Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Biopharm Drug Dispos ; 45(3): 138-148, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38823029

ABSTRACT

Bumetanide is used widely as a tool and off-label treatment to inhibit the Na-K-2Cl cotransporter NKCC1 in the brain and thereby to normalize intra-neuronal chloride levels in several brain disorders. However, following systemic administration, bumetanide only poorly penetrates into the brain parenchyma and does not reach levels sufficient to inhibit NKCC1. The low brain penetration is a consequence of both the high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, and of brain efflux transport. In previous studies, bumetanide was determined in the whole brain or a few brain regions, such as the hippocampus. However, the blood-brain barrier and its efflux transporters are heterogeneous across brain regions, so it cannot be excluded that bumetanide reaches sufficiently high brain levels for NKCC1 inhibition in some discrete brain areas. Here, bumetanide was determined in 14 brain regions following i.v. administration of 10 mg/kg in rats. Because bumetanide is much more rapidly eliminated by rats than humans, its metabolism was reduced by pretreatment with piperonyl butoxide. Significant, up to 5-fold differences in regional bumetanide levels were determined with the highest levels in the midbrain and olfactory bulb and the lowest levels in the striatum and amygdala. Brain:plasma ratios ranged between 0.004 (amygdala) and 0.022 (olfactory bulb). Regional brain levels were significantly correlated with local cerebral blood flow. However, regional bumetanide levels were far below the IC50 (2.4 µM) determined previously for rat NKCC1. Thus, these data further substantiate that the reported effects of bumetanide in rodent models of brain disorders are not related to NKCC1 inhibition in the brain.


Subject(s)
Brain , Bumetanide , Animals , Bumetanide/pharmacology , Bumetanide/pharmacokinetics , Bumetanide/administration & dosage , Brain/metabolism , Brain/drug effects , Male , Rats , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Rats, Sprague-Dawley , Tissue Distribution , Solute Carrier Family 12, Member 2/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
2.
Neuropharmacology ; 205: 108910, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34883135

ABSTRACT

The Na-K-2Cl cotransporter NKCC1 and the neuron-specific K-Cl cotransporter KCC2 are considered attractive CNS drug targets because altered neuronal chloride regulation and consequent effects on GABAergic signaling have been implicated in numerous CNS disorders. While KCC2 modulators are not yet clinically available, the loop diuretic bumetanide has been used in clinical studies to treat brain disorders and as a tool for NKCC1 inhibition in preclinical models. Bumetanide is known to have anticonvulsant and neuroprotective effects under some pathophysiological conditions. However, as shown in several species from neonates to adults (mice, rats, dogs, and by extrapolation in humans), at the low clinical doses of bumetanide approved for diuresis, this drug has negligible access into the CNS, reaching levels that are much lower than what is needed to inhibit NKCC1 in cells within the brain parenchyma. Several drug discovery strategies have been used over the last ∼15 years to develop brain-permeant compounds that, ideally, should be selective for NKCC1 to eliminate the diuresis mediated by inhibition of renal NKCC2. The strategies employed to improve the pharmacokinetic and pharmacodynamic properties of NKCC1 blockers include evaluation of other clinically approved loop diuretics; development of lipophilic prodrugs of bumetanide; development of side-chain derivatives of bumetanide; and unbiased high-throughput screening approaches of drug discovery based on large chemical compound libraries. The main outcomes are that (1), non-acidic loop diuretics such as azosemide and torasemide may have advantages as NKCC1 inhibitors vs. bumetanide; (2), bumetanide prodrugs achieve significantly higher brain levels of the parent drug and have lower diuretic activity; (3), the novel bumetanide side-chain derivatives do not exhibit any functionally relevant improvement of CNS accessibility or NKCC1 selectivity vs. bumetanide; (4) novel compounds discovered by high-throughput screening may resolve some of the inherent problems of bumetanide, but as yet this has not been achieved. Thus, further research is needed to optimize the design of brain-permeant NKCC1 inhibitors. Another major challenge is to identify the mechanisms whereby various NKCC1-expressing cellular targets of these drug within (e.g., neurons, oligodendrocytes or astrocytes) and outside the brain parenchyma (e.g., blood-brain barrier, choroid plexus, endocrine and immune system), as well as molecular off-target effects, might contribute to their reported therapeutic and adverse effects.


Subject(s)
Bumetanide/pharmacokinetics , Central Nervous System/drug effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Solute Carrier Family 12, Member 2/drug effects , Animals , Humans
3.
Epilepsia ; 62(6): 1460-1471, 2021 06.
Article in English | MEDLINE | ID: mdl-33955541

ABSTRACT

OBJECTIVES: Bumetanide was suggested as an adjunct to phenobarbital for suppression of neonatal seizures. This suggestion was based on the idea that bumetanide, by reducing intraneuronal chloride accumulation through inhibition of the Na-K-2Cl cotransporter NKCC1, may attenuate or abolish depolarizing γ-aminobutyric acid (GABA) responses caused by birth asphyxia. However, a first proof-of-concept clinical trial failed. This could have had several reasons, including bumetanide's poor brain penetration, the wide cellular NKCC1 expression pattern in the brain, and problems with the general concept of NKCC1's role in neonatal seizures. We recently replicated the clinical failure of bumetanide to potentiate phenobarbital's effect in a novel rat model of birth asphyxia. In this study, a clinically relevant dose (0.3 mg/kg) of bumetanide was used that does not lead to NKCC1-inhibitory brain levels. The aim of the present experiments was to examine whether a much higher dose (10 mg/kg) of bumetanide is capable of potentiating phenobarbital in this rat model. Furthermore, the effects of the two lipophilic bumetanide derivatives, the ester prodrug N,N-dimethylaminoethylester of bumetanide (DIMAEB) and the benzylamine derivative bumepamine, were examined at equimolar doses. METHODS: Intermittent asphyxia was induced for 30 min by exposing male and female P11 rat pups to three 7 + 3 min cycles of 9% and 5% O2 at constant 20% CO2 . All control pups exhibited neonatal seizures after the asphyxia. RESULTS: Even at 10 mg/kg, bumetanide did not potentiate the effect of a submaximal dose (15 mg/kg) of phenobarbital on seizure incidence, whereas a significant suppression of neonatal seizures was determined for combinations of phenobarbital with DIMAEB or, more effectively, bumepamine, which, however, does not inhibit NKCC1. Of interest, the bumepamine/phenobarbital combination prevented the neurodegenerative consequences of asphyxia and seizures in the hippocampus. SIGNIFICANCE: Both bumepamine and DIMAEB are promising tools that may help to develop more effective lead compounds for clinical trials.


Subject(s)
Anticonvulsants/pharmacology , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/drug therapy , Benzylamines/therapeutic use , Bumetanide/therapeutic use , Hippocampus/pathology , Nerve Degeneration/pathology , Phenobarbital/pharmacology , Seizures/drug therapy , Seizures/etiology , Animals , Animals, Newborn , Anticonvulsants/pharmacokinetics , Benzylamines/pharmacokinetics , Brain/metabolism , Bumetanide/analogs & derivatives , Bumetanide/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Female , Male , Phenobarbital/pharmacokinetics , Pregnancy , Rats , Solute Carrier Family 12, Member 2/biosynthesis
4.
Drug Metab Dispos ; 48(9): 788-795, 2020 09.
Article in English | MEDLINE | ID: mdl-32587098

ABSTRACT

Bumetanide, a sulfamyl loop diuretic, is used for the treatment of edema in association with congestive heart failure. Being a polar, anionic compound at physiologic pH, bumetanide uptake and efflux into different tissues is largely transporter-mediated. Of note, organic anion transporters (SLC22A) have been extensively studied in terms of their importance in transporting bumetanide to its primary site of action in the kidney. The contribution of one of the less-studied bumetanide transporters, monocarboxylate transporter 6 (MCT6; SLC16A5), to bumetanide pharmacokinetics (PK) and pharmacodynamics (PD) has yet to be characterized. The affinity of bumetanide for murine Mct6 was evaluated using Mct6-transfected Xenopus laevis oocytes. Furthermore, bumetanide was intravenously and orally administered to wild-type mice (Mct6+/+) and homozygous Mct6 knockout mice (Mct6-/-) to elucidate the contribution of Mct6 to bumetanide PK/PD in vivo. We demonstrated that murine Mct6 transports bumetanide at a similar affinity compared with human MCT6 (78 and 84 µM, respectively, at pH 7.4). After bumetanide administration, there were no significant differences in plasma PK. Additionally, diuresis was significantly decreased by ∼55% after intravenous bumetanide administration in Mct6-/- mice. Kidney cortex concentrations of bumetanide were decreased, suggesting decreased Mct6-mediated bumetanide transport to its site of action in the kidney. Overall, these results suggest that Mct6 does not play a major role in the plasma PK of bumetanide in mice; however, it significantly contributes to bumetanide's pharmacodynamics due to changes in kidney concentrations. SIGNIFICANCE STATEMENT: Previous evidence suggested that MCT6 transports bumetanide in vitro; however, no studies to date have evaluated the in vivo contribution of this transporter. In vitro studies indicated that mouse and human MCT6 transport bumetanide with similar affinities. Using Mct6 knockout mice, we demonstrated that murine Mct6 does not play a major role in the plasma pharmacokinetics of bumetanide; however, the pharmacodynamic effect of diuresis was attenuated in the knockout mice, likely because of the decreased bumetanide concentrations in the kidney.


Subject(s)
Bumetanide/pharmacokinetics , Diuresis/drug effects , Monocarboxylic Acid Transporters/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Bumetanide/administration & dosage , Drug Evaluation, Preclinical , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Male , Mice , Mice, Knockout , Monocarboxylic Acid Transporters/genetics , Oocytes , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Xenopus laevis
5.
Pharmacol Ther ; 205: 107422, 2020 01.
Article in English | MEDLINE | ID: mdl-31626872

ABSTRACT

Epilepsies represent one of the most common neurological diseases worldwide. They are characterized by recurrent spontaneous seizures with severe impact on a patient's life. An imbalance in excitatory and inhibitory signalling is considered the main underlying pathophysiological mechanism. Therefore, GABA-mimetic drugs, strengthening the main inhibitory signalling system in the CNS, are frequently used as antiepileptic or anticonvulsant drugs. However, the therapeutic effect of such treatment depends on the chloride gradient along the plasma membrane. Impairment of chloride homeostasis, caused by alterations in the functional balance of chloride transporters, was implicated in the pathophysiology of epilepsy and numerous other diseases. Breakdown or even inversion of the chloride gradient may result in ineffective or in worst cases proconvulsant effects of GABA-mimetics. Unfortunately, such situations are reported in considerable number. Consequently, bumetanide, an inhibitor of Na-K-Cl cotransporters gained interest as potential add-on therapy re-establishing the chloride gradient and thereby the hyperpolarizing effects of GABA-mimetic drugs. Indeed, preclinical studies yielded encouraging results, especially when applied in combination with GABA-mimetics in epilepsy models. However, bumetanide induces a strong diuretic effect and displays poor penetration across the blood-brain barrier, two adverse features for chronic antiepileptic treatment. Therefore, new compounds overcoming these limitations are under development. This review focuses on alterations in chloride homeostasis and its underlying molecular mechanisms in epilepsy, on the potential impact of impaired chloride homeostasis on the treatment of epilepsy and on concepts to overcome this problem including recent development of bumetanide derivatives with improved pharmacological profile.


