Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Med Chem ; 65(8): 6001-6016, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35239336

ABSTRACT

3,3-Disubstituted oxetanes have been utilized as bioisosteres for gem-dimethyl and cyclobutane functionalities. We report the discovery of a novel class of oxetane indole-amine 2,3-dioxygenase (IDO1) inhibitors suitable for Q3W (once every 3 weeks) oral and parenteral dosing. A diamide class of IDO inhibitors was discovered through an automated ligand identification system (ALIS). Installation of an oxetane and fluorophenyl dramatically improved the potency. Identification of a biaryl moiety as an unconventional amide isostere addressed the metabolic liability of amide hydrolysis. Metabolism identification (Met-ID)-guided target design and the introduction of polarity resulted in the discovery of potent IDO inhibitors with excellent pharmacokinetic (PK) profiles in multiple species. To enable rapid synthesis of the key oxetane intermediate, a novel oxetane ring cyclization was also developed, as well as optimization of a literature route on kg scale. These IDO inhibitors may enable unambiguous proof-of-concept testing for the IDO1 inhibition mechanism for oncology.


Subject(s)
Enzyme Inhibitors , Ethers, Cyclic , Amides , Cyclization , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
2.
J Med Chem ; 64(22): 16770-16800, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34704436

ABSTRACT

Proprotein convertase subtilisin-like/kexin type 9 (PCSK9) is a key regulator of plasma LDL-cholesterol (LDL-C) and a clinically validated target for the treatment of hypercholesterolemia and coronary artery disease. Starting from second-generation lead structures such as 2, we were able to refine these structures to obtain extremely potent bi- and tricyclic PCSK9 inhibitor peptides. Optimized molecules such as 44 demonstrated sufficient oral bioavailability to maintain therapeutic levels in rats and cynomolgus monkeys after dosing with an enabled formulation. We demonstrated target engagement and LDL lowering in cynomolgus monkeys essentially identical to those observed with the clinically approved, parenterally dosed antibodies. These molecules represent the first report of highly potent and orally bioavailable macrocyclic peptide PCSK9 inhibitors with overall profiles favorable for potential development as once-daily oral lipid-lowering agents. In this manuscript, we detail the design criteria and multiparameter optimization of this novel series of PCSK9 inhibitors.


Subject(s)
PCSK9 Inhibitors/pharmacology , Peptides, Cyclic/pharmacology , Administration, Oral , Animals , Biological Availability , Crystallography, X-Ray , Macaca fascicularis , Molecular Structure , PCSK9 Inhibitors/chemistry , PCSK9 Inhibitors/pharmacokinetics , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Rats , Structure-Activity Relationship
3.
J Am Psychoanal Assoc ; 69(2): 291-313, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34039067

ABSTRACT

Dylann Roof killed nine people in a Black church in 2015 in order to start a "war" between the white and Black races. This case is used here to develop a psychoanalytic theory of murderous racism, and even genocide. Major concepts from Freud (self-preservation, hatred, narcissism, life and death drives, delusion), Klein (projective identification), and Bion (psychotic and nonpsychotic parts of the personality) are employed. Particular attention is given to the hearing to determine Roof's capacity to represent himself after he dismissed his lawyers, who wanted to use an insanity defense to avoid the death penalty. This hearing was confronted with an undecidable dilemma, which led to legal cooperation with Roof's suicidal impulses. The deconstructive approach to undecidability, and its use in questions of law, especially as concerns the relation between psychoanalysis and the death penalty, leads to tentative recommendations for the prevention of racist murder.


Subject(s)
Psychoanalysis , Psychotic Disorders , Racism , Homicide , Humans , Male , Narcissism , Psychoanalytic Theory
4.
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
5.
Toxicol Appl Pharmacol ; 406: 115216, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32871117

ABSTRACT

Indoleamine-2,3-dioxygenase 1 (IDO1) and tryptophan-2,3-dioxygenase 2 (TDO2) degrade tryptophan (Trp) to kynurenine (Kyn), and these enzymes have promise as therapeutic targets. A comprehensive characterization of potential safety liabilities of IDO1 and TDO2 inhibitors using knockout (KO) mice has not been assessed, nor has the dual Ido1/Tdo2 KO been reported. Here we characterized male and female mice with KOs for Ido1, Tdo2, and Ido1/Tdo2 and compared findings to the wild type (WT) mouse strain, evaluated for 14 days, using metabolomics, transcriptional profiling, behavioral analysis, spleen immunophenotyping, comprehensive histopathological analysis, and serum clinical chemistry. Multiple metabolomic changes were seen in KO mice. For catabolism of Trp to Kyn and anthranilic acid, both substrates were decreased in liver of Tdo2 and dual KO mice. Metabolism of Trp to serotonin and its metabolites resulted in an increase in 5-Hydroxyindole-3-acetic acid in the Tdo2 and dual KO mice. Ido1 and dual KO mice displayed a Kyn reduction in plasma but not in liver. Nicotinamide synthesis and conversion of glucose to lactic acid were not impacted. A slight decrease in serum alkaline phosphatase was seen in all KOs, and small changes in liver gene expression of genes unrelated to tryptophan metabolism were observed. Regarding other parameters, no genotype-specific changes were observed. In summary, this work shows metabolomic pathway changes for metabolites downstream of tryptophan in these KO mice, and suggests that inhibition of the IDO1 and TDO2 enzymes would be well tolerated whether inhibited individually or in combination since no safety liabilities were uncovered.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Tryptophan Oxygenase/genetics , Tryptophan/metabolism , Animals , Female , Kynurenine/metabolism , Liver/metabolism , Male , Metabolic Networks and Pathways , Metabolomics , Mice, Knockout , Serotonin/metabolism , Spleen/immunology , ortho-Aminobenzoates/metabolism
6.
J Pharmacol Toxicol Methods ; 105: 106917, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32866658

ABSTRACT

In nonclinical toxicology the highest dose or exposure without test article-related adverse effects, known as the No Observed Adverse Effect Level (NOAEL), is a variable that may be determined. In safety pharmacology the vast majority of the endpoints measured are quantitative numeric functional endpoints such as changes in heart rate, blood pressure or respiratory frequency, endpoints that are usually not assessed using a defined framework of adversity. Therefore, we asked the question: is there a role for the NOAEL in safety pharmacology? To help answer this question, we conducted a survey via the Safety Pharmacology Society. We found that within safety pharmacology there is no formal definition of adversity and no guidance on defining NOAEL. We also found, perhaps unsurprisingly, there is no agreed rubric for using a NOAEL in safety pharmacology and we learned that the NOAEL is not a requirement in order to progress a new investigational drug through the regulatory process. Thus, a summary label such as NOAEL lacks nuance and disregards context in relation to the nature and the severity of the safety pharmacology findings. Consequently, defining 'adversity' and determining a NOAEL in safety pharmacology studies are not recommended since the range of functional endpoints investigated do not conform to a binary 'toxic/non-toxic' rubric. Focusing on describing test article-related effects on safety pharmacology endpoints, using reasoned arguments as part of an integrated risk assessment, will ensure that the clinical pharmacologists and regulatory bodies see a clear description of relevant findings at each dose or exposure level.


Subject(s)
Drugs, Investigational/adverse effects , Pharmacology/methods , Toxicity Tests/methods , Animals , Drug-Related Side Effects and Adverse Reactions , Humans , No-Observed-Adverse-Effect Level , Risk Assessment/methods
7.
Bioorg Med Chem Lett ; 30(1): 126715, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31757666

ABSTRACT

A high-throughput screening (HTS) campaign identified a class of heteroaryl piperazines with excellent baseline affinity and selectivity for phosphoinositide 3-kinase δ (PI3Kδ) over closely related isoforms. Rapid evaluation and optimization of structure-activity relationships (SAR) for this class, leveraging the modular nature of this scaffold, facilitated development of this hit class into a series of potent and selective inhibitors of PI3Kδ. This effort culminated in the identification of 29, which displayed excellent potency in enzyme and cell-based assays, as well as favorable pharmacokinetic and off-target profiles.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , High-Throughput Screening Assays/methods , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Humans , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
8.
J Pharmacol Toxicol Methods ; 98: 106593, 2019.
Article in English | MEDLINE | ID: mdl-31158459

ABSTRACT

This commentary highlights and expands upon the thoughts conveyed in the lecture by Dr. Alan S. Bass, recipient of the 2017 Distinguished Service Award from the Safety Pharmacology Society, given on 27 September 2017 in Berlin, Germany. The lecture discussed the societal, scientific, technological, regulatory and economic events that dramatically impacted the pharmaceutical industry and ultimately led to significant changes in the strategic operations and practices of safety pharmacology. It focused on the emerging challenges and opportunities, and considered the lessons learned from drug failures and the influences of world events, including the financial crisis that ultimately led to a collapse of the world economies from which we are now recovering. Events such as these, which continue to today, challenge the assumptions that form the foundation of our discipline and dramatically affect the way that safety pharmacology is practiced. These include the latest scientific and technological developments contributing to the design and advancement of safe medicines. More broadly, they reflect the philosophical mission of safety pharmacology and the roles and responsibilities served by safety pharmacologists. As the discipline of Safety Pharmacology continues to evolve, develop and mature, the reader is invited to reflect on past experiences as a framework towards a vision of the future of the field.


Subject(s)
Drug Evaluation, Preclinical/methods , Drug Industry/methods , Drug-Related Side Effects and Adverse Reactions/prevention & control , Animals , Humans , Societies
9.
J Med Chem ; 62(9): 4370-4382, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30986068

ABSTRACT

PI3Kδ catalytic activity is required for immune cell activation, and has been implicated in inflammatory diseases as well as hematological malignancies in which the AKT pathway is overactive. A purine PI3Kδ inhibitor bearing a benzimidazolone-piperidine motif was found to be poorly tolerated in dog, which was attributed to diffuse vascular injury. Several strategies were implemented to mitigate this finding, including reconstruction of the benzimidazolone-piperidine selectivity motif. Structure-based design led to the identification of O- and N-linked heterocycloalkyls, with pyrrolidines being particularly ligand efficient and kinome selective, and having an improved safety pharmacology profile. A representative was advanced into a dog tolerability study where it was found to be well tolerated, with no histopathological evidence of vascular injury.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Pyrrolidines/pharmacology , Animals , Dogs , Drug Design , HeLa Cells , Humans , Male , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/toxicity , Purines/chemical synthesis , Purines/toxicity , Pyrrolidines/chemical synthesis , Pyrrolidines/toxicity , Rats, Wistar
10.
J Pharmacol Exp Ther ; 369(2): 223-233, 2019 05.
Article in English | MEDLINE | ID: mdl-30804001

ABSTRACT

We leveraged a clinical pharmacokinetic (PK)/pharmacodynamics (PD)/efficacy relationship established with an oral phosphatidylinositol 3-kinase (PI3K)δ inhibitor (Idelalisib) in a nasal allergen challenge study to determine whether a comparable PK/PD/efficacy relationship with PI3Kδ inhibitors was observed in preclinical respiratory models of type 2 T helper cell (TH2) and type 1 T helper cell (TH1) inflammation. Results from an in vitro rat blood basophil (CD63) activation assay were used as a PD biomarker. IC50 values for PI3Kδ inhibitors, MSD-496486311, MSD-126796721, Idelalisib, and Duvelisib, were 1.2, 4.8, 0.8, and 0.5 µM. In the ovalbumin Brown Norway TH2 pulmonary inflammation model, all PI3Kδ inhibitors produced a dose-dependent inhibition of bronchoalveolar lavage eosinophils (maximum effect between 80% and 99%). In a follow-up experiment designed to investigate PK attributes [maximum (or peak) plasma concentration (Cmax), area under the curve (AUC), time on target (ToT)] that govern PI3Kδ efficacy, MSD-496486311 [3 mg/kg every day (QD) and 100 mg/kg QD] produced 16% and 93% inhibition of eosinophils, whereas doses (20 mg/kg QD, 10 mg/kg twice per day, and 3 mg/kg three times per day) produced 54% to 66% inhibition. Our profiling suggests that impact of PI3Kδ inhibitors on eosinophils is supported by a PK target with a ToT over the course of treatment close to the PD IC50 rather than strictly driven by AUC, Cmax, or Cmin (minimum blood plasma concentration) coverage. Additional studies in an Altenaria alternata rat model, a sheep Ascaris-sensitive sheep model, and a TH1-driven rat ozone exposure model did not challenge our hypothesis, suggesting that an IC50 level of TE (target engagement) sustained for 24 hours is required to produce efficacy in these traditional models. We conclude that the PK/PD observations in our animal models appear to align with clinical results associated with a TH2 airway disease.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacokinetics , Respiratory Tract Diseases/drug therapy , Respiratory Tract Diseases/immunology , Th1 Cells/drug effects , Th2 Cells/drug effects , Animals , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Male , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Rats , Respiratory Tract Diseases/metabolism
11.
Br J Pharmacol ; 172(16): 4002-11, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26031452

ABSTRACT

Evaluating whether a new medication prolongs QT intervals is a critical safety activity that is conducted in a sensitive animal model during non-clinical drug development. The importance of QT liability detection has been reinforced by non-clinical [International Conference on Harmonization (ICH) S7B] and clinical (ICH E14) regulatory guidance from the International Conference on Harmonization. A key challenge for the cardiovascular safety community is to understand how the finding from a non-clinical in vivo QT assay in animals predicts the outcomes of a clinical QT evaluation in humans. The Health and Environmental Sciences Institute Pro-Arrhythmia Working Group performed a literature search (1960-2011) to identify both human and non-rodent animal studies that assessed QT signal concordance between species and identified drugs that prolonged or did not prolong the QT interval. The main finding was the excellent agreement between QT results in humans and non-rodent animals. Ninety-one percent (21 of 23) of drugs that prolonged the QT interval in humans also did so in animals, and 88% (15 of 17) of drugs that did not prolong the QT interval in humans had no effect on animals. This suggests that QT interval data derived from relevant non-rodent models has a 90% chance of predicting QT findings in humans. Disagreement can occur, but in the limited cases of QT discordance we identified, there appeared to be plausible explanations for the underlying disconnect between the human and non-rodent animal QT outcomes.


Subject(s)
Long QT Syndrome/chemically induced , Animals , Drug Evaluation, Preclinical , Humans , Sensitivity and Specificity
12.
Handb Exp Pharmacol ; 229: 3-45, 2015.
Article in English | MEDLINE | ID: mdl-26091634

ABSTRACT

Professor Gerhard Zbinden recognized in the 1970s that the standards of the day for testing new candidate drugs in preclinical toxicity studies failed to identify acute pharmacodynamic adverse events that had the potential to harm participants in clinical trials. From his vision emerged the field of safety pharmacology, formally defined in the International Conference on Harmonization (ICH) S7A guidelines as "those studies that investigate the potential undesirable pharmacodynamic effects of a substance on physiological functions in relation to exposure in the therapeutic range and above." Initially, evaluations of small-molecule pharmacodynamic safety utilized efficacy models and were an ancillary responsibility of discovery scientists. However, over time, the relationship of these studies to overall safety was reflected by the regulatory agencies who, in directing the practice of safety pharmacology through guidance documents, prompted transition of responsibility to drug safety departments (e.g., toxicology). Events that have further shaped the field over the past 15 years include the ICH S7B guidance, evolution of molecular technologies leading to identification of new therapeutic targets with uncertain toxicities, introduction of data collection using more sophisticated and refined technologies, and utilization of transgenic animal models probing critical scientific questions regarding novel targets of toxicity. The collapse of the worldwide economy in the latter half of the first decade of the twenty-first century, continuing high rates of compound attrition during clinical development and post-approval and sharply increasing costs of drug development have led to significant strategy changes, contraction of the size of pharmaceutical organizations, and refocusing of therapeutic areas of investigation. With these changes has come movement away from dedicated internal safety pharmacology capability to utilization of capabilities within external contract research organizations. This movement has created the opportunity for the safety pharmacology discipline to come "full circle" and return to the drug discovery arena (target identification through clinical candidate selection) to contribute to the mitigation of the high rate of candidate drug failure through better compound selection decision making. Finally, the changing focus of science and losses in didactic training of scientists in whole animal physiology and pharmacology have revealed a serious gap in the future availability of qualified individuals to apply the principles of safety pharmacology in support of drug discovery and development. This is a significant deficiency that at present is only partially met with academic and professional society programs advancing a minimal level of training. In summary, with the exception that the future availability of suitably trained scientists is a critical need for the field that remains to be effectively addressed, the prospects for the future of safety pharmacology are hopeful and promising, and challenging for those individuals who want to assume this responsibility. What began in the early part of the new millennium as a relatively simple model of testing to assure the safety of Phase I clinical subjects and patients from acute deleterious effects on life-supporting organ systems has grown with experience and time to a science that mobilizes the principles of cellular and molecular biology and attempts to predict acute adverse events and those associated with long-term treatment. These challenges call for scientists with a broad range of in-depth scientific knowledge and an ability to adapt to a dynamic and forever changing industry. Identifying individuals who will serve today and training those who will serve in the future will fall to all of us who are committed to this important field of science.


Subject(s)
Drug Evaluation, Preclinical/trends , Drug-Related Side Effects and Adverse Reactions , Data Collection , Humans , Safety
13.
J Med Chem ; 58(4): 1929-39, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25625541

ABSTRACT

Development of a series of highly kinome-selective spleen tyrosine kinase (Syk) inhibitors with favorable druglike properties is described. Early leads were discovered through X-ray crystallographic analysis, and a systematic survey of cores within a selected chemical space focused on ligand binding efficiency. Attenuation of hERG ion channel activity inherent within the initial chemotype was guided through modulation of physicochemical properties including log D, PSA, and pKa. PSA proved most effective for prospective compound design. Further profiling of an advanced compound revealed bacterial mutagenicity in the Ames test using TA97a Salmonella strain, and subsequent study demonstrated that this mutagenicity was pervasive throughout the series. Identification of intercalation as a likely mechanism for the mutagenicity-enabled modification of the core scaffold. Implementation of a DNA binding assay as a prescreen and models in DNA allowed resolution of the mutagenicity risk, affording molecules with favorable potency, selectivity, pharmacokinetic, and off-target profiles.


Subject(s)
Amides/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Spleen/enzymology , Amides/chemical synthesis , Amides/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Models, Molecular , Molecular Structure , Mutagenicity Tests , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/metabolism , Spleen/drug effects , Structure-Activity Relationship
14.
Bioorg Med Chem Lett ; 23(3): 791-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265904

ABSTRACT

Inhibitors based on a benzo-fused spirocyclic oxazepine scaffold were discovered for stearoyl-coenzyme A (CoA) desaturase 1 (SCD1) and subsequently optimized to potent compounds with favorable pharmacokinetic profiles and in vivo efficacy in reducing the desaturation index in a mouse model. Initial optimization revealed potency preferences for the oxazepine core and benzylic positions, while substituents on the piperidine portions were more tolerant and allowed for tuning of potency and PK properties. After preparation and testing of a range of functional groups on the piperidine nitrogen, three classes of analogs were identified with single digit nanomolar potency: glycine amides, heterocycle-linked amides, and thiazoles. Responding to concerns about target localization and potential mechanism-based side effects, an initial effort was also made to improve liver concentration in an available rat PK model. An advanced compound 17m with a 5-carboxy-2-thiazole substructure appended to the spirocyclic piperidine scaffold was developed which satisfied the in vitro and in vivo requirements for more detailed studies.


Subject(s)
Dibenzoxazepines/chemical synthesis , Drug Delivery Systems , Enzyme Inhibitors/chemical synthesis , Liver/enzymology , Stearoyl-CoA Desaturase/antagonists & inhibitors , Animals , Cyclization , Dibenzoxazepines/pharmacokinetics , Dibenzoxazepines/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mice , Models, Animal , Molecular Structure , Rats , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology
15.
Rev. CEP-PA ; 20: 153-163, 2013.
Article in Portuguese | Index Psychology - journals | ID: psi-59876
16.
J Pharmacol Toxicol Methods ; 64(1): 7-15, 2011.
Article in English | MEDLINE | ID: mdl-21689769

ABSTRACT

In recognition of the tenth anniversary of the Safety Pharmacology Society (SPS), this review summarizes the significant events of the past 10years that have led to the birth, growth and evolution the SPS and presents a roadmap to the immediate-, intermediate- and long-term future of the SPS. The review discusses (i) the rationale for an optimal non-clinical Safety Pharmacology testing, (ii) the evolution of Safety Pharmacology over the last decade, (iii) its impact on drug discovery and development, (iv) the merits of adopting an integrated risk assessment approach, (v) the translation of non-clinical findings to humans and finally (vi) the future challenges and opportunities facing this discipline. Such challenges include the emergence of new molecular targets and new approaches to treat diseases, the rapid development of science and technologies, the growing regulatory concerns and associated number of guidance documents, and the need to train and educate the next generation of safety pharmacologist.


Subject(s)
Drug Evaluation, Preclinical/trends , Drug-Related Side Effects and Adverse Reactions , Animals , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Humans , Risk Assessment/methods , Societies, Pharmaceutical , Toxicity Tests/trends
17.
J Pharmacol Toxicol Methods ; 60(1): 69-78, 2009.
Article in English | MEDLINE | ID: mdl-19422924

ABSTRACT

Identification of novel new molecules which hold the greatest promise of safe and effective therapies remains a continuous challenge to the pharmaceutical industry. This has led the industry to implement strategies for identification of the most promising candidates during the discovery phase and for their safe and expeditious advancement through development. Testing for potential liable properties in the discovery phase has included the evaluation of major areas of pharmaceutics that have led to failure such as its physical and pharmaceutical properties, drug metabolism and pharmacokinetic characteristics, various safety endpoints including pre-development safety pharmacology, general toxicology and genetic toxicology and interrogation of counter-screen data to identify off-target affinities (i.e., receptors, ion channels, transporters, kinases, etc.) that pose a concern. Amongst the many important areas of concern is the potential for toxicities of the major organ systems. To mitigate this concern, a strategy pursued is to identify the prominent toxicological properties of the candidate prior to its recommendation for development. The results of these studies in discovery allow exclusion of the candidate before the expenditure of resources and time typical of development. In addition, the discovery phase toxicology studies serve to address key questions that may have arisen from the study of another molecule, the phenotypic profile from pre-clinical models where the therapeutic target has been genetically modified or concerns that have been raised as a result of other investigations. Importantly, the results of the exploratory drug safety studies will be used by the sponsor to judge the potential risks associated with continued pursuit of a potential development candidate. In many ways, pre-clinical toxicological investigations in discovery serve the important objective of identifying the most promising candidates to progress into development and onto registration.


Subject(s)
Drug Discovery , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions , Toxicology/methods , Animals , Drug Industry , Pharmaceutical Preparations/metabolism , Research Design
18.
J Pharmacol Toxicol Methods ; 60(1): 51-7, 2009.
Article in English | MEDLINE | ID: mdl-19447184

ABSTRACT

OBJECTIVE: Drug-induced torsades de pointes (TdP) arrhythmia is a serious public health concern that has significantly slowed the advancement of promising new therapeutic agents to the marketplace. Modeling for the potential to produce TdP has relied in part on the surrogate biomarker QT interval prolongation, measured in vivo in animals and in the clinic in man. This study was a comparison of the effects of PNU-142093, a selective 5HT1D-serotonin receptor agonist, on QT interval prolongation under restraint and non-restraint conditions in conscious cynomolgus non-human primates. METHODS: Lead II electrocardiograms (ECG) were collected following an oral single-dose (non-restraint conditions using radio-telemetry) and single- and multiple-doses for 14 days (restraint conditions using electrodes applied to the surface) at doses of 0, 5, 15, and 25 mg/kg. ECG were collected from non-restrained animals predose and for up to 5 hrs, and again at 7 hrs, postdose on 4 different days in a Latin-square crossover design; N=4/sex/dose level. ECG were collected from restrained animals on days 1, 7, and 13, predose and at approximately 4 hrs postdose; N=2/sex/group. RESULTS: Non-restrained animal heart rate ranged from 159+/-22.1 to 168+/-21.4 beats/minute when compared to restrained animal heart rate (ranging from 242+/-17.2 to 246+/-11.5 beats/minute), suggesting that non-restrained animals were under less stress. In non-restrained animals, PNU-142093 produced a non-dose related decrease in heart rate, associated with a dose-related increase in QT and QTc (QT interval corrected for changes in heart rate) intervals, which was accompanied by alterations in T-wave morphology (e.g., widening and notching of the T wave). In restrained non-human primates, PNU-142093 had no effect on heart rate or ECG morphology on any day of dosing and no statistically significant effect on QT or QTc intervals on days 1 or 7 of dosing. By day 13 there were statistically significant increases in QT and QTc intervals at 15 and 25 mg/kg. The increase in QTc interval in restrained animals on day 13 was 29+/-12 and 30+/-19 msec at 15 and 25 mg/kg/day, respectively, and that in non-restrained animals was 65+/-23 and 73+/-28 msec. DISCUSSION: These data demonstrate an ability to detect problematic drugs in conscious cynomolgus non-human primates using both restraint and non-restraint procedures. They further show that the sensitivity of these assays to identify this signal of cardiac risk is significantly improved under the condition of non-restraint.


Subject(s)
Anti-Arrhythmia Agents/adverse effects , Benzopyrans/adverse effects , Electrocardiography , Piperazines/adverse effects , Restraint, Physical , Serotonin 5-HT1 Receptor Agonists , Torsades de Pointes/chemically induced , Animals , Anti-Arrhythmia Agents/pharmacology , Benzopyrans/pharmacology , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Heart Rate/drug effects , Long QT Syndrome/chemically induced , Macaca fascicularis , Piperazines/pharmacology , Risk Assessment , Telemetry/methods
19.
J Pharmacol Toxicol Methods ; 58(2): 99-109, 2008.
Article in English | MEDLINE | ID: mdl-18692579

ABSTRACT

INTRODUCTION: The objectives of this survey were to obtain a global information update regarding current industry perspectives that describe Safety Pharmacology programs as they relate to the ICH S7A and S7B regulatory guidelines but also to obtain a broader perspective of other practises practices in the field currently used by companies. Preliminary findings were presented at the 7th Annual Meeting of the Safety Pharmacology Society (SPS) (Edinburgh, Scotland, Sept 19-21, 2007). METHODS: The survey was distributed by the SPS to 125 pharmaceutical companies. Survey topics included (a) an update on ICH S7A and S7B practices, (b) frontloading Safety Pharmacology studies prior to selection of candidate drugs, (c) abuse and dependence-liability studies and (d) an extended evaluation of industry practises practices as assessed by Contract Research Organizations (CROs). RESULTS: Respondents (>94%) include GLP core battery (CV, CNS and respiratory) studies in the drug package submitted to regulatory agencies, and approximately 40% also submit studies on gastrointestinal and renal function. Respondents to the ICH S7B aspects indicate approximately 98% include the hERG assay and QT interval (in vivo) data in submissions, 63% include APD in vitro data and another 23% APD in vivo and other cardiac channel data (26%). SP frontloading is performed by 78% of all responding companies. Respondents indicate that 39% of these non-GLP CV studies are conducted before lead optimization (LO) and 85% during LO and before candidate drug selection. The hERG, CNS selectivity binding screens and rodent behavioral studies are frontloaded by 100%, 90% and 74% of respondents. Responding CROs (26) were surveyed on the services offered including Irwin or Functional Observational Battery (FOB) tests (70%), respiratory studies (85%), in vivo telemeterized dogs (69%) and in vitro CV studies (50%). Only 38% of SP studies are combined with toxicology studies at the CROs. DISCUSSION: The survey results indicate that ICH S7A core battery studies are implemented by most of the responding companies with a clear trend of an enhanced submission of renal and GI studies. The impact of ICH S7B is clear since, all respondents assess cardiac repolarization using cellular hERG (I(Kr)) and whole animal (QT interval) assays as a component of their safety assessment. Responses indicate a diversity of approaches for conducting abuse liability studies, which primarily use the methods of self-administration and drug discrimination. While early SP frontloading of studies seems to vary, the methods used appear to be generic to some extent and include in vitro 'off-target' evaluations and in vivo tests to determine the potential for CNS and cardiovascular issues.


Subject(s)
Benchmarking/methods , Drug Industry/standards , Drug-Related Side Effects and Adverse Reactions , Data Collection , Drug Evaluation, Preclinical , Practice Guidelines as Topic , Surveys and Questionnaires
20.
J Pharmacol Toxicol Methods ; 58(2): 72-6, 2008.
Article in English | MEDLINE | ID: mdl-18508287

ABSTRACT

Biological therapeutic agents (biologicals), such as monoclonal antibodies (mAbs), are increasingly important in the treatment of human disease, and many types of biologicals are in clinical development. During preclinical drug development, cardiovascular safety pharmacology studies are performed to assess cardiac safety in accord with the ICH S7A and S7B regulations that guide these studies. The question arises, however, whether or not it is appropriate to apply these guidelines, which were devised primarily to standardize small molecule drug testing, to the cardiovascular evaluation of biologicals. We examined the scientific literature and formed a consensus of scientific opinion to determine if there is a rational basis for conducting an in vitro hERG assay as part of routine preclinical cardiovascular safety testing for biologicals. We conclude that mAb therapeutics have very low potential to interact with the extracellular or intracellular (pore) domains on hERG channel and, therefore, are highly unlikely to inhibit hERG channel activity based on their targeted, specific binding properties. Furthermore, mAb are large molecules (>140,000 Da) that cannot cross plasma membranes and therefore would be unable to access and block the promiscuous inner pore of the hERG channel, in contrast with typical small molecule drugs. Consequently, we recommend that it is not appropriate to conduct an in vitro hERG assay as part of a preclinical strategy for assessing the heart rate corrected QT interval (QTc) prolongation risk of mAbs and other types of biologicals. It is more appropriate to assess QTc risk by integrating cardiovascular endpoints into repeat-dose general toxicology studies performed in an appropriate non-rodent species. These recommendations should help shape future regulatory strategy and discussions for the cardiovascular safety pharmacology testing of mAbs as well as other biologicals and provide guidance for the preclinical cardiovascular evaluation of such agents.


Subject(s)
Biological Products/adverse effects , Cardiovascular Diseases/chemically induced , Drug Evaluation, Preclinical/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...