Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
Forensic Sci Med Pathol ; 20(1): 100-105, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37060536

ABSTRACT

Ramipril is a popular angiotensin-converting enzyme inhibitor applied in the treatment of hypertension. Its therapeutic effect is oriented on the concentration of the active metabolite ramiprilat. The information about toxic drug levels is missing in the literature. Therefore, the aim of this work was an indication of possible toxic ranges based on the analysis of real samples with high ramiprilat concentrations. For these purposes, an appropriate analytical LC-MS/MS method was developed and validated according to forensic guidelines and applied in the routine. Most real samples targeted for ramipril/ramiprilat were associated with the typical therapeutic drug range of 1-40 ng/mL described in the literature. However, higher drug levels with ramiprilat concentrations above 100 ng/mL could also be observed infrequently in cases of driving under the influence of drugs or attempted suicides. To the best of the author's knowledge, this is the first time antemortem ramipril and ramiprilat concentrations associated with driving under the influence of drugs and suicide attempts were discussed from a forensic point of view. The collected data enabled an indication of the ramiprilat toxic concentration range from about 600 ng/mL to at least 3500 ng/mL. The toxic concentration range discussed can be applied in the forensic practice as a reference for future cases.


Subject(s)
Ramipril/analogs & derivatives , Tandem Mass Spectrometry , Humans , Chromatography, Liquid , Forensic Toxicology
2.
J Pharmacol Exp Ther ; 350(1): 5-13, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24741076

ABSTRACT

This study aims to investigate the effects of ramipril (RPL) on endothelial dysfunction associated with diabetes mellitus using cultured human aortic endothelial cells (HAECs) and a type 2 diabetic animal model. The effect of RPL on vasodilatory function in fat-fed, streptozotocin-treated rats was assessed. RPL treatment of 8 weeks alleviated insulin resistance and inhibited the decrease in endothelium-dependent vasodilation in diabetic rats. RPL treatment also reduced serum advanced glycation end products (AGE) concentration and rat aorta reactive oxygen species formation and increased aorta endothelium heme oxygenase-1 (HO-1) expression. Exposure of HAECs to high concentrations of glucose induced prolonged oxidative stress, apoptosis, and accumulation of AGEs. These effects were abolished by incubation of ramiprilat (RPT), the active metabolite of RPL. However, treatment of HAECs with STO-609, a CaMKKß (Ca(2+)/calmodulin-dependent protein kinase kinase-ß) inhibitor; compound C, an AMPK (AMP-activated protein kinase) inhibitor; and Zn(II)PPIX, a selective HO-1 inhibitor, blocked these beneficial effects of RPT. In addition, RPT increased nuclear factor erythroid 2-related factor-2 (Nrf-2) nuclear translocation and activation in a CaMKKß/AMPK pathway-dependent manner, leading to increased expression of the Nrf-2-regulated antioxidant enzyme, HO-1. The inhibition of CaMKKß or AMPK by pharmaceutical approach ablated RPT-induced HO-1 expression. Taken together, RPL ameliorates insulin resistance and endothelial dysfunction in diabetes via reducing oxidative stress. These effects are mediated by RPL activation of CaMKK-ß, which in turn activates the AMPK-Nrf-2-HO-1 pathway for enhanced endothelial function.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Diabetes Mellitus, Experimental/drug therapy , Endothelial Cells/drug effects , Glucose/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Ramipril/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/antagonists & inhibitors , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Activation/drug effects , Glycation End Products, Advanced/blood , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Humans , Insulin Resistance/physiology , Male , NF-E2-Related Factor 2/metabolism , Naphthalimides/pharmacology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protoporphyrins/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Ramipril/analogs & derivatives , Ramipril/antagonists & inhibitors , Ramipril/therapeutic use , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vasodilation/drug effects , Vasodilation/physiology
3.
Acta Pharm ; 74(2): 315-328, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38815200

