1 / 68

Predictive value of PK/PD drug modelling: application to analgesic drugs

ECOLE NATIONALE VETERINAIRE T O U L O U S E. Predictive value of PK/PD drug modelling: application to analgesic drugs. PL Toutain UMR 181 Physiopathologie et Toxicologie Expérimentales INRA, ENVT. Satellite symposium: Validity and Quality of Animal Models for Measurement of Pain.

lavi
Télécharger la présentation

Predictive value of PK/PD drug modelling: application to analgesic drugs

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. ECOLE NATIONALE VETERINAIRE T O U L O U S E Predictive value of PK/PD drug modelling: application to analgesic drugs PL Toutain UMR 181 Physiopathologie et Toxicologie Expérimentales INRA, ENVT Satellite symposium: Validity and Quality of Animal Models for Measurement of Pain

  2. Objectives of the presentation • Overview on the concept of PK/PD • Predictive value of PK/PD modeling for analgesics

  3. What is PK/PD modeling? • PK-PD modeling is a scientific tool to quantify, in vivo, thekey PD parameters (efficacy, potency and sensitivity) of a drug, which allows to predict the time course of drug effects under physiological and pathological conditions (intensity and duration)

  4. What are the main practical applications of a PK/PD trial • Preclinical investigations: It is an alternative to dose-titration studies to discover a dosage regimen • Clinical setting: • It is a tool to optimize dosage regimen in a clinical setting (pop PK/PD)

  5. 1-An overview on the concept of PK/PD

  6. Dose titration Dose Response Black box PK/PD Response PK PD Dose Plasma concentration

  7. Why is plasma concentration profile a better explicative (independent) variable than dose for determining a dosage regimen ?

  8. Dose vs. plasma concentration profile as independent variable Dose Dose F% Clearance Time X Mass (no biological information) Concentration profile (biological information)

  9. Why to prefer a PK/PD approach to a classical dose-titration?

  10. PD PK The determination of an ED50 or any ED% ED50 = ED50 - is a hybrid parameter (PK and PD) - is not a genuine PD drug parameter Clearancextarget EC50 Bioavailability

  11. The 3 structural PD parameters: Dose titration (DT) vs. PK/PD Slope Sensitivity ED50/EC50 Emax 1 1 1 Emax 1 2 2 Emax 2 steep Emax/2 shallow 2 • Range of useful concentrations • Selectivity ED501 ED502 Efficacy Potency

  12. Why to prefer a PK/PD approach to a classical dose-titration? • The separation of PK and PD variability

  13. PK/PD variability • Consequence for dosage adjustment PK PD Effect BODY Receptor Dose Plasma concentration Kidney function Liver function ... • Clinical covariables • Pain severity or duration PK/PD population approach

  14. 2-Predictive value of PK/PD for analgesics

  15. Predictive value of PK/PD modeling rely on: • The question: • Mechanistic question vs. Clinical drug development • Selection of a pain model & In life validation of the selected model • Appropriate study design & conduct • Appropriate PK & PD data • Appropriate PK/PD modeling • Population PK/PD (clinical setting)

  16. The question: a mechanistic question Drug discovery

  17. Questions for a veterinary rational drug development: find an optimal dosage regimen for a target species • What is the typical Dosage regimen • Time information and decision • Onset of drug action: fentanyl vs. morphine • Duration of drug action: time of remedication ( Dosage interval) • Extrapolation • Between species • assumption of the same PD parameters • Within the same species: between route of administration • Assumption: different PK profile but same qualitative metabolic profile • Dosage adjustment • Population investigations

  18. 2-Selection of a pain model: experimental pain models vs. clinical pain for PK/PD investigations

  19. Pain model selection for PK/PD investigation: value & validity • Validity: • to be discussed by the pain’ specialist • refers to whether a study is able to scientifically answer the questions it is intended to answer • Regarding the ultimate objective: • To investigate neurophysiologic mechanisms of pain or complicate drug mechanism of action • Preclinical determination of a dosage regimen • Simple but reproducible antinociceptive model are often sufficient • Validityof a model =capacity to find a useful dose • Value: • to be demonstrated by the PK/PD trialist

  20. Pain model selection for PK/PD investigation: value & validity • Validity • Value • Ethical • Metrological performances • Reliable • Sensitive • Robust & transferable • Convenience • Etc.

  21. Models using pressure noxious stimulus or thermal noxious stimulus are considered as valuable in veterinary medicine to approximate a starting dose

  22. Inflammatory pressure noxious stimulus. (here a kaolin inflammation model)

  23. Measure of vertical forces exerted on force plate • To measure the vertical forces, a corridor of walk is used with a force plate placed in its center. • The cat walks on the force plate on leach. Video

  24. Measure of vertical forces exerted on force plate • The measure of vertical force and video control are recorded • Vertical forces (Kg) Video

  25. Measure of pain with analgesiometer • The time for the cat to withdraw its paw of the ray is measured. • withdrawal time of the paws (second) • Sensitive and specific model to activate C-fibers Video

  26. Validation of the selected model

  27. Validation of the model • A priori validation makes sure the method is suitable for its intended use • When developing a new method • In life validation (routine validation for any new trial) • Animal selection • Investigator skill • Reproducibility & repeatability of selected animals • etc

  28. Validation of the model is tedious

  29. Predictive value of PK/PD modeling rely on: • The question: • Selection of a pain model & In life validation of the selected model • Appropriate study design & conduct • Crossover design and placebo period • Appropriate PK & PD data • Appropriate PK/PD modeling • Population PK/PD (clinical setting)

  30. 4-Appropriate data for PK/PD modeling

  31. Full concentration time curve experimental setting Cmax , Cmin Clinical setting Biomarkers Surrogate Clinical outcomes Measuring variables in PK/PD trials Measuring drug exposure Measuring drug response

  32. Measuring exposure • Generally straightforward. • May be more complicate if: • presence of an active metabolite • Tramadol • Racemates • Profens

  33. Tramadol plasma concentration (ng/mL) vs. time (min) after an IM administration of tramadol (circa 8 mg/kg);

  34. pharmacokinetics of (±)-trans-T and M1 are stereoselective in vivo • Trans-tramadol [(±)-trans-T] hydrochloride is a chiral compound • (+)-, (-)-Trans-T take as the action mainly through inhibiting the reuptake of serotonin and norepinephrine, respectively • The drug is metabolized in the liver to form five phase I metabolites, with the main pathways (in man and rats) being O-demethylation to O-demethyltramadol (M1) • Among the metabolites, M1 is an only active metabolite, and (+)-M1 has a high affinity to the opioid receptor

  35. Pharmacodynamic parameters of tramadol in the rat

  36. Tramadol and tramadol metabolite M1 concentration (ng/mL) vs. time (min) in 8 dogs after an IM administration of tramadol (circa 8 mg/kg) ; Spaghetti plot; semilogarithmic scale No CYP2D6 in dogs but an ortholog i.e CYP2D15

  37. Plasma concentrations of R- and S-ketoprofen after intramuscular administration of ketoprofen ( 6 mg/kg)

  38. Time development of the plasma concentration of ketoprofen and the mechanical nociceptive thresholds before kaolin injection (negative control), after kaolin injection (positive control) and after ketoprofen administration R-keto S-Keto Kaolin Nociception EC50 R-keto=2.0±05 µg/mL S-ket=38.8±10.8 T. K. FOSSE et al JVPT in press

  39. Full concentration time curve AUC Cmax , Cmin Biomarkers Surrogate Clinical outcomes Measuring variables in PK/PD trials Measuring drug exposure Measuring drug response

  40. EC50 in vivo effect EC50action whole blood assay Inhibition of PGE2 production Suppression of lameness Inhibition of COX NSAID plasma concentration Requires 90% PGE2 inhibition EC50 response Which dependent variable for PK/PD modeling ? EC50 response >> EC50 effect

  41. 5-PK/PD modelling

  42. Modeling options regarding presence or not of a delay between PK and PD time development Emax x Cobserved EC50 + Cobservedl No PK modeling E = NO Emax x C(t)model EC50 + C(t)model PK modeling E = PK and PD delay Effect compartment model PK origin YES Indirect response model PD origin

  43. Thermal threshold Plasma Fentanyl • No hysteresis for fentanyl • Direct incorporation of plasma fentanyl concentration in an Emax model

  44. hysteresis loop IV Oral ΔT(ºC) Buprenorphine concentration

  45. Modeling strategies when there is a delay of PK origin

  46. The “effect compartment model” Dose Cp(t) Effect(t) Ce(t) Ke0 Ke0 Effect Concentration Effect effect Time Ce Time K10 3:PD model Parametric (Emax, Hill) Non parametric (spline) 1:PK model Parametric (Exponential) Non parametric (Spline) 2:Link model Ke0 Estimation of EC50 and Ke0

  47. A mechanistic class of PK/PD models

  48. An example of dose determination using a PK/PD modeling approach:Tramadol in dogs

More Related