Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Stem Cell Reports ; 8(2): 226-234, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28111276

ABSTRACT

To predict drug-induced serious adverse events (SAE) in clinical trials, a model using a panel of cells derived from human induced pluripotent stem cells (hiPSCs) of individuals with different susceptibilities could facilitate major advancements in translational research in terms of safety and pharmaco-economics. However, it is unclear whether hiPSC-derived cells can recapitulate interindividual differences in drug-induced SAE susceptibility in populations not having genetic disorders such as healthy subjects. Here, we evaluated individual differences in SAE susceptibility based on an in vitro model using hiPSC-derived cardiomyocytes (hiPSC-CMs) as a pilot study. hiPSCs were generated from blood samples of ten healthy volunteers with different susceptibilities to moxifloxacin (Mox)-induced QT prolongation. Different Mox-induced field potential duration (FPD) prolongation values were observed in the hiPSC-CMs from each individual. Interestingly, the QT interval was significantly positively correlated with FPD at clinically relevant concentrations (r > 0.66) in multiple analyses including concentration-QT analysis. Genomic analysis showed no interindividual significant differences in known target-binding sites for Mox and other drugs such as the hERG channel subunit, and baseline QT ranges were normal. The results suggest that hiPSC-CMs from healthy subjects recapitulate susceptibility to Mox-induced QT prolongation and provide proof of concept for in vitro preclinical trials.


Subject(s)
Electrophysiological Phenomena/drug effects , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Action Potentials/drug effects , Alleles , Cell Differentiation , ERG1 Potassium Channel/genetics , ERG1 Potassium Channel/metabolism , Electrocardiography , Gene Expression Profiling , Gene Frequency , Healthy Volunteers , Heart Conduction System/drug effects , Humans , Male , Mutation , Myocytes, Cardiac/cytology , Polymorphism, Single Nucleotide
2.
Int J Neuropsychopharmacol ; 20(4): 285-294, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28031269

ABSTRACT

Despite large unmet medical needs in the field for several decades, CNS drug discovery and development has been largely unsuccessful. Biomarkers, particularly those utilizing neuroimaging, have played important roles in aiding CNS drug development, including dosing determination of investigational new drugs (INDs). A recent working group was organized jointly by CINP and Japanese Society of Neuropsychopharmacology (JSNP) to discuss the utility of biomarkers as tools to overcome issues of CNS drug development.The consensus statement from the working group aimed at creating more nuanced criteria for employing biomarkers as tools to overcome issues surrounding CNS drug development. To accomplish this, a reverse engineering approach was adopted, in which criteria for the utilization of biomarkers were created in response to current challenges in the processes of drug discovery and development for CNS disorders. Based on this analysis, we propose a new paradigm containing 5 distinct tiers to further clarify the use of biomarkers and establish new strategies for decision-making in the context of CNS drug development. Specifically, we discuss more rational ways to incorporate biomarker data to determine optimal dosing for INDs with novel mechanisms and targets, and propose additional categorization criteria to further the use of biomarkers in patient stratification and clinical efficacy prediction. Finally, we propose validation and development of new neuroimaging biomarkers through public-private partnerships to further facilitate drug discovery and development for CNS disorders.


Subject(s)
Biomarkers , Central Nervous System Agents , Drug Discovery/methods , Neuroimaging , Neuropharmacology/methods , Psychopharmacology/methods , Drug Discovery/standards , Humans , Neuropharmacology/standards , Psychopharmacology/standards
3.
J Hum Genet ; 60(6): 319-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855068

ABSTRACT

Japan Pharmacogenomics Data Science Consortium (JPDSC) has assembled a database for conducting pharmacogenomics (PGx) studies in Japanese subjects. The database contains the genotypes of 2.5 million single-nucleotide polymorphisms (SNPs) and 5 human leukocyte antigen loci from 2994 Japanese healthy volunteers, as well as 121 kinds of clinical information, including self-reports, physiological data, hematological data and biochemical data. In this article, the reliability of our data was evaluated by principal component analysis (PCA) and association analysis for hematological and biochemical traits by using genome-wide SNP data. PCA of the SNPs showed that all the samples were collected from the Japanese population and that the samples were separated into two major clusters by birthplace, Okinawa and other than Okinawa, as had been previously reported. Among 87 SNPs that have been reported to be associated with 18 hematological and biochemical traits in genome-wide association studies (GWAS), the associations of 56 SNPs were replicated using our data base. Statistical power simulations showed that the sample size of the JPDSC control database is large enough to detect genetic markers having a relatively strong association even when the case sample size is small. The JPDSC database will be useful as control data for conducting PGx studies to explore genetic markers to improve the safety and efficacy of drugs either during clinical development or in post-marketing.


Subject(s)
HLA Antigens/genetics , Databases, Genetic , Female , Gene Frequency , Genetic Association Studies , Genotype , Healthy Volunteers , Humans , Japan , Male , Pharmacogenetics , Polymorphism, Single Nucleotide
4.
Yakugaku Zasshi ; 134(4): 485-90, 2014.
Article in Japanese | MEDLINE | ID: mdl-24694807

ABSTRACT

The Human Genome Project was completed in 2003. A catalog of common genetic variants in humans was built at the International HapMap Project. These variants, known as single nucleotide polymorphisms (SNPs), occur in human DNA and distributed among populations in different parts of the world. By using the Linkage Disequilibrium and mapping blocks are able to define quantitative characters of inherited diseases. Currently 50 K-5.0 M microarray are available commercially, which based on the results of following the ENCODE & 1000 genome projects. Therefore the genome wide association study (GWAS) has become a key tool for discovering variants that contribute to human diseases and provide maximum coverage of the genome, in contrast to the traditional approach in which only a few candidates genes was targeted. The available public GWAS databases provided valuable biological insights and new discovery for many common diseases, due to the availability of low cost microarray. The GWAS has the potential to provide a solution for the lack of new drug targets and reducing drug failure due to adverse drug reactions either. These are critical issues for pharmaceutical companies. Here, the Japan PGx Data Science Consortium (JPDSC), which was established on February 20, 2009 by six leading pharmaceutical companies in Japan, was introduced. We believe that the efforts of stakeholders including the regulatory authorities, health providers, and pharmaceutical companies to understand the potential and ethical risk of using genetic information including GWAS will bring benefits to patients in the future.


Subject(s)
Drug Discovery , Genome-Wide Association Study , Biomarkers/analysis , Clinical Trials as Topic , Databases, Genetic , Genome, Human , Humans
6.
Drug Metab Pharmacokinet ; 27(1): 2-8, 2012.
Article in English | MEDLINE | ID: mdl-22214937

ABSTRACT

Over the last two decades, identification of polymorphisms that influence human diseases has begun to have an impact on the provision of medical care. The promise of genetics lies in its ability to provide insight into an individual's susceptibility to disease, the likely nature of the disease and the most appropriate therapy. For much of its history, pharmacogenomics (PGx) has been limited to relatively simple phenotypes such as plasma drug levels. Progress in genetic technologies has broadened the scope of exploratory PGx and its implementation into safety and efficacy studies, impacting a broad spectrum of drug discovery and development activities. Recent PGx data show the ability of this approach to generate information that can be applied to target selection, dose selection, efficacy determination and safety issues. This in turn will lead to significant opportunities to influence the approaches to drug discovery, clinical development and the probability of success. In particular, adverse drug reactions are critical issues for pharmaceutical companies and for the patients who will benefit from these new medicines. In this review, we outline current progress in PGx, using examples to highlight the influence of polymorphisms, and discuss contemporary challenges for both researchers and clinicians.


Subject(s)
Biomedical Research , Drug Discovery , Drugs, Investigational/pharmacokinetics , Pharmacogenetics/methods , Biomedical Research/trends , Drug Discovery/trends , Drugs, Investigational/adverse effects , Drugs, Investigational/therapeutic use , Humans , Technology, Pharmaceutical
7.
J Biol Chem ; 277(7): 4816-22, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11719510

ABSTRACT

We have attempted to elucidate an involvement of cathepsin E (CE) in major histocompatibility complex class II-mediated antigen presentation by microglia. In primary cultured murine microglia, CE was localized mainly in early endosomes and its expression level was markedly increased upon stimulation with interferon-gamma. Pepstatin A, a specific inhibitor of aspartic proteases, significantly inhibited interleukin-2 production from an OVA-(266-281)-specific T helper cell hybridomas upon stimulation with native OVA presented by interferon-gamma-treated microglia. However, pepstatin A failed to inhibit the presentation of OVA-(266-281) peptide. The possible involvement of CE in the processing of native OVA into antigenic peptide was further substantiated by that digested fragments of native OVA by CE could be recognized by OVA-specific Th cells. Cathepsin D also degraded native OVA into antigenic peptide, whereas microglia prepared from cathepsin D-deficient mice retained an ability for antigen presentation. On the other hand, the requirement for cysteine proteases such as cathepsins S and B in the processing of invariant chain (Ii) was confirmed by immunoblot analyses in the presence of their specific inhibitors. In conclusion, CE is required for the generation of an antigenic epitope from OVA but not for the processing of Ii in microglia.


Subject(s)
Antigens/metabolism , Cathepsin E/metabolism , Cathepsin E/physiology , Neuroglia/cytology , Animals , Antigen Presentation , Bone Marrow Cells/metabolism , Cathepsin B/metabolism , Cathepsins/metabolism , Cells, Cultured , Epitopes/metabolism , Genes, MHC Class II , Hybridomas/metabolism , Immunoblotting , Immunohistochemistry , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pepstatins/metabolism , Peptides/chemistry , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...