15 results on '"Ware JA"'
Search Results
2. Chewing through challenges: Exploring the evolutionary pathways to wood-feeding in insects.
- Author
-
Beza-Beza CF, Wiegmann BM, Ware JA, Petersen M, Gunter N, Cole ME, Schwarz M, Bertone MA, Young D, and Mikaelyan A
- Subjects
- Animals, Feeding Behavior physiology, Mastication, Phylogeny, Biological Evolution, Gastrointestinal Microbiome, Insecta classification, Insecta microbiology, Insecta physiology, Wood
- Abstract
Decaying wood, while an abundant and stable resource, presents considerable nutritional challenges due to its structural rigidity, chemical recalcitrance, and low nitrogen content. Despite these challenges, certain insect lineages have successfully evolved saproxylophagy (consuming and deriving sustenance from decaying wood), impacting nutrient recycling in ecosystems and carbon sequestration dynamics. This study explores the uneven phylogenetic distribution of saproxylophagy across insects and delves into the evolutionary origins of this trait in disparate insect orders. Employing a comprehensive analysis of gut microbiome data, from both saproxylophagous insects and their non-saproxylophagous relatives, including new data from unexplored wood-feeding insects, this Hypothesis paper discusses the broader phylogenetic context and potential adaptations necessary for this dietary specialization. The study proposes the "Detritivore-First Hypothesis," suggesting an evolutionary pathway to saproxylophagy through detritivory, and highlights the critical role of symbiotic gut microbiomes in the digestion of decaying wood., (© 2024 The Authors. BioEssays published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
3. Bioequivalence and Relative Bioavailability Studies to Assess a New Acalabrutinib Formulation That Enables Coadministration With Proton-Pump Inhibitors.
- Author
-
Sharma S, Pepin X, Burri H, Zheng L, Kuptsova-Clarkson N, de Jong A, Yu T, MacArthur HL, Majewski M, Byrd JC, Furman RR, Ware JA, Mann J, Ramies D, Munugalavadla V, Sheridan L, and Tomkinson H
- Subjects
- Adult, Humans, Biological Availability, Therapeutic Equivalency, Tablets, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Proton Pump Inhibitors adverse effects, Proton Pump Inhibitors pharmacokinetics, Pyrazines adverse effects, Pyrazines pharmacokinetics
- Abstract
Acalabrutinib is a Bruton tyrosine kinase (BTK) inhibitor approved to treat adults with chronic lymphocytic leukemia, small lymphocytic lymphoma, or previously treated mantle cell lymphoma. As the bioavailability of the acalabrutinib capsule (AC) depends on gastric pH for solubility and is impaired by acid-suppressing therapies, coadministration with proton-pump inhibitors (PPIs) is not recommended. Three studies in healthy subjects (N = 30, N = 66, N = 20) evaluated the pharmacokinetics (PKs), pharmacodynamics (PDs), safety, and tolerability of acalabrutinib maleate tablet (AT) formulated with pH-independent release. Subjects were administered AT or AC (orally, fasted state), AT in a fed state, or AT in the presence of a PPI, and AT or AC via nasogastric (NG) route. Acalabrutinib exposures (geometric mean [% coefficient of variation, CV]) were comparable for AT versus AC (AUC
inf 567.8 ng h/mL [36.9] vs 572.2 ng h/mL [38.2], Cmax 537.2 ng/mL [42.6] vs 535.7 ng/mL [58.4], respectively); similar results were observed for acalabrutinib's active metabolite (ACP-5862) and for AT-NG versus AC-NG. The geometric mean Cmax for acalabrutinib was lower when AT was administered in the fed versus the fasted state (Cmax 255.6 ng/mL [%CV, 46.5] vs 504.9 ng/mL [49.9]); AUCs were similar. For AT + PPI, geometric mean Cmax was lower (371.9 ng/mL [%CV, 81.4] vs 504.9 ng/mL [49.9]) and AUCinf was higher (AUCinf 694.1 ng h/mL [39.7] vs 559.5 ng h/mL [34.6]) than AT alone. AT and AC were similar in BTK occupancy. Most adverse events were mild with no new safety concerns. Acalabrutinib formulations were comparable and AT could be coadministered with PPIs, food, or via NG tube without affecting the PKs or PDs., (© 2022 AstraZeneca. Clinical Pharmacology in Drug Development published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.)- Published