ABSTRACT

In patients with chronic heart failure (CHF), the use of angiotensin-converting enzyme inhibitors, including ramipril, is recommended to reduce the risk of heart failure worsening, hospitalisation, and death. Our aim was to investigate the influence of body composition on the pharmacokinetics of ramipril and its active metabolite ramiprilat and to evaluate the changes in pharmacokinetics after prolonged therapy. Twenty-three patients with CHF who were on regular therapy with ramipril participated at the first study visit ( median age 77 years, 65 % male, and 70 % New York Heart Association Class II); 19 patients attended the second study visit and the median time between the two visits was 8 months. Pharmacokinetics were assessed using a nonlinear mixed-effects parent-metabolite model comprising two compartments for ramipril and one compartment for ramiprilat. The influence of body size and composition was best described by an allometric relationship with fat-free mass. In addition, ramipril clearance was related to patient age and daily ramipril dose, while clearance of ramiprilat was influenced by glome rular filtration rate and daily ramipril dose. There were no clinically relevant changes in the pharmacokinetics of ramipril and ramiprilat between the study visits. Due to the relatively stable pharmacokinetics of ramipril, regular outpatient visits at 6-month intervals seem appropriate to evaluate ramipril therapy.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors , Heart Failure , Ramipril , Humans , Ramipril/pharmacokinetics , Ramipril/administration & dosage , Ramipril/analogs & derivatives , Heart Failure/drug therapy , Male , Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Aged , Female , Longitudinal Studies , Chronic Disease , Aged, 80 and over , Middle Aged , Body Composition
4.
Am J Physiol Lung Cell Mol Physiol ; 304(1): L70-81, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23087017

ABSTRACT

Proximal arterial stiffening is an important predictor of events in systemic and pulmonary hypertension, partly through its contribution to downstream vascular abnormalities. However, much remains undetermined regarding the mechanisms involved in the vascular changes induced by arterial stiffening. We therefore addressed the hypothesis that high pulsatility flow, caused by proximal arterial stiffening, induces downstream pulmonary artery endothelial cell (EC) dysfunction that in turn leads to phenotypic change of smooth muscle cells (SMCs). To test the hypothesis, we employed a model pulmonary circulation in which upstream compliance regulates the pulsatility of flow waves imposed onto a downstream vascular mimetic coculture composed of pulmonary ECs and SMCs. The effects of high pulsatility flow on SMCs were determined both in the presence and absence of ECs. In the presence of ECs, high pulsatility flow increased SMC size and expression of the contractile proteins, smooth muscle α-actin (SMA) and smooth muscle myosin heavy chain (SM-MHC), without affecting proliferation. In the absence of ECs, high pulsatility flow decreased SMC expression of SMA and SM-MHC, without affecting SMC size or proliferation. To identify the molecular signals involved in the EC-mediated SMC responses, mRNA and/or protein expression of vasoconstrictors [angiotensin-converting enzyme (ACE) and endothelin (ET)-1], vasodilator (eNOS), and growth factor (TGF-ß1) in EC were examined. Results showed high pulsatility flow decreased eNOS and increased ACE, ET-1, and TGF-ß1 expression. ACE inhibition with ramiprilat, ET-1 receptor inhibition with bosentan, and treatment with the vasodilator bradykinin prevented flow-induced, EC-dependent SMC changes. In conclusion, high pulsatility flow stimulated SMC hypertrophy and contractile protein expression by altering EC production of vasoactive mediators and cytokines, supporting the idea of a coupling between proximal vascular stiffening, flow pulsatility, and downstream vascular function.


Subject(s)
Endothelial Cells/metabolism , Myocytes, Smooth Muscle/pathology , Pulsatile Flow , Actins/biosynthesis , Animals , Bosentan , Cattle , Cells, Cultured , Coculture Techniques , Endothelial Cells/pathology , Hypertrophy/metabolism , Mechanotransduction, Cellular , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Myosin Heavy Chains/biosynthesis , Peptidyl-Dipeptidase A/biosynthesis , Pulmonary Artery , Ramipril/analogs & derivatives , Ramipril/pharmacology , Sulfonamides/pharmacology , Transforming Growth Factor beta1/biosynthesis , Vascular Stiffness/physiology
6.
Biomed Chromatogr ; 25(6): 664-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20812207

ABSTRACT

In this paper, we present a validated UPLC-MS/MS assay for determination of ramipril and ramiprilat from human plasma samples. The assay is capable of isolating phase II metabolites (acylglucornides) of ramipril from in vivo study samples which is otherwise not possible using conventional HPLC conditions. Both analytes were extracted from human plasma using solid-phase extraction technique. Chromatographic separation of analytes and their respective internal standards was carried out using an Acquity UPLC BEH C(18) (2.1 × 100 mm), 1.7 µm column followed by mass spectrometric detection using an Waters Quattro Premier XE. The method was validated over the range 0.35-70.0 ng/mL for ramipril and 1.0-40.0 ng/mL for ramiprilat.


Subject(s)
Chromatography, High Pressure Liquid/methods , Ramipril/analogs & derivatives , Ramipril/pharmacokinetics , Tandem Mass Spectrometry/methods , Glucuronides , Humans , Ramipril/blood , Reproducibility of Results , Sensitivity and Specificity , Solid Phase Extraction , Spectrometry, Mass, Electrospray Ionization , Therapeutic Equivalency
7.
Biomed Chromatogr ; 24(5): 528-34, 2010 May.
Article in English | MEDLINE | ID: mdl-19795361

ABSTRACT

Sensitivity enhancement via summation of multiple MRM transition pairs is gaining popularity in tandem mass spectrometric assays. Numerous validation experiments describing the assays for two model substrates, clopidogrel and ramiprilat, were performed. The quantitation of clopidogrel was achieved by the summation of transition pairs m/z 322.2 to m/z 212.0 and m/z 322.2 to m/z 184.0, while that of ramiprilat was achieved by the summation of transition pairs m/z 389.2 to m/z 206.1 and m/z 389.2 to m/z156.1. The use of summation approach achieved sensitivities of >2 fold for both compounds as compared with the reported single MRM transition pair assays. The validation experiments addressed some important assay development issues, such as: (a) lack of impact of matrix effect; (b) unequivocal verification of the percentage contribution of each MRM transition pair towards sensitivity; (c) sensitivity enhancement factor achieved by summation approach of MRM transition pairs; and (d) accurate prediction of quality control samples using summation approach vs a single MRM transition pair. In summary, the appropriateness of using two MRM transition pairs for quantitation was demonstrated for both clopidogrel and ramiprilat. Additionally, pharmacokinetic application of the MRM transition pair assays using a summation approach was established for the two compounds.


Subject(s)
Ramipril/analogs & derivatives , Tandem Mass Spectrometry/methods , Ticlopidine/analogs & derivatives , Chromatography, Liquid/methods , Clopidogrel , Humans , Ramipril/blood , Ramipril/chemistry , Sensitivity and Specificity , Ticlopidine/blood , Ticlopidine/chemistry
8.
Am J Physiol Renal Physiol ; 297(5): F1324-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726543

ABSTRACT

To understand how rapid changes in blood pressure can regulate Na-K-ATPase in the kidney cortex, we tested the hypothesis that a short-term (5 min) decrease in renal perfusion pressure will increase the amount of Na-K-ATPase in the plasma membranes by an angiotensin II-dependent mechanism. The abdominal aorta of anesthetized Sprague-Dawley rats was constricted with a ligature between the renal arteries, and pressure was monitored on either side during acute constriction. Left renal perfusion pressure was reduced to 70 +/- 1 mmHg (n = 6), whereas right renal perfusion pressure was 112 +/- 4 mmHg. In control (nonconstricted) rats (n = 5), pressure to both kidneys was similar at 119 +/- 6 mmHg. After 5 min of reduced perfusion, femoral venous samples were taken for plasma renin activity (PRA) and the kidneys excised. The cortex was dissected, minced, sieved, and biotinylated. Lower perfusion left kidneys showed a 41% increase (P < 0.003) in the amount of Na-K-ATPase in the plasma membrane compared with right kidneys. In controls, there was no difference in cell surface Na-K-ATPase between left and right kidneys (P = 0.47). PRA was 57% higher in experimental animals compared with controls. To test the role of angiotensin II in mediating the increase in Na-K-ATPase, we repeated the experiments (n = 6) in rats treated with ramiprilat. When angiotensin-converting enzyme was inhibited, the cell surface Na-K-ATPase of the two kidneys was equal (P =0.46). These results confirm our hypothesis: rapid changes in blood pressure regulate trafficking of Na-K-ATPase in the kidney cortex.


Subject(s)
Angiotensin II/physiology , Cell Membrane/enzymology , Kidney Cortex/blood supply , Kidney Cortex/enzymology , Renal Circulation/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Angiotensin II/biosynthesis , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Biotin/metabolism , Blood Pressure/physiology , In Vitro Techniques , Male , Microsomes/enzymology , Ramipril/analogs & derivatives , Ramipril/pharmacology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Renin/blood , Renin/physiology
9.
J Clin Pharmacol ; 48(1): 85-95, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17986525

ABSTRACT

We conducted 3 open-label, multiple-dose, 3-period, randomized, crossover studies in healthy subjects to assess the potential pharmacokinetic interaction between vildagliptin, a novel dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes, and representatives of 3 commonly prescribed antihypertensive drug classes: (1) the calcium channel blocker, amlodipine; (2) the angiotensin receptor blocker, valsartan; and (3) the angiotensin-converting enzyme inhibitor, ramipril. Coadministration of vildagliptin 100 mg with amlodipine 5 mg, valsartan 320 mg, or ramipril 5 mg had no clinically significant effect on the pharmacokinetics of these drugs. The 90% confidence intervals of the geometric mean ratios for area under the plasma concentration-time curve from time zero to 24 hours (AUC0-24h) and maximum plasma concentration (Cmax) for vildagliptin, amlodipine, and ramipril (and its active metabolite, ramiprilat) were contained within the acceptance range for bioequivalence (0.80-1.25). Valsartan AUC0-24h and Cmax increased by 24% and 14%, respectively, following coadministration of vildagliptin, but this was not considered clinically significant. Vildagliptin was generally well tolerated when given alone or in combination with amlodipine, valsartan, or ramipril in healthy subjects at steady state. No adjustment in dosage based on pharmacokinetic considerations is required should vildagliptin be coadministered with amlodipine, valsartan, or ramipril in patients with type 2 diabetes and hypertension.


Subject(s)
Adamantane/analogs & derivatives , Amlodipine/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Nitriles/pharmacokinetics , Pyrrolidines/pharmacokinetics , Ramipril/analogs & derivatives , Tetrazoles/pharmacokinetics , Valine/analogs & derivatives , Adamantane/administration & dosage , Adamantane/adverse effects , Adamantane/pharmacokinetics , Administration, Oral , Adult , Amlodipine/administration & dosage , Amlodipine/adverse effects , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Area Under Curve , Cross-Over Studies , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Drug Interactions , Female , Half-Life , Headache/chemically induced , Humans , Male , Middle Aged , Nausea/chemically induced , Nitriles/administration & dosage , Nitriles/adverse effects , Pyrrolidines/administration & dosage , Pyrrolidines/adverse effects , Ramipril/administration & dosage , Ramipril/adverse effects , Ramipril/pharmacokinetics , Tablets , Tetrazoles/administration & dosage , Tetrazoles/adverse effects , Valine/administration & dosage , Valine/adverse effects , Valine/pharmacokinetics , Valsartan , Vildagliptin
10.
J Vet Pharmacol Ther ; 31(4): 349-58, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18638296

ABSTRACT

The pharmacokinetics of ramipril and its active metabolite, ramiprilat, was determined in cats following single and repeated oral doses of ramipril (Vasotop tablets) (once daily for 9 days) at dose rates of 0.125, 0.25, 0.5 and 1.0 mg/kg. The pharmacodynamic effects were assessed by measuring plasma angiotensin-converting enzyme (ACE) activity. Maximum ramipril concentrations were attained within 30 min following a single dose and declined rapidly (concentrations were below the limit of quantification 4 h after treatment). Peak ramiprilat concentrations were detected at approximately 1.5 h. The apparent terminal half-life (t((1/2)beta)) was > or =20 h irrespective of the dose. Ramiprilat accumulated in plasma (ratio of accumulation 1.3 to 1.9 depending on the dose rate) following repeated administration. Steady-state conditions were attained after the second dose. Excretion was predominant in faeces (87%) and to a lesser extent in urine (11%). The rate and extent of absorption of ramipril as well as its conversion to ramiprilat were not significantly influenced by the presence of food in the gastrointestinal tract. Plasma-ACE activity was almost completely abolished 0.5-2.0 h after treatment, irrespective of the dose rate. Significant inhibition of ACE activity of 54.7 to 82.6% (depending on the dosage) was still present 24 h after treatment. Treatment was well-tolerated in all cats. Ramipril at a dose rate of 0.125 mg/kg once daily produces significant and long-lasting inhibition of ACE activity in healthy cats. The appropriateness of this dosage regime needs to be confirmed in diseased cats.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Peptidyl-Dipeptidase A/drug effects , Ramipril/analogs & derivatives , Ramipril/pharmacokinetics , Administration, Oral , Angiotensin-Converting Enzyme Inhibitors/blood , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Area Under Curve , Cats , Dose-Response Relationship, Drug , Female , Half-Life , Male , Metabolic Clearance Rate , Peptidyl-Dipeptidase A/blood , Peptidyl-Dipeptidase A/metabolism , Ramipril/blood , Ramipril/pharmacology , Reference Values
11.
J Clin Invest ; 92(6): 3003-7, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8254054

ABSTRACT

The effects of extracellular matrix proteins and mechanical strain on the mitogenic activity of angiotensins I and II (AI and AII) were examined in cultured rat vascular smooth muscle (VSM) cells. VSM cells on various extracellular matrices were exposed to AII (1 microM) for 48 h. On plastic, AII induced only a 1.6-fold increase in [3H]thymidine incorporation, but on fibronectin- or type I collagen-coated plastic, the response to AII was enhanced from two- to fourfold. On a type I collagen-coated silicone elastomer, to which mechanical strain was applied, [3H]thymidine incorporation dramatically increased to a maximum of 53-fold. Dup 753 (10(-5) M) blocked the AII-induced increase in DNA synthesis. AI also increased DNA synthesis in VSM cells, and this response was also enhanced by mechanical strain. Mitogenic activity of AI was blocked by ramiprilat (10(-5) M), indicating that its mitogenic activity was via conversion to AII. The synergy between AII and strain was completely eliminated by neutralizing antibodies to PDGF AB (3 micrograms/ml). Furthermore, the mitogenic effect of AII in unstrained cells was also synergistic with submaximal concentrations of PDGF AB (1 ng/ml). Thus, the synergy between AII and mechanical strain probably results from synergism between AII and PDGF secreted in response to strain.


Subject(s)
Angiotensin II/pharmacology , Cell Division/physiology , Collagen/pharmacology , Muscle, Smooth, Vascular/cytology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Animals, Newborn , Biphenyl Compounds/pharmacology , Cell Division/drug effects , Cells, Cultured , DNA/biosynthesis , Drug Synergism , Fibronectins/pharmacology , Gelatin/pharmacology , Imidazoles/pharmacology , Losartan , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Ramipril/analogs & derivatives , Ramipril/pharmacology , Rats , Recombinant Proteins/pharmacology , Stress, Mechanical , Tetrazoles/pharmacology , Thymidine/metabolism
12.
FASEB J ; 19(9): 1172-4, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15860541

ABSTRACT

Tissue kallikrein (TK), a major kinin-forming enzyme, is synthesized in the heart and arteries. We tested the hypothesis that TK plays a protective role in myocardial ischemia by performing ischemia-reperfusion (IR) injury, with and without ischemic preconditioning (IPC) or ACE inhibitor (ramiprilat) pretreatment, in vivo in littermate wild-type (WT) or TK-deficient (TK-/-) mice. IR induced similar infarcts in WT and TK-/-. IPC reduced infarct size by 65% in WT, and by 40% in TK-/- (P<0.05, TK-/- vs WT). Ramiprilat also reduced infarct size by 29% in WT, but in TK-/- its effect was completely suppressed. Pretreatment of WT with a B2, but not a B1, kinin receptor antagonist reproduced the effects of TK deficiency. However, B2 receptor-deficient mice (B2-/-) unexpectedly responded to IPC or ramiprilat like WT mice. But pretreatment of the B2-/- mice with a B1 antagonist suppressed the cardioprotective effects of IPC and ramiprilat. In B2-/-, B1 receptor gene expression was constitutively high. In WT and TK-/- mice, both B2 and B1 mRNA levels increased several fold during IR, and even more during IPC+IR. Thus TK and the B2 receptor play a critical role in the cardioprotection afforded by two experimental maneuvers of potential clinical relevance, IPC and ACE inhibition, during ischemia.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/prevention & control , Ramipril/analogs & derivatives , Tissue Kallikreins/physiology , Animals , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , RNA, Messenger/analysis , Ramipril/pharmacology , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/physiology
13.
Circ Res ; 94(1): 60-7, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14615289

ABSTRACT

Not all of the cardiovascular effects of angiotensin-converting enzyme (ACE) inhibitors can be attributed to changes in angiotensin II and bradykinin levels. Because the cytoplasmic tail of ACE is phosphorylated, we determined whether ACE inhibitors affect the phosphorylation of ACE and whether ACE possesses the characteristics of a signal transduction molecule. The ACE inhibitors ramiprilat and perindoprilat, and the substrate bradykinin (but not angiotensin I), enhanced the activity of ACE-associated CK2 and the phosphorylation of ACE Ser1270 in cultured endothelial cells. Mitogen-activated protein kinase kinase 7 and c-Jun N-terminal kinase (JNK) coprecipitated with ACE, and stimulation of endothelial cells with ACE inhibitors increased the activity of ACE-associated JNK and elicited the accumulation of phosphorylated c-Jun in the nucleus. Ramiprilat was however unable to activate JNK or to stimulate the nuclear accumulation of c-Jun in endothelial cells expressing a S1270A ACE mutant or in ACE-deficient cells. Because the ACE inhibitor-induced increase in ACE expression has been linked to the formation of c-Jun homodimers, we investigated whether ACE signaling via JNK contributes to this response in vitro and in vivo. Prolonged ramiprilat treatment increased ACE expression in primary cultures of human endothelial cells and in vivo (mouse lung), a response that was prevented by pretreatment with the JNK inhibitor SP600125. Thus, ACE is involved in outside-in signaling in endothelial cells and "ACE signaling" may be an important cellular mechanism contributing to the beneficial effects of ACE inhibitors.


Subject(s)
Endothelium, Vascular/enzymology , Peptidyl-Dipeptidase A/physiology , Ramipril/analogs & derivatives , Signal Transduction , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bradykinin/pharmacology , Casein Kinase II , Cells, Cultured , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Peptidyl-Dipeptidase A/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Ramipril/pharmacology , Swine
14.
BMC Clin Pharmacol ; 6: 1, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16398929

ABSTRACT

BACKGROUND: The angiotensin-converting enzyme (ACE) inhibitors have complicated and poorly characterized pharmacokinetics. There are two binding sites per ACE (high affinity "C", lower affinity "N") that have sub-nanomolar affinities and dissociation rates of hours. Most inhibitors are given orally in a prodrug form that is systemically converted to the active form. This paper describes the first human physiologically based pharmacokinetic (PBPK) model of this drug class. METHODS: The model was applied to the experimental data of van Griensven et. al for the pharmacokinetics of ramiprilat and its prodrug ramipril. It describes the time course of the inhibition of the N and C ACE sites in plasma and the different tissues. The model includes: 1) two independent ACE binding sites; 2) non-equilibrium time dependent binding; 3) liver and kidney ramipril intracellular uptake, conversion to ramiprilat and extrusion from the cell; 4) intestinal ramipril absorption. The experimental in vitro ramiprilat/ACE binding kinetics at 4 degrees C and 300 mM NaCl were assumed for most of the PBPK calculations. The model was incorporated into the freely distributed PBPK program PKQuest. RESULTS: The PBPK model provides an accurate description of the individual variation of the plasma ramipril and ramiprilat and the ramiprilat renal clearance following IV ramiprilat and IV and oral ramipril. Summary of model features: Less than 2% of total body ACE is in plasma; 35% of the oral dose is absorbed; 75% of the ramipril metabolism is hepatic and 25% of this is converted to systemic ramiprilat; 100% of renal ramipril metabolism is converted to systemic ramiprilat. The inhibition was long lasting, with 80% of the C site and 33% of the N site inhibited 24 hours following a 2.5 mg oral ramipril dose. The plasma ACE inhibition determined by the standard assay is significantly less than the true in vivo inhibition because of assay dilution. CONCLUSION: If the in vitro plasma binding kinetics of the ACE inhibitor for the two binding sites are known, a unique PBPK model description of the Griensven et. al. experimental data can be obtained.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Models, Biological , Ramipril/analogs & derivatives , Administration, Oral , Adult , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Binding Sites , Biological Availability , Humans , Injections, Intravenous , Male , Peptidyl-Dipeptidase A/blood , Ramipril/administration & dosage , Ramipril/pharmacokinetics , Randomized Controlled Trials as Topic , Reproducibility of Results , Substrate Specificity
15.
Int J Clin Pharmacol Ther ; 44(2): 93-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16502769

ABSTRACT

OBJECTIVE: To assess the bioequivalence of a ramipril 5 mg tablet formulation (ramipril test formulation from Laboratórios Biosintética Ltda (Sao Paulo, Brazil) and Triatec from Aventis Pharma (Sueano, Brazil) standard reference formulation) in 26 healthy volunteers of both sexes. METHODS: The study was conducted using an open, randomized, 2-period crossover design with a 2-week washout interval. Plasma samples were obtained over a 36-hour period. Plasma ramipril and ramiprilat concentrations were analyzed by liquid chromatography coupled to tandem mass spectrometry (LC-MS-MS) with positive ion electrospray ionization using multiple reaction monitoring (MRM). From the ramipril and ramiprilat plasma concentration vs. time curves, the following pharmacokinetic parameters were obtained: AUClast, AUCinf and Cmax. RESULTS: The limit of quantification was 0.2 ng x ml(-1) and 1.0 ng x ml(-1) for ramipril and ramiprilat, respectively. The geometric means and 90% confidence intervals (CI) for Ramipril/Triatec and Ramiprilat/Triatec percent ratios were: 104.69% (90% CI = 93.21-117.59%) for Cmax, 102.49% (90% CI = 92.76-113.24%) for AUClast, 103.60% (90% CI = 93.56 114.73%) for AUCinf, 108.48% (90% CI = 98.86-119.03%) for Cmax, 105.88% (90% CI = 101.55-110.39%) for AUClast, 97.30% (90% CI = 90.17-104.99%) for AUCinf, respectively. CONCLUSION: Since the 90% CI for AUClast, AUCinf and Cmax ratios were within the 80-125% interval proposed by the U.S. FDA, it was concluded that the ramipril formulation produced by Laboratórios Biosintética Ltda is bioequivalent to the Triatec formulation in both rate and extent of absorption.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Ramipril/pharmacokinetics , Administration, Oral , Adolescent , Adult , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/blood , Area Under Curve , Biological Availability , Chemistry, Pharmaceutical/methods , Chromatography, Liquid/methods , Cross-Over Studies , Female , Humans , Male , Mass Spectrometry/methods , Middle Aged , Ramipril/administration & dosage , Ramipril/analogs & derivatives , Ramipril/blood , Reference Values , Spectrometry, Mass, Electrospray Ionization/methods , Therapeutic Equivalency , Time Factors
16.
J Pharm Biomed Anal ; 40(3): 794-8, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16242284

ABSTRACT

In the course of development and validation of a gas chromatography-mass spectrometry (GC-MS) method for ramipril and its biologically active metabolite ramiprilat, evidence was found for an unknown interfering metabolite. Sample treatment included isolation from plasma or urine by solid-phase extraction, methylation with trimethylsilyldiazomethane and acylation with trifluoroacetic anhydride (TFAA). When liquid chromatography was used to fractionate plasma extracts prior to derivatization, the alkyl, acyl-derivative of ramipril was obtained from two separate LC fractions. Electrospray ionization mass spectral data, together with circumstances for the derivatization, were consistent with the presence of an N-glucuronide of ramipril. Interference from the metabolite was eliminated by including a wash step after extraction/alkylation, prior to acylation. The final assay had a lower limit of quantification at 1.0 nmol/L and a linear range of 1-300 nmol/L. Intra- and inter-batch precision for ramipril and ramiprilat in plasma or urine were better than 10 and 5% at 2 and 80 nmol/L, respectively.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/blood , Angiotensin-Converting Enzyme Inhibitors/urine , Ramipril/analogs & derivatives , Calibration , Gas Chromatography-Mass Spectrometry , Glucuronides/blood , Glucuronides/urine , Humans , Indicators and Reagents , Mass Spectrometry , Methylation , Ramipril/blood , Ramipril/urine , Reference Standards , Reproducibility of Results
17.
J Pharm Biomed Anal ; 40(2): 478-83, 2006 Feb 13.
Article in English | MEDLINE | ID: mdl-16181761

ABSTRACT

This study presents a rapid, specific and sensitive LC-MS/MS assay for the determination of ramipril and ramiprilat in human serum using enalapril as an internal standard (IS). A Waters Atlantis C18 column (2.1 mm x 100 mm, 3 microm) and a mobile phase consisting of 0.1% formic acid-methanol (25:75, v/v) were used for separation. The analysis was performed by the selected reaction monitoring (SRM) method, and the peak areas of the m/z 417.3-->234.3 and m/z 389.3-->206.2 transition for ramipril and ramiprilat, respectively, were measured versus that of the m/z 377.3-->234.2 for IS to generate the standard curves. The assay linearities of ramipril and ramiprilat were confirmed over the range 0.10-100 ngml(-1) and 0.25-100 ngml(-1), respectively, and limits of quantitation for them were 0.10 and 0.25 ngml(-1), respectively. The linear ranges correspond well with the serum concentrations of the analytes obtained in clinical pharmacokinetic studies. Intraday and interday relative standard deviations of ramipril and ramiprilat were 2.8-6.4% and 4.3-4.6%, 4.4-6.7% and 3.5-4.7%, respectively. The recoveries of ramipril and ramiprilat from serum were in the range of 81.0-98.2%. The developed LC-MS procedures were applied for the determination of the pharmacokinetic parameters of ramipril and ramiprilat following a single oral administration of 10mg ramipril tablets in 18 Chinese healthy male volunteers.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/blood , Chromatography, High Pressure Liquid/methods , Ramipril/analogs & derivatives , Administration, Oral , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , China , Drug Stability , Formates , Humans , Male , Methanol , Product Surveillance, Postmarketing , Ramipril/administration & dosage , Ramipril/blood , Ramipril/pharmacokinetics , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Temperature
18.
J Vet Intern Med ; 20(3): 499-507, 2006.
Article in English | MEDLINE | ID: mdl-16734081

