalopram AEs were not associated with CYP2C19 status in the primary analysis. The mechanism underlying this pediatric-specific finding is unknown but may be related to physiologic differences of adolescence. Further research is required to inform genotype-guided prescribing for these drugs in children.
Collapse
Affiliation(s)
| | - Ida T. Aka
- Departments of Pediatrics,,Contributed equally as co-first authors
| | | | - Dan M. Roden
- Medicine,,Pharmacology, and,Biomedical Informatics, School of Medicine, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
13
|
Dosage adjustment in obese children, even for common drugs, is largely unclear and a treat-to-effect approach may work best. DRUGS & THERAPY PERSPECTIVES 2020. [DOI: 10.1007/s40267-020-00734-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
14
|
Ameer B, Weintraub MA. Dosing Common Medications in Hospitalized Pediatric Patients with Obesity: A Review. Obesity (Silver Spring) 2020; 28:1013-1022. [PMID: 32441477 DOI: 10.1002/oby.22739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 12/17/2019] [Indexed: 12/31/2022]
Abstract
Medication management in children and adolescents with obesity is challenging because both developmental and pathophysiological changes may impact drug disposition and response. Evidence to date indicates an effect of obesity on drug disposition for certain drugs used in this population. This work identified published studies evaluating drug dosing, pharmacokinetics (PK), and effect in pediatric patients with obesity, focusing on 70 common medications used in a pediatric network of 42 US medical centers. A PubMed search revealed 33 studies providing PK and/or effectiveness data for 23% (16 of 70) of medications, 44% of which have just one study and can be considered exploratory. This work appraising 4 decades of literature shows several promising approaches: greater use of PK models applied to prospective clinical studies, dosing recommendations derived from both PK and safety, and multiyear effectiveness data on drugs for chronic conditions (e.g., asthma). Most studies make dose recommendations but are weakened by retrospective study design, small study populations, and no controls or historic controls. Dosing decisions continue to rely on extrapolating knowledge, including targeting systemic drug exposure typically achieved in adults. Optimal weight-based dosing strategies vary by drug and warrant prospective, controlled studies incorporating PK and modeling and simulation to complement clinical assessment.
Collapse
Affiliation(s)
- Barbara Ameer
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Michael A Weintraub
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Kyler KE, Wagner J, Hosey-Cojocari C, Watt K, Shakhnovich V. Drug Dose Selection in Pediatric Obesity: Available Information for the Most Commonly Prescribed Drugs to Children. Paediatr Drugs 2019; 21:357-369. [PMID: 31432433 PMCID: PMC7681556 DOI: 10.1007/s40272-019-00352-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity rates continue to rise in children, and little guidance exists regarding the need for adjustment away from total body weight-based doses for those prescribing drugs to this population of children. A majority of drugs prescribed to children with obesity result in either sub-therapeutic or supra-therapeutic concentrations, placing these children at risk for treatment failure and drug toxicities. In this review, we highlight available obesity-specific pharmacokinetic and dosing information for the most frequently prescribed drugs to children in the inpatient and outpatient clinical settings. We also comment on available dosing recommendations for drugs prescribed to treat common pediatric obesity-related comorbidities. This review highlights that there is no safe or proven 'rule of thumb,' for dosing drugs for children with obesity, and a striking lack of pharmacokinetic data to support the creation of dosing guidelines for children with obesity for the most commonly prescribed drugs. It is important that those prescribing for children with obesity are aware of these gaps in knowledge and of potential drug treatment failure or adverse events related to drug toxicity as a result of these knowledge gaps. Until more data are available, we recommend close monitoring of drug response and adverse events in children with obesity receiving commonly prescribed drugs.
Collapse
Affiliation(s)
- Kathryn E Kyler
- Children's Mercy Kansas City, 2401 Gillham Rd., Kansas City, MO, 64108, USA.
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA.
| | - Jonathan Wagner
- Children's Mercy Kansas City, 2401 Gillham Rd., Kansas City, MO, 64108, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | | | - Kevin Watt
- Duke University Medical Center, Durham, NC, USA
| | - Valentina Shakhnovich
- Children's Mercy Kansas City, 2401 Gillham Rd., Kansas City, MO, 64108, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| |
Collapse
|