- 2022
- Full Text
- View/download PDF
4. Evaluation of the Pharmacokinetics and Safety of a Single Dose of Acalabrutinib in Subjects With Hepatic Impairment.
- Author
-
Xu Y, Izumi R, Nguyen H, Kwan A, Kuo H, Madere J, Slatter JG, Podoll T, Vishwanathan K, Marbury T, Smith W, Preston RA, Sharma S, and Ware JA
- Subjects
- Adult, Area Under Curve, Benzamides adverse effects, Humans, Pyrazines adverse effects, Liver Diseases metabolism
- Abstract
Acalabrutinib received approval for the treatment of adult patients with mantle cell lymphoma who received at least 1 prior therapy and adult patients with chronic lymphocytic leukemia or small lymphocytic lymphoma. This study investigated the impact of hepatic impairment (HI) on acalabrutinib pharmacokinetics (PK) and safety at a single 50-mg dose in fasted subjects. This study was divided into 2 parts: study 1, an open-label, parallel-group study in Child-Pugh class A or B subjects and healthy subjects; and study 2, an open-label, parallel-group study in Child-Pugh class C subjects and healthy subjects. Baseline characteristics and safety profiles were similar across groups. Acalabrutinib exposure (area under the plasma concentration-time curve) increased slightly (1.90- and 1.48-fold) in subjects with mild (Child-Pugh class A) and moderate (Child-Pugh class B) hepatic impairment compared with healthy subjects. In severe hepatic impairment (Child-Pugh class C), acalabrutinib exposure (area under the plasma concentration-time curve and maximum plasma concentration) increased ≈5.0- and 3.6-fold, respectively. Results were consistent across total and unbound exposures. Severe hepatic impairment did not impact total/unbound metabolite (ACP-5862) exposures; the metabolite-to-parent ratio decreased to 0.6 to 0.8 (vs 3.1-3.6 in healthy subjects). In summary, single oral dose of 50-mg acalabrutinib was safe and well tolerated in subjects with mild, moderate, and severe hepatic impairment and in healthy control subjects. In subjects with severe hepatic impairment, mean acalabrutinib exposure increased by up to 5-fold and should be avoided. Acalabrutinib does not require dose adjustment in patients with mild or moderate hepatic impairment., (© 2021 The Authors. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.)
- Published
- 2022
- Full Text
- View/download PDF
5. Quantitative systems pharmacology model-based investigation of adverse gastrointestinal events associated with prolonged treatment with PI3-kinase inhibitors.
- Author
-
Gadkar K, Friedrich C, Hurez V, Ruiz ML, Dickmann L, Kumar Jolly M, Schutt L, Jin J, Ware JA, and Ramanujan S
- Subjects
- Diarrhea chemically induced, Humans, Network Pharmacology, Phosphoinositide-3 Kinase Inhibitors, Protein Isoforms, Colitis chemically induced, Phosphatidylinositol 3-Kinases
- Abstract
Several PI3K inhibitors are in clinical development for the treatment of various forms of cancers, including pan-PI3K inhibitors targeting all four PI3K isoforms (α, β, γ, and δ), and isoform-selective inhibitors. Diarrhea and immune-mediated colitis are among the adverse events observed with PI3K inhibition which limits the maximal tolerated dose. A quantitative systems pharmacology model was developed to investigate PI3K-inhibitor-induced colitis. The effects of individual PI3K isoforms on relevant cellular pathways were incorporated into a mechanistic representation of mucosal inflammation. A virtual clinical population captures the observed clinical variability in the onset timing and rates of diarrhea and colitis for seven clinically tested PI3K inhibitors. Model-based analysis suggests that colitis development is governed by both the inhibition of PI3Kδ, which drives T cell differentiation and proliferation, and PI3Kα, which regulates epithelial barrier integrity. Specifically, when PI3Kα is inhibited below a given threshold, epithelial barrier dysfunction precipitates an exaggerated T effector response due to PI3Kδ-inhibition, leading to risk of diarrhea and colitis. This synergy explains why the lowest diarrhea and colitis rates are seen with the weakest PI3Kδ inhibition (alpelisib), and higher rates are seen with strong PI3Kδ inhibition if PI3Kα is even mildly inhibited (e.g., idelalisib), whereas strong PI3Kδ inhibition in the absence of PI3Kα inhibition does not result in high colitis rates (umbralisib). Thus, the model-based analysis suggests that PI3Kα and δ inhibition play unique but synergistic roles in driving colitis. Finally, we explore if and how dose-regimen might influence colitis rates for molecules that inhibit both PI3Kα and PI3Kδ., (© 2021 Genentech, Inc. CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2022
- Full Text
- View/download PDF
6. ITC Commentary on Metformin Clinical Drug-Drug Interaction Study Design That Enables an Efficacy- and Safety-Based Dose Adjustment Decision.
