scholarly journals A Foreword from the Editor

2016 ◽  
Vol 2 (1) ◽  
pp. 1
Author(s):  
Jean-Marie Boeynaems

Welcome to the 3rd issue of the Journal of Medicines Development Sciences. Like previous issues it contains articles covering the entire process of drug development from target identification to drug registration.

Gut ◽  
2016 ◽  
Vol 65 (8) ◽  
pp. 1233-1239 ◽  
Author(s):  
Silvio Danese ◽  
Claudio Fiocchi ◽  
Julián Panés

2013 ◽  
Vol 88 (3) ◽  
pp. 357-361
Author(s):  
B. Saikia ◽  
C.C. Barua ◽  
S. Hazarika ◽  
L.C. Lahon ◽  
D. Saikia ◽  
...  

AbstractThe neuromuscular system of helminths is an important area for target identification and drug development. Many anthelmintics, namely ivermectin, levamisole, piperazine, pyrantel, praziquantel and organophosphates, produce paralysis of helminths by affecting their neuromuscular systems. The neuromuscular system of helminths is also an important area of research to identify some of the important differences between the neuromuscular physiology of helminths and mammals. The identification of differences would help in developing newer target-specific, safe and effective anthelmintics. The present study was carried out to investigate the effects of different adrenergic neurotransmitters (epinephrine, norepinephrine, dopamine, l-dopa) and their antagonists (propranolol and haloperidol) on the spontaneous muscular activity of isometrically mounted Paramphistomum cervi.


2018 ◽  
Author(s):  
Fiona M Behan ◽  
Francesco Iorio ◽  
Emanuel Gonçalves ◽  
Gabriele Picco ◽  
Charlotte M Beaver ◽  
...  

SummaryFunctional genomics approaches can overcome current limitations that hamper oncology drug development such as lack of robust target identification and clinical efficacy. Here we performed genome-scale CRISPR-Cas9 screens in 204 human cancer cell lines from 12 cancer-types and developed a data-driven framework to prioritise cancer therapeutic candidates. We integrated gene cell fitness effects with genomic biomarkers and target tractability for drug development to systematically prioritise new oncology targets in defined tissues and genotypes. Furthermore, we took one of our most promising dependencies, Werner syndrome RecQ helicase, and verified it as a candidate target for tumours with microsatellite instability. Our analysis provides a comprehensive resource of cancer dependencies, a framework to prioritise oncology targets, and nominates specific new candidates. The principles described in this study can transform the initial stages of the drug development process contributing to a new, diverse and more effective portfolio of oncology targets.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Aroon D. Hingorani ◽  
Valerie Kuan ◽  
Chris Finan ◽  
Felix A. Kruger ◽  
Anna Gaulton ◽  
...  

AbstractLack of efficacy in the intended disease indication is the major cause of clinical phase drug development failure. Explanations could include the poor external validity of pre-clinical (cell, tissue, and animal) models of human disease and the high false discovery rate (FDR) in preclinical science. FDR is related to the proportion of true relationships available for discovery (γ), and the type 1 (false-positive) and type 2 (false negative) error rates of the experiments designed to uncover them. We estimated the FDR in preclinical science, its effect on drug development success rates, and improvements expected from use of human genomics rather than preclinical studies as the primary source of evidence for drug target identification. Calculations were based on a sample space defined by all human diseases – the ‘disease-ome’ – represented as columns; and all protein coding genes – ‘the protein-coding genome’– represented as rows, producing a matrix of unique gene- (or protein-) disease pairings. We parameterised the space based on 10,000 diseases, 20,000 protein-coding genes, 100 causal genes per disease and 4000 genes encoding druggable targets, examining the effect of varying the parameters and a range of underlying assumptions, on the inferences drawn. We estimated γ, defined mathematical relationships between preclinical FDR and drug development success rates, and estimated improvements in success rates based on human genomics (rather than orthodox preclinical studies). Around one in every 200 protein-disease pairings was estimated to be causal (γ = 0.005) giving an FDR in preclinical research of 92.6%, which likely makes a major contribution to the reported drug development failure rate of 96%. Observed success rate was only slightly greater than expected for a random pick from the sample space. Values for γ back-calculated from reported preclinical and clinical drug development success rates were also close to the a priori estimates. Substituting genome wide (or druggable genome wide) association studies for preclinical studies as the major information source for drug target identification was estimated to reverse the probability of late stage failure because of the more stringent type 1 error rate employed and the ability to interrogate every potential druggable target in the same experiment. Genetic studies conducted at much larger scale, with greater resolution of disease end-points, e.g. by connecting genomics and electronic health record data within healthcare systems has the potential to produce radical improvement in drug development success rate.


2016 ◽  
Author(s):  
Chris Finan ◽  
Anna Gaulton ◽  
Felix Kruger ◽  
Tom Lumbers ◽  
Tina Shah ◽  
...  

Target identification (identifying the correct drug targets for each disease) and target validation (demonstrating the effect of target perturbation on disease biomarkers and disease end-points) are essential steps in drug development. We showed previously that biomarker and disease endpoint associations of single nucleotide polymorphisms (SNPs) in a gene encoding a drug target accurately depict the effect of modifying the same target with a pharmacological agent; others have shown that genomic support for a target is associated with a higher rate of drug development success. To delineate drug development (including repurposing) opportunities arising from this paradigm, we connected complex disease- and biomarker-associated loci from genome wide association studies (GWAS) to an updated set of genes encoding druggable human proteins, to compounds with bioactivity against these targets and, where these were licensed drugs, to clinical indications. We used this set of genes to inform the design of a new genotyping array, to enable druggable genome-wide association studies for drug target selection and validation in human disease.


Author(s):  
Masturah Bte Mohd Abdul Rashid

The inverse relationship between the cost of drug development and the successful integration of drugs into the market has resulted in the need for innovative solutions to overcome this burgeoning problem. This problem could be attributed to several factors, including the premature termination of clinical trials, regulatory factors, or decisions made in the earlier drug development processes. The introduction of artificial intelligence (AI) to accelerate and assist drug development has resulted in cheaper and more efficient processes, ultimately improving the success rates of clinical trials. This review aims to showcase and compare the different applications of AI technology that aid automation and improve success in drug development, particularly in novel drug target identification and design, drug repositioning, biomarker identification, and effective patient stratification, through exploration of different disease landscapes. In addition, it will also highlight how these technologies are translated into the clinic. This paradigm shift will lead to even greater advancements in the integration of AI in automating processes within drug development and discovery, enabling the probability and reality of attaining future precision and personalized medicine.


Author(s):  
Suzanne F. Jones ◽  
Andrew J. McKenzie

As researchers learn more about tumor biology and the molecular mechanisms involved in tumorigenesis, metastasis, and tumor evolution, clinical trials are growing more complex and patient selection for clinical trials is becoming more specific. Rather than exploit certain phenotypic characteristics of tumor cells (e.g., rapid cell division and uncontrolled cell growth), pharmaceuticals targeting the genotypic causes of tumorigenesis are emerging. The sequencing of the human genome, advances in chemical techniques, and increased efficiency in drug target identification have changed the way drugs are developed. Now, more precise drugs targeting specific mutations within individual genes are being used to treat narrow patient populations harboring these specific driver mutations, often with greater efficacy and lower toxicity than traditional chemotherapeutic agents. This precision in drug development relies not only on the ability to design exquisitely specific pharmaceuticals but also to identify (with the same level of precision) the patients who are most likely to respond to those therapies. Robust screening techniques and adequate molecular oncology education are required to match the appropriate patient to precision therapies, and these same screening techniques provide the data necessary to advance to the next generation of drug development.


2017 ◽  
Vol 9 (383) ◽  
pp. eaag1166 ◽  
Author(s):  
Chris Finan ◽  
Anna Gaulton ◽  
Felix A. Kruger ◽  
R. Thomas Lumbers ◽  
Tina Shah ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document