Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Rev Neurol (Paris) ; 179(9): 967-974, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37460332

ABSTRACT

Cannabis may have therapeutic benefits to relieve symptoms of amyotrophic lateral sclerosis (ALS) thanks to its pleiotropic pharmacological activity. This study is the first to present a large questionnaire-based survey about the "real-life" situation regarding cannabis use in the medical context in ALS patients in France. There were 129 respondents and 28 reported the use of cannabis (21.7%) to relieve symptoms of ALS. Participants mostly reported the use of cannabidiol (CBD) oil and cannabis weed and declared benefits both on motor (rigidity, cramps, fasciculations) and non-motor (sleep quality, pain, emotional state, quality of life, depression) symptoms and only eight reported minor adverse reactions (drowsiness, euphoria and dry mouth). Even if cannabis is mostly used outside medical pathways and could expose patients to complications (street and uncontrolled drugs, drug-drug interactions, adverse effects…), most of the participants reported "rational" consumption (legal cannabinoids, with only few combustion and adverse reactions). Despite some limitations, this study highlights the need for further research on the potential benefits of cannabis use for the management of ALS motor and non-motor symptoms. Indeed, there is an urgent need and call for and from patients to know more about cannabis and secure its use in a medical context.


Subject(s)
Amyotrophic Lateral Sclerosis , Cannabinoids , Cannabis , Humans , Cannabis/adverse effects , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/complications , Quality of Life , Cannabinoids/adverse effects , Pain
2.
Eur J Clin Pharmacol ; 76(9): 1281-1289, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32495084

ABSTRACT

PURPOSE: Beta-lactams (BL), the most commonly prescribed class of antibiotics, are recommended as the first-line therapy for multiple indications in infectious disease guidelines. Meropenem (MERO) is frequently used in intensive care units (ICU) to treat bacterial infections with or without sepsis. The pharmacokinetics of MERO display a large variability in patients admitted to ICUs due to altered pathophysiology. The aim of this study was to perform an external evaluation of published population pharmacokinetic models of MERO in order to test their predictive performance in a cohort of ICU adult patients. METHODS: A literature search in PubMed/Medline database was made following the PRISMA statement. External evaluation was performed using NONMEM software, and the bias and inaccuracy values were calculated. RESULTS: An external validation dataset from the Timone Hospital in Marseille, France, included 84 concentration samples from 27 patients. Four models of MERO were identified according to the inclusion criteria of the study. None of the models presented acceptable values of bias and inaccuracy. CONCLUSION: While performing external evaluations on some populations may confirm a model's suitability to diverse groups of patients, there is still some variability that cannot be explained nor solved by the procedure. This brings to light the difficulty to develop only one model for ICU patients and the need to develop one specific model to each population of critically ill patients.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Intensive Care Units , Meropenem/pharmacokinetics , Models, Biological , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Critical Illness , Female , Humans , Male , Meropenem/administration & dosage , Middle Aged , Prospective Studies , Young Adult
3.
Encephale ; 44(6S): S34-S38, 2018 Dec.
Article in French | MEDLINE | ID: mdl-30935485

ABSTRACT

Despite the lack of progress in the curative treatment of mental illness, especially schizophrenia, the accumulation of neuroscience data over the past decade suggests the re-conceptualization of schizophrenia. With the advent of new biomarkers and cognitive tools, new neuroscience technologies such as functional dynamic connectivity and the identification of subtle clinical features; it is now possible to detect early stages at risk or prodromes of a first psychotic episode. Current concepts reconceptualizes schizophrenia as a neurodevelopmental disorder at early onset, with polygenic risk and only symptomatic treatment for positive symptoms at this time. The use of such technologies in the future suggests new diagnostic and therapeutic options. Next steps include new pharmacological perspectives and potential contributions of new technologies such as quantitative system pharmacology brain computational modeling approach.


Subject(s)
Antipsychotic Agents , Early Medical Intervention/methods , Pharmacology, Clinical/methods , Schizophrenia/drug therapy , Adolescent , Adult , Age Factors , Age of Onset , Antipsychotic Agents/classification , Antipsychotic Agents/therapeutic use , Brain , Child , Child, Preschool , Continuity of Patient Care , Early Diagnosis , Female , Humans , Infant , Infant, Newborn , Middle Aged , Pregnancy , Schizophrenia/diagnosis , Schizophrenia/epidemiology , Schizophrenia/pathology , Schizophrenic Psychology , Systems Integration , Young Adult
4.
Encephale ; 42(6S): S2-S6, 2016 Dec.
Article in French | MEDLINE | ID: mdl-28236988

ABSTRACT

Clinical trials in psychiatry allow to build the regulatory dossiers for market authorization but also to document the mechanism of action of new drugs, to build pharmacodynamics models, evaluate the treatment effects, propose prognosis, efficacy or tolerability biomarkers and altogether to assess the impact of drugs for patient, caregiver and society. However, clinical trials have shown some limitations. Number of recent dossiers failed to convince the regulators. The clinical and biological heterogeneity of psychiatric disorders, the pharmacokinetic and pharmacodynamics properties of the compounds, the lack of translatable biomarkers possibly explain these difficulties. Several breakthrough options are now available: quantitative system pharmacology analysis of drug effects variability, pharmacometry and pharmacoepidemiology, Big Data analysis, brain modelling. In addition to more classical approaches, these opportunities lead to a paradigm change for clinical trials in psychiatry.


Subject(s)
Clinical Trials as Topic , Mental Disorders/therapy , Psychiatry/methods , Psychiatry/trends , Brain/pathology , Clinical Trials as Topic/methods , Clinical Trials as Topic/organization & administration , Clinical Trials as Topic/standards , Computer Simulation , Humans , Mental Disorders/epidemiology , Pharmacoepidemiology , Pharmacogenomic Testing/methods , Pharmacogenomic Testing/trends , Research Design/standards , User-Computer Interface
5.
Methods Find Exp Clin Pharmacol ; 32(10): 737-43, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21225010

ABSTRACT

Advances in mass spectrometry instruments have led to increased utilization of high performance liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) and it would be necessary to standardize blood quantification of immunosuppressant drugs. The aim of the study was to validate and assess the robustness of an LC-MS/MS method for quantification of tacrolimus in whole blood using the Waters Masstrak™ Immunosuppressant Kit. After protein precipitation from whole blood samples, chromatographic separation was performed in 2 min. Detection was performed with a Waters Tandem Quadrupole MS Quattro Premier XE, operated in multiple-reaction monitoring in positive electrospray ionization mode. This method was validated and compared to Enzyme Multiplied Immunoassay Technique (EMIT) method in accordance with actual guidelines. The limit of quantification was 1.0 ng/mL and the calibration curve was linear to 27.6 ng/mL. Between-day and within-day trueness and precision were < 15% at three concentrations spanning the linear range. The EMIT assay showed an average positive bias of 28.3% compared with the LC-MS/MS. Internal and external quality control were always accepted and demonstrated the robustness of this method. In conclusion, we validated a rapid, simple and robust quantification of tacrolimus in blood samples with the Waters Masstrak™ Immunosuppressant Kit.


Subject(s)
Chromatography, High Pressure Liquid/methods , Immunosuppressive Agents/blood , Tacrolimus/blood , Tandem Mass Spectrometry/methods , Calibration , Drug Monitoring/methods , Enzyme Multiplied Immunoassay Technique , Humans , Quality Control
6.
Therapie ; 75(4): 363-370, 2020.
Article in English | MEDLINE | ID: mdl-32473812

ABSTRACT

Chloroquine and hydroxychloroquine are drugs that have shown in vitro activity on the replication of certain coronaviruses. In the context of the SARS-Cov-2 epidemic, the virus responsible for the novel coronavirus disease (COVID-19), these two drugs have been proposed as possible treatments. The results of the first clinical studies evaluating the effect of hydroxychloroquine do not support any efficacy of this drug in patients with COVID-19, due to major methodological weaknesses. Yet, these preliminary studies have aroused considerable media interest, raising fears of massive and uncontrolled use. In the absence of evidence of clinical benefits, the main risk is of exposing patients unnecessarily to the well-known adverse effects of hydroxychloroquine, with a possibly increased risk in the specific setting of COVID-19. In addition, widespread use outside of any recommendation risks compromising the completion of good quality clinical trials. The chloroquine hype, fueled by low-quality studies and media announcements, has yielded to the implementation of more than 150 studies worldwide. This represents a waste of resources and a loss of opportunity for other drugs to be properly evaluated. In the context of emergency, rigorous trials are more than ever needed in order to have, as soon as possible, reliable data on drugs that are possibly effective against the disease. Meanwhile, serious adverse drug reactions have been reported in patients with COVID-19 receiving hydroxychloroquine, justifying to limit its prescription, and to perform suitable cardiac and therapeutic drug monitoring.


Subject(s)
Chloroquine/administration & dosage , Coronavirus Infections/drug therapy , Hydroxychloroquine/administration & dosage , Pneumonia, Viral/drug therapy , COVID-19 , Chloroquine/adverse effects , Coronavirus Infections/virology , Drug Monitoring , Humans , Hydroxychloroquine/adverse effects , Pandemics , Pneumonia, Viral/virology , Treatment Outcome , COVID-19 Drug Treatment
7.
Int J Antimicrob Agents ; 56(4): 106124, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32739478

ABSTRACT

Modifications of antibiotic pharmacokinetic parameters have been reported in critically ill patients, resulting in a risk of treatment failure. We aimed to determine optimised amikacin (AMK), gentamicin (GEN) and tobramycin (TOB) intravenous dosing regimens in this patient population. Patients admitted to the medical ICU and treated with AMK, GEN or TOB were included. Analyses were performed using a parametric population approach. Monte Carlo simulations were performed and the probability of target attainment (PTA) was calculated using Cmax/MIC ≥ 8 and trough concentrations as targets. A total of 117 critically ill hospitalised patients were studied. Median values (interindividual variability, É·2) of clearance were 3.51 (0.539), 3.53 (0.297), 2.70 (0.339) and 5.07 (0.339) L/h for AMK, GEN, TOB, and TOB in cystic fibrosis (CF), respectively. Median values (É·2) of central volume of distribution were 30.2 (0.215), 20.0 (0.109) and 25.6 (0.177) L for AMK, GEN and TOB, respectively. Simulations showed that doses should be adjusted to actual body weight and creatinine clearance (CLCR) for AMK and GEN, and according to CLCR and presence of CF for TOB. In conclusion, our recommendations for treating Pseudomonas aeruginosa infections in this population include using initial doses of 35 mg/kg for AMK or 10 mg/kg for TOB (CF and non-CF patients). GEN demonstrated the best rates of target attainment against Staphylococcus aureus infections with a dose of 5 mg/kg. As high aminoglycoside doses are required in this population, efficacy and safety targets are conflicting and therapeutic drug monitoring remains an important tool to manage this issue.


Subject(s)
Aminoglycosides/therapeutic use , Anti-Bacterial Agents/therapeutic use , Pseudomonas Infections/drug therapy , Sepsis/drug therapy , Staphylococcal Infections/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Amikacin/administration & dosage , Amikacin/therapeutic use , Aminoglycosides/administration & dosage , Anti-Bacterial Agents/administration & dosage , Critical Illness , Female , Gentamicins/administration & dosage , Gentamicins/therapeutic use , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Prospective Studies , Pseudomonas aeruginosa/drug effects , Sepsis/microbiology , Staphylococcus aureus/drug effects , Tobramycin/administration & dosage , Tobramycin/therapeutic use , Young Adult
8.
Paediatr Drugs ; 20(4): 375-381, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29736878

ABSTRACT

BACKGROUND: The application of population pharmacokinetic models and Bayesian methods offers the potential to develop individualized therapeutic approaches. OBJECTIVES: The current study presents an external evaluation of a vancomycin pharmacokinetic model in a pediatric cancer population and proposes an easy-to-use chart for clinicians for a priori vancomycin schedule adaptation to achieve target concentration. METHODS: External evaluation of a population pharmacokinetic model of vancomycin administered via continuous infusion was realized in a new retrospective dataset of pediatric patients with cancer. The published population pharmacokinetic model was implemented in NONMEM 7.3 with the structural and variance parameter values set equal to estimates previously reported. Predictive performance was assessed by quantifying bias and accuracy of model prediction. Normalized prediction distribution errors were also evaluated. Dosage simulations were performed according to the target concentration. RESULTS: A total of 77 patients were included in this study, representing 146 vancomycin courses and 289 concentrations. The model adequately predicted vancomycin concentrations (median prediction error % of - 9.4%, median |PE|% of 24.1%). Based on simulation results, vancomycin dosage (mg/kg) should be adapted for each child on the basis of body weight and cyclosporine coadministration. CONCLUSION: The model previously proposed by Guilhaumou et al. in pediatric patients with solid or hematological malignant disease was externally validated. Simulations have enabled the description of new dosage schedules and creation of a chart to help clinicians adapt vancomycin dosage.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Drug Dosage Calculations , Hematologic Neoplasms/blood , Models, Biological , Vancomycin/pharmacokinetics , Anti-Bacterial Agents/administration & dosage , Bayes Theorem , Child , Child, Preschool , Drug Monitoring , Emollients , Female , Humans , Male , Neoplasms/blood , Retrospective Studies , Vancomycin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL