1
|
Emerging role of carbonic anhydrase inhibitors. Clin Sci (Lond) 2021; 135:1233-1249. [PMID: 34013961 DOI: 10.1042/cs20210040] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
Inhibition of carbonic anhydrase (CA, EC 4.2.1.1) was clinically exploited for decades, as most modern diuretics were obtained considering as lead molecule acetazolamide, the prototypical CA inhibitor (CAI). The discovery and characterization of multiple human CA (hCA) isoforms, 15 of which being known today, led to new applications of their inhibitors. They include widely clinically used antiglaucoma, antiepileptic and antiobesity agents, antitumor drugs in clinical development, as well as drugs for the management of acute mountain sickness and idiopathic intracranial hypertension (IIH). Emerging roles of several CA isoforms in areas not generally connected to these enzymes were recently documented, such as in neuropathic pain, cerebral ischemia, rheumatoid arthritis, oxidative stress and Alzheimer's disease. Proof-of-concept studies thus emerged by using isoform-selective inhibitors, which may lead to new clinical applications in such areas. Relevant preclinical models are available for these pathologies due to the availability of isoform-selective CAIs for all human isoforms, belonging to novel classes of compounds, such as coumarins, sulfocoumarins, dithiocarbamates, benzoxaboroles, apart the classical sulfonamide inhibitors. The inhibition of CAs from pathogenic bacteria, fungi, protozoans or nematodes started recently to be considered for obtaining anti-infectives with a new mechanism of action.
Collapse
|
2
|
Casanova AG, Fuentes-Calvo I, Hernández-Sánchez MT, Quintero M, Toral P, Caballero MT, Martínez-Salgado C, Morales AI, Layton AT, Eleno N, López-Hernández FJ. The furosemide stress test and computational modeling identify renal damage sites associated with predisposition to acute kidney injury in rats. Transl Res 2021; 231:76-91. [PMID: 33253980 DOI: 10.1016/j.trsl.2020.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI) diagnosis relies on plasma creatinine concentration (Crpl), a relatively insensitive, surrogate biomarker of glomerular filtration rate that increases only after significant damage befalls. However, damage in different renal structures may occur without increments in Crpl, a condition known as subclinical AKI. Thus, detection of alterations in other aspects of renal function different from glomerular filtration rate must be included in an integral diagnosis of AKI. With this aim, we adapted to and validated in rats (for preclinical research) the furosemide stress test (FST), a tubular function test hitherto performed only in humans. We also tested its sensitivity in detecting subclinical tubular alterations. In particular, we predisposed rats to AKI with 3 mg/kg cisplatin and subsequently subjected them to a triggering insult (ie, 50 mg/kg/d gentamicin for 6 days) that had no effect on nonpredisposed animals but caused an overt AKI in predisposed rats. The FST was performed immediately before adding the triggering insult. Predisposed animals showed a reduced response to the FST (namely, reduced furosemide-induced diuresis and K+ excretion), whereas nonpredisposed animals showed no alteration, compared to the controls. Computational modeling of epithelial transport of solutes and water along the nephrons applied to experimental data suggested that proximal tubule transport was only minimally reduced, the sodium-chloride symporter was upregulated by 50%, and the renal outer medullary potassium channel was downregulated by 85% in predisposed animals. In conclusion, serial coupling of the FST and computational modeling may be used to detect and localize subclinical tubular alterations.
Collapse
Affiliation(s)
- Alfredo G Casanova
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain; Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Fuentes-Calvo
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain
| | - María T Hernández-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Quintero
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula Toral
- Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain
| | - María T Caballero
- Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martínez-Salgado
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain; Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain; Disease and Theranostic Modeling (DisMOD) International Consortium, Salamanca, Spain
| | - Ana I Morales
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain; Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain; Disease and Theranostic Modeling (DisMOD) International Consortium, Salamanca, Spain
| | - Anita T Layton
- Departments of Applied Mathematics and Biology, and Schools of Computer Science and Pharmacology, University of Waterloo, Waterloo, Ontario, Canada; Disease and Theranostic Modeling (DisMOD) International Consortium, Salamanca, Spain.
| | - Nélida Eleno
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain.
| | - Francisco J López-Hernández
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain; Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain; Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain; Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain, National Network for Kidney Research REDINREN, RD016/0009/0025, Instituto de Salud Carlos III, Madrid, Spain; Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain; Disease and Theranostic Modeling (DisMOD) International Consortium, Salamanca, Spain.
| |
Collapse
|
3
|
Kutovaya IV, Zarezin DP, Shmatova OI, Nenajdenko VG. Six-Component Azido-Ugi Reaction: from Cyclic Ketimines to Bis-Tetrazole-Derived 5-7-Membered Amines. European J Org Chem 2019. [DOI: 10.1002/ejoc.201900244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Irina V. Kutovaya
- Department of Chemistry; Moscow State University; Leninskie Gory Moscow 119992 Russia
| | - Danil P. Zarezin
- Department of Chemistry; Moscow State University; Leninskie Gory Moscow 119992 Russia
| | - Olga I. Shmatova
- Department of Chemistry; Moscow State University; Leninskie Gory Moscow 119992 Russia
| | | |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW The kidney, like most other organs, has a reserve capacity that can be utilized in times of increased physiologic demand. The ability to quantify this renal reserve function across various parts of the nephron (glomerular and tubular) has been an area of increased investigation over the past several years. In this review, we discuss several techniques that have been developed to interrogate the maximal physiologic capacity of the injured kidney. RECENT FINDINGS Although protein loading has been established as an ideal method to investigate glomerular filtration capacity in healthy kidneys, other methods such as the antagonism of the renin-angiotensin-aldosterone system have demonstrated promise as a method to determine underlying glomerular disease in those with acute kidney injury and other comorbidities (e.g., congestive heart failure and chronic kidney disease). The furosemide stress test has been demonstrated to be a useful clinical tool to ascertain tubular integrity in the setting of acute kidney injury. SUMMARY Although various methods to interrogate the reserve capacity of the several nephron segments (glomerulus and tubules) have been investigated, none of these techniques have had wide-spread clinical implementation. Further research into acute kidney injury stress testing is warranted.
Collapse
|
5
|
McMahon BA, Koyner JL, Novick T, Menez S, Moran RA, Lonze BE, Desai N, Alasfar S, Borja M, Merritt WT, Ariyo P, Chawla LS, Kraus E. The prognostic value of the furosemide stress test in predicting delayed graft function following deceased donor kidney transplantation. Biomarkers 2017; 23:61-69. [PMID: 29034718 DOI: 10.1080/1354750x.2017.1387934] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES AND METHODS The Furosemide Stress Test (FST) is a novel dynamic assessment of tubular function that has been shown in preliminary studies to predict patients who will progress to advanced stage acute kidney injury, including those who receive renal replacement therapy (RRT). The aim of this study is to investigate if the urinary response to a single intraoperative dose of intravenous furosemide predicts delayed graft function (DGF) in patients undergoing deceased donor kidney transplant. RESULTS On an adjusted multiple logistic regression, a single 100 mg dose of intraoperative furosemide after the anastomosis of the renal vessels (FST) predicted the need for RRT at 2 and 6 h post kidney transplantation (KT). Recipient urinary output was measured at 2 and 6 h post furosemide administration. In receiver-operating characteristic (ROC) analysis, the FST predicted DGF with an area-under-the curve of 0.85 at an optimal urinary output cut-off of <600 mls at 6 h with a sensitivity of and a specificity of 83% and 74%, respectively. CONCLUSIONS The FST is a predictor of DGF post kidney transplant and has the potential to identify patients requiring RRT early after KT.
Collapse
Affiliation(s)
- Blaithin A McMahon
- a Division of Nephrology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Jay L Koyner
- b Section of Nephrology, Department of Medicine , University of Chicago , Chicago , IL , USA
| | - Tessa Novick
- a Division of Nephrology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Steve Menez
- a Division of Nephrology , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Robert A Moran
- c Division of Gastroenterology, Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Bonnie E Lonze
- d Comprehensive Renal Transplantation Unit , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Niraj Desai
- d Comprehensive Renal Transplantation Unit , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Sami Alasfar
- a Division of Nephrology , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Comprehensive Renal Transplantation Unit , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Marvin Borja
- d Comprehensive Renal Transplantation Unit , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - William T Merritt
- e Johns Hopkins Anesthesiology and Critical Care Medicine , Baltimore , MD , USA
| | - Promise Ariyo
- e Johns Hopkins Anesthesiology and Critical Care Medicine , Baltimore , MD , USA
| | - Lakhmir S Chawla
- f Department of Medicine, Division of Intensive Care Medicine and Division of Nephrology , Veterans Affairs Medical Center , Washington DC , USA
| | - Edward Kraus
- a Division of Nephrology , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,d Comprehensive Renal Transplantation Unit , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
6
|
Malvia H, Sharma A, Sharma P, Mishra R. A Micellar Liquid Chromatographic Method for the Determination of Azosemide in Solubilized System. J SURFACTANTS DETERG 2017. [DOI: 10.1007/s11743-017-2008-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
7
|
Chawla LS, Ronco C. Renal Stress Testing in the Assessment of Kidney Disease. Kidney Int Rep 2016; 1:57-63. [PMID: 29142914 PMCID: PMC5678605 DOI: 10.1016/j.ekir.2016.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 11/02/2022] Open
Abstract
As part of human evolutionary development, many human organ systems have innate mechanisms to adapt to increased "work demand" or stress. This reserve capacity can be informative and is used commonly in cardiology to assess cardiac function (e.g., treadmill test). Similarly, the kidney possesses reserve capacity, which can be demonstrated in at least 2 of the following renal domains: glomerular and tubular. When appropriate stimulants are used, healthy patients with intact kidneys can significantly increase their glomerular filtration rate and their tubular secretion. This approach has been used to develop diagnostics for the assessment of renal function. This article reviews both glomerular and tubular kidney stress tests and their respective diagnostic utility.
Collapse
Affiliation(s)
- Lakhmir S Chawla
- Department of Medicine, Veterans Affairs Medical Center, Washington, DC, USA.,Department of Medicine, George Washington University, Washington, DC, USA
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Vicenza, Italy.,International Renal Research Institute of Vicenza, Vicenza, Italy
| |
Collapse
|
8
|
Chawla LS, Davison DL, Brasha-Mitchell E, Koyner JL, Arthur JM, Shaw AD, Tumlin JA, Trevino SA, Kimmel PL, Seneff MG. Development and standardization of a furosemide stress test to predict the severity of acute kidney injury. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R207. [PMID: 24053972 PMCID: PMC4057505 DOI: 10.1186/cc13015] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 09/20/2013] [Indexed: 12/24/2022]
Abstract
Introduction In the setting of early acute kidney injury (AKI), no test has been shown to definitively predict the progression to more severe stages. Methods We investigated the ability of a furosemide stress test (FST) (one-time dose of 1.0 or 1.5 mg/kg depending on prior furosemide-exposure) to predict the development of AKIN Stage-III in 2 cohorts of critically ill subjects with early AKI. Cohort 1 was a retrospective cohort who received a FST in the setting of AKI in critically ill patients as part of Southern AKI Network. Cohort 2 was a prospective multicenter group of critically ill patients who received their FST in the setting of early AKI. Results We studied 77 subjects; 23 from cohort 1 and 54 from cohort 2; 25 (32.4%) met the primary endpoint of progression to AKIN-III. Subjects with progressive AKI had significantly lower urine output following FST in each of the first 6 hours (p<0.001). The area under the receiver operator characteristic curves for the total urine output over the first 2 hours following FST to predict progression to AKIN-III was 0.87 (p = 0.001). The ideal-cutoff for predicting AKI progression during the first 2 hours following FST was a urine volume of less than 200mls(100ml/hr) with a sensitivity of 87.1% and specificity 84.1%. Conclusions The FST in subjects with early AKI serves as a novel assessment of tubular function with robust predictive capacity to identify those patients with severe and progressive AKI. Future studies to validate these findings are warranted.
Collapse
|
9
|
Kim YC, Lee MG, Ko SH, Kim SH. Effects of the rate and composition of fluid replacement on the pharmacokinetics and pharmacodynamics of intravenous torasemide. J Pharm Pharmacol 2010; 55:1515-22. [PMID: 14713362 DOI: 10.1211/0022357022034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
The effects of differences in the rate and composition of intravenous fluid replacement for urine loss on the pharmacokinetics and pharmacodynamics of torasemide were evaluated in rabbits. Each rabbit received 2-h constant intravenous infusion of 1 mg kg−1 torasemide with 0% replacement (treatment 1, n = 6), 50% replacement (treatment 2, n = 9), 100% replacement with lactated Ringer's solution (treatment 3, n = 8), and 100% replacement with 5% dextrose in water (treatment 4, n = 6). Total body (4.53, 5.72, 10.0 and 4.45 mL min−1 kg−1 for treatments 1–4, respectively) and renal clearance (1.44, 1.87, 6.78 and 1.72 mL min−1 kg−1) of torasemide, and total amount of unchanged torasemide excreted in 8-h urine (Ae 0–8 h: 694, 780, 1310 and 1040 μg) in treatment 3 were considerably faster and greater compared with treatments 1, 2 and 4. Although the difference in Ae 0–8 h between treatments 1 and 3 was only 88.8%, the diuretic and/or natriuretic effects of torasemide were markedly different among the four treatments. For example, the mean 8-h urine output was 101, 185, 808 and 589 mL for treatments 1–4, respectively, and the corresponding values for sodium excretion were 10.1, 20.6, 89.2 and 29.9 mmol, and for chloride excretion were 14.5, 27.9, 94.0 and 37.2 mmol. Although full fluid replacement was used in both treatments 3 and 4, the 8-h diuretic, natriuretic and chloruretic effects in treatment 3 were significantly greater compared with treatment 4, indicating the importance of the composition of fluid replacement. Both treatments 1 and 4 received no sodium replacement, however, the 8-h diuretic, natriuretic and chloruretic effects were significantly greater in treatment 4 compared with treatment 1, indicating the importance of rate of fluid replacement for the diuretic effects. Therefore, the 8-h diuretic, natriuretic and chloruretic effects were significantly greater in treatment 3 compared with treatments 1, 2 and 4, indicating the importance of full fluid and electrolyte replacement. Some implications for the bioequivalence evaluation of dosage forms of torasemide are discussed.
Collapse
Affiliation(s)
- Yu Chul Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, San 56-1, Shinlim-Dong, Kwanak-Gu, Seoul 151-742, Korea
| | | | | | | |
Collapse
|
10
|
Kim EJ, Lee AK, Kim SH, Kim SG, Lee MG. Pharmacokinetics and pharmacodynamics of intravenous azosemide in mutant Nagase analbuminemic rats. Drug Metab Dispos 2003; 31:194-201. [PMID: 12527700 DOI: 10.1124/dmd.31.2.194] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This paper reports 1) the increase in expression of CYP1A2 in mutant Nagase analbuminemic rats (NARs), 2) the role of globulin binding of azosemide in circulating blood in its urinary excretion and hence its diuretic effects in NARs, and 3) the significantly faster renal (CL(R)) and nonrenal (CL(NR)) clearances of azosemide in NARs. Azosemide (mainly metabolized via CYP1A2 in rats), 10 mg/kg, was intravenously administered to control rats and NARs. Northern and Western blot analyses revealed that the expression of CYP1A2 increased approximately 3.5-fold in NARs as compared with control. The plasma protein binding of azosemide in control rats and NARs was 97.9 and 84.6%, respectively. In NARs, plasma protein binding (84.6%) was due to binding to alpha- (82.6%) and beta- (68.9%) globulins. In NARs, the amount of unchanged azosemide excreted in 8-h urine was significantly greater (37.7 versus 21.0% of intravenous dose) than that in control rats due to an increase in intrinsic renal active secretion of azosemide. Accordingly, the 8-h urine output was significantly greater in NARs. The area under the plasma concentration-time curve of azosemide was significantly smaller (505 versus 2790 microg. min/ml) in NARs because of markedly faster CL(R) (7.36 versus 0.772 ml/min/kg, secondary to a significant increase in urinary excretion of azosemide and intrinsic renal active secretion). Additionally, CL(NR) was significantly faster (12.4 versus 3.05 ml/min/kg, because of approximately 3.5 fold increase in CYP1A2) in NARs compared with control. Based on in vitro hepatic microsomal studies, the intrinsic M1 [a metabolite of azosemide; 5-(2-amino-4-chloro-5-sulfamoylphenyl)-tetrazole] formation clearance was significantly faster (67.0% increase) in NARs than that in control rats, and this supports significantly faster CL(NR) in NARs. Renal sensitivity to azosemide was significantly greater in NARs than in control rats with respect to 8-h urine output (385 versus 221 ml/kg) and 8-h urinary excretions of sodium, potassium, and chloride. This study supports that in NARs, binding of azosemide to alpha- and beta-globulins in circulating blood play an important role in its diuretic effects.
Collapse
Affiliation(s)
- Eun J Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, San 56-1, Shinlim-Dong, Kwanak-Gu, Seoul 151-742, South Korea
| | | | | | | | | |
Collapse
|
11
|
Abstract
Azosemide is used in the treatment of oedematous states and hypertension. The exact mechanism of action is not fully understood, but it mainly acts on both the medullary and cortical segments of the thick ascending limb of the loop of Henle. Delayed tolerance was demonstrated in humans by homeostatic mechanisms (principally an increase in aldosterone secretion and perhaps also an increase in the reabsorption of solute in the proximal tubule). After oral administration to healthy humans in the fasting state, the plasma concentration of azosemide reached its peak at 3-4 h with an absorption lag time of approximately 1 h and a terminal half-life of 2-3 h. The estimated extent of absolute oral bioavailability in humans was approximately 20.4%. After oral administration of the same dose of azosemide and furosemide, the diuretic effect was similar between the two drugs, but after intravenous administration, the effect of azosemide was 5.5-8 times greater than that in furosemide. This could be due to the considerable first-pass effect of azosemide. The protein binding to 4% human serum albumin was greater than 95% at azosemide concentrations ranging from 10 to 100 microg/ml using an equilibrium dialysis technique. The poor affinity of human tissues to azosemide was supported by the relatively small value of the apparent post-pseudodistribution volume of distribution (Vdbeta), 0.262 l/kg. Eleven metabolites (including degraded products) of azosemide including M1, glucuronide conjugates of both M1 and azosemide, thiophenemethanol, thiophencarboxylic acid and its glycine conjugate were obtained in rats. Only azosemide and its glucuronide were detected in humans. In humans, total body clearance, renal clearance and terminal half-life of azosemide were 112 ml/min, 41.6 ml/min and 2.03 h, respectively. Azosemide is actively secreted in the renal proximal tubule possibly via nonspecific organic acid secretory pathway in humans. Thus, the amount of azosemide that reaches its site of action could be significantly modified by changes in the capacity of this transport system. This capacity, in turn, could be predictably changed in disease states, resulting in decreased delivery of the diuretic to the transport site, as well as in the presence of other organic acids such as nonsteroidal anti-inflammatory drugs which could compete for active transport of azosemide. The urinary excretion rate of azosemide could be correlated well to its diuretic effects since the receptors are located in the loop of Henle. The diuretic effects of azosemide were dependent on the rate and composition of fluid replacement in rabbits; therefore, this factor should be considered in the evaluation of bioequivalence assessment.
Collapse
Affiliation(s)
- Ok K Suh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | | | | |
Collapse
|
12
|
Kim HJ, Lee AK, Kim YG, Bu SC, Kim SH, Yang CH, Suh OK, Kim SG, Lee MG. Influence of 4-week and 8-week exercise training on the pharmacokinetics and pharmacodynamics of intravenous and oral azosemide in rats. Life Sci 2002; 70:2299-319. [PMID: 12005188 DOI: 10.1016/s0024-3205(02)01516-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Cytochrome P450 expression was determined in the livers of control, 4-week exercised (4WE) and 8-week exercised (8WE) rats. Even though the 4-week and 8-week exercise training caused 53 and 25% increases, respectively, in total cytochrome P450 contents in the liver, exercise training did not cause any changes in the levels of P450 1A2 (which primarily metabolizes azosemide), 2E1 and 3A23 in the liver, as assessed by both Western and Northern blot analyses. Also, exercise training failed to alter the activity of NADPH-dependent cytochrome P450 reductase. The plasma concentrations of norepinephrine and epinephrine were significantly (2 to 3 folds) higher in 4WE rats than in controls, presumably due to physical stress, but the catecholamine levels in 8 WE rats returned to control levels. After intravenous administration (10 mg/kg of azosemide), the amount of unchanged azosemide excreted in 8-h urine (Ae(Azo, 0-8 h)) was significantly greater (46% increase) in 4WE rats than that in control rats. This resulted in a significantly faster (82% increase) renal clearance of azosemide. However, the nonrenal clearances were not significantly different between control and 4WE rats. The significantly greater Ae(Azo, 0-8 h) in 4WE rats was mainly due to a significant increase in intrinsic active secretion of azosemide in renal tubules and not due to a decrease in the metabolism of azosemide. After oral administration (20 mg/kg), Ae(Azo, 0-8 h) was also significantly greater (264%) in 4WE rats and this again was due to a significant increase in intrinsic active renal secretion of azosemide and not due to an increase in gastrointestinal absorption. After both intravenous and oral administration, the 8-h urine output was not significantly different between control and 4WE rats although Ae(Azo, 0-8 h) increased significantly in 4WE rats. This could be due to the fact that the urine output reached a plateau at 10 mg/kg after intravenous administration and 20 mg/kg after oral administration of azosemide to rats and possibly due to increase in plasma antidiuretic hormone levels and aldosterone production in 4WE rats.
Collapse
Affiliation(s)
- Hee J Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Kwanak-Gu, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Gender differences in pharmacokinetics and pharmacodynamics of azosemide were evaluated after intravenous, 10 mg kg(-1), and oral, 10 mg kg(-1), administration to male and female rats. After intravenous administration to male rats, the percentages of intravenous dose of azosemide recovered from entire gastrointestinal tract at 24 h (13.2 versus 3.93%) was significantly greater than those in female rats. In male rats, the nonrenal clearance of azosemide tended (p<0.066) to be faster and kidney weight tended (p<0.068) to be greater than those in female rats. After oral administration of azosemide to male rats, the 8-h urinary excretion of potassium (0.395 versus 0.766 mmol g(-1) kidney) and 8-h kaluretic efficiency (55.9 versus 284 mmol mg(-1)) decreased significantly compared with female rats.
Collapse
Affiliation(s)
- Y S Lee
- College of Pharmacy, Seoul National University, San 56-1, Shinlim-Dong, Kwanak-Gu, Seoul 151-742, Republic of Korea
| | | | | |
Collapse
|
14
|
Han KS, Lee MG. Circadian changes in the pharmacokinetics and pharmacodynamics of azosemide in rats. J Pharm Pharmacol 1998; 50:767-74. [PMID: 9720626 DOI: 10.1111/j.2042-7158.1998.tb07138.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The circadian changes in the pharmacokinetics and pharmacodynamics of azosemide were investigated after intravenous and oral administration of the drug (10 mg kg(-1)) to rats at 1000 or 2200 h. After intravenous administration of azosemide the percentage of the dose excreted in 8-h urine as unchanged azosemide was significantly higher in the 1000 h group than in the 2200 h group (41.7 compared with 28.9%) and this resulted in a significant increase in 8-h urine output (84.7 compared with 36.6 mL/100 g). After intravenous administration the time-averaged renal clearance (CLR) of azosemide was significantly faster (2.86 compared with 1.76 mL min(-1) kg(-1)) and urinary excretion of sodium (46.4 compared with 25.9 mmol/100 g) and chloride (35.6 compared with 18.8 mmol/100 g) increased significantly in the 1000 h group. However, after oral administration, the percentages of oral dose of azosemide excreted in 8-h urine as unchanged azosemide were significantly higher (1.88 compared with 0.67%) and the CL(R) of azosemide was significantly faster (3.64 compared with 0.79 mL min(-1) kg(-1)) in the 2200 h group. This could be at least partly because of increased absorption of azosemide from the gastrointestinal tract in the 2200 h group; the percentages of oral dose of azosemide recovered from the gastrointestinal tract in 8 h as unchanged azosemide was significantly smaller (5.7 compared with 13.2%) in the 2200 h group. The pharmacodynamic parameters of azosemide were not significantly different after oral administration of the drug to both groups of rats. If these data could be extrapolated to man, the intravenous dose of azosemide could be modified on the basis of circadian time.
Collapse
Affiliation(s)
- K S Han
- College of Pharmacy, Seoul National University, Korea
| | | |
Collapse
|
15
|
Abstract
Since considerable first-pass effects of azosemide have been reported after oral administration of the drug to rats and man, first-pass effects of azosemide were evaluated after intravenous, intraportal and oral administration, and intraduodenal instillation of the drug, to rats. The total body clearances of azosemide after intravenous (5 mg kg-1) and intraportal (5 and 10 mg kg-1) administration of the drug to rats were considerably smaller than the cardiac output of rats suggesting that the lung or heart first-pass effect (or both) of azosemide after oral administration of the drug to rats was negligible. The total area under the plasma concentration-time curve from time zero to time infinity (AUC) after intraportal administration (5 mg kg-1) of the drug was significantly lower than that after intravenous administration (5 mg kg-1) of the drug (1000 vs 1270 micrograms min mL-1) suggesting that the liver first-pass effect of azosemide was approximately 20% in rats. The AUC from time 0 to 8 h (AUC0-8 h) after oral administration (5 mg kg-1) of the drug was considerably smaller than that after intraportal administration (5 mg kg-1) of the drug (27.1 vs 1580 micrograms min mL-1) suggesting that there are considerable gastrointestinal first-pass effects of azosemide after oral administration of azosemide to rats. Although the AUC0-8 h after oral administration (5 mg kg-1) of azosemide was approximately 15% lower than that after intraduodenal instillation (5 mg kg-1) of the drug (27.1 vs 32.0 micrograms min mL-1), the difference was not significant, suggesting that the gastric first-pass effect of azosemide was not considerable in rats. Azosemide was stable in human gastric juices and pH solutions ranging from 2 to 13. Almost complete absorption of azosemide from whole gastrointestinal tract was observed after oral administration of the drug to rats. The above data indicated that most of the orally administered azosemide disappeared (mainly due to metabolism) following intestinal first-pass in rats.
Collapse
Affiliation(s)
- J Kim
- College of Pharmacy, Seoul National University, Korea
| | | | | |
Collapse
|
16
|
Lee SH, Lee MG. Pharmacokinetics and pharmacodynamics of azosemide after intravenous and oral administration to rats: absorption from various GI segments. JOURNAL OF PHARMACOKINETICS AND BIOPHARMACEUTICS 1996; 24:551-68. [PMID: 9300350 DOI: 10.1007/bf02353480] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Azosemide, 5, 10, 20, and 30 mg/kg, was administered both intravenously and orally to determine the pharmacokinetics and pharmacodynamics of azosemide in rats (n = 7-12). The absorption of azosemide from various segments of GI tract and the reasons for the appearance of multiple peaks in plasma concentrations of azosemide after oral administration were also investigated. After intravenous (iv) dose, the pharmacokinetic parameters of azosemide such as t1/2. MRT, VSS, CL, CLR, and CLNR were found to be dose-dependent in the dose ranges studied. The percentages of the iv dose excreted in 8-hr urine as azosemide, MI (a metabolite of azosemide), glucuronide of azosemide, and glucuronide of MI-expressed in terms of azosemide-were also dose-dependent in the dose ranges studied. The data above suggest saturable metabolism of azosemide in rats. The measurements taken after the iv administrations such as the 8 hr urine output, the total amount of sodium and chloride excreted in 8-hr urine per 100 g body weight, and diuretic, natriuretic, kaluretic, and chloruretic efficiencies were also shown to be dose-dependent. However, the total amount of potassium excreted in 8-hr urine per 100 g body weight was dose-independent. Similar dose-dependency was also observed following oral administration. Azosemide was absorbed from all regions of GI tract studied and approximately 93.5, 79.1, 86.1, and 71.5% of the doses (5, 10, 20, and 30 mg/kg, respectively) were absorbed between 1 and 24 hr after oral administration. The appearance of multiple peaks after oral administration is suspected to be due mainly to the gastric emptying pattern. The percentages of azosemide absorbed from the GI tract as unchanged azosemide for up to 24 hr after oral doses of 5, 10, 20, and 30 mg/kg were significantly different with doses (decreased with increasing doses), suggesting that the problem of azosemide precipitating in acidic gastric juices or dissolution may have at least partially influenced the absorption of azosemide after oral administration.
Collapse
Affiliation(s)
- S H Lee
- College of Pharmacy, Seoul National University, Korea
| | | |
Collapse
|
17
|
Park KJ, Yoon WH, Shin WG, Lee MG. Pharmacokinetics and pharmacodynamics of azosemide after intravenous and oral administration to rats with alloxan-induced diabetes mellitus. J Pharm Pharmacol 1996; 48:1093-7. [PMID: 8953515 DOI: 10.1111/j.2042-7158.1996.tb05906.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Because physiological changes occurring in diabetes mellitus patients could alter the pharmacokinetics and pharmacodynamics of the drugs used to treat the disease, the pharmacokinetics and pharmacodynamics of azosemide were investigated after intravenous and oral administration of the drug (10 mg kg-1) to control and alloxan-induced diabetes mellitus rats (AIDRs). After intravenous administration of azosemide to the AIDRs, the area under the plasma concentration-time curve (AUC) increased considerably (3120 compared with 2520 micrograms min mL-1; P < 0.135) and the total body clearance decreased considerably (3.20 compared with 3.96 mL min-1 kg-1; P < 0.0593). The considerable reduction in time-averaged total body clearance in the AIDRs was a result of the significant decrease in renal clearance (1.01 compared with 1.55 mL min-1 kg-1) in the AIDRs, the non-renal clearance being comparable between the two groups of rats. After intravenous administration, the 8-h urinary excretion of azosemide (29.5 compared with 40% of intravenous dose; P < 0.0883) and one of its metabolites, M1 (2.15 compared with 2.60% of intravenous dose, expressed in terms of azosemide; P < 0.05) decreased in the AIDRs because of the impaired kidney function. The diuretic, natriuretic, kaliuretic and chloruretic efficiencies increased significantly in the AIDRs. After oral administration of azosemide, AUC decreased significantly in the AIDRs (115 compared with 215 micrograms min mL-1) possibly because of the reduced gastrointestinal absorption of azosemide in the AIDRs. After oral administration of azosemide, the 8-h urine output decreased significantly in the AIDRs (9.32 compared with 16.1 mL per 100 g body weight) because of the significantly reduced 8-h urinary excretion of azosemide (3.00 compared with 9.14% of oral dose). After both intravenous and oral administration some pharmacokinetic and pharmacodynamic parameters of azosemide were significantly different in AIDRs.
Collapse
Affiliation(s)
- K J Park
- College of Pharmacy, Seoul National University, Korea
| | | | | | | |
Collapse
|
18
|
Lee SH, Lee MG. Stability, tissue metabolism, tissue distribution and blood partition of azosemide. Biopharm Drug Dispos 1995; 16:547-61. [PMID: 8785379 DOI: 10.1002/bdd.2510160704] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Stability of azosemide after incubation in various pH solutions, human plasma, human gastric juice, and rat liver homogenates, metabolism of azosemide after incubation in 9000 g supernatant fraction of various rat tissue homogenates in the presence of NADPH, tissue distribution of azosemide and M1 after intravenous (i.v.) administration of azosemide, 20 mg kg-1, to rats, and blood partition of azosemide between plasma and blood cells from rabbit blood were studied. Azosemide seemed to be stable for up to 48 h incubation in various pH solutions ranging from two to 13 at an azosemide concentration of 10 micrograms mL-1; more than 93.4% of azosemide was recovered, and a metabolite of azosemide, M1, was not detected. However, the drug was unstable in pH1 solution: 75.8% of azosemide was recovered and 2.16 micrograms mL-1 of M1 (expressed in terms of azosemide) was formed after 48 h incubation in pH 1 solution at an azosemide concentration of 10 micrograms mL-1. Azosemide was stable in both human plasma and rat liver homogenates for up to 24 h incubation at an azosemide concentration of 1 microgram mL-1, and in human gastric juice for up to 4 h incubation at an azosemide concentration of 10 micrograms mL-1. However, all rat tissues studied had metabolic activity for azosemide in the presence of NADPH, with heart having a considerable metabolic activity: approximately 22% of azosemide disappeared and 9.32 micrograms of M1 was formed per gram of heart (expressed in terms of azosemide) after 30 min incubation of 50 micrograms of azosemide in 9000 g supernatant fraction of heart homogenates. The tissue to plasma ratios of azosemide (T/P) were greater than unity only in the liver (1.26) and kidney (1.74); however, M1 showed high affinity for all tissues studied except the brain and spleen when each tissue was collected at 30 min after i.v. administration of azosemide to rats. The equilibrium plasma to blood cell concentration ratios of azosemide were independent of azosemide blood concentrations: the values were 2.78-4.25 at azosemide blood concentrations of 1, 10, and 20 micrograms mL-1 in three rabbits. There was negligible 'blood storage effect' of azosemide, especially at low blood concentrations of azosemide, such as 1 and 10 micrograms mL-1.
Collapse
Affiliation(s)
- S H Lee
- College of Pharmacy, Seoul National University, Korea
| | | |
Collapse
|
19
|
Puschett JB, Winaver J. Effects of Diuretics on Renal Function. Compr Physiol 1992. [DOI: 10.1002/cphy.cp080250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
20
|
Abstract
25 years have elapsed since the introduction of the first effective oral diuretic, chlorothiazide. Diuretics are now amongst the most widely prescribed drugs in clinical practice worldwide. Availability of these drugs has not only brought therapeutic benefit to countless numbers of patients but it has at the same time provided valuable research tools with which to investigate the functional behaviour of the kidney and other electrolyte-transporting tissues. Despite many remaining gaps in our knowledge of the biochemical processes involved in diuretic drug action, available compounds can be divided into 5 groups on the basis of their preferential effects on different segments of the nephron involved in tubular reabsorption of sodium chloride and water. Firstly, there is heterogeneous group of chemicals that share the common property of powerful, short-lived diuretic effects that are complete within 4 to 6 hours. These agents act on the thick ascending limb of Henle's loop and are known as 'high ceiling' or 'loop' diuretics. The second group are the benzothiadiazines and their many related heterocyclic variants, all of which localise their effects to the early portion of the distal tubule. The third group comprises the potassium-sparing diuretics which act exclusively on the Na+-K+/H+ exchange mechanisms in the late distal tubule and cortical collecting duct. The action of drugs in groups 2 and 3 is prolonged to between 12 and 24 hours. The fourth group consists of diuretics that are chemically related to ethacrynic acid but have the unusual property of combining within the same molecule the property of saluresis and uricosuria. These compounds have actions, to different individual extents, in the proximal tubule, thick ascending limb, and early distal tubule and are known as 'polyvalent' diuretics. Finally, there is a mixed group of weak or adjunctive diuretics which includes the vasodilator xanthines such as aminophylline, and the osmotically active compounds such as mannitol. Available evidence on the molecular mechanisms of action of diuretics in each group is reviewed. The haemodynamic, humoral and physical factors involved in control of electrolyte and fluid handling by the kidney in normal conditions and pathological states are discussed in relation to rational choices of different diuretics in the treatment of various oedematous and non-oedematous conditions.
Collapse
|
21
|
Rarey KE, Ross MD. A survey of the effects of loop diuretics on the zonulae occludentes of the perilymph-endolymph barrier by freeze fracture. Acta Otolaryngol 1982; 94:307-16. [PMID: 7148443 DOI: 10.3109/00016488209128918] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The acute (30 min) effects of the loop diuretics piretanide, ethacrynic acid, bumetanide, furosemide, and azosemide, and the chronic (8 days) effects of furosemide and bumetanide on the zonulae occludentes (tight junctions) of the perilymph-endolymph barrier in the stria vascularis and in Reissner's membrane of the basal cochlear turns were studied by freeze-fracture. Quantitative analysis of their effects indicated that the structural integrity of the barrier was modified by either as increase or a decrease in the number, depth, and density of the strands of the tight junctions of the strial marginal cells. Only azosemide appeared to modify the tight junctions between the epithelial cells of Reissner's membrane, but it had little effect on the strial junctions. The tight junctions between the basal cells of the stria appeared to be the least affected by the various loop diuretics, although piretanide appeared to increase randomly the intercellular spaces lying between the strands.
Collapse
|
22
|
Brater DC, Fox WR, Chennavasin P. Electrolyte excretion patterns. Intravenous and oral doses of bumetanide compared to furosemide. J Clin Pharmacol 1981; 21:599-603. [PMID: 7338569 DOI: 10.1002/j.1552-4604.1981.tb05670.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The response to bumetanide and furosemide administered orally and intravenously was compared in normal subjects. Doses of bumetanide were 0.5 and 1 mg intravenously and 0.5, 1 and 2 mg orally. Doses of furosemide were 20 and 40 mg intravenously and 20, 40, and 80 mg orally. After intravenous doses of both drugs, peak natriuresis occurred during the first collection period of 30 minutes and the response returned to baseline by 3.5 hours. After oral dosing, peak effect occurred at 75 minutes and returned to baseline within 4 hours. Cumulative response was assessed for volume, sodium, potassium, and chloride excretion. The response did not differ substantially between drugs. On a milligram basis, bumetanide was approximately 50 times as potent as furosemide. For both drugs, the intravenous dose was approximately three times as potent as the oral preparation. The time course of response shows that the curves are virtually superimposable. The diuretic effect was short-lived for both diuretics. The urine volume and sodium and chloride excretion were parallel. The sodium/potassium ratio was calculated as follows: for every 200 mEq sodium excreted in 4 hours, bumetanide caused about 35 mEq and furosemide caused about 50 mEq potassium to be eliminated. Even if statistically significant, this difference does not appear to be clinically remarkable. If bumetanide offers therapeutic advantages in this regard, studies in patients with various disease states would have to be performed.
Collapse
|
23
|
Horký K, Gregorová I, Dvoráková J. The effect of renin and aldosterone inhibition by beta-adrenergic blockade on the response to the new diuretic azosemide. Eur J Pharmacol 1981; 69:439-46. [PMID: 6113965 DOI: 10.1016/0014-2999(81)90447-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The effect of repeated Azosemide infusions (20 mg in 500 ml 5% glucose for one h) on urine volume and electrolyte excretion, and on the activity of the renin-angiotensin-aldosterone system (RAAS) was studied in a group of 15 patients with benign essential hypertension before and during treatment with the beta-adrenergic blocker Trimepranol. Azosemide alone had a marked but short-lasting diuretic and natriuretic effect. Repeated administration on three consecutive days led, however, to a progressive decrease in the natriuretic effectiveness of Azosemide, associated with an increase in plasma renin activity (from 0.413 o.032 to 1.631 0.438 pmol/l). Treatment with Trimepranol 20 mg/day enhanced and prolonged the diuretic and natriuretic response to Azosemide concomitantly with a reduction of its stimulatory effect of RAAS. There results suggest that stimulation of the RAAS might be responsible for the diminishing effectiveness of repeated Azosemide infusions and that the stimulation could be, at least partly, inhibited by a beta-blocker Trimepranol, resulting in a greater diuretic and natriuretic effect of Azosemide.
Collapse
|
24
|
Seiwell R, Brater C. Separation and analysis of azosemide in urine and in serum by high-performance liquid chromatography. JOURNAL OF CHROMATOGRAPHY 1980; 182:257-61. [PMID: 7380919 DOI: 10.1016/s0378-4347(00)81633-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|