Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 226
Filter
1.
J Am Heart Assoc ; 10(11): e020633, 2021 06.
Article in English | MEDLINE | ID: mdl-34032129

ABSTRACT

Pulmonary hypertension (PH) attributable to left heart disease (LHD) is believed to be the most common form of PH and is strongly associated with increased mortality and morbidity in this patient population. Specific therapies for PH-LHD have not yet been identified and the use of pulmonary artery hypertension-targeted therapies in PH-LHD are not recommended. Endothelin receptor antagonists, phosphodiesterase-5 inhibitors, guanylate cyclase stimulators, and prostacyclins have all been studied in PH-LHD with conflicting results. Understanding the mechanisms underlying PH-LHD could potentially provide novel therapeutic targets. Fibrosis, oxidative stress, and metabolic syndrome have been proposed as pathophysiological components of PH-LHD. Genetic associations have also been identified, offering additional mechanisms with biological plausibility. This review summarizes the evidence and challenges for treatment of PH-LHD and focuses on underlying mechanisms on the horizon that could develop into potential therapeutic targets for this disease.


Subject(s)
Disease Management , Hypertension, Pulmonary/therapy , Pulmonary Wedge Pressure/physiology , Ventricular Dysfunction, Left/therapy , Ventricular Function, Left/physiology , Endothelin Receptor Antagonists/therapeutic use , Guanylate Cyclase/drug effects , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Oxidative Stress , Phosphodiesterase 5 Inhibitors/therapeutic use , Prognosis , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/etiology
2.
J Psychopharmacol ; 34(4): 429-440, 2020 04.
Article in English | MEDLINE | ID: mdl-31913077

ABSTRACT

BACKGROUND: Endocannabinoid neurotransmission in the bed nucleus of the stria terminalis is involved in the control of cardiovascular responses to stress. However, the local mechanisms involved is this regulation are not known. AIMS: The purpose of this study was to assess an interaction of bed nucleus of the stria terminalis endocannabinoid neurotransmission with local nitrergic signaling, as well as to investigate the involvement of local N-methyl-D-aspartate glutamate receptor and nitric oxide signaling in the control of cardiovascular responses to acute restraint stress by bed nucleus of the stria terminalis endocannabinoid neurotransmission in rats. METHODS: The first protocol evaluated the effect of intra-bed nucleus of the stria terminalis microinjection of the selective cannabinoid receptor type 1 receptor antagonist AM251 in nitrite/nitrate content in the bed nucleus of the stria terminalis following restraint stress. The other protocols evaluated the impact of local pretreatment with the selective N-methyl-D-aspartate glutamate receptor antagonist LY235959, the selective neuronal nitric oxide synthase inhibitor Nω-propyl-L-arginine, the soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or the protein kinase G inhibitor KT5823 in restraint-evoked cardiovascular changes following bed nucleus of the stria terminalis treatment with AM251. RESULTS: Bilateral microinjection of AM251 into the bed nucleus of the stria terminalis increased local nitric oxide release during restraint stress. Bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist also enhanced the tachycardia caused by restraint stress, but without affecting arterial pressure increase and sympathetic-mediated cutaneous vasoconstriction. The facilitation of restraint-evoked tachycardia following bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist was completely inhibited by local pretreatment with LY235959, Nω-propyl-L-arginine, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or KT5823. CONCLUSIONS: Our results provide evidence that bed nucleus of the stria terminalis endocannabinoid neurotransmission inhibits local N-methyl-D-aspartate/neuronal nitric oxide synthase/soluble guanylate cyclase/protein kinase G signaling, and this mechanism is involved in the control of the cardiovascular responses to stress.


Subject(s)
Hemodynamics/drug effects , Receptor, Cannabinoid, CB1/drug effects , Septal Nuclei/drug effects , Signal Transduction/drug effects , Stress, Psychological/complications , Stress, Psychological/drug therapy , Animals , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/drug effects , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/drug effects , Male , Microinjections , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/drug effects , Piperidines/administration & dosage , Piperidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Restraint, Physical , Synaptic Transmission/drug effects
3.
Postepy Hig Med Dosw (Online) ; 71(0): 577-588, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28791952

ABSTRACT

Pulmonary hypertension (PH) is a rare disorder associated with abnormally elevated pulmonary pressures that, if untreated, leads to right heart failure and premature death. Special population include patents with pulmonary arterial hypertension (PAH). A greater understanding of the epidemiology, pathogenesis, and pathophysiology of PAH has led to significant advances over the past few years. Modern drug therapy provides a significant improvement in patient symptomatic status and a slower rate of clinical deterioration. Despite this, PAH remains a chronic disease without a cure. There is a need for the development of novel therapies and therapeutic strategies, as treatment options are neither universally available nor always effective, possibly due to the large number of mediator and signaling pathways with downstream effectors which are implicated in the pathobiology of PH, and which are not fully reversed during PAH therapy. In the following pages, we review novel strategies for treatment of PAH. For this purpose we summarized the role of specific drug therapies that involve: endothelin receptor antagonists (ERA), phosphodiesterase type 5 inhibitors (PDE-5i) and prostacyclin and prostanoids (PGI2). We focused on novel molecular mechanisms in PAH of recently approved: Guanylate cyclase stimulator and non-prostanoid IP receptor agonist. We discussed novel approach to combined therapy, as well as a new generation of investigational drugs and promising PAH-associated signaling pathways, such as, PDGF, RhoA/ROCK RAAS, HT-5 and others.


Subject(s)
Hypertension, Pulmonary/drug therapy , Drug Therapy, Combination , Endothelin Receptor Antagonists/therapeutic use , Guanylate Cyclase/drug effects , Humans , Phosphodiesterase 5 Inhibitors/therapeutic use , Prostaglandins/therapeutic use
4.
Respir Med ; 129: 124-129, 2017 08.
Article in English | MEDLINE | ID: mdl-28732819

ABSTRACT

Riociguat is a soluble guanylate cyclase stimulator that has been approved for the treatment of pulmonary arterial hypertension and inoperable chronic thromboembolic pulmonary hypertension or persistent/recurrent pulmonary hypertension following pulmonary endarterectomy. Riociguat is administered using an 8-week individual dose-adjustment scheme whereby a patient initially receives riociguat 1.0 mg three times daily (tid), and the dose is then increased every 2 weeks in the absence of hypotension, indicated by systolic blood pressure measurements and symptoms, up to a maximum dose of 2.5 mg tid. The established riociguat dose-adjustment scheme allows the dose of riociguat to be individually optimized in terms of tolerability and efficacy. The majority of patients in the phase III clinical trials and their long-term extension phases achieved the maximum riociguat dose, whereas some patients remained on lower doses. There is evidence that these patients may experience benefits at riociguat doses lower than 2.5 mg tid, with improvement in exercise capacity being observed after only 2-4 weeks of treatment in the phase III studies and in the exploratory 1.5 mg-maximum patient group of PATENT-1. This review aims to provide an overview of the rationale behind the riociguat dose-adjustment scheme and examine its application to both clinical trials and real-life clinical practice.


Subject(s)
Guanylate Cyclase/drug effects , Hypertension, Pulmonary/drug therapy , Pulmonary Embolism/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Activators/therapeutic use , Exercise Tolerance/drug effects , Guanylate Cyclase/metabolism , Humans , Natriuretic Peptide, Brain/drug effects , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrimidines/administration & dosage , Pyrimidines/blood , Vascular Resistance/drug effects , Walk Test/methods
5.
Am J Physiol Endocrinol Metab ; 313(3): E321-E334, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28679626

ABSTRACT

Bradykinin (BK) promotes insulin sensitivity and glucose uptake in adipocytes and other cell types. We demonstrated that in rat adipocytes BK enhances insulin-stimulated glucose transport via endothelial nitric oxide synthase, nitric oxide (NO) generation, and decreased activity of the mitogen-activated protein kinase (MAPK) JNK (c-Jun NH2-terminal kinase). In endothelial cells, NO increases soluble guanylate cyclase (sGC) activity, which, in turn, activates protein kinase G (PKG) by increasing cGMP levels. In this study, we investigated whether BK acts via the sGC-cGMP-PKG pathway to inhibit the negative effects of JNK on insulin signaling and glucose uptake in rat adipocytes. BK augmented cGMP concentrations. The BK-induced enhancement of insulin-stimulated glucose uptake was mimicked by the sGC activator YC-1 and a cell-permeable cGMP analog, CPT-cGMP, and inhibited by the sGC inhibitor ODQ and the PKG inhibitor KT 5823. Transfection of dominant-negative PKG reduced the BK augmentation of insulin-induced Akt phosphorylation. The activation of JNK and ERK1/2 by insulin was attenuated by BK, which was mediated by the sGC-cGMP-PKG pathway. Whereas insulin-stimulated phosphorylation of upstream activators of JNK and ERK, i.e., MKK4 and MEK1/2, was unaffected, BK augmented insulin-mediated induction of MKP-5 mRNA and protein levels. Furthermore, zaprinast, a phosphodiesterase inhibitor, enhanced cGMP and MKP-5 and prolonged the action of BK. These data indicate that BK enhances insulin action by inhibition of negative feedback by JNK and ERK via upregulation of MKP-5, mediated by the sGC-cGMP-PKG signaling pathway.


Subject(s)
Adipocytes/drug effects , Bradykinin/pharmacology , Cyclic GMP-Dependent Protein Kinases/drug effects , Dual-Specificity Phosphatases/drug effects , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Phosphatases/drug effects , RNA, Messenger/drug effects , Adipocytes/metabolism , Animals , Blotting, Western , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Glucose/metabolism , Guanylate Cyclase/drug effects , Guanylate Cyclase/metabolism , Immunoprecipitation , JNK Mitogen-Activated Protein Kinases/drug effects , Male , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphodiesterase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Purinones/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
6.
Ann Thorac Surg ; 104(1): 122-129, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28110808

ABSTRACT

BACKGROUND: Spasm of arterial grafts is still a clinical problem in coronary artery bypass surgery. The present study was designed to examine the effect of particulate guanylyl cyclase activator (carperitide) as an antispastic agent in internal thoracic artery and gastroepiploic artery grafts. METHODS: Isolated arterial grafts taken during surgery were studied in organ bath in three ways: the relaxing effect of carperitide on vasoconstrictor-induced precontraction; the inhibitory effect of pretreatment with carperitide on subsequent vasoconstrictor-induced contraction; and the effect of carperitide and nitroglycerin on increase of intracellular cyclic guanosine monophosphate levels. RESULTS: Carperitide produced a concentration-related, endothelium-independent relaxation contracted with potassium chloride, phenylephrine, prostaglandin F2α, or endothelin-1. Carperitide showed significantly higher potency and efficacy than nitroglycerin and nifedipine. Pretreatment with carperitide significantly attenuated the subsequent vasoconstrictor-induced contraction. Carperitide produced more cyclic guanosine monophosphate than nitroglycerin. CONCLUSIONS: Carperitide has a potent inhibitory effect on the vasoconstriction mediated by different vasoconstrictors in human internal thoracic artery and gastroepiploic artery grafts. The use of carperitide in patients during and after coronary artery bypass surgery is favored for the prevention and reversal of graft spasm.


Subject(s)
Atrial Natriuretic Factor/therapeutic use , Coronary Artery Bypass/adverse effects , Graft Occlusion, Vascular/prevention & control , Guanylate Cyclase/drug effects , Mammary Arteries/drug effects , Vasoconstriction/drug effects , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/physiopathology , Guanylate Cyclase/metabolism , Humans , Mammary Arteries/physiopathology , Mammary Arteries/transplantation
7.
Proc Natl Acad Sci U S A ; 113(17): E2355-62, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27071111

ABSTRACT

Asthma is defined by airway inflammation and hyperresponsiveness, and contributes to morbidity and mortality worldwide. Although bronchodilation is a cornerstone of treatment, current bronchodilators become ineffective with worsening asthma severity. We investigated an alternative pathway that involves activating the airway smooth muscle enzyme, soluble guanylate cyclase (sGC). Activating sGC by its natural stimulant nitric oxide (NO), or by pharmacologic sGC agonists BAY 41-2272 and BAY 60-2770, triggered bronchodilation in normal human lung slices and in mouse airways. Both BAY 41-2272 and BAY 60-2770 reversed airway hyperresponsiveness in mice with allergic asthma and restored normal lung function. The sGC from mouse asthmatic lungs displayed three hallmarks of oxidative damage that render it NO-insensitive, and identical changes to sGC occurred in human lung slices or in human airway smooth muscle cells when given chronic NO exposure to mimic the high NO in asthmatic lung. Our findings show how allergic inflammation in asthma may impede NO-based bronchodilation, and reveal that pharmacologic sGC agonists can achieve bronchodilation despite this loss.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Benzoates/pharmacology , Biphenyl Compounds/pharmacology , Bronchodilator Agents/pharmacology , Guanylate Cyclase/drug effects , Hydrocarbons, Fluorinated/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Animals , Anti-Asthmatic Agents/therapeutic use , Asthma/enzymology , Asthma/physiopathology , Benzoates/therapeutic use , Biphenyl Compounds/therapeutic use , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/enzymology , Bronchodilator Agents/therapeutic use , Coculture Techniques , Cyclic GMP/metabolism , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Female , Humans , Hydrocarbons, Fluorinated/therapeutic use , Lung/enzymology , Mice , Mice, Inbred BALB C , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Nitric Oxide/pharmacology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Solubility , Trachea/drug effects
8.
Invest Ophthalmol Vis Sci ; 57(3): 1317-26, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26998718

ABSTRACT

PURPOSE: The nitric oxide (NO)-cyclic guanosine-3',5'-monophosphate (cGMP) pathway regulates aqueous humor outflow and therefore, intraocular pressure. We investigated the pharmacologic effects of the soluble guanylate cyclase (sGC) stimulator IWP-953 on primary human trabecular meshwork (HTM) cells and conventional outflow facility in mouse eyes. METHODS: Cyclic GMP levels were determined in vitro in HEK-293 cells and four HTM cell strains (HTM120/HTM123: predominantly myofibroblast-like phenotype, HTM130/HTM141: predominantly endothelial-like phenotype), and in HTM cell culture supernatants. Conventional outflow facility was measured following intracameral injection of IWP-953 or DETA-NO using a computerized pressure-controlled perfusion system in enucleated mouse eyes ex vivo. RESULTS: IWP-953 markedly stimulated cGMP production in HEK-293 cells in the presence and absence of DETA-NO (half maximal effective concentrations: 17 nM, 9.5 µM). Similarly, IWP-953 stimulated cGMP production in myofibroblast-like HTM120 and HTM123 cells, an effect that was greatly amplified by the presence of DETA-NO. In contrast, IWP-953 stimulation of cGMP production in endothelial-like HTM130 and HTM141 cells was observed, but was markedly less prominent than in HTM120 and HTM123 cells. Notably, cGMP was found in all HTM culture supernatants, following IWP-953/DETA-NO stimulation. In paired enucleated mouse eyes, IWP-953 at 10, 30, 60, and 100 µM concentration-dependently increased outflow facility. This effect (89.5%) was maximal at 100 µM (P = 0.002) and in magnitude comparable to DETA-NO at 100 µM (97.5% increase, P = 0.030). CONCLUSIONS: These data indicate that IWP-953, via modulation of the sGC-cGMP pathway, increases aqueous outflow facility in mouse eyes, suggesting therapeutic potential for sGC stimulators as novel ocular hypotensive drugs.


Subject(s)
Aqueous Humor/chemistry , Cyclic GMP/metabolism , Enzyme Inhibitors/therapeutic use , Glaucoma, Open-Angle/drug therapy , Guanylate Cyclase/drug effects , Intraocular Pressure/drug effects , Trabecular Meshwork/metabolism , Adult , Animals , Cells, Cultured , Child, Preschool , Disease Models, Animal , Glaucoma, Open-Angle/pathology , Glaucoma, Open-Angle/physiopathology , Guanylate Cyclase/metabolism , Humans , Infant , Mice , Mice, Inbred C57BL , Trabecular Meshwork/pathology
9.
PLoS One ; 11(1): e0146212, 2016.
Article in English | MEDLINE | ID: mdl-26727368

ABSTRACT

BACKGROUND: Autologous arteriovenous (AV) fistulas are the first choice for vascular access but have a high risk of non-maturation due to insufficient vessel adaptation, a process dependent on nitric oxide (NO)-signaling. Chronic kidney disease (CKD) is associated with oxidative stress that can disturb NO-signaling. Here, we evaluated the influence of CKD on AV fistula maturation and NO-signaling. METHODS: CKD was established in rats by a 5/6th nephrectomy and after 6 weeks, an AV fistula was created between the carotid artery and jugular vein, which was followed up at 3 weeks with ultrasound and flow assessments. Vessel wall histology was assessed afterwards and vasoreactivity of carotid arteries was studied in a wire myograph. The soluble guanylate cyclase (sGC) activator BAY 60-2770 was administered daily to CKD animals for 3 weeks to enhance fistula maturation. RESULTS: CKD animals showed lower flow rates, smaller fistula diameters and increased oxidative stress levels in the vessel wall. Endothelium-dependent relaxation was comparable but vasorelaxation after sodium nitroprusside was diminished in CKD vessels, indicating NO resistance of the NO-receptor sGC. This was confirmed by stimulation with BAY 60-2770 resulting in increased vasorelaxation in CKD vessels. Oral administration of BAY 60-2770 to CKD animals induced larger fistula diameters, however; flow was not significantly different from vehicle-treated CKD animals. CONCLUSIONS: CKD induces oxidative stress resulting in NO resistance that can hamper AV fistula maturation. sGC activators like BAY 60-2770 could offer therapeutic potential to increase AV fistula maturation.


Subject(s)
Arteriovenous Shunt, Surgical , Nitric Oxide Donors/pharmacology , Nitric Oxide/physiology , Nitroprusside/pharmacology , Renal Insufficiency, Chronic/therapy , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Animals , Benzoates/therapeutic use , Biphenyl Compounds/therapeutic use , Carotid Arteries/drug effects , Carotid Arteries/surgery , Drug Resistance , Guanylate Cyclase/drug effects , Guanylate Cyclase/physiology , Hydrocarbons, Fluorinated/therapeutic use , Jugular Veins/drug effects , Jugular Veins/surgery , NG-Nitroarginine Methyl Ester/pharmacology , Nephrectomy/adverse effects , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitroprusside/therapeutic use , Oxidative Stress , Phenylephrine/pharmacology , Rats , Rats, Wistar , Renal Insufficiency, Chronic/physiopathology , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/therapeutic use
10.
Annu Rev Med ; 67: 229-43, 2016.
Article in English | MEDLINE | ID: mdl-26473417

ABSTRACT

The intracellular nucleotide cyclic guanosine monophosphate (cGMP) is found in many human organ tissues. Its concentration increases in response to the activation of receptor enzymes called guanylyl cyclases (GCs). Different ligands bind GCs, generating the second messenger cGMP, which in turn leads to a variety of biological actions. A deficit or dysfunction of this pathway at the cardiac, vascular, and renal levels manifests in cardiovascular diseases such as heart failure, arterial hypertension, and pulmonary arterial hypertension. An impairment of the cGMP pathway also may be involved in the pathogenesis of obesity as well as dementia. Therefore, agents enhancing the generation of cGMP for the treatment of these conditions have been intensively studied. Some have already been approved, and others are currently under investigation. This review discusses the potential of novel drugs directly or indirectly targeting cGMP as well as the progress of research to date.


Subject(s)
Cardiovascular Diseases/drug therapy , Cyclic GMP/biosynthesis , Guanylate Cyclase/drug effects , Metabolic Diseases/metabolism , Natriuretic Peptides/metabolism , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Benzoates/therapeutic use , Cardiovascular Diseases/metabolism , Cyclic GMP/deficiency , Enzyme Activators/therapeutic use , Guanylate Cyclase/metabolism , Humans , Ligands , Metabolic Diseases/drug therapy , Natriuretic Peptides/therapeutic use , Neprilysin/antagonists & inhibitors , Phosphodiesterase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Signal Transduction/drug effects
12.
JAMA ; 314(21): 2251-62, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26547357

ABSTRACT

IMPORTANCE: Worsening chronic heart failure (HF) is a major public health problem. OBJECTIVE: To determine the optimal dose and tolerability of vericiguat, a soluble guanylate cyclase stimulator, in patients with worsening chronic HF and reduced left ventricular ejection fraction (LVEF). DESIGN, SETTING, AND PARTICIPANTS: Dose-finding phase 2 study that randomized 456 patients across Europe, North America, and Asia between November 2013 and January 2015, with follow-up ending June 2015. Patients were clinically stable with LVEF less than 45% within 4 weeks of a worsening chronic HF event, defined as worsening signs and symptoms of congestion and elevated natriuretic peptide level requiring hospitalization or outpatient intravenous diuretic. INTERVENTIONS: Placebo (n = 92) or 1 of 4 daily target doses of oral vericiguat (1.25 mg [n = 91], 2.5 mg [n = 91], 5 mg [n = 91], 10 mg [n = 91]) for 12 weeks. MAIN OUTCOMES AND MEASURES: The primary end point was change from baseline to week 12 in log-transformed level of N-terminal pro-B-type natriuretic peptide (NT-proBNP). The primary analysis specified pooled comparison of the 3 highest-dose vericiguat groups with placebo, and secondary analysis evaluated a dose-response relationship with vericiguat and the primary end point. RESULTS: Overall, 351 patients (77.0%) completed treatment with the study drug with valid 12-week NT-proBNP levels and no major protocol deviation and were eligible for primary end point evaluation. In primary analysis, change in log-transformed NT-proBNP levels from baseline to week 12 was not significantly different between the pooled vericiguat group (log-transformed: baseline, 7.969; 12 weeks, 7.567; difference, -0.402; geometric means: baseline, 2890 pg/mL; 12 weeks, 1932 pg/mL) and placebo (log-transformed: baseline, 8.283; 12 weeks, 8.002; difference, -0.280; geometric means: baseline, 3955 pg/mL; 12 weeks, 2988 pg/mL) (difference of means, -0.122; 90% CI, -0.32 to 0.07; ratio of geometric means, 0.885, 90% CI, 0.73-1.08; P = .15). The exploratory secondary analysis suggested a dose-response relationship whereby higher vericiguat doses were associated with greater reductions in NT-proBNP level (P < .02). Rates of any adverse event were 77.2% and 71.4% among the placebo and 10-mg vericiguat groups, respectively. CONCLUSIONS AND RELEVANCE: Among patients with worsening chronic HF and reduced LVEF, compared with placebo, vericiguat did not have a statistically significant effect on change in NT-proBNP level at 12 weeks but was well-tolerated. Further clinical trials of vericiguat based on the dose-response relationship in this study are needed to determine the potential role of this drug for patients with worsening chronic HF. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT01951625.


Subject(s)
Guanylate Cyclase/drug effects , Heart Failure/drug therapy , Heterocyclic Compounds, 2-Ring/therapeutic use , Pyrimidines/therapeutic use , Ventricular Dysfunction, Left/drug therapy , Aged , Aged, 80 and over , Chronic Disease , Female , Heart Failure/complications , Hospitalization , Humans , Male , Middle Aged , Natriuretic Peptide, Brain , Peptide Fragments , Treatment Outcome , Ventricular Dysfunction, Left/complications
14.
Ann Rheum Dis ; 74(8): 1621-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25817717

ABSTRACT

OBJECTIVES: Stimulators of the soluble guanylate cyclase (sGC) have recently been shown to inhibit transforming growth factor-ß signalling. Here, we aimed to demonstrate that riociguat, the drug candidate for clinical trials in systemic sclerosis (SSc), is effective in experimental fibrosis and to compare its efficacy to that of phosphodiesterase V inhibitors that also increase the intracellular levels of cyclic guanosine monophosphate. METHODS: The antifibrotic effects of riociguat and sildenafil were compared in the tight-skin 1 model, in bleomycin-induced fibrosis and in a model of sclerodermatous chronic graft-versus-host-disease (cGvHD). Doses of 0.1-3 mg/kg twice a day for riociguat and of 3-10 mg/kg twice a day for sildenafil were used. RESULT: Riociguat dose-dependently reduced skin thickening, myofibroblast differentiation and accumulation of collagen with potent antifibrotic effects at 1 and 3 mg/kg. Riociguat also ameliorated fibrosis of the gastrointestinal tract in the cGvHD model. The antifibrotic effects were associated with reduced phosphorylation of extracellular signal-regulated kinases. Sildenafil at doses of 3 and 10 mg/kg exerted mild antifibrotic effects that were significantly less pronounced compared with 1 and 3 mg/kg riociguat. CONCLUSIONS: These data demonstrated potent antifibrotic effects of riociguat on experimental skin and organ fibrosis. These findings suggest a role for riociguat for the treatment of fibrotic diseases, especially for the treatment of SSc. A phase II study with riociguat in patients with SSc is currently starting.


Subject(s)
Guanylate Cyclase/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Skin/pathology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrosis , Mice , Mice, Inbred Strains , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Scleroderma, Systemic/drug therapy , Sildenafil Citrate/pharmacology
15.
Expert Rev Cardiovasc Ther ; 13(5): 467-76, 2015 May.
Article in English | MEDLINE | ID: mdl-25797801

ABSTRACT

Chronic thromboembolic pulmonary hypertension (CTEPH) is an insidious, progressive disease with a poor prognosis. The treatment of choice is pulmonary thromboendarterectomy, although not all patients benefit from surgery at a specialized center. Riociguat, an oral soluble guanylate cyclase (sGC) stimulator is the first pharmacotherapeutic agent that has been shown to improve exercise capacity and hemodynamics in a large multicenter, double-blind, randomized placebo-controlled trial for the treatment of patients with inoperable or persistent CTEPH. Riociguat stimulates sGC directly in a nitric oxide (NO)-independent manner, thereby increasing the sensitivity of sGC to NO, and also in synergy with NO, leading to increased production of cyclic guanosine monophosphate, an intracellular messenger involved in regulating vascular tone, smooth muscle cell proliferation, fibrosis and inflammation. This review will summarize the pharmacodynamics, pharmacokinetics as well as safety and efficacy data of riociguat in inoperable or persistent CTEPH.


Subject(s)
Hypertension, Pulmonary/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Thromboembolism/drug therapy , Animals , Chronic Disease , Guanylate Cyclase/drug effects , Humans , Hypertension, Pulmonary/etiology , Nitric Oxide/metabolism , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Thromboembolism/complications
16.
Clin Pharmacol Ther ; 97(1): 88-102, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25670386

ABSTRACT

Nitric oxide (NO) activates soluble guanylate cyclase (sGC) by binding its prosthetic heme group, thereby catalyzing cyclic guanosine monophosphate (cGMP) synthesis. cGMP causes vasodilation and may inhibit smooth muscle cell proliferation and platelet aggregation. The NO-sGC-cGMP pathway is disordered in pulmonary arterial hypertension (PAH), a syndrome in which pulmonary vascular obstruction, inflammation, thrombosis, and constriction ultimately lead to death from right heart failure. Expression of sGC is increased in PAH but its function is reduced by decreased NO bioavailability, sGC oxidation and the related loss of sGC's heme group. Two classes of sGC modulators offer promise in PAH. sGC stimulators (e.g., riociguat) require heme-containing sGC to catalyze cGMP production, whereas sGC activators (e.g., cinaciguat) activate heme-free sGC. Riociguat is approved for PAH and yields functional and hemodynamic benefits similar to other therapies. Its main serious adverse effect is dose-dependent hypotension. Riociguat is also approved for inoperable chronic thromboembolic pulmonary hypertension.


Subject(s)
Drug Design , Guanylate Cyclase/drug effects , Hypertension, Pulmonary/drug therapy , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , Benzoates/adverse effects , Benzoates/pharmacology , Benzoates/therapeutic use , Chronic Disease , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/physiopathology , Nitric Oxide/metabolism , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/adverse effects , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Thromboembolism/drug therapy , Thromboembolism/physiopathology
17.
Med Clin (Barc) ; 144(12): 566-70, 2015 Jun 22.
Article in Spanish | MEDLINE | ID: mdl-25070518

ABSTRACT

Treatment of pulmonary arterial hypertension has achieved significant progress over the past 20 years. Currently, 3 groups of drugs have proven useful for the treatment of this disease: endothelin receptor antagonist, phosphodiesterase inhibitors and prostacyclin and its analogues. It is recommended to initiate treatment with one of these drugs, the choice depending on the initial severity of patient disease and the preferences of the treating physician. When the patient does not have a satisfactory response, new drugs acting at a different pathway are most commonly added. At this time, considering referral for lung transplantation could be an alternative. Most experts recommend grouping maximum experience in what is known as expert centers. Treatment has led to better survival in these patients, but there is still a long way to cure this life-threatening disease.


Subject(s)
Endothelin Receptor Antagonists/therapeutic use , Hypertension, Pulmonary/therapy , Lung Transplantation , Phosphodiesterase Inhibitors/therapeutic use , Prostaglandins I/therapeutic use , Clinical Trials as Topic , Disease Management , Drug Therapy, Combination , Exercise Tolerance , Guanylate Cyclase/drug effects , Heart Atria/surgery , Heart Septum/surgery , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/surgery , Life Style , Palliative Care , Prostaglandins I/agonists
18.
Am J Health Syst Pharm ; 71(21): 1839-44, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25320133

ABSTRACT

PURPOSE: The pharmacology, pharmacokinetics, clinical efficacy, safety, and role in therapy for riociguat are reviewed. SUMMARY: Riociguat is the first member of a new class of medications, soluble guanylate cyclase stimulators. Riociguat is indicated for patients with resistant or recurrent chronic thromboembolic pulmonary hypertension (CTEPH) after pulmonary endarterectomy who have World Health Organization (WHO) functional class IV pulmonary arterial hypertension (PAH) and in patients with inoperable CTEPH, regardless of WHO functional class, to improve exercise capacity and WHO functional class. Riociguat is indicated in patients with WHO functional class II PAH to improve exercise capacity, improve functional class, and delay clinical worsening. The mechanism of action of riociguat is within the nitric oxide pathway in the pulmonary vasculature. Clinical trials have demonstrated improvements in exercise capacity as measured by the six-minute walk distance test and in pulmonary arterial hemodynamics as measured by invasive pulmonary monitoring. Riociguat must be administered three times daily and requires dosage adjustments. Riociguat is a pregnancy category X drug and interacts with numerous medications. The two most serious adverse effects related to riociguat are hypotension and bleeding. Riociguat's role in the therapy of both PAH and CTEPH will be determined as more clinical experience and data are collected. Riociguat will likely cost approximately $90,000 annually. CONCLUSION: Riociguat is a soluble guanylate cyclase stimulator approved for the treatment of CTEPH and PAH. It can be considered first-line therapy for the treatment of CTEPH and should be considered as an alternative to phosphodiesterase type-5 inhibitors in patients with PAH.


Subject(s)
Hypertension, Pulmonary/drug therapy , Pulmonary Embolism/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Thromboembolism/complications , Chronic Disease , Female , Guanylate Cyclase/drug effects , Humans , Hypertension, Pulmonary/etiology , Pregnancy , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology
19.
Bratisl Lek Listy ; 115(6): 334-9, 2014.
Article in English | MEDLINE | ID: mdl-25023422

ABSTRACT

OBJECTIVES: It is aimed to investigate the effects of guanylyl cyclase activation and inhibition on acute morphine antinociception and the development of tolerance to its effect. BACKGROUND: Nitric oxide-soluble guanylyl cyclase signal transduction cascade suggested to play an important role in the development of tolerance to antinociceptive effects of morphine. METHODS: Nociception was evaluated by tail flick and hot plate tests in male Wistar rats. The analgesic effects of intraperitoneal protoporphyrin IX (PPIX; an activator of soluble guanylyl cyclase), 3-morpholinosydnonimine hydrochloride (SIN-1; NO donor and activator of guanylyl cyclase), S-Nitroso-N-acetylpenicillamine (SNAP; an activator of guanylyl cyclase), 3,3-Bis (amino ethyl)-1-hydroxy-2-oxo-1-triazene (NOC-18; NO donor activating guanylyl cyclase) and 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; an inhibitor of guanylyl cyclase) alone or in combination with subcutaneous morphine injection were evaluated. Their effects on morphine tolerance development were evaluated by giving these agents 20 minutes prior to twice daily morphine injection during tolerance development for 5 days. On day 6, the expression of morphine tolerance was determined. RESULTS: PPIX, SIN-1, SNAP and NOC-18 significantly increased expression of morphine tolerance while ODQ decreased. CONCLUSION: These data suggested that sGC activators have a significant role in tolerance to the analgesic effect of morphine (Tab. 1, Fig. 4, Ref. 29).


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/metabolism , Morphine/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Guanylate Cyclase/drug effects , Male , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Oxadiazoles/pharmacology , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/drug effects , Restriction Mapping , S-Nitroso-N-Acetylpenicillamine/pharmacology , Soluble Guanylyl Cyclase
20.
Pulm Pharmacol Ther ; 28(2): 130-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24657506

ABSTRACT

Riociguat is a new soluble guanylate cyclase stimulator under development for pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. So far, the interaction potential of riociguat with other drugs is nearly unknown. Therefore, we assessed in vitro the potency of riociguat to inhibit important drug metabolising enzymes (cytochrome P450 (CYP) 3A4, CYP2C19, and CYP2D6) and drug transporters (P-glycoprotein (P-gp/ABCB1), breast cancer resistance protein (BCRP/ABCG2), and organic anion transporting polypeptides (OATP) 1B1 and 1B3). In addition we evaluated its substrate characteristics for P-gp, BCRP, and the multidrug resistance-associated protein 1 (MRP1/ABCC1). We also assessed riociguat's inducing properties on important drug metabolising enzymes and transporters and investigated its ability to activate the pregnane-X-receptor (PXR). Riociguat was identified as a weak to moderate inhibitor of P-gp (f2-value: 11.7 ± 4.8 µM), BCRP (IC50 = 46.2 ± 20.3 µM), OATP1B1 (IC50 = 34.1 ± 3.15 µM), OATP1B3 (IC50 = 50.3 ± 7.5 µM), CYP2D6 (IC50 = 12.4 ± 0.74 µM), and CYP2C19 (IC50 = 46.1 ± 7.14 µM). Furthermore, it induced mRNA expression of BCRP/ABCG2 (3-fold at 20 µM) and to a lesser extent of CYP3A4 (2.3-fold at 20 µM), UGT1A4, and ABCB11. The only weak inducing properties were confirmed by weak activation of PXR. Considering its systemic concentrations its interaction potential as a perpetrator drug seems to be low. In contrast, our data suggest that riociguat is a P-gp substrate and might therefore act as a victim drug when co-administered with strong P-gp inductors or inhibitors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Gene Expression Regulation/drug effects , Guanylate Cyclase/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cell Line , Cell Line, Tumor , Dogs , Drug Interactions , Humans , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Mice , Pharmacokinetics , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , RNA, Messenger/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL