1 / 19

The Ideal DMPK Profile versus Lead Optimization

Clinical DMPK Profile. Rationale. High oral bioavailability: Half-life between 12 and 24 hr: Multiple elimination pathways: No reactive metabolites: No human-specific metabolites: No inhibition of CYP450 enzymes: No induction of CYP enzymes:. Low inter-subject variability/ cost of goods

dara
Download Presentation

The Ideal DMPK Profile versus Lead Optimization

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Clinical DMPK Profile Rationale High oral bioavailability: Half-life between 12 and 24 hr: Multiple elimination pathways: No reactive metabolites: No human-specific metabolites: No inhibition of CYP450 enzymes: No induction of CYP enzymes: Low inter-subject variability/ cost of goods QD dosing/ acceptable accumulation Drug-drug interactions (DDI) less likely Avoid safety issues/ idiosyncratic AEs Simplifies safety program & risk assessment Drug unlikely to cause DDIs Avoid autoinduction or DDIs Drug Safety DMPK Profile Good PK with developable form: Acceptable exposure multiples: Stable and predictable exposure: Clean in AMES test: Crystalline form - reduced bioavailability? Human risk assessment Reliably target appropriate exposure Avoid mutagens The Ideal DMPK Profile versus Lead Optimization

  2. Discovery DMPK In Vitro Assays Assay Rationale CYP 450 Inhibition: CYP 450 Profiling: Rat liver slice - induction: hPXR : Hepatocyte clearance: Inter-species hepatocyte met ID : Plasma stability: Plasma protein binding: Mock hERG drug assay: Caco-2: Avoid drug-drug interactions (DDI)? Avoid Polymorphism? Avoid DDI? Predict in vivo rat enzyme induction. Predict CYP 3A4 induction in humans. Predict human metabolic rate. Look for human-specific metabolites. Ex vivo degradation? Normalize exposure based on ‘free’ drug Confirm actual concentration. Predict human absorption potential.

  3. Study Rationale Predict human PK Full” PK (IV & PO): Adequate exposure for safety testing Single-rising dose: Steady-state; tumorigenic induction 14-Day Rat multiple dose: Absorption and circulating metabolites ‘Hot’ PK (3H-SCH) : Identify metabolites in ‘safety’ species Metabolite Pathway Elucidation: Avoid unusual retention of metabolites Mass Balance of 3H-SCH: Melanin Binding (LE rats): Assess phototoxicity potential PK/PD: Estimate efficacious drug exposure Developable form acceptability: Adequate exposure for safety testing Discovery DMPK In Vivo Studies

  4. Pharmacodynamics (PD) Effect vs Concentration Pharmacokinetics (PK) Concentration vs Time PK/PD Effect vs Time Pharmacokinetics and Pharmacodynamics Assumption: The magnitude of the desired effect (or side effect) is a function of the drug concentration at the site of action

  5. Drug excretion (Metabolism-Elimination) Drug at Effect Site Drug at Absorption Site Drug in Tissues (Distribution) Response!! Drug at Effect Site Pharmacodynamics Pharmacokinetics PK-PD relationship PK What your body does to the drug PD What the drug does to your body

  6. Different potency in different species

  7. The test compound has a full effect at the dose of 10 mg/kg po Pharmacological effect Plasma level 16 cutoff 800 14 700 ng/ml 12 600 10 500 8 400 6 300 4 200 2 100 Veh 0 0 10 mpk, po 0 30 60 120 180 240 360 Time (min) At 10 mg/kg po the temporal profile of the plasma level and the pharmacological activity profile are similar The PK-PD relationship

  8. Effect on the principal physiological system (cardiovascular, renal, nervous, etc). Safety Safety pharmacology Toxic effects could mechanism-based or compound-based. • To establish : • safe dose • MTD (maximum tolerated dose) • terapeutic window • target organs • Reversibility of toxicity Precinical toxicology Safety and preclinical toxicology

  9. Preclinical toxicology • Acute toxicity profile • Chronic toxicity profile • 14 days toxicity test in one rodent and one non-rodent species before use in man. • 3 months study read out at 28 days • longer studies (12 & 24 month) • Mutagenicity tests in vitro and in vivo

  10. Tolerability Therapeutic effect Sedation Ansiolitic effect What a therapeutic window is? Toxicity Respiratory depression

  11. It is possible to obtain a clear separation between antithrombotic and bleeding effects P2Y12 antagonists Therapeutic index evaluation (van Giezen and HumphriesSemin Thrombosis Hemost2005)

  12. Reasons for Failure in Development

  13. Moving from Animals to Man • Humans and animals have different biochemistry, physiology and anatomy • Predictions of a drug’s PK profile in humans using animal PK data must account for these differences • Allometric scaling is used to predict differences based only on size. • The relationship of some PK parameters across species can be correlated with body weight. • One can determine an empirical relationship log PK parameters and log Body Weight • These parameters can be used to extrapolate PK parameters in humans when parameters have been determined in lower species (mouse, rat, dog, monkey, etc.) • The relationship is not always predictive, but it can often give a good estimate.

  14. Summary The information collected in ADME and DMPK study are necessary to establish the dose that will be used in human

  15. Questions??

  16. Pre-marketing Registration R&D process for a new drug CANDIDATE POC DRUG Exploratory development Full development Developpability Safety Therapeutic efficacy Fase 0 or Preclinical development Fase II Study in the patient Fase III Study in the patient Fase I (A and B) Fase IV Post marketing Surveillance

  17. Formulation study • Pharmaceutical form: tablet , capsule, cream, injection, etc • To achieve the best effect is necessary to identify not only the • best form but also the most suitable formulation. • Example of composition of a tablet: • Active principle, filler, binder, • lubricant, disintegrant, surfactant.

  18. Objectives of Clinical Trials • Phase I: First in man  safety e tolerability • Phase II: First in patient IIa  safety and tolerability IIb dose, dosage form • Phase III: Value (is better than existing treatments) • Post marketing surveillance or Phase IV : Monitor the drug in the real clinical setting

  19. Clinical trials Uncontrolled Controlled Randomized Open or blind Sequential or cross-over

More Related