- Author
-
Zamek-Gliszczynski MJ, Chu X, Cook JA, Custodio JM, Galetin A, Giacomini KM, Lee CA, Paine MF, Ray AS, Ware JA, Wittwer MB, and Zhang L
- Subjects
- Animals, Biomarkers blood, Blood Glucose drug effects, Blood Glucose metabolism, Computer Simulation, Dose-Response Relationship, Drug, Drug Dosage Calculations, Drug Interactions, Glucose Tolerance Test, Humans, Hypoglycemic Agents adverse effects, Hypoglycemic Agents pharmacokinetics, Lactic Acid blood, Metformin adverse effects, Metformin pharmacokinetics, Models, Biological, Pharmacogenetics, Polypharmacy, Renal Elimination, Risk Assessment, Drug Development methods, Hypoglycemic Agents administration & dosage, Metformin administration & dosage, Research Design
- Abstract
Metformin drug-drug interaction (DDI) studies are conducted during development of drugs that inhibit organic cation transporters and/or multidrug and toxin extrusion proteins (OCTs/MATEs). Monitoring solely changes in systemic exposure, the typical DDI study endpoint appears inadequate for metformin, which is metabolically stable, has poor passive membrane permeability, and undergoes transporter-mediated tissue distribution and clearance. Evaluation of renal clearance, antihyperglycemic effects, and potentially lactate as an exploratory safety marker, can support rational metformin dose adjustment. The proposed DDI study design aims to adequately inform metformin dosing during comedication., (© 2018, The American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2018
- Full Text
- View/download PDF
7. Disease-Associated Changes in Drug Transporters May Impact the Pharmacokinetics and/or Toxicity of Drugs: A White Paper From the International Transporter Consortium.
- Author
-
Evers R, Piquette-Miller M, Polli JW, Russel FGM, Sprowl JA, Tohyama K, Ware JA, de Wildt SN, Xie W, and Brouwer KLR
- Subjects
- Animals, Drug Interactions, Drug-Related Side Effects and Adverse Reactions etiology, Drug-Related Side Effects and Adverse Reactions metabolism, Humans, Membrane Transport Modulators metabolism, Membrane Transport Proteins genetics, Models, Biological, Risk Assessment, Acute Disease, Chronic Disease drug therapy, Membrane Transport Modulators pharmacology, Membrane Transport Proteins drug effects, Membrane Transport Proteins metabolism, Pharmaceutical Preparations metabolism, Pharmacokinetics
- Abstract
Drug transporters are critically important for the absorption, distribution, metabolism, and excretion (ADME) of many drugs and endogenous compounds. Therefore, disruption of these pathways by inhibition, induction, genetic polymorphisms, or disease can have profound effects on overall physiology, drug pharmacokinetics, drug efficacy, and toxicity. This white paper provides a review of changes in transporter function associated with acute and chronic disease states, describes regulatory pathways affecting transporter expression, and identifies opportunities to advance the field., (© 2018, The American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2018
- Full Text
- View/download PDF
8. Combining "Bottom-up" and "Top-down" Approaches to Assess the Impact of Food and Gastric pH on Pictilisib (GDC-0941) Pharmacokinetics.
- Author
-
Lu T, Fraczkiewicz G, Salphati L, Budha N, Dalziel G, Smelick GS, Morrissey KM, Davis JD, Jin JY, and Ware JA
- Subjects
- Administration, Oral, Biological Availability, Computer Simulation, Cross-Over Studies, Diet, High-Fat, Female, Healthy Volunteers, Humans, Hydrogen-Ion Concentration, Male, Models, Biological, Random Allocation, Anti-Ulcer Agents administration & dosage, Indazoles administration & dosage, Indazoles pharmacokinetics, Intestines chemistry, Rabeprazole administration & dosage, Sulfonamides administration & dosage, Sulfonamides pharmacokinetics
- Abstract
Pictilisib, a weakly basic compound, is an orally administered, potent, and selective pan-inhibitor of phosphatidylinositol 3-kinases for oncology indications. To investigate the significance of high-fat food and gastric pH on pictilisib pharmacokinetics (PK) and enable label recommendations, a dedicated clinical study was conducted in healthy volunteers, whereby both top-down (population PK, PopPK) and bottom-up (physiologically based PK, PBPK) approaches were applied to enhance confidence of recommendation and facilitate the clinical development through scenario simulations. The PopPK model identified food (for absorption rate constant (K
a )) and proton pump inhibitors (PPI, for relative bioavailability (Frel ) and Ka ) as significant covariates. Food and PPI also impacted the variability of Frel . The PBPK model accounted for the supersaturation tendency of pictilisib, and gastric emptying physiology successfully predicted the food and PPI effect on pictilisib absorption. Our research highlights the importance of applying both quantitative approaches to address critical drug development questions., (© 2017 The Authors CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.)- Published
- 2017
- Full Text
- View/download PDF
9. A multicenter, single-arm, open-label, phase 2 study of apitolisib (GDC-0980) for the treatment of recurrent or persistent endometrial carcinoma (MAGGIE study).
- Author
-
Makker V, Recio FO, Ma L, Matulonis UA, Lauchle JO, Parmar H, Gilbert HN, Ware JA, Zhu R, Lu S, Huw LY, Wang Y, Koeppen H, Spoerke JM, Lackner MR, and Aghajanian CA
- Abstract
Background: The current single-arm, open-label trial was designed to evaluate the activity of apitolisib (GDC-0980), a dual phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) inhibitor, in patients with advanced endometrial cancer (EC)., Methods: Patients with recurrent or persistent EC who were treated with 1 to 2 prior lines of chemotherapy but no prior PI3K/mTOR inhibitor received oral apitolisib at a dose of 40 mg daily during 28-day cycles until disease progression or intolerable toxicity occurred. Patients with type I/II diabetes who required insulin were excluded. The primary endpoints were progression-free survival (PFS) at 6 months and objective response rate., Results: A total of 56 women were enrolled, including 13 (23%) with well-controlled diabetes. Reasons for discontinuation were disease progression (24 patients; 43%), adverse events (13 patients; 23%), and withdrawal by subject (12 patients; 21%). Grade 3/4 apitolisib-related adverse events were hyperglycemia (46%), rash (30%), colitis (5%), and pneumonitis (4%) (toxicities were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). The PFS rate at 6 months was 20% (Kaplan-Meier estimate; 95% confidence interval [95% CI], 7%-33%). The objective response rate was 6% (confirmed). The median PFS was 3.5 months (95% CI, 2.7-3.7 months) and the median overall survival was 15.7 months (95% CI, 9.2-17.0 months). Nineteen patients discontinued the study before the first tumor assessment. Dose reductions were required for 4 diabetic (31%) and 18 nondiabetic (42%) patients. Comprehensive molecular profiling of 46 evaluable archival tumor samples demonstrated that 57% of patients had at least 1 alteration in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), phosphatase and tensin homolog (PTEN), or AKT1. All 3 patients with a confirmed response had at least 1 alteration in a PI3K pathway gene., Conclusions: The antitumor activity noted with the use of a dose of 40 mg of apitolisib daily was limited by tolerability, especially in diabetic patients. Patients with PI3K pathway mutations may have derived enhanced benefit from apitolisib. Cancer 2016;122:3519-28. © 2016 American Cancer Society., (© 2016 American Cancer Society.)
- Published
- 2016
- Full Text
- View/download PDF
10. Response to "Drug interactions produced by proton pump inhibitors: not simply a pH effect".
- Author
-
Budha NR, Benet LZ, and Ware JA
- Subjects
- Humans, Anticarcinogenic Agents therapeutic use, Gastroesophageal Reflux drug therapy, Neoplasms drug therapy, Proton Pump Inhibitors therapeutic use
- Published
- 2013
- Full Text
- View/download PDF
11. Drug absorption interactions between oral targeted anticancer agents and PPIs: is pH-dependent solubility the Achilles heel of targeted therapy?
- Author
-
Budha NR, Frymoyer A, Smelick GS, Jin JY, Yago MR, Dresser MJ, Holden SN, Benet LZ, and Ware JA
- Subjects
- Anticarcinogenic Agents pharmacokinetics, Drug Interactions, Gastric Acid, Gastric Acidity Determination, Gastroesophageal Reflux metabolism, Humans, Hydrogen-Ion Concentration, Intestinal Absorption, Neoplasms metabolism, Proton Pump Inhibitors pharmacokinetics, Solubility, Anticarcinogenic Agents therapeutic use, Gastroesophageal Reflux drug therapy, Neoplasms drug therapy, Proton Pump Inhibitors therapeutic use
- Abstract
A majority of the novel orally administered, molecularly targeted anticancer therapies are weak bases that exhibit pH-dependent solubility, and suppression of gastric acidity with acid-reducing agents could impair their absorption. In addition, a majority of cancer patients frequently take acid-reducing agents to alleviate symptoms of gastroesophageal reflux disease, thereby raising the potential for a common but underappreciated drug-drug interaction (DDI) that could decrease the exposure of anticancer medication and result in subsequent failure of therapy. This article is a review of the available clinical literature describing the extent of the interaction between 15 orally administered, small-molecule targeted anticancer therapies and acid-reducing agents. The currently available clinical data suggest that the magnitude of this DDI is largest for compounds whose in vitro solubility varies over the pH range 1-4. This range represents the normal physiological gastric acidity (pH ~1) and gastric acidity while on an acid-reducing agent (pH ~4).
- Published
- 2012
- Full Text
- View/download PDF
12. Drug-drug interactions mediated through P-glycoprotein: clinical relevance and in vitro-in vivo correlation using digoxin as a probe drug.
- Author
-
Fenner KS, Troutman MD, Kempshall S, Cook JA, Ware JA, Smith DA, and Lee CA
- Subjects
- Amiodarone metabolism, Amiodarone pharmacology, Caco-2 Cells, Digoxin pharmacology, Drug Interactions, Humans, Pharmaceutical Preparations metabolism, Retrospective Studies, ATP Binding Cassette Transporter, Subfamily B, Member 1 antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Digoxin metabolism
- Abstract
The clinical pharmacokinetics and in vitro inhibition of digoxin were examined to predict the P-glycoprotein (P-gp) component of drug-drug interactions. Coadministered drugs (co-meds) in clinical trials (N = 123) resulted in a small,
0.1 is predictive of clinical digoxin interactions (AUC and C(max)). - Published
- 2009
- Full Text
- View/download PDF
13. Pharmacogenetic characterization of sulfasalazine disposition based on NAT2 and ABCG2 (BCRP) gene polymorphisms in humans.
- Author
-
Yamasaki Y, Ieiri I, Kusuhara H, Sasaki T, Kimura M, Tabuchi H, Ando Y, Irie S, Ware J, Nakai Y, Higuchi S, and Sugiyama Y
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2, ATP-Binding Cassette Transporters metabolism, Adult, Arylamine N-Acetyltransferase metabolism, Humans, Male, Neoplasm Proteins metabolism, Pharmacogenetics, Polymorphism, Genetic drug effects, Sulfasalazine blood, ATP-Binding Cassette Transporters genetics, Arylamine N-Acetyltransferase genetics, Neoplasm Proteins genetics, Polymorphism, Genetic genetics, Sulfasalazine pharmacokinetics
- Abstract
The role of breast cancer resistance protein (BCRP), an efflux ABC transporter, in the pharmacokinetics of substrate drugs in humans is unknown. We investigated the impact of genetic polymorphisms of ABCG2 (421C>A) and NAT2 on the pharmacokinetics of sulfasalazine (SASP), a dual substrate, in 37 healthy volunteers, taking 2,000 mg of conventional SASP tablets. In ABCG2, SASP AUC(0-48) of C/C, C/A, and A/A subjects was 171 +/- 85, 330 +/- 194, and 592 +/- 275 microg h/ml, respectively, with significant differences among groups. In contrast, AUC(0-48) of sulfapyridine (SP) tended to be lower in subjects with the ABCG2-A allele as homozygosity. In NAT2, AUC(AcSP)/AUC(SP) was significantly higher in rapid than in intermediate and slow acetylator (SA) genotypes. We successfully described the pharmacokinetics of SASP, SP, and N -acetylsulfapyridine (AcSP) simultaneously by nonlinear mixed-effects modeling (NONMEM) analysis with regard to both gene polymorphisms. The data indicate that SASP is a candidate probe of BCRP, particularly in its role in intestinal absorption.
- Published
- 2008
- Full Text
- View/download PDF
14. Disposition of procainamide in patients with chronic congestive heart failure receiving medical therapy.
- Author
-
Tisdale JE, Rudis MI, Padhi ID, Borzak S, Svensson CK, Webb CR, Acciaioli J, Ware JA, Krepostman A, and Zarowitz BJ
- Subjects
- Adult, Aged, Angiotensin-Converting Enzyme Inhibitors therapeutic use, Anti-Arrhythmia Agents adverse effects, Anti-Arrhythmia Agents therapeutic use, Digoxin therapeutic use, Diuretics therapeutic use, Female, Heart Failure drug therapy, Humans, Male, Middle Aged, Procainamide adverse effects, Procainamide therapeutic use, Anti-Arrhythmia Agents pharmacokinetics, Heart Failure metabolism, Procainamide pharmacokinetics
- Abstract
Dosage reduction of procainamide has been recommended in patients with congestive heart failure (CHF). However, these recommendations are based primarily on studies with unmatched control groups, suboptimal blood sampling, and in patients not receiving angiotensin-converting enzyme (ACE) inhibitors. These agents increase renal blood flow, which theoretically may offset alterations in drug disposition in patients with CHF. The pharmacokinetics of procainamide in patients with chronic CHF and in matched controls were compared. A single intravenous dose of 750 mg of procainamide was administered to 9 patients with chronic New York Heart Association (NYHA) class II or III CHF (mean +/- SD left ventricular ejection fraction 22 +/- 9%) receiving medical therapy and 7 control subjects matched for age and gender. Blood and urine samples were collected at intervals over a period of 48 and 72 hours, respectively. Patients with CHF and control subjects were demographically similar, with the exception of concomitant medications, including ACE inhibitors (8/9 versus 1/7, respectively). There were no significant differences between patients with CHF and control subjects in mean +/- SD peak serum concentrations (Cmax), area under the serum concentration-time curve (AUC0-infinity), total clearance, renal clearance, half-life (t1/2), or volume of distribution (Vd) of procainamide. Similarly, there were no significant differences between patients with CHF and control subjects in the mean +/- SD Cmax, AUC0-infinity, renal clearance, or t1/2 of N-acetylprocainamide (NAPA). Procainamide dosage reduction may not be necessary in patients with chronic stable CHF who are receiving medical therapy.
- Published
- 1996
- Full Text
- View/download PDF
15. Inhibition of N-acetylation of procainamide and renal clearance of N-acetylprocainamide by para-aminobenzoic acid in humans.
- Author
-
Tisdale JE, Rudis MI, Padhi ID, Svensson CK, Webb CR, Borzak S, Ware JA, Krepostman A, and Zarowitz BJ
- Subjects
- Acecainide metabolism, Acetylation drug effects, Administration, Oral, Adult, Anti-Arrhythmia Agents pharmacokinetics, Cross-Over Studies, Drug Interactions, Female, Humans, Injections, Intravenous, Kidney drug effects, Male, Procainamide pharmacokinetics, 4-Aminobenzoic Acid pharmacology, Acecainide pharmacokinetics, Anti-Arrhythmia Agents metabolism, Kidney metabolism, Procainamide metabolism, Sunscreening Agents pharmacology
- Abstract
Procainamide administration often results in excessively high serum N-acetylprocainamide (NAPA) concentrations and subtherapeutic serum procainamide concentrations. Inhibition of N-acetylation of procainamide may prevent accumulation of excessive NAPA while maintaining therapeutic serum procainamide concentrations. The purpose of this randomized, two-way crossover study was to determine if para-aminobenzoic acid (PABA) inhibits N-acetylation of procainamide in healthy volunteers. Eleven (7 female, 4 male) fast acetylators of caffeine received, in random order, PABA 1.5 g orally every 6 hours for 5 days, with a single intravenous dose of procainamide 750 mg administered over 30 minutes on the third day, or intravenous procainamide alone. Blood samples were collected during a 48-hour period after initiation of the infusion. Urine was collected over a 72-hour period. Serum procainamide and NAPA concentrations were analyzed using fluorescence polarization immunoassay. Urine procainamide and NAPA concentrations were measured with high performance liquid chromatography. PABA did not significantly influence total or renal procainamide clearance, elimination rate constant, AUC0-00, amount of procainamide excreted unchanged in the urine, or volume of distribution. However, concomitant PABA administration with procainamide resulted in increases in NAPA AUC0-00 and t1/2 and reductions in NAPA Ke, procainamide acetylation (NAPA formation) clearance, and NAPA renal clearance. Although PABA inhibits metabolic conversion of procainamide to NAPA, it also impairs the renal clearance of NAPA (but not procainamide) in healthy subjects. Therefore, PABA may not be useful for optimizing the safety of efficacy of procainamide in patients.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.