Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Regul Toxicol Pharmacol ; 113: 104624, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32126256

ABSTRACT

An international expert working group representing 37 organisations (pharmaceutical/biotechnology companies, contract research organisations, academic institutions and regulatory bodies) collaborated in a data sharing exercise to evaluate the utility of two species within regulatory general toxicology studies. Anonymised data on 172 drug candidates (92 small molecules, 46 monoclonal antibodies, 15 recombinant proteins, 13 synthetic peptides and 6 antibody-drug conjugates) were submitted by 18 organisations. The use of one or two species across molecule types, the frequency for reduction to a single species within the package of general toxicology studies, and a comparison of target organ toxicities identified in each species in both short and longer-term studies were determined. Reduction to a single species for longer-term toxicity studies, as used for the development of biologicals (ICHS6(R1) guideline) was only applied for 8/133 drug candidates, but might have been possible for more, regardless of drug modality, as similar target organ toxicity profiles were identified in the short-term studies. However, definition and harmonisation around the criteria for similarity of toxicity profiles is needed to enable wider consideration of these principles. Analysis of a more robust dataset would be required to provide clear, evidence-based recommendations for expansion of these principles to small molecules or other modalities where two species toxicity testing is currently recommended.


Subject(s)
Drug Development , Drug Evaluation, Preclinical/adverse effects , Toxicity Tests , Animals , Databases, Factual , Humans , Risk Assessment
2.
Lab Chip ; 20(3): 468-476, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31989145

ABSTRACT

The human kidney contains approximately one million nephrons. As the functional unit of the kidney, the nephron affords an opportunity to approximate the kidney at a microphysiological scale. Recent emergence of physiologically accurate human tissue models has radically advanced the possibilities of mimicking organ biology and multi-organ combinations in vitro. Anatomically, the nephron is one of the most complex, sequentially integrated microfluidic units in the body making the miniaturized microfluidic systems excellent candidates for capturing the kidney biology in vitro. While these models are promising, there are a number of considerations for practical implementation into a drug development paradigm. Opportunities for pharmaceutical industry applications of new MPS models often start with drug safety testing. As such, the intent of this article is to focus on safety and ADME applications. This article reviews biological functions of the kidney and options for characterizing known roles in nephrotoxicity. The concept of "context-of-use" is introduced as a framework for describing and verifying the specific features of an MPS platform for use in drug development. Overall, we present a perspective on key attributes of microphysiological kidney models, which the pharmaceutical industry could leverage to improve confident safety and ADME evaluations of experimental therapies.


Subject(s)
Kidney/drug effects , Pharmaceutical Preparations/metabolism , Drug Development , Drug Evaluation, Preclinical/adverse effects , Drug Industry , Humans , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Models, Biological , Pharmaceutical Preparations/chemistry
3.
Pharmacol Res Perspect ; 7(6): e00547, 2019 12.
Article in English | MEDLINE | ID: mdl-31832205

ABSTRACT

Activation of MrgX2, an orphan G protein-coupled receptor expressed on mast cells, leads to degranulation and histamine release. Human MrgX2 binds promiscuously to structurally diverse peptides and small molecules that tend to have basic properties (basic secretagogues), resulting in acute histamine-like adverse drug reactions of injected therapeutic agents. We set out to identify MrgX2 orthologues from other mammalian species used in nonclinical stages of drug development. Previously, the only known orthologue of human MrgX2 was from mouse, encoded by Mrgprb2. MrgX2 genes of rat, dog (beagle), minipig, pig, and Rhesus and cynomolgus monkey were identified by bioinformatic approaches and verified by their ability to mediate calcium mobilization in transfected cells in response to the classical MrgX2 agonist, compound 48/80. The peptide GSK3212448 is an inhibitor of the PRC2 epigenetic regulator that caused profound anaphylactoid reactions upon intravenous infusion to rat. We showed GSK3212448 to be a potent MrgX2 agonist particularly at rat MrgX2. We screened sets of drug-like molecules and peptides to confirm the highly promiscuous nature of MrgX2. Approximately 20% of drug-like molecules activated MrgX2 (pEC50 ranging from 4.5 to 6), with the principle determinant being basicity. All peptides tested of net charge +3 or greater exhibited agonist activity, including the cell penetrating peptides polyarginine (acetyl-Arg9-amide) and TAT (49-60), a fragment of HIV-1 TAT protein. Finally, we showed that the glycopeptide antibiotic vancomycin, which is associated with clinical pseudo-allergic reactions known as red man syndrome, is an agonist of MrgX2.


Subject(s)
Anaphylaxis/chemically induced , Mast Cells/drug effects , Nerve Tissue Proteins/agonists , Peptide Fragments/adverse effects , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide/agonists , Vancomycin/adverse effects , Anaphylaxis/immunology , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/adverse effects , Disease Models, Animal , Drug Evaluation, Preclinical/adverse effects , HEK293 Cells , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Mast Cells/immunology , Mast Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Peptide Fragments/administration & dosage , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Syndrome , Vancomycin/administration & dosage , p-Methoxy-N-methylphenethylamine/pharmacology
4.
Regul Toxicol Pharmacol ; 96: 18-29, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29679677

ABSTRACT

Toxicogenomics held great promise as an approach to enable early detection of toxicities induced by xenobiotics; however, there remain questions regarding the impact of the discipline on pharmaceutical nonclinical safety assessment. To understand the current state of toxicogenomics in the sector, an industry group surveyed companies to determine the frequency of toxicogenomics use in in vivo studies at various stages of drug discovery and development and to assess how toxicogenomics use has evolved over time. Survey data were compiled during 2016 from thirteen pharmaceutical companies. Toxicogenomic analyses were infrequently conducted in the development phase and when performed were done to address specific mechanistic questions. Prior to development, toxicogenomics use was more frequent; however, there were significant differences in approaches among companies. Across all phases, gaining mechanistic insight was the most frequent reason cited for pursing toxicogenomics with few companies using toxicogenomics to predict toxicities. These data were consistent with the commentary submitted in response to survey questions asking companies to describe the evolution of their toxicogenomics strategy. Overall, these survey data indicate that toxicogenomics is not widely used as a predictive tool in the pharmaceutical industry but is used regularly by some companies and serves a broader role in mechanistic investigations and as a complement to other technologies.


Subject(s)
Drug Evaluation, Preclinical/adverse effects , Drug Industry , Toxicogenetics , Animals , Humans
5.
Methods Mol Biol ; 1641: 213-228, 2017.
Article in English | MEDLINE | ID: mdl-28748467

ABSTRACT

An in-depth evaluation of target safety is an invaluable resource throughout drug discovery and development. The goal of a target safety evaluation is to identify potential unintended adverse consequences of target modulation, and to propose a risk evaluation and mitigation strategy to shepherd compounds through the discovery and development pipeline, to confirm and characterize unavoidable on-target toxicities in a timely manner to assist in early program advancement decisions, and to anticipate, monitor, and manage potential clinical adverse events. The role of an experienced discovery toxicologist in synthesizing the available information into an actionable set of recommendations for a safety evaluation strategy is critical to its successful application in early discovery programs. This chapter presents a summary of some of the information types and sources that should be investigated, and approaches that can be taken to generate an early assessment of potential safety liabilities.


Subject(s)
Drug Evaluation, Preclinical/adverse effects , Drug Discovery , Drug-Related Side Effects and Adverse Reactions , Humans
6.
Regul Toxicol Pharmacol ; 87 Suppl 3: S1-S15, 2017 Jul 31.
Article in English | MEDLINE | ID: mdl-28483710

ABSTRACT

The transition from nonclinical to First-in-Human (FIH) testing is one of the most challenging steps in drug development. In response to serious outcomes in a recent Phase 1 trial (sponsored by Bial), IQ Consortium/DruSafe member companies reviewed their nonclinical approach to progress small molecules safely to FIH trials. As a common practice, safety evaluation begins with target selection and continues through iterative in silico and in vitro screening to identify molecules with increased probability of acceptable in vivo safety profiles. High attrition routinely occurs during this phase. In vivo exploratory and pivotal FIH-enabling toxicity studies are then conducted to identify molecules with a favorable benefit-risk profile for humans. The recent serious incident has reemphasized the importance of nonclinical testing plans that are customized to the target, the molecule, and the intended clinical plan. Despite the challenges and inherent risks of transitioning from nonclinical to clinical testing, Phase 1 studies have a remarkably good safety record. Given the rapid scientific evolution of safety evaluation, testing paradigms and regulatory guidance must evolve with emerging science. The authors posit that the practices described herein, together with science-based risk assessment and management, support safe FIH trials while advancing development of important new medicines.


Subject(s)
Clinical Trials, Phase I as Topic , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/adverse effects , Humans , Risk Assessment/methods , Safety
7.
Stud Hist Philos Biol Biomed Sci ; 61: 35-45, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28039775

ABSTRACT

According to the received view, the transportation view, animal extrapolation consists in inductive prediction of the outcome of a mechanism in a target, based on an analogical mechanism in a model. Through an analysis of the failure of preclinical studies of TGN1412, an innovative drug, to predict the tragic consequences of its first-in-man trial in 2006, the received view is challenged by a proposed view of animal extrapolation, the chimera view. According to this view, animal extrapolation is based on a hypothesis about how human organisms work, supported by the amalgamation of results drawn from various experimental organisms, and only predicting the 'predictive grid', that is, a global framework of the effects to be expected.


Subject(s)
Animal Experimentation , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Drug Evaluation, Preclinical , Animals , Drug Evaluation, Preclinical/adverse effects , Humans
8.
Curr Top Med Chem ; 15(18): 1827-36, 2015.
Article in English | MEDLINE | ID: mdl-26002591

ABSTRACT

The increasing use of Nanoparticles (NPs) in a wide range of applications has led to a rising concern on the possible toxicological effects that this use may have on human health and the environment. Since experimental toxicity evaluation for the different types of NPs already available, is often expensive and time consuming, several computational approaches are proposed for the risk assessment of NPs. In this work, we have developed a predictive classification model for the toxicological assessment of iron oxide NPs with different core, coating and surface modification based on a number of different properties including size, relaxivities, zeta potential and type of coating. The model was fully validated based on several validation measurements and was released online via Enalos InSilicoNano Platform (http://enalos.insilicotox.com/QNAR_IronOxide_Toxicity/). The developed web service gives the interested user the opportunity to insert the indicated properties and get a toxicity prediction accompanied by an indication of its reliability based on the domain of applicability. This newly introduced web service complements our previously reported efforts to extract important information from available datasets and develop user friendly applications for the toxicity assessment of NPs.


Subject(s)
Drug Evaluation, Preclinical/adverse effects , Metals/adverse effects , Nanoparticles/adverse effects , Nanotechnology , Oxides/adverse effects , Humans , Quantitative Structure-Activity Relationship , Risk Assessment
9.
J Pharmacol Toxicol Methods ; 70(3): 246-54, 2014.
Article in English | MEDLINE | ID: mdl-25087753

ABSTRACT

INTRODUCTION: Detection of drug-induced pro-arrhythmic risk is a primary concern for pharmaceutical companies and regulators. Increased risk is linked to prolongation of the QT interval on the body surface ECG. Recent studies have shown that multiple ion channel interactions can be required to predict changes in ventricular repolarisation and therefore QT intervals. In this study we attempt to predict the result of the human clinical Thorough QT (TQT) study, using multiple ion channel screening which is available early in drug development. METHODS: Ion current reduction was measured, in the presence of marketed drugs which have had a TQT study, for channels encoded by hERG, CaV1.2, NaV1.5, KCNQ1/MinK, and Kv4.3/KChIP2.2. The screen was performed on two platforms - IonWorks Quattro (all 5 channels, 34 compounds), and IonWorks Barracuda (hERG & CaV1.2, 26 compounds). Concentration-effect curves were fitted to the resulting data, and used to calculate a percentage reduction in each current at a given concentration. Action potential simulations were then performed using the ten Tusscher and Panfilov (2006), Grandi et al. (2010) and O'Hara et al. (2011) human ventricular action potential models, pacing at 1Hz and running to steady state, for a range of concentrations. RESULTS: We compared simulated action potential duration predictions with the QT prolongation observed in the TQT studies. At the estimated concentrations, simulations tended to underestimate any observed QT prolongation. When considering a wider range of concentrations, and conventional patch clamp rather than screening data for hERG, prolongation of ≥5ms was predicted with up to 79% sensitivity and 100% specificity. DISCUSSION: This study provides a proof-of-principle for the prediction of human TQT study results using data available early in drug development. We highlight a number of areas that need refinement to improve the method's predictive power, but the results suggest that such approaches will provide a useful tool in cardiac safety assessment.


Subject(s)
Action Potentials/drug effects , Drug Evaluation, Preclinical/adverse effects , Electrocardiography/drug effects , Ion Channels/metabolism , Long QT Syndrome/chemically induced , Dose-Response Relationship, Drug , Humans
10.
Toxicol Sci ; 123(1): 281-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21693436

ABSTRACT

Improved in vitro systems for predicting drug-induced toxicity are needed in the pharmaceutical and biotechnology industries to decrease late-stage drug attrition. One unmet need is an early screen for cardiotoxicity, which accounts for about one third of safety-based withdrawn pharmaceuticals. Herein, the first published report of a high-throughput functional assay employing a monolayer of beating human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is described, detailing a model that accurately detects drug-induced cardiac abnormalities. Using 96-well plates with interdigitated electrode arrays that assess impedance, the rhythmic, synchronous contractions of the iPSC-CMs were detected. Treatment of the iPSC-CMs with 28 different compounds with known cardiac effects resulted in compound-specific changes in the beat rate and/or the amplitude of the impedance measurement. Changes in impedance for the compounds tested were comparable with the results from a related technology, electric field potential assessment obtained from microelectrode arrays. Using the results from the set of compounds, an index of drug-induced arrhythmias was calculated, enabling the determination of a drug's proarrhythmic potential. This system of interrogating human cardiac function in vitro opens new opportunities for predicting cardiac toxicity and studying cardiac biology.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Drug Evaluation, Preclinical/adverse effects , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Drug Discovery/methods , Humans , Pharmaceutical Preparations
12.
Bioorg Med Chem Lett ; 19(8): 2224-9, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19289280

ABSTRACT

This paper documents a serious problem met during the testing of Gi protein-activating properties of a new series of synthetic compounds by measuring the induced binding of [(35)S]GTPgammaS to different subtypes of Gi protein. The problem arose from the strong affinity between [(35)S]GTPgammaS and the tested compounds, that are characterized by several (2-4) positive charges and high lipophilicity. Apparently, such affinity yields insoluble, labelled complexes that, also in the absence of Gi protein, are retained on the filters and give rise to false positive results.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Surface-Active Agents/metabolism , Surface-Active Agents/pharmacology , Drug Evaluation, Preclinical/adverse effects , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Hydrophobic and Hydrophilic Interactions , Protein Binding/drug effects , Protein Binding/physiology , Solubility , Sulfur Radioisotopes/metabolism , Surface Tension/drug effects , Surface-Active Agents/chemistry
14.
Br J Clin Pharmacol ; 67(2): 153-60, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19076987

ABSTRACT

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Recent regulatory guidance has highlighted the importance of using pharmacokinetic-pharmacodynamic (PK-PD) modelling in the selection of starting doses in first-in-human trials of high-risk biologics. However, limited examples exist in literature illustrating this procedure. WHAT THIS STUDY ADDS: An interpretation of the recommended dose-selection methodology and the minimum anticipated biological effect level (MABEL) principle, contained in the updated European Medicines Agency guidance on risk-mitigation strategies for first-in-human studies, is presented. Some literature and simulation-based examples of the application of PK-PD modelling principles to starting dose selection using in vitro and in vivo data under the MABEL paradigm are highlighted, along with the advantages and limitations of this approach. AIMS: To illustrate the use of pharmacokinetic-pharmacodynamic (PK-PD) models to select rational starting doses in clinical trials within the minimum anticipated biological effect level (MABEL) principle using literature data and through simulations. METHODS: The new European Medicines Agency guidance on starting dose selection of high-risk biologics was analysed considering the basic pharmacological properties and preclinical testing limitations of many biologics. The MABEL approach to dose selection was illustrated through simulations and through literature-reported examples on the selection of starting doses for biologics such as antibodies based on in vitro biomarker data, in vivo PK and PK-PD data. RESULTS: Literature reports indicating the use of preclinical pharmacological and toxicological data to select successfully safe starting doses in line with the MABEL principle are summarized. PK-PD model-based simulations of receptor occupancy for an anti-IgE antibody system indicate that the relative abundance of IgE in animal models and patients and the turnover rate of the IgE-antibody complex relative to the off-rate of the antibody from IgE are important determinants of in vivo receptor occupancy. CONCLUSIONS: Mechanistic PK-PD models are capable of integrating preclinical in vitro and in vivo data to select starting doses rationally in first-in-human trials. Biological drug-receptor interaction dynamics is complex and multiple factors affect the dose-receptor occupancy relationship. Thus, these factors should be taken into account when selecting starting doses.


Subject(s)
Antibodies, Anti-Idiotypic/metabolism , Biomarkers, Pharmacological , Pharmaceutical Preparations/metabolism , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/adverse effects , Drug-Related Side Effects and Adverse Reactions , Humans , Models, Animal , Models, Biological
15.
Aten. prim. (Barc., Ed. impr.) ; 40(11): 555-558, nov. 2008. tab
Article in Es | IBECS | ID: ibc-69729

ABSTRACT

Objetivo. Evaluar el impacto de lasrecomendaciones que el Comité Técnico delSistema Español de Farmacovigilancia envióa los editores de revistas médicas españolasen el año 2004 sobre la información mínimaque deberían contener los casos publicadosde reacciones adversas a medicamentos.Diseño. Estudio observacional.Emplazamiento. Base de datos nacional desospechas de reacciones adversas (FEDRA).Participantes. Notificaciones de publicacionesde sospechas de reacciones adversasregistradas en FEDRA y publicadas enlos años previo (2003) y posterior (2005)al envío de las recomendaciones.Mediciones principales. Información en losapartados: sexo, edad, dosis, motivo de laprescripción, duraciones del tratamiento y dela reacción, secuencia temporal, efecto de laretirada y causas alternativas de laspublicaciones de sospechas de reaccionesadversas registradas en FEDRA. Secompararon los resultados de los 2 años.Resultados. No se observaron diferenciassignificativas en la información contenidaen las publicaciones de casos de reaccionesadversas entre los años 2003 y 2005.Los apartados que con mayor frecuenciapresentaban información incompleta eranlas dosis y duraciones de los tratamientosy de la reacción adversa. Sólo un tercio,aproximadamente, de las publicacionescontenían información completa.Conclusiones. Es necesario mejorarel contenido de la información de laspublicaciones de casos de reacciones adversasa medicamentos para que esta mejordocumentación contribuya a mejorarla evaluación de las señales de alerta


Objective. To assess the impact of therecommendations that, the TechnicalCommittee of the SpanishPharmacovigilance System sent, in 2004,to the editors of Spanish medical journalson the minimum information required forpublication of adverse drug reaction casereports.Design. Observational study.Setting. National suspected adverse drugreactions database (FEDRA).Participants. Published adverse drug reactionreports registered in FEDRA, publishedin the years before (2003) and after (2005)the recommendations were issued.Main measurements. The following dataelements were analysed: sex, age, dose,disease treated with the suspecteddrug, length of treatment and adverse drugreaction, temporal sequence, withdrawaleffect, and alternative causes. The resultsof the 2 years were compared.Results. The information in the case reportspublished between years 2003 and 2005was not significantly different. The dataelements more often incomplete were dose,length of treatment, as well as length ofadverse reaction. Approximately one thirdof the published case reports included fullinformation.Conclusions. There seems to be a needto improve the data elements contentof published adverse drug reactions casereports, so that such documentation can contribute to improve the assessment of alert signals (AU)


Subject(s)
Drug and Narcotic Control/economics , Drug and Narcotic Control/methods , Drug Compounding/adverse effects , Signs and Symptoms , Health Surveillance/methods , Epidemiological Monitoring , Drug Evaluation/adverse effects , Drug Evaluation, Preclinical/adverse effects , Quality of Homeopathic Remedies , Health Status Indicators , Economics, Pharmaceutical/legislation & jurisprudence , Economics, Pharmaceutical/organization & administration , Economics, Pharmaceutical/standards
18.
In Vivo ; 19(1): 103-7, 2005.
Article in English | MEDLINE | ID: mdl-15796161

ABSTRACT

BACKGROUND: The detection of molecular targets for flavonoids in cell signalling has opened new perspectives for their application in medicine. Both tangeretin, a citrus methoxyflavone, and xanthohumol, the main prenylated chalcone present in hops (Humulus lupulus L.), act on the mitogen-activated protein kinase pathway and await further investigation for administration in vivo. MATERIALS AND METHODS: A safety study was designed in laboratory mice orally administered concentrates of purified tangeretin (1 x 10(-4) M) or xanthohumol (5 x 10(-4) M) at libitum for 4 weeks. Blood samples were collected for the analysis of a variety of haematological and biochemical parameters. RESULTS: A reduction of the circulating lymphocyte number was noticed for tangeretin, while all other parameters were unaffected by treatment with either tangeretin or xanthohumol. The parameters encompassed an integrity check of the following tissues and organs: bone marrow, liver, exocrine pancreas, kidneys, muscles, thyroid, ovaries and surrenal cortex. Furthermore, no differences were noted in the metabolism of proteins, lipids, carbohydrates and uric acid, as well as in ion concentrations. CONCLUSION: All data indicate that oral administration of tangeretin or xanthohumol to laboratory mice does not affect major organ functions and opens the gate for further safety studies in humans.


Subject(s)
Animals, Laboratory , Drug Evaluation, Preclinical/adverse effects , Flavones/adverse effects , Plant Extracts/adverse effects , Propiophenones/adverse effects , Administration, Oral , Animals , Body Weight , Female , Flavones/administration & dosage , Flavones/pharmacology , Flavonoids , Mice , Mice, Inbred C3H , Mice, Nude , Organ Size , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Propiophenones/administration & dosage , Propiophenones/pharmacology
20.
J Pharmacol Toxicol Methods ; 49(3): 145-51, 2004.
Article in English | MEDLINE | ID: mdl-15172010

ABSTRACT

The origins of safety pharmacology are grounded upon observations that organ functions (like organ structures) can be toxicological targets in humans exposed to novel therapeutic agents, and that drug effects on organ functions (unlike organ structures) are not readily detected by standard toxicological testing. Safety pharmacology is " em leader those studies that investigate the potential undesirable pharmacodynamic effects of a substance on physiological functions in relationship to exposure in the therapeutic range and above em leader " [International Conference on Harmonization (ICH) S7A guidelines; Safety Pharmacology Studies for Human Pharmaceuticals]. This publication provides a comprehensive review of the history of safety pharmacology, international regulatory guidelines that govern the practices of this important field, and the scientific challenges that are being faced by its rapid emergence in pharmaceutical development. The criticality of identifying undesired adverse effects of new drugs in nonclinical models, which reflect the overall human condition, is reflected in the importance of generating an integrated and accurate assessment of possible human risk. The conundrum posed by the challenge of formulating a reliable risk assessment is the importance of improving and enhancing the safe progression of new drugs to the marketplace, while preventing unnecessary delays (or discontinuances), based on nonclinical findings that are not relevant or interpretable in terms of clinical response or human risk.


Subject(s)
Drug Evaluation, Preclinical/adverse effects , Drugs, Investigational/adverse effects , Pharmacology/methods , Animals , Clinical Trials as Topic , Forecasting , Guidelines as Topic , Humans , Pharmacology/standards , Pharmacology, Clinical , Risk Assessment , Safety , Toxicity Tests/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...