Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain
the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in
Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles
and JavaScript.
Articles in this series provide an accessible discussion of a particular aspect of the process of turning ideas into drugs. The aim is to allow readers with limited knowledge of a given area to become familiar with the key concepts and techniques involved. Written by those closely involved in the discovery process, these articles aim to provide insights that will aid in future drug discovery programs.
Traditional cell-based disease models often fail to adequately represent key disease characteristics, increasing the risk of subsequent attrition in clinical trials. This article presents a set of principles for disease-relevant assays, and discusses new opportunities for exploiting advances in cell-based assay technologies in drug discovery, including induced pluripotent stem cells as well as 3D co-culture and organ-on-a-chip systems, which are being complemented by progress with single-cell imaging and gene editing technologies.
The recent growth in the number of academic drug discovery centres is providing new opportunities to couple the curiosity-driven research culture in academia with rigorous preclinical drug discovery practices used in industry. To realize the potential of these opportunities, it is important that academic researchers understand the risks in several key areas — including organization, target selection, assay design, medicinal chemistry and preclinical pharmacology — which are discussed in this article.
Many clinical trial failures can be traced back to the limited predictive value of preclinical models of disease. Plenge and colleagues discuss how knowledge from human genetics, such as naturally occurring mutations in humans that affect the activity of particular proteins, can be used as a tool to more effectively prioritize molecular targets in drug development.
Allosteric ligands bind to G protein-coupled receptors at a site distinct from the endogenous ligand. This Review discusses the potential advantages that allosteric ligands could hold, and highlights how the complexity of their actions provides both challenges and opportunities for drug screening.
Animal models are vital tools in the development of therapies for orphan diseases, given the small populations of patients available to evaluate the therapies. Here, Sepodes and colleagues from the European Medicines Agency's Committee for Orphan Medicinal Products harness their experience to provide an overview of the animal models used to support regulatory applications for metabolic, neuromuscular and ophthalmological rare diseases.
In vitropharmacological profiling is playing an increasing part in identifying undesirable off-target effects of candidate drugs earlier in the drug discovery process. In this article, authors from four large pharmaceutical companies share their views on the rationale, strategies and methodologies forin vitropharmacological profiling, and recommend a minimal panel of targets for screening.
The therapeutic index of drug candidates — a quantitative relationship between their safety and efficacy, such as the ratio of the highest exposure to a drug that does not cause toxicity to the exposure that has the desired pharmacological effects — is widely used to aid decision-making in drug discovery and development. Muller and Milton discuss key issues in the calculation and interpretation of therapeutic indices at different stages of the process.
Extensive analyses of successful and failed compounds in drug discovery and development have improved our understanding of the role of physicochemical properties in attrition. They have also clarified the difficulties in finding the 'sweet spot' in medicinal chemistry programmes. Hann and Keserü discuss scientific, strategic and cultural considerations for medicinal chemistry practices, with the aim of promoting more effective use of what is already known, and a wider appreciation of the risks of pursuing suboptimal compounds.
Dysregulated cell death occurs in many human diseases, and modulating the associated pathways has proved effective in the treatment of cancer, stroke and neurodegenerative disorders. In their review, Kepp and colleagues provide an overview of assays capable of accurately quantifying distinct cell death pathways, focusing on those techniques that are applicable to high-throughput screening.
Data on the fraction of protein-bound drug are frequently used to guide chemical structure design and to prioritize compounds forin vivostudies. Here, the authors highlight how these practices are misleading and could result in the wrong compounds being progressed through discovery programmes.
Orloff and colleagues describe how moving from the traditional approach to clinical trials based on sequential, distinct phases towards a more integrated strategy that increases flexibility and maximizes the use of accumulated knowledge could have a key role in improving the efficiency and cost-effectiveness of drug development. Using examples in which novel trial designs have been successfully applied, they also illustrate the use of the tools involved, such as Bayesian methodologies, and discuss the advantages and challenges for their more widespread implementation.
The importance of pharmacovigilance — the ongoing assessment of the safety of a marketed medicine — has been increasingly appreciated in recent years. Breckenridge and colleagues summarize the key tools that are available for pharmacovigilance, discuss which might be the most appropriate to use in different situations and consider the future directions of the field.
Here, the authors summarize the current state of multi-parameter profiling technologies and how phenotypic profiling of small molecules provides important insights into their mechanisms of action, as well as a systems level understanding of biological pathways and their responses to drug treatments.
Despite improvements in European regulatory standards and procedures, the occurrence of failed or problematic marketing authorization applications for biotechnological products remains common. Here, Schäffner-Dallmann and Schneider identify common regulatory concerns and consider strategies that may help prevent such issues.
Ion channels remain an under-exploited drug target class owing to the low-throughput nature of patch-clamp electrophysiology. In this Review, Dunlop and colleagues evaluate automated electrophysiology platforms and discuss their impact in terms of ion-channel screening, lead optimization and the assessment of cardiac ion-channel safety liability.
The number and frequency of use of protein therapeutics has increased dramatically since the introduction of the first recombinant protein therapeutic — human insulin — 25 years ago. Golan and colleagues overview some of the key characteristics of protein therapeutics, summarize the more than 130 protein therapeutics used currently and suggest a new classification of these proteins based on their pharmacological action.
Toxicity is a leading cause of attrition at all stages of the drug development process. In their Review, Kramer and colleagues discuss how the early application of preclinical safety assessment can aid the design of safer pharmaceutical lead candidates.
Bayesian statistical methods are being increasingly used in clinical research owing to the advantages they can offer compared with conventional approaches. Berry explains the underlying rationale, and discusses the potential of Bayesian trials to improve the effectiveness of drug development.