Subject(s)
Anticonvulsants/pharmacology , Chlorides/metabolism , Epilepsy/drug therapy , Animals , Anticonvulsants/adverse effects , Blood-Brain Barrier , Bumetanide/adverse effects , Bumetanide/pharmacokinetics , Bumetanide/pharmacology , Diuretics/adverse effects , Diuretics/pharmacokinetics , Diuretics/pharmacology , Drug Development , Epilepsy/physiopathology , Humans , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Tissue Distribution
6.
Neuropharmacology ; 143: 186-204, 2018 12.
Article in English | MEDLINE | ID: mdl-30248303

ABSTRACT

Based on the potential role of Na-K-Cl cotransporters (NKCCs) in epileptic seizures, the loop diuretic bumetanide, which blocks the NKCC1 isoforms NKCC1 and NKCC2, has been tested as an adjunct with phenobarbital to suppress seizures. However, because of its physicochemical properties, bumetanide only poorly penetrates through the blood-brain barrier. Thus, concentrations needed to inhibit NKCC1 in hippocampal and neocortical neurons are not reached when using doses (0.1-0.5 mg/kg) in the range of those approved for use as a diuretic in humans. This prompted us to search for a bumetanide derivative that more easily penetrates into the brain. Here we show that bumepamine, a lipophilic benzylamine derivative of bumetanide, exhibits much higher brain penetration than bumetanide and is more potent than the parent drug to potentiate phenobarbital's anticonvulsant effect in two rodent models of chronic difficult-to-treat epilepsy, amygdala kindling in rats and the pilocarpine model in mice. However, bumepamine suppressed NKCC1-dependent giant depolarizing potentials (GDPs) in neonatal rat hippocampal slices much less effectively than bumetanide and did not inhibit GABA-induced Ca2+ transients in the slices, indicating that bumepamine does not inhibit NKCC1. This was substantiated by an oocyte assay, in which bumepamine did not block NKCC1a and NKCC1b after either extra- or intracellular application, whereas bumetanide potently blocked both variants of NKCC1. Experiments with equilibrium dialysis showed high unspecific tissue binding of bumetanide in the brain, which, in addition to its poor brain penetration, further reduces functionally relevant brain concentrations of this drug. These data show that CNS effects of bumetanide previously thought to be mediated by NKCC1 inhibition can also be achieved by a close derivative that does not share this mechanism. Bumepamine has several advantages over bumetanide for CNS targeting, including lower diuretic potency, much higher brain permeability, and higher efficacy to potentiate the anti-seizure effect of phenobarbital.


Subject(s)
Anticonvulsants/pharmacology , Benzylamines/pharmacology , Bumetanide/pharmacology , Phenobarbital/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Benzylamines/chemical synthesis , Benzylamines/chemistry , Benzylamines/pharmacokinetics , Brain/drug effects , Brain/metabolism , Bumetanide/analogs & derivatives , Bumetanide/chemistry , Bumetanide/pharmacokinetics , Drug Evaluation, Preclinical , Drug Synergism , Epilepsy/drug therapy , Epilepsy/metabolism , Female , Mice , Oocytes , Phenobarbital/pharmacokinetics , Rats, Wistar , Seizures/drug therapy , Seizures/metabolism , Sodium Potassium Chloride Symporter Inhibitors/chemistry , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Tissue Culture Techniques , Xenopus laevis
7.
J Am Heart Assoc ; 7(4)2018 02 10.
Article in English | MEDLINE | ID: mdl-29440005

ABSTRACT

BACKGROUND: Dapagliflozin inhibits the sodium-glucose-linked transporter 2 in the renal proximal tubule, thereby promoting glycosuria to reduce hyperglycemia in type 2 diabetes mellitus. Because these patients may require loop diuretics, and sodium-glucose-linked transporter 2 inhibition causes an osmotic diuresis, we evaluated the diuretic interaction between dapagliflozin and bumetanide. METHODS AND RESULTS: Healthy subjects (n=42) receiving a fixed diet with ≈110 mmol·d-1 of Na+ were randomized to bumetanide (1 mg·d-1), dapagliflozin (10 mg·d-1), or both for 7 days, followed by 7 days of both. There were no meaningful pharmacokinetic interactions. Na+ excretion increased modestly with the first dose of dapagliflozin (22±6 mmol·d-1; P<0.005) but by more (P<0.005) with the first dose of bumetanide (74±7 mmol·d-1; P<0.005), which was not significantly different from both diuretics together (80±5 mmol·d-1; P<0.005). However, Na+ excretion with dapagliflozin was 190% greater (P<0.005) when added after 1 week of bumetanide (64±6 mmol·d-1), and Na+ excretion with bumetanide was 36% greater (P<0.005) when added after 1 week of dapagliflozin (101±8 mmol·d-1). Serum urate was increased 4% by bumetanide but reduced 40% by dapagliflozin or 20% by combined therapy (P<0.05). CONCLUSIONS: First-dose Na+ excretion with bumetanide and dapagliflozin is not additive, but the weekly administration of one diuretic enhances the initial Na+ excretion with the other, thereby demonstrating mutual adaptive natriuretic synergy. Combined therapy reverses bumetanide-induced hyperuricemia. This requires further study in diabetic patients with hyperglycemia who have enhanced glycosuria and natriuresis with dapagliflozin. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00930865.