ABSTRACT

Ramipril, an angiotensin-converting enzyme (ACE) inhibitor for use in dogs, is converted in vivo to its active form, ramiprilat, which is eliminated in the bile and urine in the dog. The objective of this study was to assess the effect of renal impairment on the pharmacokinetics (PKs) and pharmacodynamics (PDs) of ramipril and ramiprilat. Ten adult Beagle dogs were used. PK/PD studies were performed before and after the induction of subclinical renal impairment. Ramiprilat was given at 0.25 mg/kg by a single IV bolus. After a 2-week washout period, ramipril was administered PO at 0.25 mg/kg once daily for 8 days. Ramipril and ramiprilat PKs were studied by using a physiologically based model. The relationship between free plasma ramiprilat concentration and ACE activity was described by using the fractional Hill model. Glomerular filtration rate was decreased by 58%. No biologically relevant changes in usual plasma variables were observed between the 1st and the 8th day of oral treatment with ramipril under either condition. After an IV bolus of ramiprilat, the only changes in renal-impaired dogs were a 14 and 49% decrease in clearance of the free fraction of ramiprilat (P < .01) and free plasma concentration required to produce 50% of the maximal effect (P < .05), respectively. After repeated PO administration of ramipril, there were no alterations in any of the PK and PD parameters in healthy or renal-impaired dogs. No adjustment of the recommended PO dosage of ramipril is needed in dogs with moderate renal impairment.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Dog Diseases/metabolism , Dogs/metabolism , Ramipril/pharmacokinetics , Renal Insufficiency/veterinary , Administration, Oral , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/blood , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Disease Models, Animal , Drug Administration Schedule , Female , Glomerular Filtration Rate/drug effects , Injections, Intravenous/veterinary , Iohexol , Ramipril/administration & dosage , Ramipril/analogs & derivatives , Ramipril/blood , Ramipril/pharmacology , Renal Insufficiency/metabolism , p-Aminohippuric Acid
19.
Vet J ; 208: 38-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26639833

ABSTRACT

The pharmacokinetics and pharmacodynamics (PK/PD) of the angiotensin-converting enzyme inhibitor (ACEI) ramiprilat after intravenous (IV) and oral (PO) administration of ramipril have not been evaluated in horses. This study was designed to establish PK profiles for ramipril and ramiprilat as well as to determine the effects of ramiprilat on serum angiotensin converting enzyme (ACE) and to select the most appropriate ramipril dose that suppresses ACE activity. Six healthy horses in a cross-over design received IV ramipril 0.050 mg/kg, PO at a dose of 0 (placebo), and 0.050, 0.10, 0.20, 0.40 and 0.80 mg/kg ramipril. Blood pressures were measured and blood samples obtained at different times. Serum ramipril and ramiprilat concentrations and serum ACE activity were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and spectrophotometry, respectively. Systemic bioavailability of ramiprilat after PO ramipril was 6-9%. Percentages of maximum ACE inhibitions from baseline were 98.88 (IV ramipril), 5.31 (placebo) and 27.68, 39.27, 46.67, 76.13 and 84.27 (the five doses of PO ramipril). Blood pressure did not change during the experiments. Although oral availability of ramiprilat was low, ramipril has sufficient enteral absorption and bioconversion to ramiprilat to induce serum ACE inhibitions of almost 85% after a dose of 0.80 mg/kg ramipril. Additional research on ramipril administration in equine patients is indicated.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Blood Pressure/drug effects , Horses/metabolism , Ramipril/analogs & derivatives , Ramipril/pharmacology , Ramipril/pharmacokinetics , Administration, Intravenous/veterinary , Administration, Oral , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/pharmacology , Dose-Response Relationship, Drug , Female , Male , Peptidyl-Dipeptidase A/blood
20.
Biochim Biophys Acta ; 1041(1): 22-30, 1990 Oct 18.
Article in English | MEDLINE | ID: mdl-2171661

ABSTRACT

The angiotensin I-converting enzyme (peptidyl-dipeptide hydrolase, EC 3.4.15.1) inhibitor, ramiprilat (2-[N-[(S)-1-ethoxycarbonyl-3-phenylpropyl]-L-Ala]-(1S,3S,5S)-2- azabicyclo[3.3.0]octane-3-carboxylic acid), is shown to exist in tow conformational isomers, cis and trans, which interconvert around the amide bond. The two conformers were separated by reversed-phase high-performance liquid chromatography. The conformers were identified by nuclear Overhauser effect measurements. From line shape analysis the isomerization rate constants were determined to be kcis----trans = 15 s-1 and ktrans----cis = 5 s-1 at 368 K in [2H]phosphate buffer (p2H 7.5). By enzyme kinetic studies using 3-(2-furylacryloyl)-L-Phe-Gly-Gly as substrate, the trans conformer was found to be the most potent enzyme inhibitor, whereas the cis conformer had a very low inhibitory effect. A new inhibition mechanism is presented for this type of slow, tight-binding inhibitors that contain an amide bond. This mechanism involves an equilibrium between the two conformers and the enzyme-bound inhibitor complex.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Pyrroles/chemistry , Ramipril/analogs & derivatives , Dimethyl Sulfoxide , Isomerism , Kinetics , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Conformation , Molecular Structure , Peptidyl-Dipeptidase A/metabolism , Protein Binding , Pyrroles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL