Discovery and development of drugs

Published on 02/03/2015 by admin

Filed under Basic Science

Last modified 02/03/2015

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1155 times

Chapter 3 Discovery and development of drugs

Making a new medicine

Discovering and developing a new medicine requires combining the skills of biology, chemistry, clinical medicine and, of course, pharmacology. There are three key decision points along the way:

Medicinal therapeutics rests on the two great supporting pillars of pharmacology:

Once a target has been selected the process of finding the right chemical to act as an antagonist, agonist, inhibitor, activator or modulator of the protein function depends on a process of screening. For decades, the rational discovery of new medicines depended on modifications of the structures of natural chemical mediators. This may still be the route to find the drug, but more often now large libraries of compounds are screened against the target in robotic high throughput screens. However, it is worth remembering that the exact molecular basis of drug action may remain unknown, and this book contains frequent examples of old drugs whose mechanism of action remains mysterious. The evolution of molecular medicine (including recombinant DNA technology) in the past 30 years has led to identification of many thousands of potential drug targets, but the function of many of these genes remains unknown. The hope was that the identification of targets identified by genetics coupled with high throughout screening would lead to a great increase in the productivity of drug discovery. This has turned out not to be the case since it is clearly important to understand both the function of the target and the nature of the interaction of the chemical drug with the target in order to make a selective and safe medicine. The skills of quantitative pharmacology are key.

The chances of discovering a truly novel medicine, i.e. one that does something valuable for patients that had previously not been possible (or that does safely what could previously have been achieved only with substantial risk), are increased when the discovery and development programme is founded on precise knowledge of the biological processes it is desired to change. The commercial rewards of a successful product are potentially enormous and provide a great incentive for developers to invest and risk huge sums of money. Most projects in drug discovery and development fail. Indeed the chances of making it through from target selection to having a medicine on the market are under 1 in 100.

The huge increase in understanding of molecular signalling – both between cells and within cells – has opened many new opportunities to develop medicines that can target discrete steps in the body’s elaborate pathways of chemical reactions.3 The challenge, of course, is to do so in a way that produces benefit without harm. The more fundamental the pathway targeted the more likely there is to be a big effect, whether beneficial, harmful or both. No benefit comes without some risk.

The molecular, industrialised and automated approach to drug discovery that followed sequencing of the genome and application of high throughput chemical approaches led to two consequences:

Pharmacogenetics has gained momentum from recent advances in molecular genetics and genome sequencing, due to:

Expectations of pharmacogenetics and its progeny, pharmacoproteomics (understanding of and drug effects on protein variants) remain high, but the applicability will not be universal. They include the following:

Consequences of these expectations include: smaller clinical trial programmes with well-defined patient groups (based on phenotypic and genotypic characterisation), better understanding of the pharmacokinetics and dynamics according to genetic variation, and improved monitoring of adverse events after marketing.

New drug development proceeds thus:

The (critical) phase of progress from the laboratory to humans is often termed translational science or experimental medicine. It was defined as ‘the application of biomedical research (pre-clinical and clinical), conducted to support drug development, which aids in the identification of the appropriate patient for treatment (patient selection), the correct dose and schedule to be tested in the clinic (dosing regimen) and the best disease in which to test a potential agent’.4

It will be obvious from the account that follows that drug development is an extremely arduous, highly technical and enormously expensive operation. Successful developments (1% of compounds that proceed to full test eventually become licensed medicines) must carry the cost of the failures (99%).5 It is also obvious that such programmes are likely to be carried to completion only when the organisations and the individuals within them are motivated overall by the challenge to succeed and to serve society, as well as to make money. A previous edition of this chapter included a quote from a paper I wrote from my time in academia and I leave it here:

Let us get one thing straight: the drug industry works within a system that demands it makes a profit to satisfy shareholders. Indeed, it has a fiduciary6 duty to do so. The best way to make a lot of money is to invent a drug that produces a dramatically beneficial clinical effect, is far more effective than existing options, and has few unwanted effects. Unfortunately most drugs fall short of this ideal.7

Techniques of discovery

(See Fig. 3.1)

The newer technologies, the impact of which has yet to be fully felt, include the following:

Proteins as medicines: biotechnology

The targets of most drugs are proteins (cell receptors, enzymes) and it is only lack of technology that has hitherto prevented the exploitation of proteins (and peptides) as medicines. This technology is now available and some of the most successful new medicines of the last few years have been biological – antibodies or petides. The practical limitations are that (1) they need to be injected, as they are digested when swallowed and (2) they target soluble factors and targets on the cell membrane but are generally unable to target intracellular proteins. However, these limitations are offset by their high degree of specificity, their often very long half-life (for antibodies) that means that the medicine may be given by monthly injections, and the more predictable toxicity when compared to the rather unpredictable effect of classical small molecule drugs (the usual ‘white pills’). Biotechnology involves the use of recombinant DNA technology/genetic engineering to clone and express human genes, for example in microbial (Escherichia coli or yeast) cells so that they manufacture proteins. Such techniques can deliver hormones and autacoids in commercial amounts (such as insulin and growth hormone, erythropoietins, cell growth factors and plasminogen activators, interferons, vaccines and – probably most important – antibodies).

Transgenic animals (that breed true for the gene) are also used as models for human disease as well as for production of medicines.

Antisense approaches
Buy Membership for Basic Science Category to continue reading. Learn more here