Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
CPT Pharmacometrics Syst Pharmacol ; 12(4): 500-512, 2023 04.
Article in English | MEDLINE | ID: covidwho-2299993

ABSTRACT

Apixaban is an oral small-molecule, direct factor Xa (FXa) inhibitor approved in adults for treatment of deep vein thrombosis and pulmonary embolism, and for reducing risk of venous thromboembolism recurrence after initial anticoagulant therapy. This phase I study (NCT01707394) evaluated the pharmacokinetics (PKs), pharmacodynamics (PDs), and safety of apixaban in pediatric subjects (<18 years), enrolled by age group, at risk of venous or arterial thrombotic disorder. A single apixaban dose, targeting adult steady-state exposure with apixaban 2.5 mg, was administered using two pediatric formulations: 0.1 mg sprinkle capsule (age <28 days); 0.4 mg/ml solution (age 28 days to <18 years; dose range, 1.08-2.19 mg/m2 ). End points included safety, PKs, and anti-FXa activity. For PKs/PDs, four to six blood samples were collected ≤26 h postdosing. A population PK model was developed with data from adults and pediatric subjects. Apparent oral clearance (CL/F) included fixed maturation function based on published data. From January 2013 to June 2019, 49 pediatric subjects received apixaban. Most adverse events were mild/moderate, and the most common was pyrexia (n = 4/15). Apixaban CL/F and apparent central volume of distribution increased less than proportionally with body weight. Apixaban CL/F increased with age, reaching adult values in subjects aged 12 to <18 years. Maturation affected CL/F most notably in subjects aged <9 months. Plasma anti-FXa activity values were linearly related to apixaban concentrations, with no apparent age-related differences. Pediatric subjects tolerated single apixaban doses well. Study data and population PK model supported phase II/III pediatric trial dose selection.


Subject(s)
Factor Xa Inhibitors , Pyridones , Adult , Humans , Child , Adolescent , Factor Xa Inhibitors/adverse effects , Pyrazoles , Anticoagulants/pharmacokinetics
2.
Clin Pharmacokinet ; 62(6): 835-847, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2301873

ABSTRACT

BACKGROUND AND OBJECTIVE: The risk of thrombotic complications in critical patients with COVID-19 remains extremely high, and multicenter trials failed to prove a survival benefit of escalated doses of low-molecular-weight heparins (nadroparin calcium) in this group. The aim of this study was to develop a pharmacokinetic model of nadroparin according to different stages of COVID-19 severity. METHODS: Blood samples were obtained from 43 patients with COVID-19 who received nadroparin and were treated with conventional oxygen therapy, mechanical ventilation, and extracorporeal membrane oxygenation. We recorded clinical, biochemical, and hemodynamic variables during 72 h of treatment. The analyzed data comprised 782 serum nadroparin concentrations and 219 anti-Xa levels. We conducted population nonlinear mixed-effects modeling (NONMEM) and performed Monte Carlo simulations of the probability of target attainment for reaching 0.2-0.5 IU/mL anti-Xa levels in study groups. RESULTS: We successfully developed a one-compartment model to describe the population pharmacokinetics of nadroparin in different stages of COVID-19. The absorption rate constant of nadroparin was 3.8 and 3.2 times lower, concentration clearance was 2.22 and 2.93 times higher, and anti-Xa clearance was 0.87 and 1.1 times higher in mechanically ventilated patients and the extracorporeal membrane oxygenation group compared with patients treated with conventional oxygen, respectively. The newly developed model indicated that 5.900 IU of nadroparin given subcutaneously twice daily in the mechanically ventilated patients led to a similar probability of target attainment of 90% as 5.900 IU of subcutaneous nadroparin given once daily in the group supplemented with conventional oxygen. CONCLUSIONS: Different nadroparin dosing is required for patients undergoing mechanical ventilation and extracorporeal membrane oxygenation to achieve the same targets as those for non-critically ill patients. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov identifier no. NCT05621915.


Subject(s)
COVID-19 , Nadroparin , Humans , Nadroparin/therapeutic use , Nadroparin/pharmacokinetics , Anticoagulants/pharmacokinetics
3.
Molecules ; 26(9)2021 Apr 29.
Article in English | MEDLINE | ID: covidwho-1217102

ABSTRACT

Hemostasis disorders play an important role in the pathogenesis, clinical manifestations, and outcome of COVID-19. First of all, the hemostasis system suffers due to a complicated and severe course of COVID-19. A significant number of COVID-19 patients develop signs of hypercoagulability, thrombocytopenia, and hyperfibrinolysis. Patients with severe COVID-19 have a tendency toward thrombotic complications in the venous and arterial systems, which is the leading cause of death in this disease. Despite the success achieved in the treatment of SARS-CoV-2, the search for new effective anticoagulants, thrombolytics, and fibrinolytics, as well as their optimal dose strategies, continues to be relevant. The wide therapeutic potential of seaweed sulfated polysaccharides (PSs), including anticoagulant, thrombolytic, and fibrinolytic activities, opens up new possibilities for their study in experimental and clinical trials. These natural compounds can be important complementary drugs for the recovery from hemostasis disorders due to their natural origin, safety, and low cost compared to synthetic drugs. In this review, the authors analyze possible pathophysiological mechanisms involved in the hemostasis disorders observed in the pathological progression of COVID-19, and also focus the attention of researchers on seaweed PSs as potential drugs aimed to correction these disorders in COVID-19 patients. Modern literature data on the anticoagulant, antithrombotic, and fibrinolytic activities of seaweed PSs are presented, depending on their structural features (content and position of sulfate groups on the main chain of PSs, molecular weight, monosaccharide composition and type of glycosidic bonds, the degree of PS chain branching, etc.). The mechanisms of PS action on the hemostasis system and the issues of oral bioavailability of PSs, important for their clinical use as oral anticoagulant and antithrombotic agents, are considered. The combination of the anticoagulant, thrombolytic, and fibrinolytic properties, along with low toxicity and relative cheapness of production, open up prospects for the clinical use of PSs as alternative sources of new anticoagulant and antithrombotic compounds. However, further investigation and clinical trials are needed to confirm their efficacy.


Subject(s)
Anticoagulants/pharmacology , COVID-19/complications , Hemostasis/drug effects , Polysaccharides/pharmacology , Seaweed , Sulfates/pharmacology , Thrombosis/complications , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacokinetics , Anticoagulants/therapeutic use , COVID-19/blood , Drug Discovery , Humans , Polysaccharides/chemistry , Polysaccharides/pharmacokinetics , Polysaccharides/therapeutic use , Seaweed/chemistry , Sulfates/chemistry , Sulfates/pharmacokinetics , Sulfates/therapeutic use , Thrombosis/blood , Thrombosis/drug therapy , COVID-19 Drug Treatment
5.
J Am Heart Assoc ; 9(14): e017529, 2020 07 21.
Article in English | MEDLINE | ID: covidwho-981044

ABSTRACT

Atrial fibrillation is a common clinical manifestation in hospitalized patients with coronavirus disease 2019 (COVID-19). Medications used to treat atrial fibrillation, such as antiarrhythmic drugs and anticoagulants, may have significant drug interactions with emerging COVID-19 treatments. Common unintended nontherapeutic target effects of COVID-19 treatment include potassium channel blockade, cytochrome P 450 isoenzyme inhibition or activation, and P-glycoprotein inhibition. Drug-drug interactions with antiarrhythmic drugs and anticoagulants in these patients may lead to significant bradycardia, ventricular arrhythmias, or severe bleeding. It is important for clinicians to be aware of these interactions, drug metabolism changes, and clinical consequences when choosing antiarrhythmic drugs and anticoagulants for COVID-19 patients with atrial fibrillation. The objective of this review is to provide a practical guide for clinicians who are managing COVID-19 patients with concomitant atrial fibrillation.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Anticoagulants/therapeutic use , Antiviral Agents/therapeutic use , Atrial Fibrillation/therapy , Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/pharmacokinetics , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Atrial Fibrillation/diagnosis , Atrial Fibrillation/epidemiology , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Drug Interactions , Host Microbial Interactions , Humans , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Risk Assessment , Risk Factors , SARS-CoV-2 , Treatment Outcome , COVID-19 Drug Treatment
8.
J Cardiovasc Pharmacol ; 76(2): 138-145, 2020 08.
Article in English | MEDLINE | ID: covidwho-433268

ABSTRACT

Coronavirus disease 2019 (COVID-19) outbreak is a public health emergency of international concerns because of a highly pathogenic human coronavirus (HCoV), actually named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite much emerging data about the epidemiological association between cardiovascular diseases and COVID-19, little is still known about atrial fibrillation and its optimal management in this clinical contest. The aim of our review is to describe the pharmacological interactions between cardiovascular drugs more commonly used in atrial fibrillation management and experimental COVID-19 therapies, based on EU and US summaries of product characteristics.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Anticoagulants/therapeutic use , Antiviral Agents/therapeutic use , Atrial Fibrillation/drug therapy , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/pharmacokinetics , Anticoagulants/adverse effects , Anticoagulants/pharmacokinetics , Antiviral Agents/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/epidemiology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Drug Interactions , Host-Pathogen Interactions , Humans , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Risk Factors , SARS-CoV-2 , Treatment Outcome , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL