Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Clin Pharmacol Ther ; 116(1): 106-116, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38709223

ABSTRACT

The ICH E14/S7B Q&As highlighted the need for best practices concerning the design, execution, analysis, interpretation, and reporting of the in vivo non-rodent QT assay as a component of the integrated risk assessment to potentially support a TQT waiver or substitute. We conducted a dog telemetry study to assess the effects on QTc of six reference compounds (five positive and one negative) previously evaluated by Darpo et al. (2015) in humans. The sensitivity of the assay to detect QTc increases was determined, and exposure-response analysis was performed, as done in clinical practice. By-timepoint analysis showed QTc prolongation induced by moxifloxacin, dofetilide, dolasetron, ondansetron, and quinine within human relevant plasma exposures ranges. Moreover, a hysteresis was observed for quinine. As expected, levocetirizine showed no statistically significant effect on QTc across a range of exposure, well exceeding the therapeutic Cmax. Power analyses confirmed the study ability to detect statistically significant QTc changes of less than 10 milliseconds with 80% probability, even with a sample size as low as n = 4 animals. Finally, concentration-QTc modeling enabled to predict the minimal plasma concentration needed to detect a 10 milliseconds QTc prolongation, including for quinine. The comparison with clinical available data supported the relevance of dogs under these experimental conditions as a robust translational predictor of drug-induced QTc prolongation in humans as a key pillar of the integrated risk assessment.


Subject(s)
Electrocardiography , Long QT Syndrome , Dogs , Animals , Prospective Studies , Long QT Syndrome/chemically induced , Long QT Syndrome/diagnosis , Electrocardiography/drug effects , Male , Female , Telemetry/methods , Risk Assessment/methods , Humans , Heart Rate/drug effects
3.
Diabetes Obes Metab ; 26(6): 2368-2378, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38560764

ABSTRACT

AIM: To describe the biomarker strategy that was applied to select survodutide (BI 456906), BI 456908 and BI 456897 from 19 dual glucagon receptor (GCGR)/ glucagon-like peptide-1 receptor (GLP-1R) agonists for in-depth pharmacological profiling, which led to the qualification of survodutide as the clinical development candidate. MATERIALS AND METHODS: Potencies to increase cyclic adenosine monophosphate (cAMP) were determined in Chinese hamster ovary (CHO)-K1 cells stably expressing human GCGR and GLP-1R. Agonism for endogenously expressed receptors was investigated in insulinoma cells (MIN6) for mouse GLP-1R, and in rat primary hepatocytes for the GCGR. In vivo potencies to engage the GLP-1R or GCGR were determined, measuring improvement in oral glucose tolerance (30 nmol/kg) and increase in plasma fibroblast growth factor-21 (FGF21) and liver nicotinamide N-methyltransferase (NNMT) mRNA expression (100 nmol/kg), respectively. Body weight- and glucose-lowering efficacies were investigated in diet-induced obese (DIO) mice and diabetic db/db mice, respectively. RESULTS: Upon acute dosing in lean mice, target engagement biomarkers for the GCGR and GLP-1R demonstrated a significant correlation (Spearman correlation coefficient with p < 0.05) to the in vitro GCGR and GLP-1R potencies for the 19 dual agonists investigated. Survodutide, BI 456908 and BI 456897 were selected for in-depth pharmacological profiling based on the significant improvement in acute oral glucose tolerance achieved (area under the curve [AUC] of 54%, 57% and 60% vs. vehicle) that was comparable to semaglutide (AUC of 45% vs. vehicle), while showing different degrees of in vivo GCGR engagement, as determined by hepatic NNMT mRNA expression (increased by 15- to 17-fold vs. vehicle) and plasma FGF21 concentrations (increased by up to sevenfold vs. vehicle). In DIO mice, survodutide (30 nmol/kg/once daily), BI 456908 (30 nmol/kg/once daily) and BI 456897 (10 nmol/kg/once daily) achieved a body weight-lowering efficacy from baseline of 25%, 27% and 26%, respectively. In db/db mice, survodutide and BI 456908 (10 and 20 nmol/kg/once daily) significantly lowered glycated haemoglobin (0.4%-0.6%); no significant effect was observed for BI 456897 (3 and 7 nmol/kg/once daily). CONCLUSIONS: Survodutide was selected as the clinical candidate based on its balanced dual GCGR/GLP-1R pharmacology, engaging the GCGR for robust body weight-lowering efficacy exceeding that of selective GLP-1R agonists, while achieving antidiabetic efficacy that was comparable to selective GLP-1R agonism. Survodutide is currently being investigated in Phase 3 clinical trials in people living with obesity.


Subject(s)
Cricetulus , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents , Receptors, Glucagon , Animals , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Mice , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , CHO Cells , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Humans , Biomarkers/blood , Male , Rats , Mice, Obese , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Diabetes Mellitus, Type 2/drug therapy
4.
J Pharmacol Toxicol Methods ; 122: 107279, 2023.
Article in English | MEDLINE | ID: mdl-37257761

ABSTRACT

Conducting safety evaluations of new drugs using conscious animals has been a specialty of our working group for thirty years. In this article, we review the various technical challenges and solutions dealt with over the years to improve both the data quality and the well being of our animal subjects. Of particular interest for us has been the use of telemetry-based data acquisition for conducting studies on cardiovascular (CV) function. This includes the evolving technical aspects of the studies, as well as the development of new applications that take advantage of this technical approach.


Subject(s)
Cardiovascular Agents , Cardiovascular System , Animals , Electrocardiography , Heart Rate , Telemetry , Cardiovascular Agents/adverse effects , Blood Pressure
5.
J Pharmacol Toxicol Methods ; 115: 107171, 2022.
Article in English | MEDLINE | ID: mdl-35398273

ABSTRACT

Cardiovascular (CV) effects represent a major safety issue during drug development. Typically, this risk is mitigated by preclinical in vivo CV studies, based on which measured CV readouts are analyzed independently. Here, we apply a regression approach to simultaneously integrate CV readouts, i.e., heart rate (HR), mean arterial pressure (MAP) and QT from five dog telemetry studies. These CV studies comprise data on verapamil, captopril, dofetilide, pimobendan, and formoterol, and are combined with the respective dog pharmacokinetic (PK) profiles. A published PK/CV model structure for rats is extended by a semi-mechanistic parameterization of the interaction between HR and QT specific to dogs. This semi-mechanistic modelling approach allows differentiation between compound-independent system-specific parameters (e.g., HR baseline) and compound-specific parameters (e.g., EC50). Compared to previous results in rodents, estimated parameters for dogs indicate stronger dependency of stroke volume on HR, slower HR response, faster QT response and steeper concentration-response relationships. In addition, we illustrate how to practically apply the PK/CV model to derive concentration-response relationships for CV readouts. This approach allows a more detailed quantitative evaluation based on the maximum effect on CV effects (Emax), the EC50, and the steepness of this relation (Hill coefficient) especially for HR-independent effects on QT interval duration (QTc) while taking the systemic feedback into account. This approach also allows to derive plasma concentrations associated with relevant CV effects ("threshold concentration"; CTHRESH). The presented modelling analysis highlights the potential of an integrative evaluation of CV data and provides a framework for obtaining quantitative insights from safety pharmacology evaluations.


Subject(s)
Cardiovascular System , Long QT Syndrome , Animals , Dogs , Drug Development , Electrocardiography , Heart Rate , Long QT Syndrome/chemically induced , Rats , Telemetry/methods , Verapamil/pharmacology
6.
Analyst ; 146(13): 4226-4234, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34095908

ABSTRACT

DNA hybridization phenomena occurring on solid supports are not understood as clearly as aqueous phase hybridizations and mathematical models cannot predict some empirically obtained results. Ongoing research has identified important parameters but remains incomplete to accurately account for all interactions. It has previously been shown that the length of the overhanging (dangling) end of the target DNA strand following hybridization to the capture probe is correlated to interactions with the complementary strand in solution which can result in unbinding of the target and its release from the surface. We have developed an instrument for real-time monitoring of DNA hybridization on spherical particles functionalized with oligonucleotide capture probes and arranged in the form of a tightly packed monolayer bead bed inside a microfluidic cartridge. The instrument is equipped with a pneumatic module to mediate displacement of fluid on the cartridge. We compared this system to both conventional (passive) and centrifugally-driven (active) microfluidic microarray hybridization on glass slides to establish performance levels for the detection of single nucleotide polymorphisms. The system was also used to study the effect of the dangling end's length in real-time when the immobilized target DNA is exposed to the complementary strand in solution. Our findings indicate that increasing the length of the dangling end leads to desorption of target amplicons from bead-bound capture probes at a rate approaching that of the initial hybridization process. Finally, bead bed hybridization was performed with Streptococcus agalactiae cfb gene amplicons obtained from randomized clinical samples, which allowed for identification of group B streptococci within 5-15 min. The methodology presented here is useful for investigating competitive hybridization mechanisms on solid supports and to rapidly validate the suitability of microarray capture probes.


Subject(s)
DNA , Microfluidics , DNA/genetics , DNA Probes/genetics , Nucleic Acid Hybridization , Oligonucleotide Probes/genetics
7.
J Pharmacol Exp Ther ; 377(2): 218-231, 2021 05.
Article in English | MEDLINE | ID: mdl-33648939

ABSTRACT

Cardiovascular adverse effects in drug development are a major source of compound attrition. Characterization of blood pressure (BP), heart rate (HR), stroke volume (SV), and QT-interval prolongation are therefore necessary in early discovery. It is, however, common practice to analyze these effects independently of each other. High-resolution time courses are collected via telemetric techniques, but only low-resolution data are analyzed and reported. This ignores codependencies among responses (HR, BP, SV, and QT-interval) and separation of system (turnover properties) and drug-specific properties (potencies, efficacies). An analysis of drug exposure-time and high-resolution response-time data of HR and mean arterial blood pressure was performed after acute oral dosing of ivabradine, sildenafil, dofetilide, and pimobendan in Han-Wistar rats. All data were modeled jointly, including different compounds and exposure and response time courses, using a nonlinear mixed-effects approach. Estimated fractional turnover rates [h-1, relative standard error (%RSE) within parentheses] were 9.45 (15), 30.7 (7.8), 3.8 (13), and 0.115 (1.7) for QT, HR, total peripheral resistance, and SV, respectively. Potencies (nM, %RSE within parentheses) were IC 50 = 475 (11), IC 50 = 4.01 (5.4), EC 50 = 50.6 (93), and IC 50 = 47.8 (16), and efficacies (%RSE within parentheses) were I max = 0.944 (1.7), Imax = 1.00 (1.3), E max = 0.195 (9.9), and Imax = 0.745 (4.6) for ivabradine, sildenafil, dofetilide, and pimobendan. Hill parameters were estimated with good precision and below unity, indicating a shallow concentration-response relationship. An equilibrium concentration-biomarker response relationship was predicted and displayed graphically. This analysis demonstrates the utility of a model-based approach integrating data from different studies and compounds for refined preclinical safety margin assessment. SIGNIFICANCE STATEMENT: A model-based approach was proposed utilizing biomarker data on heart rate, blood pressure, and QT-interval. A pharmacodynamic model was developed to improve assessment of high-resolution telemetric cardiovascular safety data driven by different drugs (ivabradine, sildenafil, dofetilide, and pimobondan), wherein system- (turnover rates) and drug-specific parameters (e.g., potencies and efficacies) were sought. The model-predicted equilibrium concentration-biomarker response relationships and was used for safety assessment (predictions of 20% effective concentration, for example) of heart rate, blood pressure, and QT-interval.


Subject(s)
Biomarkers, Pharmacological/blood , Blood Pressure , Cardiovascular Agents/toxicity , Heart Rate , Animals , Cardiotoxicity/blood , Cardiotoxicity/etiology , Cardiotoxicity/physiopathology , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/pharmacokinetics , Ivabradine/administration & dosage , Ivabradine/pharmacokinetics , Ivabradine/toxicity , Male , Phenethylamines/administration & dosage , Phenethylamines/pharmacokinetics , Phenethylamines/toxicity , Pyridazines/administration & dosage , Pyridazines/pharmacokinetics , Pyridazines/toxicity , Rats , Rats, Wistar , Sildenafil Citrate/administration & dosage , Sildenafil Citrate/pharmacokinetics , Sildenafil Citrate/toxicity , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity
8.
Clin Pharmacol Ther ; 109(2): 310-318, 2021 02.
Article in English | MEDLINE | ID: mdl-32866317

ABSTRACT

Defining an appropriate and efficient assessment of drug-induced corrected QT interval (QTc) prolongation (a surrogate marker of torsades de pointes arrhythmia) remains a concern of drug developers and regulators worldwide. In use for over 15 years, the nonclinical International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) S7B and clinical ICH E14 guidances describe three core assays (S7B: in vitro hERG current & in vivo QTc studies; E14: thorough QT study) that are used to assess the potential of drugs to cause delayed ventricular repolarization. Incorporating these assays during nonclinical or human testing of novel compounds has led to a low prevalence of QTc-prolonging drugs in clinical trials and no new drugs having been removed from the marketplace due to unexpected QTc prolongation. Despite this success, nonclinical evaluations of delayed repolarization still minimally influence ICH E14-based strategies for assessing clinical QTc prolongation and defining proarrhythmic risk. In particular, the value of ICH S7B-based "double-negative" nonclinical findings (low risk for hERG block and in vivo QTc prolongation at relevant clinical exposures) is underappreciated. These nonclinical data have additional value in assessing the risk of clinical QTc prolongation when clinical evaluations are limited by heart rate changes, low drug exposures, or high-dose safety considerations. The time has come to meaningfully merge nonclinical and clinical data to enable a more comprehensive, but flexible, clinical risk assessment strategy for QTc monitoring discussed in updated ICH E14 Questions and Answers. Implementing a fully integrated nonclinical/clinical risk assessment for compounds with double-negative nonclinical findings in the context of a low prevalence of clinical QTc prolongation would relieve the burden of unnecessary clinical QTc studies and streamline drug development.


Subject(s)
Drugs, Investigational/adverse effects , Long QT Syndrome/chemically induced , Animals , Arrhythmias, Cardiac/chemically induced , Drug Development/methods , Drug Industry/methods , Electrocardiography/methods , Humans , Risk Assessment , Torsades de Pointes/chemically induced
9.
Stem Cells Transl Med ; 8(8): 822-832, 2019 08.
Article in English | MEDLINE | ID: mdl-31037857

ABSTRACT

We previously demonstrated that intracardiac delivery of autologous peripheral blood-derived CD34+ stem cells (SCs), mobilized by granulocyte-colony stimulating factor (G-CSF) and collected by leukapheresis after myocardial infarction, structurally and functionally repaired the damaged myocardial area. When used for cardiac indication, CD34+ cells are now considered as Advanced Therapy Medicinal Products (ATMPs). We have industrialized their production by developing an automated device for ex vivo CD34+ -SC expansion, starting from a whole blood (WB) sample. Blood samples were collected from healthy donors after G-CSF mobilization. Manufacturing procedures included: (a) isolation of total nuclear cells, (b) CD34+ immunoselection, (c) expansion and cell culture recovery in the device, and (d) expanded CD34+ cell immunoselection and formulation. The assessment of CD34+ cell counts, viability, and immunophenotype and sterility tests were performed as quality tests. We established graft acceptance criteria and performed validation processes in three cell therapy centers. 59.4 × 106 ± 36.8 × 106 viable CD34+ cells were reproducibly generated as the final product from 220 ml WB containing 17.1 × 106 ± 8.1 × 106 viable CD34+ cells. CD34+ identity, genetic stability, and telomere length were consistent with those of basal CD34+ cells. Gram staining and mycoplasma and endotoxin analyses were negative in all cases. We confirmed the therapeutic efficacy of both CD34+ -cell categories in experimental acute myocardial infarct (AMI) in immunodeficient rats during preclinical studies. This reproducible, automated, and standardized expansion process produces high numbers of CD34+ cells corresponding to the approved ATMP and paves the way for a phase I/IIb study in AMI, which is currently recruiting patients. Stem Cells Translational Medicine 2019;8:822&832.


Subject(s)
Antigens, CD34/genetics , Automation, Laboratory/methods , Flow Cytometry/methods , Myocardial Infarction/therapy , Peripheral Blood Stem Cell Transplantation/methods , Peripheral Blood Stem Cells/cytology , Adult , Animals , Antigens, CD34/metabolism , Cells, Cultured , Clinical Trials as Topic , Humans , Immunophenotyping/methods , Male , Middle Aged , Peripheral Blood Stem Cells/metabolism , Primary Cell Culture/methods , Rats
11.
Sci Rep ; 6: 37948, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27901061

ABSTRACT

Safety pharmacology aims to predict rare side effects of new drugs. We explored whether rare pro-arrhythmic effects could be linked to the variability of the effects of these drugs on ion currents and whether taking into consideration this variability in computational models could help to better detect and predict cardiac side effects. For this purpose, we evaluated how intra- and inter-individual variability influences the effect of hERG inhibition on both the action potential duration and the occurrence of arrhythmias. Using two computer simulation models of human action potentials (endocardial and Purkinje cells), we analyzed the contribution of two biological parameters on the pro-arrhythmic effects of several hERG channel blockers: (i) spermine concentration, which varies with metabolic status, and (ii) L-type calcium conductance, which varies due to single nucleotide polymorphisms or mutations. By varying these parameters, we were able to induce arrhythmias in 1 out of 16 simulations although conventional modeling methods to detect pro-arrhythmic molecules failed. On the basis of our results, taking into consideration only 2 parameters subjected to intra- and inter-individual variability, we propose that in silico computer modeling may help to better define the risks of new drug candidates at early stages of pre-clinical development.


Subject(s)
Action Potentials , Arrhythmias, Cardiac , Computer Simulation , ERG1 Potassium Channel/antagonists & inhibitors , Models, Cardiovascular , Potassium Channel Blockers/pharmacology , Action Potentials/drug effects , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/physiopathology , Endocardium/metabolism , Humans , Purkinje Cells/metabolism
12.
J Feline Med Surg ; 15(12): 1104-13, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23813147

ABSTRACT

This study compared indirect blood pressure measurements using a non-invasive method, high-definition oscillometry (HDO), with direct measurements using a radio-telemetry device in awake cats. Paired measurements partitioned to five sub-ranges were collected in six cats using both methods. The results were analysed for assessment of correlation and agreement between the two methods, taking into account all pressure ranges, and with data separated in three sub-groups, low, normal and high ranges of systolic (SBP) and diastolic (DBP) blood pressure. SBP data displayed a mean correlation coefficient of 0.92 ± 0.02 that was reduced for low SBP. The agreement level evaluated from the whole data set was high and slightly reduced for low SBP values. The mean correlation coefficient of DBP was lower than for SBP (ie, 0.81 ± 0.02). The bias for DBP between the two methods was 22.3 ± 1.6 mmHg, suggesting that HDO produced lower values than telemetry. These results suggest that HDO met the validation criteria defined by the American College of Veterinary Internal Medicine consensus panel and provided a faithful measurement of SBP in conscious cats. For DBP, results suggest that HDO tended to underestimate DBP. This finding is clearly inconsistent with the good agreement reported in dogs, but is similar to outcomes achieved in marmosets and cynomolgus monkeys, suggesting that this is not related to HDO but is species related. The data support that the HDO is the first and only validated non-invasive blood pressure device and, as such, it is the only non-invasive reference technique that should be used in future validation studies.


Subject(s)
Blood Pressure Monitoring, Ambulatory/veterinary , Blood Pressure Monitors/veterinary , Cats/physiology , Telemetry/veterinary , Animals , Blood Pressure Monitoring, Ambulatory/instrumentation , Blood Pressure Monitoring, Ambulatory/methods , Heart Rate , Telemetry/instrumentation , Telemetry/methods
13.
J Pharmacol Toxicol Methods ; 68(1): 166-74, 2013.
Article in English | MEDLINE | ID: mdl-23435316

ABSTRACT

INTRODUCTION: Power spectral analysis of heart rate variability is a tool known to provide information of interest on the autonomic control of heart rate in human. However, its use and its conditions of application and interpretation for safety purposes are not well defined for cardiovascular safety pharmacology studies. Likewise, data of power spectral analysis of heart rate variability in cynomolgus monkeys, a species often appropriate for use as second non rodent species in preclinical safety programmes, are not available. This study was designed to evaluate the relevance of this biomarker in this non human primate species, and to compare results with those from beagle dogs under the conditions of safety evaluation studies. METHODS: Power spectral analysis of heart rate variability was performed on data collected in both species by telemetry following a standard design for cardiovascular safety pharmacology studies. Various pharmacological agents were tested in order to compare the profile of responses in both species after modifying the autonomic nervous balance. RESULTS: Heart rate variability in cynomolgus monkeys is mainly driven by the parasympathetic nervous system as in beagle dogs although vagal tone is less than in dogs. Power spectral analysis of heart rate variability allows detection of interaction with the autonomic nervous system in both species in all investigated situations, i.e. sympatholytic/sympathomimetic and parasympatholytic/parasympathomimetic drug induced effects. However, due to species difference in the autonomic control of heart rate, cynomolgus monkeys are likely to be more sensitive than beagle dogs for assessment of sympatholytic properties. DISCUSSION: This study confirms that power spectral analysis of heart rate variability from data derived from ECG recordings in telemetry studies is applicable in cardiovascular safety pharmacology studies and may provide relevant information about possible interaction with the autonomic nervous system when new drug entities are evaluated in either species. However, interspecies differences in autonomic control must be taken into account when interpreting possible drug effects.


Subject(s)
Autonomic Nervous System/drug effects , Heart Rate/drug effects , Telemetry/methods , Toxicity Tests/methods , Animals , Autonomic Nervous System/metabolism , Dogs , Electrocardiography , Female , Macaca fascicularis , Male , Models, Animal , Species Specificity
15.
PLoS One ; 7(12): e52653, 2012.
Article in English | MEDLINE | ID: mdl-23300736

ABSTRACT

Pancreatic tumors are the gastrointestinal cancer with the worst prognosis in humans and with a survival rate of 5% at 5 years. Nowadays, no chemotherapy has demonstrated efficacy in terms of survival for this cancer. Previous study focused on the development of a new therapy by non thermal plasma showed significant effects on tumor growth for colorectal carcinoma and glioblastoma. To allow targeted treatment, a fibered plasma (Plasma Gun) was developed and its evaluation was performed on an orthotopic mouse model of human pancreatic carcinoma using a MIA PaCa2-luc bioluminescent cell line. The aim of this study was to characterize this pancreatic carcinoma model and to determine the effects of Plasma Gun alone or in combination with gemcitabine. During a 36 days period, quantitative BLI could be used to follow the tumor progression and we demonstrated that plasma gun induced an inhibition of MIA PaCa2-luc cells proliferation in vitro and in vivo and that this effect could be improved by association with gemcitabine possibly thanks to its radiosensitizing properties.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/therapy , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/therapy , Plasma Gases/therapeutic use , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Tracking , Combined Modality Therapy , Deoxycytidine/pharmacology , Disease Models, Animal , Female , Humans , Luciferases, Firefly/biosynthesis , Mice , Pancreatic Neoplasms/pathology , Tumor Burden , Whole Body Imaging , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
16.
Carcinogenesis ; 33(2): 260-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22114075

ABSTRACT

The aim of this study was to determine how n-3 polyunsaturated fatty acid (PUFAs) counteracted tumor chemoresistance by restoring a functional vascularization. Rats with chemically induced mammary tumors were divided into two nutritional groups: a control group and a group fed with an n-3 PUFA-enriched diet. Both groups were treated with docetaxel. Functional vascular parameters (ultrasounds, interstitial fluid pressure) were determined for both nutritional groups before (W(0)) and during docetaxel treatment [every 2 h up to 1 week (W(+1)) for interstitial fluid pressure, at W(+1) for Evans blue extravasation and at W(+2) and W(+6) for ultrasounds]. In vitro n-3 PUFA-induced changes in endothelial cell migration, permeability and phosphorylation of endothelial nitric oxide synthase were evaluated using human umbilical vein endothelial cells. Whereas docetaxel stabilized tumor growth in the rat control group, it induced a 50% tumor regression in the n-3 PUFA group. Ultrasounds parameters were consistently lower in the n-3 PUFA group at all time points measured, down to ∼50% at W(+6). A single dose of docetaxel in the n-3 PUFA group markedly reduced interstitial fluid pressure from 2 h after injection up to W(+1) when Evans blue extravasation was increased by 3-fold. A decreased activation of endothelial nitric oxide synthase in tumors of the n-3 PUFA group, and in human umbilical vein endothelial cell cultured with n-3 PUFA, points toward a PUFA-induced disruption of nitric oxide signaling pathway. This normalization of tumor vasculature functions under n-3 PUFA diet indicates that such a supplementation, by improving drug delivery in mammary tumors, could be a complementary clinical strategy to decrease anticancer drug resistance.


Subject(s)
Endothelial Cells/drug effects , Extracellular Fluid/drug effects , Fatty Acids, Omega-3/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Coloring Agents , Disease Progression , Docetaxel , Docosahexaenoic Acids/metabolism , Drug Resistance, Neoplasm , Eicosapentaenoic Acid/metabolism , Endothelial Cells/metabolism , Evans Blue , Extracellular Fluid/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mammary Neoplasms, Animal/metabolism , Neoplasm Metastasis/pathology , Neovascularization, Physiologic/drug effects , Permeability/drug effects , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Taxoids/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
17.
Pharmacol Ther ; 132(2): 125-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21420430

ABSTRACT

Common arrhythmias, particularly atrial fibrillation (AF) and ventricular tachycardia/fibrillation (VT/VF) are a major public health concern. Classic antiarrhythmic (AA) drugs for AF are of limited effectiveness, and pose the risk of life-threatening VT/VF. For VT/VF, implantable cardiac defibrillators appear to be the unique, yet unsatisfactory, solution. Very few AA drugs have been successful in the last few decades, due to safety concerns or limited benefits in comparison to existing therapy. The Vaughan-Williams classification (one drug for one molecular target) appears too restrictive in light of current knowledge of molecular and cellular mechanisms. New AA drugs such as atrial-specific and/or multichannel blockers, upstream therapy and anti-remodeling drugs, are emerging. We focus on the cellular mechanisms related to abnormal Na⁺ and Ca²âº handling in AF, heart failure, and inherited arrhythmias, and on novel strategies aimed at normalizing ionic homeostasis. Drugs that prevent excessive Na⁺ entry (ranolazine) and aberrant diastolic Ca²âº release via the ryanodine receptor RyR2 (rycals, dantrolene, and flecainide) exhibit very interesting antiarrhythmic properties. These drugs act by normalizing, rather than blocking, channel activity. Ranolazine preferentially blocks abnormal persistent (vs. normal peak) Na⁺ currents, with minimal effects on normal channel function (cell excitability, and conduction). A similar "normalization" concept also applies to RyR2 stabilizers, which only prevent aberrant opening and diastolic Ca²âº leakage in diseased tissues, with no effect on normal function during systole. The different mechanisms of action of AA drugs may increase the therapeutic options available for the safe treatment of arrhythmias in a wide variety of pathophysiological situations.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Heart Failure/drug therapy , Animals , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/metabolism , Heart Failure/metabolism , Humans , Ion Channels/drug effects , Ion Channels/metabolism
18.
J Pharmacol Toxicol Methods ; 63(3): 269-78, 2011.
Article in English | MEDLINE | ID: mdl-21224008

ABSTRACT

INTRODUCTION: In a previous study, two electrophysiological patterns for torsadogenic drugs were characterised in the model of isolated canine Purkinje fibres from their respective effects on action potential. This study was designed to elucidate the possible mechanisms underlying these two electrophysiological profiles. METHODS: Effects of representative torsadogenic agents and non torsadogenic drugs on I(Kr), I(Ks), I(K1), I(Na) and I(CaL) were studied in transfected HEK 293 cells using the path-clamp method as well as in conscious beagle dogs and cynomolgus monkeys by telemetry. RESULTS: Patch-clamp studies confirmed that torsadogenic molecules could be discriminated into at least two subgroups. The first subgroup can be defined as apparently pure I(Kr) blockers. The second subgroup can be defined as I(Kr) blockers with ancillary properties on sodium and/or calcium channels which counterbalance the I(Kr) prolongation component. This discrimination is transposable to the telemetered cynomolgus monkey model in terms of QT prolongation but not to the telemetered beagle dog model. This latter inter-species difference could be related to the sympathetic/parasympathetic balance and could involve reserve repolarisation dependent mechanisms. DISCUSSION: The confirmation that torsadogenic drugs might have at least two different electrophysiological profiles should be taken into consideration in preclinical safety pharmacology studies because it increases the value of the cynomolgus monkey model in two particular situations: firstly when an NCE causes sympathetic activation and secondly, when an NCE exhibits a pure I(Kr) blocker pattern independently of its potency to block HERG channels.


Subject(s)
Action Potentials/drug effects , Calcium Channel Blockers/pharmacology , Potassium Channel Blockers/pharmacology , Purkinje Fibers/drug effects , Sodium Channel Blockers/pharmacology , Torsades de Pointes/chemically induced , Action Potentials/physiology , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Channels/physiology , Cell Line , Dogs , Electrocardiography , Female , Humans , Macaca fascicularis , Male , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels/physiology , Purkinje Fibers/physiology , Sodium Channels/genetics , Sodium Channels/metabolism , Sodium Channels/physiology , Telemetry , Torsades de Pointes/metabolism , Torsades de Pointes/physiopathology , Transfection
19.
J Pharmacol Toxicol Methods ; 61(3): 251-63, 2010.
Article in English | MEDLINE | ID: mdl-20172034

ABSTRACT

INTRODUCTION: QT interval assessment by telemetry has become one of the most useful models in testing strategies adopted for detection of drug induced QT prolongation in non-clinical safety pharmacology studies. This study reports experimental data showing that the autonomic nervous system might influence drug induced QT prolongation. METHODS: Animals were instrumented with telemetric transmitters and epicardial ECG leads. Effects on QT interval of reference drugs such as thioridazine and terfenadine were analysed with different approaches, the Holzgrefe's probabilistic method, the QT shift method and an individual analysis of beat-to-beat QT/RR pair distribution visualised as points-cloud. RESULTS: Two cases of unexpected absence of QT interval prolongation are reported with thioridazine and terfenadine in conscious beagle dogs under conditions of concomitant tachycardia. The pro-arrhythmic properties of these two molecules were unmasked by co-treatment with sympatholytic agents, atenolol and clonidine respectively suggesting that sympathetic activation and/or parasympathetic withdrawal might impair a drug induced QT prolongation. DISCUSSION: The apparent absence of changes in the QT interval due to novel drug candidates should be interpreted cautiously under conditions of concomitant tachycardia or elevated heart rate levels in non-clinical safety studies.


Subject(s)
Autonomic Nervous System/physiopathology , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Animals , Autonomic Nervous System/drug effects , Dogs , Electrocardiography/drug effects , Female , Long QT Syndrome/diagnosis , Male , Retrospective Studies , Terfenadine/pharmacology , Terfenadine/toxicity , Thioridazine/pharmacology , Thioridazine/toxicity
20.
J Pharmacol Toxicol Methods ; 59(2): 73-85, 2009.
Article in English | MEDLINE | ID: mdl-19135537

ABSTRACT

INTRODUCTION: Drug-induced QT interval prolongation is a major concern in new drug candidate development. This study presents a method of assessment of drug-induced QT interval prolongation without need for QT correction in conscious Beagle dogs and Cynomolgus monkeys monitored by telemetry. Accuracy and reliability are analysed by comparison with a reference QT correction method (Holzgrefe) from experiments performed with reference substances terfenadine, thioridazine and sotalol. METHODS: The QT shift method principle is assessment of any drug-induced QT interval shift directly from the individual QT/RR relationship. The individual QT/RR relationship is built from a treatment-free 24-hour recording period. QT and RR intervals are determined from a beat-to-beat analysis. A probabilistic method is used to define the individual QT/RR relationships. Checks were performed to compare results obtained with the QT shift method and the QT correction methods. The robustness of the QT shift method was tested under various conditions of drug-induced heart rate change (i.e. normal, bradycardia and tachycardia). RESULTS: The extent of agreement with the used reference QT correction method, Holzgrefe formula, was excellent (3-4 ms) in both animal species under the various drug induced effects on heart rate. The statistical sensitivity threshold for detection of QT prolongation according to a standard safety pharmacology study design was 7-8 ms. DISCUSSION: When combined with the probabilistic determination of individual QT/RR relationships, this simple method provides a direct assessment of a drug-induced effect on QT interval, without any curve fitting or application of correction formula. Despite noticeably different shapes in QT/RR relationships, the QT shift method is applicable to both Beagle dogs and Cynomolgus monkeys. It is likely that the QT shift method will be particularly helpful in problematic cases, enabling detection of drug-induced prolongation of less than 10 ms.


Subject(s)
Drug Evaluation, Preclinical/methods , Electrocardiography/instrumentation , Electrocardiography/standards , Heart Rate/physiology , Long QT Syndrome/chemically induced , Animals , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/pharmacology , Dogs , Dopamine Antagonists/adverse effects , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Electrocardiography/drug effects , Female , Heart Rate/drug effects , Histamine H1 Antagonists, Non-Sedating/adverse effects , Histamine H1 Antagonists, Non-Sedating/pharmacology , Macaca fascicularis , Male , Models, Statistical , Phenethylamines/adverse effects , Phenethylamines/pharmacology , Reference Standards , Sensitivity and Specificity , Sotalol/adverse effects , Sotalol/pharmacology , Sulfonamides/adverse effects , Sulfonamides/pharmacology , Telemetry , Terfenadine/adverse effects , Terfenadine/pharmacology , Thioridazine/adverse effects , Thioridazine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL