Drug Discovery Module 21 PDF

Loading...
Loading...
Loading...
Loading...
Loading...
Loading...
Loading...

Summary

This document discusses drug discovery, highlighting the importance of target identification, validation, and druggability in the development process. It also touches on factors like disease targeting and preclinical testing, which are vital steps in the process before human trials.

Full Transcript

MODULE 21: DRUG DISCOVERY NOW Choosing Disease Unmet Serious condition and existing therapy is inadequate medical - Serious = health is deteriorating need - Inadequate: Drugs are resistant, the effective dose has many adverse effect Economic Size of Target...

MODULE 21: DRUG DISCOVERY NOW Choosing Disease Unmet Serious condition and existing therapy is inadequate medical - Serious = health is deteriorating need - Inadequate: Drugs are resistant, the effective dose has many adverse effect Economic Size of Target - Orphan diseases have additional challenges viability Population - Definition of orphan in terms of population number is different in different countries - Large target population = large market = large return for companies Purchasing Power - Less in low- and middle-income countries - Or if disease is higher in third world countries but not in developed countries Feasability of drug - Lower risk draws more funding development - Information about the disease What makes a drug a good target - Involved in pathophysiology of disease - Primarily expressed on tissue of interest à Otherwise can cause on target adverse effects on off target tissues Known 3D structure - Druggable – target can be altered in desired manner by low molecular weight compounds - Assays available for high-throughput screening or assays can be developed Druggability - Likelihood that target can be modulated by low molecular weight compounds with ‘drug-like’ properties o So its orally active and bioactive o Has high affinity o High potency = less potential for adverse effects - Can be predicted o Use information about structure and endogenous ligand § Small drug with one target = good target § Large protein with multiple binding sites = bad target o Use information about its family § Previous successfully targeted proteins? o Not perfect - Human genome ≈20,000-22,000 protein encoding genes o Druggable genome ≈ 3,000 o Drug target ≈ 600-1500 o Disease modifying genes ≈ 3,000 Target identification and validation - Evidence that target is involved in disease pathogenesis - Evidence that modifying the target alters disease state - Target identification and validation: o requires understanding of molecular mechanisms of disease o sometimes relatively straightforward (monogenic diseases) but often not (polygenic diseases) § monogenic = C-C gene for cystic fibrosis o can use big data § some gens are upregulated/downregulated during disease Assay development – Screening – Hits & Leads - Target Based Drug Discovery Development of assay to high- Screening Hit and lead identification throughput screening are often biochemical or cellular - large small molecular library = costly - confirm chemical hits - can assess binding and/or functional - focused library = can be aided by - physiochemical properties and selectivity activity structure based drug design or ligand (often in silico) - ideally surrogate for clinical based design = cost effective - PD = rand potency endpoint - Toxicity Target-based vs phenotype-based Target Phenotype You have identified and validated a target. You - You have a compound eliciting a desired phenotype. You identify the target develop assays and screen compound libraries to retrospectively. identify ‘hits’. - Don’t need to know the target mechanism, will figure out after - Was used most before the human genome was fully sequenced What information about a drug might be important before starting clinical trial? - Side effects - EC50 - Toxicity - Lipid solubility - Drug-drug interactions - Half-life - Efficacious dose - Vd - Bioavailability - Metabolism Preclinical development: What’s on the IND dossier - IND dossier o Submission made to FDA to allow the drug to be considered for clinical testing - Information includes o Toxicity profile o Pharmacological profile = in vitro and in vivo § Pharmacodynamic and pharmacokinetic data (ADME), information about dose, frequency, route of administration o Chemical/manufacturing info, composition/source of natural products o Formulation § Combination of drug substance with other substances, often to aid administration/compliance, stability/shelf life § Dose and administration route determined using animals before testing on humans àHelps understand toxicity o Proposed clinical plans/protocols Toxicity testing Systematic and hierarchical Acute Chronic Length of study (exposure to drug) (in vitro, ex vivo, in vivo) effect of single exposure to high effect of long-term exposure to depends on intended clinical use dose over 6-24 months Unsafe drugs - catalysts for toxicity testing - Thalidomide: indicated for morning sickness - Discovered to be teratogenic (led to malformed infants) o Was originally tested in rats which were more resistant to the drugs - Changed regulatory landscape Mandatory testing of drugs for teratogenic potential Previously, testing was not mandatory (and not regularly performed) - So now we have to test drugs in 1 rodent and 1 non-rodent species before testing on humans Characterising pharmacological profiles - Systematic and hierarchical! o (in vitro, ex vivo, in vivo – rodents to primates) o pharmacological assays should reflect disease - Can be used to assess safety by identifying pharmacodynamic effects of a substance on physiological functions. Adverse effects may be, o Predictable § related to mechanism of action on desired target in intended tissue or unintended tissue § related to off-target effect associated with chemical class o unpredictable § unrelated to mechanism of action Clinical trials - Test the compound in humans to determine o tolerability o effectiveness o whether the compound elicits adverse effects o whether the compound elicits other beneficial effects Phase 1à in humans 2 3 4à Drug has been approved No. of Small group of healthy Larger group of healthy Large number of patients General population People volunteers (20-80) volunteers (several with the disease (several - How effective is the drug in the Enrolled Not always healthy, if hundred) or hundred to several general population? we are testing drugs Small group of diseased thousand) - Greater interpatient variability. such as chemotherapy patients - Not controlled environment. Study - Is the drug safe? - Is the drug safe? - How effective is the Larger population. Purpose - What are the - Dose - how much and drug (is it working as - Continued monitoring of safety adverse effects? how often? intended in the specific and adverse effects - Is the drug - How effective is the target population)? - Can uncover rare adverse effects tolerable? drug (is it working as - Often compared to due to larger population. - Pharmacokinetics/ intended in the standard intervention, - Drugs can be withdrawn if adverse Pharmacodynamics specific target another experimental effects are severe. population)? intervention or non- - May prompt changes in mitigation interventional standard strategies (e.g. changes in the care. drugs regulation such as labelling) Duration Several months Months to 2 years Continued monitoring of Forever adverse effects 1-3years

Use Quizgecko on...
Browser
Browser