Subject(s)
Benzhydryl Compounds/administration & dosage , Bumetanide/administration & dosage , Glucosides/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Adolescent , Adult , Benzhydryl Compounds/adverse effects , Benzhydryl Compounds/pharmacokinetics , Bumetanide/adverse effects , Bumetanide/pharmacokinetics , Drug Interactions , Female , Glucosides/adverse effects , Glucosides/pharmacokinetics , Healthy Volunteers , Humans , Hyperuricemia/blood , Hyperuricemia/chemically induced , Hyperuricemia/prevention & control , Male , Middle Aged , Natriuresis/drug effects , Renal Elimination/drug effects , Risk Assessment , Sodium Potassium Chloride Symporter Inhibitors/adverse effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium, Dietary/urine , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics , Uric Acid/blood , Young Adult
8.
Biopharm Drug Dispos ; 39(3): 125-134, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29319897

ABSTRACT

Bumetanide is a loop diuretic that is proposed to possess a beneficial effect on disorders of the central nervous system, including neonatal seizures. Therefore, prediction of unbound bumetanide concentrations in the brain is relevant from a pharmacological prospective. A physiologically-based pharmacokinetic (PBPK) model was developed for the prediction of bumetanide disposition in plasma and brain in adult and paediatric populations. A compound file was built for bumetanide integrating physicochemical data and in vitro data. Bumetanide concentration profiles were simulated in both plasma and brain using the Simcyp PBPK model. Simulations of plasma bumetanide concentrations were compared against plasma levels published in the literature. The model performance was verified with data from adult studies before predictions in the paediatric population were undertaken. The adult and paediatric intravenous models predicted pharmacokinetic factors, namely area under the concentration-time curve, maximum concentration in plasma and time to maximum plasma concentration, within two-fold of observed values. However, predictions of plasma concentrations within the neonatal intravenous model did not produce a good fit with the observed values. The PBPK approach used in this study produced reasonable predictions of plasma concentrations of bumetanide, except in the critically ill neonatal population. This PBPK model requires more information regarding metabolic intrinsic clearance and transport parameters prior to further validation of drug disposition predictions in the neonatal population. Given the lack of information surrounding certain parameters in this special population, the model is not appropriately robust to support the recommendation of a suitable dose of bumetanide for use as an adjunct antiepileptic in neonates.


Subject(s)
Brain/metabolism , Bumetanide/blood , Bumetanide/pharmacokinetics , Models, Biological , Adult , Child , Child, Preschool , Computer Simulation , Diuretics/blood , Diuretics/pharmacokinetics , Female , Humans , Infant , Male
9.
Neuropharmacology ; 117: 182-194, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28192112

ABSTRACT

There is accumulating evidence that bumetanide, which has been used over decades as a potent loop diuretic, also exerts effects on brain disorders, including autism, neonatal seizures, and epilepsy, which are not related to its effects on the kidney but rather mediated by inhibition of the neuronal Na-K-Cl cotransporter isoform NKCC1. However, following systemic administration, brain levels of bumetanide are typically below those needed to inhibit NKCC1, which critically limits its clinical use for treating brain disorders. Recently, active efflux transport at the blood-brain barrier (BBB) has been suggested as a process involved in the low brain:plasma ratio of bumetanide, but it is presently not clear which transporters are involved. Understanding the processes explaining the poor brain penetration of bumetanide is needed for developing strategies to improve the brain delivery of this drug. In the present study, we administered probenecid and more selective inhibitors of active transport carriers at the BBB directly into the brain of mice to minimize the contribution of peripheral effects on the brain penetration of bumetanide. Furthermore, in vitro experiments with mouse organic anion transporter 3 (Oat3)-overexpressing Chinese hamster ovary cells were performed to study the interaction of bumetanide, bumetanide derivatives, and several known inhibitors of Oats on Oat3-mediated transport. The in vivo experiments demonstrated that the uptake and efflux of bumetanide at the BBB is much more complex than previously thought. It seems that both restricted passive diffusion and active efflux transport, mediated by Oat3 but also organic anion-transporting polypeptide (Oatp) Oatp1a4 and multidrug resistance protein 4 explain the extremely low brain concentrations that are achieved after systemic administration of bumetanide, limiting the use of this drug for targeting abnormal expression of neuronal NKCC1 in brain diseases.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , Bumetanide/pharmacokinetics , Multidrug Resistance-Associated Proteins/physiology , Organic Anion Transporters, Sodium-Independent/physiology , Organic Cation Transport Proteins/physiology , Animals , Biological Transport/drug effects , Blood-Brain Barrier/drug effects , Brain/drug effects , Bumetanide/analogs & derivatives , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Diffusion , Female , Membrane Transport Modulators/pharmacology , Mice , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Probenecid/pharmacology
10.
Eur J Heart Fail ; 19(8): 1014-1022, 2017 08.
Article in English | MEDLINE | ID: mdl-28105769

ABSTRACT

BACKGROUND: Loop diuretic resistance is a common barrier to effective decongestion in acute heart failure (AHF), and is associated with poor outcome. Specific mechanisms underlying diuretic resistance are currently unknown in contemporary AHF patients. We therefore aimed to determine the relative importance of defects in diuretic delivery vs. renal tubular response in determining diuretic response (DR) in AHF. METHODS AND RESULTS: Fifty AHF patients treated with intravenous bumetanide underwent a 6-h timed urine collection for sodium and bumetanide clearance. Whole-kidney DR was defined as sodium excreted per doubling of administered loop diuretic and represents the sum of defects in drug delivery and renal tubular response. Tubular DR, defined as sodium excreted per doubling of renally cleared (urinary) loop diuretic, captures resistance specifically in the renal tubule. Median administered bumetanide dose was 3.0 (2.0-4.0) mg with 52 (33-77)% of the drug excreted into the urine. Significant between-patient variability was present as the administered dose only explained 39% of variability in the quantity of bumetanide in urine. Cumulatively, factors related to drug delivery such as renal bumetanide clearance, administered dose, and urea clearance explained 28% of the variance in whole-kidney DR. However, resistance at the level of the renal tubule (tubular DR) explained 71% of the variability in whole-kidney DR. CONCLUSION: Defects at the level of the renal tubule are substantially more important than reduced diuretic delivery in determining diuretic resistance in patients with AHF.


Subject(s)
Bumetanide/administration & dosage , Drug Resistance , Glomerular Filtration Rate/physiology , Heart Failure/drug therapy , Kidney Tubules/drug effects , Sodium/urine , Acute Disease , Administration, Intravenous , Biomarkers/urine , Bumetanide/pharmacokinetics , Dose-Response Relationship, Drug , Female , Glomerular Filtration Rate/drug effects , Heart Failure/urine , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Kidney Tubules/metabolism , Male , Middle Aged , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics
11.
Eur J Pharmacol ; 770: 117-25, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26673740

ABSTRACT

Recently, it has been suggested that bumetanide, an inhibitor of the Na-K-2Cl co-transporter (NKCC1), may be useful in the treatment of central nervous system (CNS) disorders. However, from a physicochemical perspective, bumetanide may not cross the blood-brain barrier to the extent that is necessary for it to be an effective brain NKCC1 inhibitor in vivo. High plasma-protein binding, potentially high brain-tissue binding and putative efflux transporters including organic anion transporter 3 (OAT3) contribute to the poor pharmacokinetic profile of bumetanide. Bidirectional permeability assays are an in vitro method to determine the impact of plasma-protein/brain tissue binding, as well as efflux transport, on the permeability of a compound. We established and validated a cell line stably overexpressing human OAT3 using lentiviral cloning techniques for use in in vitro bidirectional permeability assays. Using efflux transport studies, we show that bumetanide is a transported substrate of human OAT3, exhibiting a transport ratio of ≥1.5, which is attenuated by OAT3 inhibitors. Bidirectional permeability assays were carried out in the presence and absence of either albumin or brain homogenate to elucidate the effect of plasma-protein/brain tissue binding. These tests confirmed the pharmacokinetic limitations for brain delivery of bumetanide. In this experiment, bumetanide is 53% bound to albumin, 77% bound to brain tissue and accumulates in brain cells. Moreover, we conclusively established that bumetanide is a transported substrate of OAT3. Taken together, these bidirectional permeability studies highlight the potential of efflux transporter inhibition as an augmentation strategy for enhanced delivery of bumetanide to the CNS.


Subject(s)
Bumetanide/metabolism , Bumetanide/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Blood-Brain Barrier/metabolism , Bumetanide/pharmacokinetics , HEK293 Cells , Humans , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Permeability
12.
J Clin Pharmacol ; 56(3): 284-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26189501

ABSTRACT

Recent experimental data suggest bumetanide as a possible therapeutic option in newborn infants with seizures after birth asphyxia. Because pharmacokinetic (PK) data are lacking in this population, who very often benefit from therapeutic cooling, which can modify the PK behavior of a drug, a PK study was conducted in term infants with seizures caused by hypoxic-ischemic encephalopathy. Fourteen infants were included, 13 of them being cooled. Forty-nine blood samples were available for the determination of the plasma concentration of bumetanide. Concentration-time data were analyzed by the use of a population approach performed with Monolix Software. Bumetanide was found to follow a 2-compartment model. The mean values were 0.063 L/h for clearance, 0.28 and 0.44 L for the central and peripheral distribution volumes, respectively, and 0.59 L/h for the distribution clearance. Birth body weight explained the interindividual variability of bumetanide clearance via an allometric model. No relationship was found between bumetanide exposure and its efficacy (reduction in seizure burden) or its toxicity (hearing loss). This study describes the first PK model of bumetanide in hypothermia-treated infants with seizures.


Subject(s)
Bumetanide/blood , Bumetanide/pharmacokinetics , Seizures/blood , Seizures/congenital , Anticonvulsants/adverse effects , Anticonvulsants/blood , Anticonvulsants/therapeutic use , Asphyxia/complications , Bumetanide/adverse effects , Bumetanide/therapeutic use , Female , Hearing Loss/chemically induced , Humans , Infant, Newborn , Male , Models, Biological , Pilot Projects , Seizures/drug therapy , Seizures/etiology
13.
Neuroscience ; 286: 203-15, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25463517

ABSTRACT

Neonatal seizures caused by perinatal asphyxia and hypoxic-ischemic encephalopathy can be refractory to conventional anticonvulsants. This may be due to the depolarizing effects of gamma-aminobutyric acid (GABA) achieved by the activity of the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1). The aim of this study is to evaluate the long-term effects of bumetanide, a NKCC1 inhibitor, on hippocampal neurogenesis and seizure susceptibility in hypoxia-induced neonatal seizure model. Wistar rats were subjected to hypoxia-induced neonatal seizures at postnatal day 10 (P10). Following acute seizures, the rats were treated with intraperitoneal injection (i.p.) of bumetanide at a dose of 0.5mg/kg for 3 weeks. In later adulthood, hypoxia-induced seizures increased the number of newborn dentate gyrus cells (DGCs), promoted mossy fiber sprouting (MFS) and reduced the apical dendritic complexity of newborn DGCs 1 month after the insults. In addition, these seizures resulted in long-lasting consequences, such as spontaneous electroencephalography (EEG) seizures, though spatial learning impairments were not seen. Bumetanide treatments significantly enhanced cell proliferation and dendritic development of newborn DGCs after neonatal seizures, accompanied by the decreased seizure activity. However, systemic administration of bumetanide resulted in much lower brain concentrations, and was incompatible with NKCC1 inhibition in blood-brain barrier (BBB)-protected brain tissue. Our results suggested that bumetanide might have long-term effects in suppressing seizure activity, and altering the neurogenesis after neonatal seizures. These effects of bumetanide may be mediated by the targets outside the BBB-protected central nerve system (CNS) or CNS-located target(s) other than NKCC1.


Subject(s)
Bumetanide/administration & dosage , Dentate Gyrus/drug effects , Dentate Gyrus/physiopathology , Hypoxia/complications , Seizures/prevention & control , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Animals , Animals, Newborn , Brain/drug effects , Brain/physiopathology , Brain Chemistry , Bumetanide/analysis , Bumetanide/pharmacokinetics , Cell Proliferation/drug effects , Electroencephalography , Neurogenesis/drug effects , Neurons/drug effects , Neurons/physiology , Rats , Rats, Wistar , Seizures/etiology , Sodium Potassium Chloride Symporter Inhibitors/analysis , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Solute Carrier Family 12, Member 2 , Spatial Learning/drug effects
14.
Eur J Pharmacol ; 746: 167-73, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25449033

ABSTRACT

Bumetanide is increasingly being used for experimental treatment of brain disorders, including neonatal seizures, epilepsy, and autism, because the neuronal Na-K-Cl cotransporter NKCC1, which is inhibited by bumetanide, is implicated in the pathophysiology of such disorders. However, use of bumetanide for treatment of brain disorders is associated with problems, including poor brain penetration and systemic adverse effects such as diuresis, hypokalemic alkalosis, and hearing loss. The poor brain penetration is thought to be related to its high ionization rate and plasma protein binding, which restrict brain entry by passive diffusion, but more recently brain efflux transporters have been involved, too. Multidrug resistance protein 4 (MRP4), organic anion transporter 3 (OAT3) and organic anion transporting polypeptide 2 (OATP2) were suggested to mediate bumetanide brain efflux, but direct proof is lacking. Because MRP4, OAT3, and OATP2 can be inhibited by probenecid, we studied whether this drug alters brain levels of bumetanide in mice. Probenecid (50 mg/kg) significantly increased brain levels of bumetanide up to 3-fold; however, it also increased its plasma levels, so that the brain:plasma ratio (~0.015-0.02) was not altered. Probenecid markedly increased the plasma half-life of bumetanide, indicating reduced elimination of bumetanide most likely by inhibition of OAT-mediated transport of bumetanide in the kidney. However, the diuretic activity of bumetanide was not reduced by probenecid. In conclusion, our study demonstrates that the clinically available drug probenecid can be used to increase brain levels of bumetanide and decrease its elimination, which could have therapeutic potential in the treatment of brain disorders.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Bumetanide/pharmacokinetics , Membrane Transport Modulators/pharmacology , Organic Anion Transporters/antagonists & inhibitors , Probenecid/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Animals , Animals, Outbred Strains , Anticonvulsants/blood , Anticonvulsants/metabolism , Anticonvulsants/pharmacokinetics , Biological Transport/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Bumetanide/blood , Bumetanide/metabolism , Bumetanide/pharmacology , Cell Line , Diuretics/blood , Diuretics/metabolism , Diuretics/pharmacokinetics , Dogs , Drug Interactions , Female , Half-Life , Humans , Kidney/drug effects , Kidney/metabolism , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sodium Potassium Chloride Symporter Inhibitors/blood , Sodium Potassium Chloride Symporter Inhibitors/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology
15.
J Pharm Pharmacol ; 67(4): 501-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25490899

ABSTRACT

OBJECTIVES: Recent data highlight the potential of bumetanide as a treatment for neonatal seizures and autism, as it facilitates the excitatory to inhibitory switch in gamma-aminobutyric acid signalling. This study examines the extent of blood-brain barrier (BBB) permeation of bumetanide, a key determinant of the efficacy of centrally acting drugs. Furthermore, the impact of efflux transporter organic anion transporter 3 (oat3) inhibition on bumetanide pharmacokinetics was investigated. METHODS: Bumetanide levels in extracellular fluid (ECF) and plasma in the presence and absence of oat3 inhibitor probenecid were monitored using integrated microdialysis. KEY FINDINGS: Following a bumetanide bolus/continuous infusion of 10 mg/kg and 6 mg/kg/h, bumetanide was detected in hippocampal ECF at the estimated concentration of 131 ± 55 ng/ml. Plasma bumetanide levels were ∼20 mg/l at steady state. Coadministration of probenecid resulted in an increase in bumetanide levels in both ECF and plasma, indicating that oat3 inhibition influences the pharmacokinetics of bumetanide primarily in the periphery. CONCLUSION: Although bumetanide reached detectable levels in hippocampal ECF, bumetanide concentration in ECF was low relative to systemic concentration. Oat3 inhibition by probenecid resulted in increased bumetanide concentrations in brain and plasma. As an acute treatment in neonatal seizures, the bumetanide/probenecid combination may hold therapeutic potential.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Blood-Brain Barrier/drug effects , Bumetanide/pharmacokinetics , Extracellular Fluid/metabolism , Hippocampus/metabolism , Organic Anion Transporters/antagonists & inhibitors , Probenecid/pharmacology , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Bumetanide/metabolism , Bumetanide/pharmacology , Drug Interactions , GABA Agents/pharmacology , Male , Microdialysis , Rats, Sprague-Dawley
16.
Eur J Neurosci ; 39(4): 673-87, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24251546

ABSTRACT

The diuretic bumetanide, which acts by blocking the Na-K-Cl cotransporter (NKCC), is widely used to inhibit neuronal NKCC1, particularly when NKCC1 expression is abnormally increased in brain diseases such as epilepsy. However, bumetanide poorly penetrates into the brain and, in rodents, is rapidly eliminated because of extensive oxidation of its N-butyl sidechain, reducing the translational value of rodent experiments. Inhibition of oxidation by piperonyl butoxide (PBO) has previously been reported to increase the half-life and diuretic activity of bumetanide in rats. Here we studied whether inhibition of bumetanide metabolism by PBO also increases brain levels of bumetanide in rats, and whether this alters pharmacodynamic effects in the kindling model of epilepsy. Furthermore, we studied the effects of PBO in mice. Mice eliminated bumetanide less rapidly than rats (elimination half-life 47 min vs. 13 min). Pretreatment with PBO increased the half-life in mice to average values (70 min) previously determined in humans, and markedly elevated brain levels of bumetanide. In rats, the increase in plasma and brain levels of bumetanide by PBO was less marked than in mice. PBO significantly increased the diuretic activity of bumetanide in rats and, less effectively, in mice. In epileptic mice, bumetanide (with PBO) did not suppress spontaneous seizures. In the rat kindling model, bumetanide (with or without PBO) did not exert anticonvulsant effects on fully kindled seizures, but dose-dependently altered kindling development. These data indicate that PBO offers a simple means to enhance the translational properties of rodent experiments with bumetanide, particularly when using mice.


Subject(s)
Brain/drug effects , Bumetanide/therapeutic use , Diuretics/therapeutic use , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Status Epilepticus/drug therapy , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/physiopathology , Bumetanide/pharmacokinetics , Bumetanide/pharmacology , Diuretics/pharmacokinetics , Diuretics/pharmacology , Kainic Acid/toxicity , Mice , Piperonyl Butoxide/pharmacology , Piperonyl Butoxide/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Status Epilepticus/chemically induced
17.
PLoS One ; 8(3): e57148, 2013.
Article in English | MEDLINE | ID: mdl-23536761

ABSTRACT

Neonatal seizures can be refractory to conventional anticonvulsants, and this may in part be due to a developmental increase in expression of the neuronal Na(+)-K(+)-2 Cl(-) cotransporter, NKCC1, and consequent paradoxical excitatory actions of GABAA receptors in the perinatal period. The most common cause of neonatal seizures is hypoxic encephalopathy, and here we show in an established model of neonatal hypoxia-induced seizures that the NKCC1 inhibitor, bumetanide, in combination with phenobarbital is significantly more effective than phenobarbital alone. A sensitive mass spectrometry assay revealed that bumetanide concentrations in serum and brain were dose-dependent, and the expression of NKCC1 protein transiently increased in cortex and hippocampus after hypoxic seizures. Importantly, the low doses of phenobarbital and bumetanide used in the study did not increase constitutive apoptosis, alone or in combination. Perforated patch clamp recordings from ex vivo hippocampal slices removed following seizures revealed that phenobarbital and bumetanide largely reversed seizure-induced changes in EGABA. Taken together, these data provide preclinical support for clinical trials of bumetanide in human neonates at risk for hypoxic encephalopathy and seizures.


Subject(s)
Bumetanide/administration & dosage , Hypoxia/complications , Phenobarbital/administration & dosage , Seizures/etiology , Animals , Animals, Newborn , Anticonvulsants/administration & dosage , Anticonvulsants/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Bumetanide/pharmacokinetics , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Cell Death/drug effects , Drug Synergism , Drug Therapy, Combination , Electroencephalography , Evoked Potentials/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Phenobarbital/pharmacokinetics , Rats , Seizures/drug therapy , Seizures/metabolism , Seizures/physiopathology , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Symporters/metabolism , K Cl- Cotransporters
18.
Eur J Drug Metab Pharmacokinet ; 38(1): 69-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22618469

ABSTRACT

This study presents application of a new linear and nonlinear fractional derivative two compartmental model to the evaluation of individual pharmacokinetics. In the model, the integer order derivatives are replaced by derivatives of real order. A specific nonlinear function is used for the fit improvement of a fractional derivative two compartmental model with the mass balance conservation. The agreement of the values predicted by the proposed model with the values obtained through experiments with bumetanide tablets in human volunteers is shown to be good. Thus, pharmacokinetics of bumetanide can be described well by a linear or a nonlinear two compartmental model with fractional derivatives of the same order proposed here. Parameters in the model are determined by the least squares method and the particle swarm optimization numerical procedure is used. The results show that the linear fractional order two compartmental model for bumetanide is useful improvement of the classical (integer order) two compartmental model and that the nonlinear fractional order model is useful improvement of the linear model in a great number of volunteers.


Subject(s)
Bumetanide/pharmacokinetics , Diuretics/pharmacokinetics , Linear Models , Models, Biological , Nonlinear Dynamics , Administration, Oral , Bumetanide/administration & dosage , Diuretics/administration & dosage , Humans , Least-Squares Analysis , Tablets
19.
Paediatr Drugs ; 14(4): 233-46, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22702741

ABSTRACT

The loop diuretics furosemide and bumetanide are used widely for the management of fluid overload in both acute and chronic disease states. To date, most pharmacokinetic studies in neonates have been conducted with furosemide and little is known about bumetanide. The aim of this article was to review the published data on the pharmacology of furosemide and bumetanide in neonates and infants in order to provide a critical analysis of the literature, and a useful tool for physicians. The bibliographic search was performed electronically using PubMed and EMBASE databases as search engines and March 2011 was the cutoff point. The half-life (t(½)) of both furosemide and bumetanide is considerably longer in neonates than in adults and consequently the clearance (CL) of these drugs is reduced at birth. In healthy volunteers, plasma t(½) of furosemide ranges from 33 to 100 minutes, whereas in neonates it ranges from 8 to 27 hours. The volume of distribution (V(d)) of furosemide undergoes little variation during neonate maturation. The dose of furosemide, administered by intermittent intravenous infusion, is 1 mg/kg and may increase to a maximum of 2 mg/kg every 24 hours in premature infants and every 12 hours in full-term infants. Comparison of continuous infusion versus intermittent infusion of furosemide showed that the diuresis is more controlled with fewer hemodynamic and electrolytic variations during continuous infusion. The appropriate infusion rate of furosemide ranges from 0.1 to 0.2 mg/kg/h and when the diuresis is <1 mL/kg/h the infusion rate may be increased to 0.4 mg/kg/h. Treatment with theophylline before administration of furosemide results in a significant increase of urine flow rate. Bumetanide is more potent than furosemide and its dose after intermittent intravenous infusion ranges from 0.005 to 0.1 mg/kg every 24 hours. The t(½) of bumetanide in neonates ranges from 1.74 to 7.0 hours. Up to now, no data are available on the continuous infusion of bumetanide. Extracorporeal membrane oxygenation (ECMO) is used for a variety of indications including sepsis, persistent pulmonary hypertension, meconium aspiration syndrome, cardiac defects and congenital diaphragmatic hernia. There are two studies of furosemide in neonates undergoing ECMO and only one on the pharmacokinetics of bumetanide under ECMO. When ECMO was conducted for 72 hours, the total amount of furosemide administered was 7.0 mg/kg, and the urine production in the 3 days of treatment was about 6 mL/kg/h, which is the target value. The t(½) of bumetanide in neonates during ECMO was extremely variable. CL, t(½), and V(d) were 0.63 mL/min/kg, 13.2 hours, and 0.45 L/kg, respectively. Furosemide may be administered by inhalation and inhibits the bronco-constrictive effect of exercise, cold air ventilation and antigen challenge. However, inhaled furosemide is not active in infants with viral bronchiolitis and its effect on broncho-pulmonary dysplasia is still uncertain. Furosemide does not significantly increase the risk of failure of patent ductus arteriosus closure when indomethacin or ibuprofen have been co-administered. Infants with low birth weight treated long-term with furosemide are at risk for the development of intra-renal calcification. Furosemide therapy above 10 mg/kg bodyweight cumulative dose had a 48-fold increased risk of nephrocalcinosis. The use of furosemide in combination with indomethacin increased the incidence of acute renal failure. The maturation of the kidney governs the pharmacokinetics of furosemide and bumetanide in the infant. CL and t(½) are influenced by development, and this must be taken into consideration when planning a dosage regimen with these drugs.


Subject(s)
Bumetanide/pharmacology , Furosemide/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Acetazolamide/therapeutic use , Bumetanide/pharmacokinetics , Bumetanide/therapeutic use , Drug Therapy, Combination , Ductus Arteriosus, Patent/drug therapy , Extracorporeal Membrane Oxygenation , Furosemide/pharmacokinetics , Furosemide/therapeutic use , Humans , Hydrocephalus/drug therapy , Hypercalcemia/drug therapy , Ibuprofen/therapeutic use , Indomethacin/therapeutic use , Infant , Infant, Newborn , Infant, Premature , Sodium Potassium Chloride Symporter Inhibitors/pharmacokinetics , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use
20.
J Biol Chem ; 287(21): 17308-17317, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22437837

ABSTRACT

The Na-K-Cl cotransporter (NKCC) plays central roles in cellular chloride homeostasis and in epithelial salt transport, but to date little is known about the mechanism by which the transporter moves ions across the membrane. We examined the functional role of transmembrane helix 3 (TM3) in NKCC1 using cysteine- and tryptophan-scanning mutagenesis and analyzed our results in the context of a structural homology model based on an alignment of NKCC1 with other amino acid polyamine organocation superfamily members, AdiC and ApcT. Mutations of residues along one face of TM3 (Tyr-383, Met-382, Ala-379, Asn-376, Ala-375, Phe-372, Gly-369, and Ile-368) had large effects on translocation rate, apparent ion affinities, and loop diuretic affinity, consistent with a proposed role of TM3 in the translocation pathway. The prediction that Met-382 is part of an extracellular gate that closes to form an occluded state is strongly supported by conformational sensitivity of this residue to 2-(trimethylammonium)ethyl methanethiosulfonate, and the bumetanide insensitivity of M382W is consistent with tryptophan blocking entry of bumetanide into the cavity. Substitution effects on residues at the intracellular end of TM3 suggest that this region is also involved in ion coordination and may be part of the translocation pathway in an inward-open conformation. Mutations of predicted pore residues had large effects on binding of bumetanide and furosemide, consistent with the hypothesis that loop diuretic drugs bind within the translocation cavity. The results presented here strongly support predictions of homology models of NKCC1 and demonstrate important roles for TM3 residues in ion translocation and loop diuretic inhibition.


Subject(s)
Bumetanide/pharmacology , Diuretics/pharmacology , Mutagenesis , Sodium-Potassium-Chloride Symporters/metabolism , Binding Sites , Bumetanide/pharmacokinetics , Diuretics/pharmacokinetics , HEK293 Cells , Humans , Ion Transport/drug effects , Mesylates/pharmacology , Mutation, Missense , Protein Structure, Secondary , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Sulfhydryl Reagents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...