Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 157
1.
JAMA Netw Open ; 4(6): e2118253, 2021 06 01.
Article En | MEDLINE | ID: mdl-34181009

Importance: A publication reported that N-nitrosodimethylamine (NDMA), a probable human carcinogen, was formed when ranitidine and nitrite were added to simulated gastric fluid. However, the nitrite concentrations used were greater than the range detected in acidic gastric fluid in prior clinical studies. Objective: To characterize NDMA formation following the addition of ranitidine to simulated gastric fluid using combinations of fluid volume, pH levels, and nitrite concentrations, including physiologic levels. Design, Setting, and Participants: One 150-mg ranitidine tablet was added to 50 or 250 mL of simulated gastric fluid with a range of nitrite concentrations from the upper range of physiologic (100 µmol/L) to higher concentrations (10 000 µmol/L) with a range of pH levels. NDMA amounts were assessed with a liquid chromatography-mass spectrometry method. Main Outcomes and Measures: NDMA detected in simulated gastric fluid 2 hours after adding ranitidine. Results: At a supraphysiologic nitrite concentration (ie, 10 000 µmol/L), the mean (SD) amount of NDMA detected in 50 mL simulated gastric fluid 2 hours after adding ranitidine increased from 222 (12) ng at pH 5 to 11 822 (434) ng at pH 1.2. Subsequent experiments with 50 mL of simulated gastric fluid at pH 1.2 with no added nitrite detected a mean (SD) of 22 (2) ng of NDMA, which is the background amount present in the ranitidine tablets. Similarly, at the upper range of physiologic nitrite (ie, 100 µmol/L) or at nitrite concentrations as much as 50-fold greater (1000 or 5000 µmol/L) only background mean (SD) amounts of NDMA were observed (21 [3] ng, 24 [2] ng, or 24 [3] ng, respectively). With 250 mL of simulated gastric fluid, no NDMA was detected at the upper physiologic range (100 µmol/L) or 10-fold physiologic (1000 µmol/L) nitrite concentrations, while NDMA was detected (mean [SD] level, 7353 [183] ng) at a 50-fold physiologic nitrite concentration (5000 µmol/L). Conclusions and Relevance: In this in vitro study of ranitidine tablets added to simulated gastric fluid with different nitrite concentrations, ranitidine conversion to NDMA was not detected until nitrite was 5000 µmol/L, which is 50-fold greater than the upper range of physiologic gastric nitrite concentrations at acidic pH. These findings suggest that ranitidine is not converted to NDMA in gastric fluid at physiologic conditions.


Dimethylnitrosamine/metabolism , Gastrointestinal Absorption/physiology , Ranitidine/analysis , Histamine H2 Antagonists/analysis , Histamine H2 Antagonists/blood , Humans , Ranitidine/blood
2.
Anal Bioanal Chem ; 413(4): 1117-1125, 2021 Feb.
Article En | MEDLINE | ID: mdl-33409672

Biocompatible and luminescent nanostructures synthesized by capping gold-carbon nanoparticles (HOOC-4-C6H4-AuNPs) with amino acids tyrosine, tryptophan, and cysteine were used for the quantitative estimation of ranitidine (RNH), a peptic ulcer and gastroesophageal reflux drug. We applied a fluorescence quenching mechanism to investigate the viability of the energy transfer based on gold-carbon nanosensors. Förster resonance energy transfer (FRET) calculations showed a donor-acceptor distance of 1.69 nm (Tyr@AuNPs), 2.27 nm (Trp@AuNPs), and 2.32 nm (Cys@AuNPs). The constant time-resolved fluorescence lifetime measurements supported the static quenching nature. This method was successfully utilized in the detection and quantification of RNH, with a limit of detection (LOD) of 0.174, 0.56, and 0.332 µM for Tyr@AuNP, Trp@AuNP, and Cys@AuNP bioconjugates, respectively. This approach was also successful in the quantification of RNH in spiked serum samples.


Amino Acids/chemistry , Anti-Ulcer Agents/blood , Fluorescent Dyes/chemistry , Gold/chemistry , Nanoparticles/chemistry , Ranitidine/blood , Carbon/chemistry , Fluorescence Resonance Energy Transfer/methods , Humans , Limit of Detection
3.
J Clin Pharmacol ; 60(3): 312-323, 2020 03.
Article En | MEDLINE | ID: mdl-31542894

Trospium chloride, a muscarinic receptor blocker, is poorly absorbed with different rates from areas in the jejunum and the cecum/ascending colon. To evaluate whether organic cation transporter (OCT) 1, OCT2 and multidrug and toxin extrusion (MATE) 1 and MATE2-K are involved in pharmacokinetics, competitions with ranitidine, a probe inhibitor of the cation transporters, were evaluated in transfected HEK293 cells. Furthermore, a drug interaction study with trospium chloride after intravenous (2 mg) and oral dosing (30 mg) plus ranitidine (300 mg) was performed in 12 healthy subjects and evaluated by noncompartmental analysis and population pharmacokinetic modeling. Ranitidine inhibited OCT1, OCT2, MATE1, and MATE2-K with half maximal inhibitory concentration values of 186 ± 25 µM, 482 ± 105 µM, 134 ± 37 µM, and 35 ± 11 µM, respectively. In contrast to our hypothesis, coadministration of ranitidine did not significantly decrease oral absorption of trospium. Instead, renal clearance was lowered by ∼15% (530 ± 99 vs 460 ± 120 mL/min; P < .05). It is possible that ranitidine was not available in competitive concentrations at the major colonic absorption site, as the inhibitor is absorbed in the small intestine and undergoes degradation by microbiota. The renal effects apparently result from inhibition of MATE1 and/or MATE2-K by ranitidine as predicted by in vitro to in vivo extrapolation. However, all pharmacokinetic changes were not of clinical relevance for the drug with highly variable pharmacokinetics. Intravenous trospium significantly lowered mean absorption time and relative bioavailability of ranitidine, which was most likely caused by muscarinic receptor blocking effects on intestinal motility and water turnover.


Benzilates/adverse effects , Benzilates/pharmacokinetics , Muscarinic Antagonists/adverse effects , Muscarinic Antagonists/pharmacokinetics , Nortropanes/adverse effects , Nortropanes/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Ranitidine/pharmacology , Ranitidine/pharmacokinetics , Administration, Intravenous , Administration, Oral , Adult , Benzilates/administration & dosage , Benzilates/blood , Biological Availability , Cells, Cultured , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Muscarinic Antagonists/administration & dosage , Muscarinic Antagonists/blood , Nortropanes/administration & dosage , Nortropanes/blood , Organic Cation Transport Proteins/antagonists & inhibitors , Ranitidine/administration & dosage , Ranitidine/blood
4.
Anal Chim Acta ; 1095: 118-128, 2020 Jan 25.
Article En | MEDLINE | ID: mdl-31864612

A novel liquid-liquid-solid membrane microextraction (LLSMME) method which integrates hollow fiber liquid phase microextraction (HF-LPME) and solid phase microextraction (SPME) was developed for bio-sample preparation. The homogeneous zeolitic imidazolate framework 8 mixed matrix membrane (ZIF-8-MMM) was prepared by in situ self-assembly of ZIF-8 on the inner surface of hollow fiber membrane and employed as a flexible LLSMME device. Incorporating the advantages of both HF-LPME and SPME, the as-fabricated ZIF-8-MMM exhibited excellent performance on the extraction and preconcentration of small molecule drugs of different polarity from complex biological matrices. As a case study, ZIF-8-MMM-based LLSMME coupled with UPLC-MS/MS were developed and validated for determination of ibuprofen, simvastatin and ranitidine at trace levels in rat plasma. The method showed good linearity (r2 > 0.99) and repeatability (RSD < 15%), low limits of detection (2-3 ng mL-1) and high relative recoveries (97.42-103.8%). The enrichment factors were between 87.3 and 112.6. Our study provided a promising strategy for developing more efficient, cost-effective and environmentally friendly technique for bio-sample pretreatment.


Ibuprofen/blood , Liquid Phase Microextraction/methods , Ranitidine/blood , Simvastatin/blood , Solid Phase Microextraction/methods , Zeolites/chemistry , Animals , Chromatography, High Pressure Liquid , Ibuprofen/isolation & purification , Imidazoles/chemistry , Limit of Detection , Membranes, Artificial , Molecular Docking Simulation , Ranitidine/isolation & purification , Rats, Sprague-Dawley , Reproducibility of Results , Simvastatin/isolation & purification , Tandem Mass Spectrometry
5.
Eur J Pharm Sci ; 107: 249-255, 2017 Sep 30.
Article En | MEDLINE | ID: mdl-28709912

The pharmaceutical excipient, polyethylene glycol 400 (PEG 400), unexpectedly alters the bioavailability of the BCS class III drug ranitidine in a sex-dependent manner. As ranitidine is a substrate for the efflux transporter P-glycoprotein (P-gp), we hypothesized that the sex-related influence could be due to interactions between PEG 400 and P-gp. In this study, we tested this hypothesis by: i) measuring the influence of PEG 400 on the oral bioavailability of another P-gp substrate (ampicillin) and of a non-P-gp substrate (metformin); and ii) measuring the effect of PEG 400 on drug bioavailability in the presence of a P-gp inhibitor (cyclosporine A) in male and female rats. We found that PEG 400 significantly increased (p<0.05) the bioavailability of ampicillin (the P-gp substrate) in male rats, but not in female ones. In contrast, PEG 400 had no influence on the bioavailability of the non-P-gp substrate, metformin in male or female rats. Inhibition of P-gp by oral pre-treatment with cyclosporine A increased the bioavailability of the P-gp substrates (ampicillin and ranitidine) in males and females (p<0.05), and to a greater extent in males, but had no influence on the bioavailability of metformin in either male or female rats. These results prove the hypothesis that the sex-specific effect of PEG 400 on the bioavailability of certain drugs is due to the interaction of PEG 400 with the efflux transporter P-gp.


Ampicillin/pharmacokinetics , Excipients/pharmacology , Metformin/pharmacokinetics , Polyethylene Glycols/pharmacology , Ranitidine/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Ampicillin/blood , Animals , Biological Availability , Cyclosporine/pharmacology , Female , Male , Metformin/blood , Ranitidine/blood , Rats, Wistar , Sex Factors
6.
J Vet Pharmacol Ther ; 40(1): 92-96, 2017 Jan.
Article En | MEDLINE | ID: mdl-27292271

The use of anti-ulcer medications, such as cimetidine, ranitidine, and omeprazole, is common in performance horses. The use of these drugs is regulated in performance horses, and as such a withdrawal time is necessary prior to competition to avoid a medication violation. To the authors' knowledge, there are no reports in the literature describing repeated oral administrations of these drugs in the horse to determine a regulatory threshold and related withdrawal time recommendations. Therefore, the objective of the current study was to describe the disposition and elimination pharmacokinetics of these anti-ulcer medications following oral administration to provide data upon which appropriate regulatory recommendations can be established. Nine exercised Thoroughbred horses were administered 20 mg/kg BID of cimetidine or 8 mg/kg BID of ranitidine, both for seven doses or 2.28 g of omeprazole SID for four doses. Blood samples were collected, serum drug concentrations were determined, and elimination pharmacokinetic parameters were calculated. The serum elimination half-life was 7.05 ± 1.02, 7.43 ± 0.851 and 3.94 ± 1.04 h for cimetidine, ranitidine, and omeprazole, respectively. Serum cimetidine and ranitidine concentrations were above the LOQ and omeprazole and omeprazole sulfide below the LOQ in all horses studied upon termination of sample collection.


Anti-Ulcer Agents/pharmacokinetics , Cimetidine/pharmacokinetics , Horses/metabolism , Omeprazole/pharmacokinetics , Ranitidine/pharmacokinetics , Administration, Oral , Animals , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/blood , Cimetidine/administration & dosage , Cimetidine/blood , Drug Administration Schedule/veterinary , Female , Half-Life , Male , Omeprazole/administration & dosage , Omeprazole/blood , Physical Conditioning, Animal , Ranitidine/administration & dosage , Ranitidine/blood
7.
BMC Pediatr ; 16: 90, 2016 07 13.
Article En | MEDLINE | ID: mdl-27412521

BACKGROUND: The aim of this study was to determine the effect of gestational age on pharmacokinetics of ranitidine in newborns with gastroesophageal reflux. METHODS: A prospective, descriptive and pharmacokinetic study was carried out in 30 pre-term and 20 full-term babies. 3 mg/kg of ranitidine was administered intravenously to all the babies and at 0.25, 0.5, 1, 2, 4, and 8 h following the administration, samples of blood were drawn to assess ranitidine levels using high performance liquid chromatographic technique. RESULTS: Pharmacokinetics of ranitidine had a bi-exponential behavior with a half-life elimination of (t1/2el) 2.79 h, area under curve (AUC) of 1688 ng/mL, volume of distribution (Vd) of 1.44 L/kg, and clearance (Cl) of 5.9 L/kg/h. The median plasmatic concentration in pre-terms was 1113 ng/mL and 280 ng/mL in full-terms. Vd, t1/2 and Cl presented high values in preterm although the correlation of Cl with glomerular filtration in term newborns was better. CONCLUSIONS: Plasma levels of ranitidine depend on the gestational age of the newborns. However, the possible relationship between after-birth age and pharmacokinetics of the neonates as their internal organs get matured without minding their gestational background.


Anti-Ulcer Agents/pharmacokinetics , Gastroesophageal Reflux/drug therapy , Infant, Premature, Diseases/drug therapy , Ranitidine/pharmacokinetics , Anti-Ulcer Agents/blood , Anti-Ulcer Agents/therapeutic use , Area Under Curve , Chromatography, High Pressure Liquid , Female , Gastroesophageal Reflux/blood , Gestational Age , Half-Life , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/blood , Injections, Intravenous , Male , Prospective Studies , Ranitidine/blood , Ranitidine/therapeutic use
8.
J Chromatogr Sci ; 54(3): 419-28, 2016 Mar.
Article En | MEDLINE | ID: mdl-26538490

A simple, efficient and reliable ion-pair chromatography (IPC) method was developed and validated for the determination of some H2 receptor antagonists including ranitidine (RAN), nizatidine (NIZ) and famotidine (FAM). The use of IPC separations provided improved peak resolution with good peak shape in short analysis time and augmented method selectivity compared with the frequently used RP-C18 methods. A simple isocratic mode with mobile phase containing acetonitrile and 20 mM acetate buffer (50 : 50, v/v) containing 20 mM sodium dodecyl sulfate was used for separation. The flow rate was set at 1.0 mL min(-1), and the effluent was monitored by UV detector at 280 nm FAM and 320 nm for NIZ and RAN. The method was validated in accordance with International Conference on Harmonization guidelines and shown to be suitable for intended applications. The limits of detections and quantitations were 0.008-0.011 and 0.025-0.033 µg mL(-1), respectively. The proposed IPC method was successfully applied for the determination of pharmaceutical dosage forms without prior need for separation. Additionally, the developed method was applied for the determination of RAN in rabbit plasma using NIZ as the internal standard. The method entailed direct injection of the plasma samples after deproteination using methanol. Finally, the proposed IPC method was applied successfully in a pharmacokinetic study for RAN in rabbits after a single oral dose of RAN.


Chromatography, High Pressure Liquid/methods , Famotidine/pharmacokinetics , Histamine H2 Antagonists/pharmacokinetics , Nizatidine/pharmacokinetics , Ranitidine/pharmacokinetics , Acetonitriles , Administration, Oral , Animals , Buffers , Chromatography, High Pressure Liquid/standards , Famotidine/blood , Female , Histamine H2 Antagonists/blood , Humans , Limit of Detection , Nizatidine/blood , Rabbits , Ranitidine/blood , Sodium Dodecyl Sulfate , Solvents
9.
Clin Pharmacokinet ; 53(9): 837-47, 2014 Sep.
Article En | MEDLINE | ID: mdl-25117183

BACKGROUND AND OBJECTIVE: Vandetanib is a selective inhibitor of vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR) and rearranged during transfection (RET) signalling, indicated for the treatment of medullary thyroid cancer. We investigated potential drug-drug interactions between vandetanib and metformin [organic cation transporter 2 (OCT2) substrate; NCT01551615]; digoxin [P-glycoprotein (P-gp) substrate; NCT01561781]; midazolam [cytochrome P450 (CYP) 3A4 substrate; NCT01544140]; omeprazole (proton pump inhibitor) or ranitidine (histamine H2-receptor antagonist; both NCT01539655). METHODS: Four open-label, phase I studies were conducted in healthy volunteers: n = 14 (metformin), n = 14 (digoxin), n = 17 (midazolam), n = 16 (omeprazole), n = 18 (ranitidine). Three of these comprised the following regimens: metformin 1000 mg ± vandetanib 800 mg, midazolam 7.5 mg ± vandetanib 800 mg, or digoxin 0.25 mg ± vandetanib 300 mg. The randomized study comprised vandetanib 300 mg alone and then either (i) omeprazole 40 mg (days 1-4), and omeprazole + vandetanib (day 5); or (ii) ranitidine 150 mg (day 1), and ranitidine + vandetanib (day 2). The primary objective assessed metformin, digoxin, midazolam and vandetanib pharmacokinetics. RESULTS: Vandetanib + metformin increased metformin area under the plasma concentration-time curve from zero to infinity (AUC0-∞) and maximum observed plasma concentration (Cmax) by 74 and 50 %, respectively, and decreased the geometric mean metformin renal clearance (CLR) by 52 % versus metformin alone. Vandetanib + digoxin increased digoxin area under the concentration-time curve from zero to the last quantifiable concentration (AUC0-last) and Cmax by 23 and 29 %, respectively, versus digoxin alone, with only a 9 % decrease in CLR. Vandetanib had no effect on midazolam exposure. Vandetanib exposure was unchanged during co-administration with omeprazole/ranitidine. Treatment combinations were generally well tolerated. CONCLUSION: Patients receiving vandetanib with metformin/digoxin may require additional monitoring of metformin/digoxin, with dose adjustments where necessary. Vandetanib with CYP3A4 substrates or omeprazole/ranitidine is unlikely to result in clinically relevant drug-drug interactions.


Digoxin/pharmacokinetics , Metformin/pharmacokinetics , Midazolam/pharmacokinetics , Omeprazole/pharmacokinetics , Piperidines/pharmacokinetics , Quinazolines/pharmacokinetics , Ranitidine/pharmacokinetics , Adolescent , Adult , Area Under Curve , Digoxin/adverse effects , Digoxin/blood , Drug Interactions , Female , Humans , Male , Metformin/adverse effects , Metformin/blood , Midazolam/adverse effects , Midazolam/blood , Middle Aged , Omeprazole/adverse effects , Omeprazole/blood , Piperidines/adverse effects , Piperidines/blood , Quinazolines/adverse effects , Quinazolines/blood , Ranitidine/adverse effects , Ranitidine/blood , Young Adult
10.
J Clin Pharmacol ; 54(2): 201-5, 2014 Feb.
Article En | MEDLINE | ID: mdl-24258773

To characterize and compare acid suppression (pharmacodynamics) and pharmacokinetics of IV famotidine and ranitidine in critically ill children at risk for stress gastritis. Single-blind, randomized study in PICU patients 6 months to 18 years requiring mechanical ventilation with continuous gastric pH monitoring, randomized to IV famotidine 12 mg/m(2) or ranitidine 60 mg/m(2) when gastric pH < 4.0 >1 hour with serial blood sampling following first dose. Twenty-four children randomized to either famotidine (n = 12) or ranitidine (n = 12). Sixteen out of twenty-four completed both PK and PD study arms (7/12 famotidine; 4.7 ± 3.4 years; 9/12 ranitidine; 6.6 ± 4.7 years; p = 0.38). Time to gastric pH 4.0 and total time pH above 4.0 similar with no difference in pH at 6 and 12 hours (p > 0.2). No difference between drugs in clearance, volume of distribution and half-life (p > 0.05). Ratio of AUC pH to AUC drug concentration 0-12 hours after first dose was significantly greater for famotidine (0.06849 ± 0.01460 SD) than ranitidine (0.02453 ± 0.01448; p < 0.001) demonstrating greater potency of famotidine. pH lowering efficacy of both drugs is similar. Greater potency of famotidine may offer clinical advantage due to lower drug exposure and less frequent dosing to achieve same pH lowering effect.


Critical Illness , Famotidine/pharmacokinetics , Ranitidine/pharmacokinetics , Child , Child, Preschool , Famotidine/administration & dosage , Famotidine/blood , Famotidine/pharmacology , Female , Gastric Acid/chemistry , Gastric Acid/metabolism , Gastric Acidity Determination , Gastritis/drug therapy , Gastritis/metabolism , Histamine H2 Antagonists/administration & dosage , Histamine H2 Antagonists/blood , Histamine H2 Antagonists/pharmacokinetics , Histamine H2 Antagonists/pharmacology , Humans , Hydrogen-Ion Concentration/drug effects , Infant , Infusions, Intravenous , Ranitidine/administration & dosage , Ranitidine/blood , Ranitidine/pharmacology , Single-Blind Method
11.
J Ethnopharmacol ; 142(2): 354-61, 2012 Jul 13.
Article En | MEDLINE | ID: mdl-22580040

AIMS OF THE STUDY: This study was to investigate the gastrokinetic activity of Morinda citrifolia aqueous fruit extract (AFE) in human subjects by examining the GI absorption of ranitidine, a putative indicator of GI motility and to elucidate its possible gastrokinetic mechanism of action in rats. MATERIALS AND METHODS: The single-dose, randomized, open-label and 2-period crossover study was performed on 20 Thai healthy volunteers with a washout period of 14 day between the doses. AFE or drinking water was administered orally 30 min prior to a single oral administration of ranitidine (300 mg). Blood samples were collected over a 12 h period after drug administration and the pharmacokinetic parameters of ranitidine were calculated. The gastrokinetic mechanism of action of AFE was elucidated by measurement of its contractile response on the isolated rat gastric fundus strip. RESULTS: The area under the plasma ranitidine concentration-time curve and the maximal plasma ranitidine concentration were significantly increased after pretreatment with AFE (p=0.001). The plasma ranitidine concentrations were significantly greater at 30-120 min after its administration. AFE produced a definite contractile response of a rat gastric fundus strip with a dose dependency. Scopoletin at the same equivalent dose present in AFE elicited a concentration-dependent contraction that amounted to 45% of the maximal response to AFE. The contractile response of both AFE and scopoletin was mediated through the 5-HT(4) receptor. CONCLUSION: AFE has a unique gastrokinetic activity in enhancement of the rate and the extent of ranitidine absorption. The underlying mechanism can be attributed, at least in part, to the ability of its active component: scopoletin to stimulate the 5-HT(4) receptor.


Gastrointestinal Motility/drug effects , Morinda/chemistry , Muscle Contraction/drug effects , Plant Extracts/pharmacology , Ranitidine/pharmacokinetics , Scopoletin/pharmacology , Stomach/drug effects , Adolescent , Adult , Animals , Area Under Curve , Cross-Over Studies , Dose-Response Relationship, Drug , Female , Fruit , Gastrointestinal Agents/pharmacology , Humans , Intestinal Absorption , Male , Middle Aged , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Ranitidine/blood , Rats, Wistar , Receptors, Serotonin/metabolism , Serotonin Receptor Agonists/pharmacology , Stomach/physiology , Young Adult
12.
J Chromatogr Sci ; 50(4): 301-6, 2012 Apr.
Article En | MEDLINE | ID: mdl-22345389

A novel precolumn derivatization reversed-phase high-performance liquid chromatography method with fluorescence detection is described for the determination of ranitidine in human plasma. The method was based on the reaction of ranitidine with 4-fluoro-7-nitrobenzo-2-oxa-1,3-diazole forming yellow colored fluorescent product. The separation was achieved on a C(18) column using methanol-water (60:40, v/v) mobile phase. Fluorescence detection was used at the excitation and emission of 458 and 521 nm, respectively. Lisinopril was utilized as an internal standard. The flow rate was 1.2 mL/min. Ranitidine and lisinopril appeared at 3.24 and 2.25 min, respectively. The method was validated for system suitability, precision, accuracy, linearity, limit of detection, limit of quantification, recovery and robustness. Intra- and inter-day precisions of the assays were in the range of 0.01-0.44%. The assay was linear over the concentration range of 50-2000 ng/mL. The mean recovery was determined to be 96.40 ± 0.02%. This method was successfully applied to a pharmacokinetic study after oral administration of a dose (150 mg) of ranitidine.


Chromatography, High Pressure Liquid/methods , Ranitidine/blood , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Adult , Female , Humans , Liquid-Liquid Extraction , Lisinopril , Ranitidine/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence
13.
Mol Pharm ; 9(1): 14-28, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-22047066

As 95% of all prescriptions are for orally administered drugs, the issue of oral absorption is central to the development of pharmaceuticals. Oral absorption is limited by a high molecular weight (>500 Da), a high log P value (>2.0) and low gastrointestinal permeability. We have designed a triple action nanomedicine from a chitosan amphiphile: quaternary ammonium palmitoyl glycol chitosan (GCPQ), which significantly enhances the oral absorption of hydrophobic drugs (e.g., griseofulvin and cyclosporin A) and, to a lesser extent, the absorption of hydrophilic drugs (e.g., ranitidine). The griseofulvin and cyclosporin A C(max) was increased 6- and 5-fold respectively with this new nanomedicine. Hydrophobic drug absorption is facilitated by the nanomedicine: (a) increasing the dissolution rate of hydrophobic molecules, (b) adhering to and penetrating the mucus layer and thus enabling intimate contact between the drug and the gastrointestinal epithelium absorptive cells, and (c) enhancing the transcellular transport of hydrophobic compounds. Although the C(max) of ranitidine was enhanced by 80% with the nanomedicine, there was no appreciable opening of tight junctions by the polymer particles.


Chitosan/analogs & derivatives , Cyclosporine/administration & dosage , Drug Carriers/administration & dosage , Griseofulvin/administration & dosage , Intestinal Absorption , Nanoparticles/chemistry , Quaternary Ammonium Compounds/chemistry , Adhesiveness , Animals , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/blood , Anti-Ulcer Agents/metabolism , Anti-Ulcer Agents/pharmacokinetics , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacokinetics , Antifungal Agents/administration & dosage , Antifungal Agents/blood , Antifungal Agents/metabolism , Antifungal Agents/pharmacokinetics , Caco-2 Cells , Chitosan/chemistry , Cyclosporine/blood , Cyclosporine/metabolism , Cyclosporine/pharmacokinetics , Drug Carriers/analysis , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Griseofulvin/blood , Griseofulvin/metabolism , Griseofulvin/pharmacokinetics , Humans , Hydrophobic and Hydrophilic Interactions , Idarubicin/administration & dosage , Idarubicin/metabolism , Idarubicin/pharmacokinetics , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Male , Nanoparticles/ultrastructure , Ranitidine/administration & dosage , Ranitidine/blood , Ranitidine/metabolism , Ranitidine/pharmacokinetics , Rats , Rats, Wistar
14.
J Pharm Biomed Anal ; 56(5): 1057-63, 2011 Dec 15.
Article En | MEDLINE | ID: mdl-21890296

A novel approach has been developed to determine ranitidine in paediatric samples using dried blood spots (DBS) on Guthrie cards (Whatman 903). A selective and sensitive HPLC-MS/MS assay has been developed and validated using small volumes of blood (30 µl). A 6 mm disc was punched from each DBS and extracted with methanolic solution of the internal standard (IS) nizatidine. This was further subjected to solid phase extraction (SPE), followed by reversed phase HPLC separation, using a XBridge™ C18 column and mobile phase 10 mM ammonium acetate/methanol (98:2 v/v) with a flow rate of 0.3 mL/min. This was combined with multiple reaction monitoring (MRM) mass detection using electrospray ionisation (ESI). The calibration curve for ranitidine was found linear over the range 10-500 ng/mL (r=0.996). The limit of quantification (LOQ) of the method was validated at 10 ng/mL. Accuracy and precision values for within and between days were <20% at the LOQ and <15% at all other concentrations. The validated DBS method was successfully applied to a clinical study employing 81 samples from 36 paediatric patients.


Chromatography, High Pressure Liquid/methods , Histamine H2 Antagonists/blood , Ranitidine/blood , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Child , Humans , Limit of Detection , Reference Standards , Reproducibility of Results
15.
Pak J Pharm Sci ; 23(4): 379-83, 2010 Oct.
Article En | MEDLINE | ID: mdl-20884450

A simple, specific and accurate reverse phase liquid chromatographic method has been developed for the simultaneous determination of naproxen and ranitidine HCl. Both the drugs are official with British Pharmacopoeia 2007, but do not involve simultaneous determination of naproxen and ranitidine HCl. The separation was carried out using 4.6 × 250 mm Symmetry Shield TM RP 18 with a particle diameter of 5 µm and mobile phase containing 0.1M orthophosphoric acid: methanol (35:65, pH 3.1) in isocratic mode. The flow rate was 1.00 ml/min and effluent was monitored at 240 nm. The retention times (average) of ranitidine HCl and naproxen were 2.36 min and 12.39 min, respectively. The linearity for naproxen and ranitidine HCl was in the range of 5-35 µg/ml and 1.5-12 µg/ml, respectively. The potencies of naproxen and ranitidine HCl were found 99.40 % and 99.48 %, respectively. The proposed method was validated and successfully applied to the estimation of naproxen and ranitidine HCl in newly formulated combined tablet and in plasma.


Anti-Inflammatory Agents, Non-Steroidal/analysis , Histamine H2 Antagonists/analysis , Naproxen/analysis , Ranitidine/analysis , Anti-Inflammatory Agents, Non-Steroidal/blood , Buffers , Calibration , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Drug Combinations , Histamine H2 Antagonists/blood , Humans , Naproxen/blood , Ranitidine/blood , Reference Standards , Reproducibility of Results , Spectrophotometry, Ultraviolet , Tablets
16.
J Chromatogr Sci ; 48(9): 721-5, 2010 Oct.
Article En | MEDLINE | ID: mdl-20875233

A new, simple, and reliable reversed-phase high-performance liquid chromatographic method has been developed and validated for the simultaneous determination of metformin (Metf), cimetidine (Cimt), famotidine (Famt), and ranitidine (Rant) in their synthetic mixtures and tablet formulations. These drugs were separated on a Purospher Star RP18 endcapped (250 mm × 4.6 mm i.d.) column packed with 5-µm particles. The mobile phase, optimized through an experimental design, consisted of methanol-water-triethylamine (20:80:0.05), whose pH was adjusted to 3.0 with phosphoric acid (85%) pumped at a flow rate of 1.0 mL/min. UV detection was performed at 229 nm. The method was validated in the sample concentration range of 5-25 µg/mL for all the drugs, where it demonstrated good linearity with r = 0.9998, 0.9979, 0.9997, and 0.9987 (n = 6), respectively. For independent 100% level samples, the intra-day and inter-day precision was in the range i.e. < 2.0 for all the drugs. The method demonstrated robustness, resisting to small deliberate changes in pH, flow rate, and composition (organic:aqueous ratio) of the mobile phase. The limit of detection values were 0.071, 0.116, 0.134, and 0.110 µg/mL, while the limit of quantitation were 0.217, 0.352, 0.405, and 0.368 µg/mL for Metf, Cimt, Famt, and Rant, respectively. The applicability of the method was demonstrated by determining the drug content in pharmaceutical formulations, where it exhibited good performance.


Chromatography, High Pressure Liquid/methods , Cimetidine/blood , Famotidine/blood , Histamine H2 Antagonists/blood , Metformin/blood , Ranitidine/blood , Spectrophotometry, Ultraviolet/methods , Adult , Ethylamines/chemistry , Humans , Hydrogen-Ion Concentration , Linear Models , Methanol/chemistry , Reproducibility of Results , Sensitivity and Specificity , Tablets/chemistry , Young Adult
17.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(31): 3953-9, 2009 Dec 01.
Article En | MEDLINE | ID: mdl-19853541

A sensitive and universal LC-MS/MS method for the simultaneous determination of famotidine, cimetidine, ranitidine and lafutidine in human plasma was presented. This is the first single LC-MS/MS method reported for the simultaneous analysis of these four H(2) antagonists in human plasma. Following liquid-liquid extraction with ethyl acetate, the separation was performed on an Agilent Zorbax SB-CN (150 mm x 2.1mm I.D., 5 microm) column using a mobile phase consisted of methanol:20 mM ammonium acetate (55:45, v/v). The total run time was 7 min per sample. Quantification was performed by electrospray ionization-triple quadrupole mass spectrometry by selected reaction monitoring (SRM) detection in the positive mode. All calibration curves showed good linear regression (r(2)>0.99) from 0.5 to 1000 ng/mL for famotidine and lafutidine, and 5-20,000 ng/mL for cimetidine and ranitidine. The method showed good precision and accuracy with overall intra- and inter-day variations of 1.37-9.29% and 3.51-9.40%, respectively. The assay was successfully applied to a bioequivalence study using ranitidine as the model compound.


Chromatography, Liquid/methods , Histamine H2 Antagonists/blood , Tandem Mass Spectrometry/methods , Acetamides/blood , Calibration , Cimetidine/blood , Famotidine/blood , Humans , Piperidines/blood , Pyridines/blood , Ranitidine/blood , Reproducibility of Results , Therapeutic Equivalency
18.
Clin Chim Acta ; 398(1-2): 105-12, 2008 Dec.
Article En | MEDLINE | ID: mdl-18796299

BACKGROUND: Acetaminophen, caffeine, phenytoin, ranitidine, and theophylline are widely used in pediatric pharmacotherapy, but only very limited information is available on the pharmacokinetics of these medications in premature neonates. As pharmacokinetic studies in this population are hampered by limitations in the number and volume of plasma samples, we developed an LC-MS/MS assay for the simultaneous determination of these medications in small volume human plasma specimens for pharmacokinetic evaluations in neonates. METHODS: Sample preparation was performed by protein precipitation with methanol after addition of internal standard to 50 microl of plasma specimen. After chromatographic separation on a C18 column using gradient elution, analytes were detected using a triple quadrupole mass spectrometer that was operated in positive ion mode with electrospray ionization. RESULTS: All 5 analytes could be simultaneously quantified in human plasma. The linear quantification range comprised 12.2 to 25,000 ng/ml for acetaminophen, phenytoin, and ranitidine, 24.4 to 25,000 ng/ml for theophylline, and 48.8 to 25,000 ng/ml for caffeine with accuracies ranging from 87.5 to 115.0%. The intra-day and inter-day precision (%CV) was between 2.8 and 11.8% and 4.5 and 13.5%, respectively. CONCLUSIONS: An accurate, sensitive, and reliable LC-MS/MS method was developed and validated to simultaneously quantify 5 drugs frequently used in neonatal pharmacotherapy.


Acetaminophen/blood , Analgesics/blood , Anticonvulsants/blood , Bronchodilator Agents/blood , Caffeine/blood , Central Nervous System Stimulants/blood , Histamine H2 Antagonists/blood , Phenytoin/blood , Ranitidine/blood , Theophylline/blood , Calibration , Chromatography, High Pressure Liquid , Humans , Infant, Newborn , Quality Control , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
19.
Biomed Chromatogr ; 22(8): 851-6, 2008 Aug.
Article En | MEDLINE | ID: mdl-18384065

Da Cheng Qi decoction (DCQD) is composed of Dahuang, Houpu, Zhishi and Mangxiao. It is a formula created under the theory of Chinese medicine to purge the 'evil heat' in the gastrointestitinal tract, which arises from the ileus and acute pancreatitis. The present study was conducted to evaluate the herb-drug interaction between DCQD and ranitidine, which are often co-administered in clinical practice. Ranitidine was administered orally alone or together with DCQD to rats, and plasma ranitidine concentrations were measured by HPLC. Following oral administration, ranitidine plasma levels revealed curves characterized by peaks at 1.8 and 4.2 h corresponding to ranitidine alone and ranitidine with DCQD at mean concentrations of 16.315 and 1.455 microg/mL, respectively. After ranitidine was orally dosed alone or with DCQD, the half-lives were 1.787 and 3.758 h, while the area under the concentration-time curve (0-12 h) was 28.083 and 9.826 microg/L h, respectively, suggesting that DCQD might significantly affect the pharmacokinetics of ranitidine in rats. When physicians or pharmacists administer DCQD and ranitidine, they must make a careful effort to adjust the dosage of the drug and Chinese decoction, or avoid the herb-drug co-administration.


Drugs, Chinese Herbal , Histamine H2 Antagonists/pharmacokinetics , Ranitidine/pharmacokinetics , Animals , Area Under Curve , Calibration , Chromatography, High Pressure Liquid , Half-Life , Histamine H2 Antagonists/blood , Male , Plant Extracts , Ranitidine/blood , Rats , Rats, Sprague-Dawley , Reproducibility of Results
20.
Eur J Pharm Biopharm ; 69(3): 977-85, 2008 Aug.
Article En | MEDLINE | ID: mdl-18424095

The present research is based on the hypothesis that leaky enteric-coated pellets formulations are able to provide sustained input for drugs that have an absorption window, such as ranitidine hydrochloride, without jeopardizing their bioavailability. Leaky enteric-coated pellets formulations are defined as enteric-coated pellets that allow some of the drug to be released from the formulation in gastric fluid. Different approaches to making leaky enteric-coated pellets were investigated using extrusion-spheronization followed by spray coating. Leaky enteric coats were formulated using a commonly used enteric polymer, Eudragit L 30 D-55, combined with soluble compounds including lactose, PEG 8000 and surfactants (Span 60 (hydrophobic) or Tween 80 (hydrophilic)). The rate of drug release from the formulations in simulated gastric fluid can be tailored by varying the additive's amount or type. All leaky enteric-coated formulations studied completely released the drugs within 30 min after changing dissolution medium to phosphate buffer, pH 6. Predictions of plasma concentration-time profiles of the model drug ranitidine hydrochloride from leaky enteric-coated pellets in fasted conditions and from immediate-release formulations were performed using computer simulations. Simulation results are consistent with a hypothesis that leaky enteric-coated pellets formulations provide sustained input for drugs shown to have an absorption window without decreasing bioavailability. The sustained input results from the combined effects of the formulation and GI transit effects on pellets. The present research demonstrates a new application of knowledge about gastrointestinal transit effects on drug formulations. It also shows that enteric-coating polymers have new applications in areas other than the usual enteric-coated formulations. The hypothesis that a leaky enteric-coated pellets formulation may maintain or increase the bioavailability of drugs that have a window of absorption is still to be confirmed by further in vivo studies.


Histamine H2 Antagonists/administration & dosage , Ranitidine/administration & dosage , Algorithms , Biological Availability , Chemistry, Pharmaceutical , Computer Simulation , Excipients , Half-Life , Histamine H2 Antagonists/blood , Histamine H2 Antagonists/pharmacokinetics , Humans , Lactose/chemistry , Models, Statistical , Polyethylene Glycols , Ranitidine/blood , Ranitidine/pharmacokinetics , Solubility , Tablets, Enteric-Coated
...