Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
SLAS Discov ; 25(7): 812-821, 2020 08.
Article in English | MEDLINE | ID: mdl-32432952

ABSTRACT

Drug discovery programs are moving increasingly toward phenotypic imaging assays to model disease-relevant pathways and phenotypes in vitro. These assays offer richer information than target-optimized assays by investigating multiple cellular pathways simultaneously and producing multiplexed readouts. However, extracting the desired information from complex image data poses significant challenges, preventing broad adoption of more sophisticated phenotypic assays. Deep learning-based image analysis can address these challenges by reducing the effort required to analyze large volumes of complex image data at a quality and speed adequate for routine phenotypic screening in pharmaceutical research. However, while general purpose deep learning frameworks are readily available, they are not readily applicable to images from automated microscopy. During the past 3 years, we have optimized deep learning networks for this type of data and validated the approach across diverse assays with several industry partners. From this work, we have extracted five essential design principles that we believe should guide deep learning-based analysis of high-content images and multiparameter data: (1) insightful data representation, (2) automation of training, (3) multilevel quality control, (4) knowledge embedding and transfer to new assays, and (5) enterprise integration. We report a new deep learning-based software that embodies these principles, Genedata Imagence, which allows screening scientists to reliably detect stable endpoints for primary drug response, assess toxicity and safety-relevant effects, and discover new phenotypes and compound classes. Furthermore, we show how the software retains expert knowledge from its training on a particular assay and successfully reapplies it to different, novel assays in an automated fashion.


Subject(s)
Drug Discovery/trends , High-Throughput Screening Assays , Molecular Imaging , Signal Transduction/genetics , Automation , Deep Learning , Humans , Image Processing, Computer-Assisted , Microscopy , Software
2.
SLAS Discov ; 22(2): 203-209, 2017 02.
Article in English | MEDLINE | ID: mdl-27789754

ABSTRACT

Surface plasmon resonance (SPR) is a powerful method for obtaining detailed molecular interaction parameters. Modern instrumentation with its increased throughput has enabled routine screening by SPR in hit-to-lead and lead optimization programs, and SPR has become a mainstream drug discovery technology. However, the processing and reporting of SPR data in drug discovery are typically performed manually, which is both time-consuming and tedious. Here, we present the workflow concept, design and experiences with a software module relying on a single, browser-based software platform for the processing, analysis, and reporting of SPR data. The efficiency of this concept lies in the immediate availability of end results: data are processed and analyzed upon loading the raw data file, allowing the user to immediately quality control the results. Once completed, the user can automatically report those results to data repositories for corporate access and quickly generate printed reports or documents. The software module has resulted in a very efficient and effective workflow through saved time and improved quality control. We discuss these benefits and show how this process defines a new benchmark in the drug discovery industry for the handling, interpretation, visualization, and sharing of SPR data.


Subject(s)
Biosensing Techniques/methods , Data Analysis , Drug Discovery , Drug Evaluation, Preclinical/trends , Drug Design , Humans , Pharmaceutical Research , Software , Surface Plasmon Resonance , Workflow
3.
J Biomol Screen ; 12(8): 1042-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18087069

ABSTRACT

Recent technological advances in high-content screening instrumentation have increased its ease of use and throughput, expanding the application of high-content screening to the early stages of drug discovery. However, high-content screens produce complex data sets, presenting a challenge for both extraction and interpretation of meaningful information. This shifts the high-content screening process bottleneck from the experimental to the analytical stage. In this article, the authors discuss different approaches of data analysis, using a phenotypic neurite outgrowth screen as an example. Distance measurements and hierarchical clustering methods lead to a profound understanding of different high-content screening readouts. In addition, the authors introduce a hit selection procedure based on machine learning methods and demonstrate that this method increases the hit verification rate significantly (up to a factor of 5), compared to conventional hit selection based on single readouts only.


Subject(s)
Neurites/metabolism , Tissue Array Analysis/standards , Cluster Analysis , Multivariate Analysis , Quality Control , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...