💊Medicinal Chemistry Unit 2 – Pharmacokinetics

Pharmacokinetics explores how drugs move through the body, focusing on absorption, distribution, metabolism, and excretion. This field helps determine optimal dosing for effective and safe drug therapy, considering factors like administration routes and patient characteristics. Key concepts include bioavailability, volume of distribution, clearance, and half-life. Understanding these principles is crucial for drug development, personalized medicine, and managing drug interactions in clinical practice.

What's Pharmacokinetics?

  • Pharmacokinetics studies how the body processes drugs and other substances over time
  • Encompasses absorption, distribution, metabolism, and excretion (ADME) of drugs
  • Quantifies drug concentration changes in different body compartments
  • Helps determine optimal dosing regimens for therapeutic efficacy and safety
  • Considers factors like route of administration, drug formulation, and patient characteristics
    • Examples include oral, intravenous, or topical administration
    • Formulations can be immediate-release or extended-release
  • Pharmacodynamics, in contrast, studies the biochemical and physiological effects of drugs on the body
  • Mathematical models describe pharmacokinetic processes and predict drug behavior
  • Clinical applications include drug development, personalized medicine, and drug interaction studies

Key Concepts and Terminology

  • Bioavailability: fraction of administered drug that reaches systemic circulation unchanged
    • Influenced by factors like first-pass metabolism and drug solubility
  • Volume of distribution (Vd): theoretical volume needed to contain the total amount of drug at the same concentration found in plasma
  • Clearance: volume of plasma cleared of drug per unit time
    • Represents the efficiency of drug elimination processes
  • Half-life (t1/2): time required for drug concentration to decrease by half
  • Steady-state: equilibrium reached when drug administration and elimination rates are equal
  • Area under the curve (AUC): total drug exposure over time, calculated from a concentration-time graph
  • Maximum concentration (Cmax): highest drug concentration achieved after administration
  • Time to maximum concentration (Tmax): time to reach Cmax after drug administration

The ADME Process

  • Absorption: process by which a drug moves from the site of administration into the bloodstream
    • Influenced by factors like pH, surface area, and blood flow at the absorption site
  • Distribution: movement of drug from the bloodstream into various tissues and organs
    • Depends on drug properties (lipophilicity, protein binding) and tissue characteristics (perfusion, permeability)
  • Metabolism: chemical modification of the drug by enzymes, primarily in the liver
    • Biotransformation reactions (Phase I and Phase II) can activate or inactivate drugs
    • Cytochrome P450 (CYP) enzymes play a crucial role in drug metabolism
  • Excretion: removal of the drug and its metabolites from the body
    • Major routes include renal excretion (via urine) and biliary excretion (via feces)
  • The ADME process determines the onset, duration, and intensity of drug action
  • Understanding ADME helps optimize drug delivery and minimize adverse effects

Factors Affecting Drug Absorption

  • Physicochemical properties of the drug (solubility, lipophilicity, ionization)
    • Lipophilic drugs tend to have higher absorption rates
    • Ionization state depends on the pH of the environment and the drug's pKa
  • Route of administration (oral, parenteral, topical, inhalation)
    • Oral route is most convenient but subject to first-pass metabolism
    • Parenteral routes (intravenous, intramuscular) bypass absorption barriers
  • Formulation and dosage form (tablets, capsules, solutions, suspensions)
    • Disintegration and dissolution rates affect drug release and absorption
  • Physiological factors (gastrointestinal motility, pH, food intake)
    • Food can delay gastric emptying and alter drug absorption
    • Gastrointestinal diseases (Crohn's, celiac) can impair absorption
  • Drug interactions (chelation, adsorption, enzyme induction/inhibition)
    • Antacids can chelate drugs and reduce their absorption
    • Grapefruit juice inhibits CYP3A4, increasing bioavailability of some drugs

Drug Distribution in the Body

  • Plasma protein binding affects drug distribution and activity
    • Albumin and α1-acid glycoprotein are major drug-binding proteins
    • Only unbound (free) drug can exert pharmacological effects
  • Tissue perfusion and permeability determine drug access to target sites
    • Highly perfused organs (liver, kidneys) receive drug more rapidly
    • Blood-brain barrier restricts entry of many drugs into the central nervous system
  • Drug reservoirs can accumulate in certain tissues (fat, bone)
    • Lipophilic drugs tend to have larger volumes of distribution
    • Redistribution from reservoirs can prolong drug effects
  • Special populations may have altered distribution patterns
    • Elderly patients often have reduced muscle mass and increased body fat
    • Pregnancy can change drug distribution due to physiological adaptations
  • Drug transporters (P-glycoprotein, organic anion transporters) influence distribution
    • Efflux transporters can limit drug access to certain tissues
    • Influx transporters can facilitate drug uptake into cells

Metabolism and Biotransformation

  • Liver is the primary site of drug metabolism, but other organs also contribute
  • Phase I reactions (oxidation, reduction, hydrolysis) modify drug structure
    • Cytochrome P450 (CYP) enzymes catalyze many Phase I reactions
    • Examples include CYP3A4, CYP2D6, and CYP2C9
  • Phase II reactions (conjugation) attach polar groups to drugs or their metabolites
    • Common conjugation reactions include glucuronidation, sulfation, and acetylation
    • Conjugation generally increases drug water solubility and facilitates excretion
  • Genetic polymorphisms can affect enzyme activity and drug metabolism rates
    • Poor metabolizers may have higher risk of adverse effects or therapeutic failure
    • Ultrarapid metabolizers may require higher doses for therapeutic effect
  • Drug-drug interactions can occur through enzyme induction or inhibition
    • Inducers (rifampin, carbamazepine) increase enzyme activity and drug metabolism
    • Inhibitors (ketoconazole, erythromycin) decrease enzyme activity and drug metabolism
  • Prodrugs are inactive compounds that undergo biotransformation to active drugs
    • Examples include codeine (metabolized to morphine) and enalapril (metabolized to enalaprilat)

Elimination and Excretion

  • Renal excretion is the primary route of drug elimination
    • Glomerular filtration, tubular secretion, and tubular reabsorption determine drug clearance
    • Renal impairment can lead to drug accumulation and toxicity
  • Biliary excretion and fecal elimination are important for some drugs
    • Drugs and their metabolites can be secreted into bile and eliminated in feces
    • Enterohepatic recirculation can prolong drug presence in the body
  • Other minor elimination routes include sweat, saliva, and breast milk
  • Elimination rate constant (ke) and half-life (t1/2) characterize drug elimination
    • Ke represents the fraction of drug eliminated per unit time
    • t1/2 is the time required for drug concentration to decrease by half
  • Clearance (CL) is the volume of plasma cleared of drug per unit time
    • Total clearance is the sum of renal clearance, hepatic clearance, and other routes
    • Clearance determines the maintenance dose rate required to achieve steady-state concentrations
  • Dosage adjustments may be necessary for patients with organ dysfunction
    • Renal dosing adjustments based on creatinine clearance or estimated glomerular filtration rate (eGFR)
    • Hepatic dosing adjustments based on Child-Pugh classification or liver function tests

Pharmacokinetic Models and Equations

  • Compartmental models describe drug distribution and elimination
    • One-compartment model assumes rapid distribution and a single elimination phase
    • Two-compartment model includes a central compartment and a peripheral compartment
    • Multi-compartment models can be used for more complex drug behavior
  • Non-compartmental analysis uses statistical moments to estimate pharmacokinetic parameters
  • Absorption rate constant (ka) describes the rate of drug absorption into the systemic circulation
  • Elimination rate constant (ke) describes the rate of drug elimination from the body
  • Volume of distribution (Vd) relates the amount of drug in the body to the plasma concentration
    • Calculated as: Vd=Dose/(AUC×ke)Vd = Dose / (AUC × ke)
  • Clearance (CL) is the volume of plasma cleared of drug per unit time
    • Calculated as: CL=ke×VdCL = ke × Vd
  • Bioavailability (F) is the fraction of administered drug that reaches systemic circulation unchanged
    • Calculated as: F=(AUCoral/AUCIV)×(DoseIV/Doseoral)F = (AUC_{oral} / AUC_{IV}) × (Dose_{IV} / Dose_{oral})
  • Half-life (t1/2) is the time required for drug concentration to decrease by half
    • Calculated as: t1/2=0.693/ket_{1/2} = 0.693 / ke

Clinical Applications and Case Studies

  • Therapeutic drug monitoring (TDM) uses pharmacokinetic principles to optimize dosing
    • Drugs with narrow therapeutic indices (digoxin, lithium) require careful monitoring
    • TDM helps maintain drug concentrations within the therapeutic range
  • Pharmacogenomics considers genetic variations in drug metabolism and response
    • Genetic testing can guide drug selection and dosing for certain medications (warfarin, clopidogrel)
    • Personalized medicine aims to tailor drug therapy based on individual genetic profiles
  • Drug interactions can be predicted and managed using pharmacokinetic knowledge
    • Dose adjustments or alternative medications may be necessary to avoid adverse interactions
    • Examples include the interaction between digoxin and clarithromycin (increased digoxin levels)
  • Special populations require pharmacokinetic considerations
    • Pediatric and geriatric patients may have altered drug absorption, distribution, and elimination
    • Obesity can affect drug distribution and dosing requirements
    • Pregnancy and lactation involve unique physiological changes and safety concerns
  • Case studies illustrate the application of pharmacokinetic principles in clinical practice
    • Example: Adjusting vancomycin dosing based on renal function and serum concentrations
    • Example: Managing drug therapy in a patient with liver cirrhosis and hepatic encephalopathy


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.
Glossary
Glossary