Clinical pharmacokinetics
Basic principles and its
       applications
     S. M. Habibur Rahman
   Department of Pharmaceutics
    PSG College of Pharmacy
     Clinical Pharmacokinetics
• The science of the rate of movement of
  drugs within biological systems, as
  affected by the absorption, distribution,
  metabolism,    and     elimination     of
  medications
Why Study Pharmacokinetics (PK)
and Pharmacodynamics (PD)?
• Individualize patient drug therapy
• Monitor medications with a narrow
  therapeutic index
• Decrease the risk of adverse effects while
  maximizing pharmacologic response of
  medications
• Evaluate PK/PD as a diagnostic tool for
  underlying disease states
      Organization of Workshop
•   PK basic principles
•   ADME factors
•   Half life, Elimination Rate and AUC
•   Models
•   Hands on Experience
                                    Clinical (Human) Testing
Preclinical testing
                                                            Population PK/PD
                                     Dose response trials
                                                            characteristics in
                                                            large efficacy trials
                      PK-guided                                                Post-
                                        Efficacy
                      Dose                                                     marketing
                                                             PK/PD in
                      escalation                                               surveillance
  In vitro PK/PD                                             Special
                                        Dosage selection     populations
  Animal PK/PD         Safety
                       Assessment      Patient variables
  Toxicity
   Animal             Phase I             Phase II              Phase III
   testing
                  Significance
• Once the target enzyme or receptor is identified,
  medicinal chemists use a variety of empirical and
  semiempirical structure-activity relationships to modify
  the chemical structure of a compound to maximize its in
  vitro activity.
• However, good in vitro activity cannot be extrapolated to
  good in vivo activity unless a drug has good
  bioavailability and a desirable duration of action.
• Key role- Pharmacokinetics and drug metabolism -has
  led many drug companies to PK and drug properties as
  part of their screening processes in the selection of drug
  candidates.
                Absorption
• Able to get medications into the patient’s
  body
• Drug characteristics that affect absorption:
  – Molecular weight, ionization, solubility, &
    formulation
• Factors affecting drug absorption related
  to patients:
  – Route of administration, gastric pH, contents
    of GI tract
Physicochemical properties
                Transport
• Bickel (1994) have shown initial uptake of
  drugs into adipose tissue is related to their
  lipophilicity, the degree of adipose tissue
  storage does not correlate with their
  lipophilicity.
• Factors such as drug binding to plasma
  and tissue proteins also play a significant
  role in drug storage in adipose tissues.
              Pgp transport
• P-glycoprotein, located on the apical surface of
  the endothelial cells of the brain capillaries
  toward the vascular lumen (Tew et al., 1993;
  Pardridge, 1991), is believed to be responsible
  for the poor BBB penetration of some highly
  lipophilic drugs.
• The poor BBB penetration of drugs may be
  related to the efflux function of p-glycoprotein.
               Distribution
• Membrane permeability
  – cross membranes to site of action
• Plasma protein binding
  – bound drugs do not cross membranes
  – malnutrition = albumin =  free drug
• Lipophilicity of drug
  – lipophilic drugs accumulate in adipose tissue
• Volume of distribution
              Metabolism
• Drugs and toxins are seen as foreign to
  patients bodies
• Drugs can undergo metabolism in the
  lungs, blood, and liver
• Body works to convert drugs to less active
  forms and increase water solubility to
  enhance elimination
              Metabolism
• Liver - primary route of drug metabolism
• Liver may be used to convert pro-drugs
  (inactive) to an active state
• Types of reactions
  – Phase I (Cytochrome P450 system)
  – Phase II
                Elimination
• Pulmonary = expired in the air
• Bile = excreted in feces
  – enterohepatic circulation
• Renal
  – glomerular filtration
  – tubular reabsorption
  – tubular secretion
    Pharmacokinetic principles
• Steady State: the amount of drug
  administered is equal to the amount of
  drug eliminated within one dosing interval
  resulting in a plateau or constant serum
  drug level
• Drugs with short half-life reach steady
  state rapidly; drugs with long half-life take
  days to weeks to reach steady state
 Steady State Pharmacokinetics
• Half-life    =      time
  required for serum                 100
                                      90
  plasma                              80
                                      70
  concentrations         to     %     60
  decrease by one-half        steady 50
                               state 40
  (50%)                               30
                                      20
• 4-5 half-lives to reach             10
                                       0
  steady state                             1   2   3       4   5
                                               Half-life
            Loading Doses
• Loading doses allow
                          40
  rapid achievement of
                          35
  therapeutic    serum
                          30
  levels
                          25   w/ bolus
• Same loading dose       20
                               w/o
  used regardless of      15   bolus
  metabolism/eliminatio   10
  n dysfunction            5
                           0
    Linear Pharmacokinetics
• Linear = rate of
                                            120
  elimination          is
                                            100
  proportional         to
                            concentration
                                             80
  amount      of    drug
  present                                    60
                                             40
• Dosage       increases
                                             20
  result in proportional
  increase in plasma                          0
  drug levels                                     dose
 Nonlinear Pharmacokinetics
• Nonlinear = rate of
                                            50
  elimination is constant                   45
  regardless of amount                      40
                                            35
                            concentration
  of drug present                           30
• Dosage        increases                   25
                                            20
  saturate binding sites                    15
  and result in non-                        10
                                             5
  proportional                               0
  increase/decrease in                           dose
  drug levels
   Michaelis-Menten Kinetics
• Follows linear kinetics
                                            30
  until        enzymes
                                            25
  become saturated
                            concentration
                                            20
• Enzymes responsible                       15
  for        metabolism                     10
  /elimination become                        5
                                             0
  saturated resulting in
  non-proportional                                dose
  increase    in    drug                         phenytoin
  levels
   Special Patient Populations
• Renal Disease: same hepatic metabolism,
  same/increased volume of distribution and
  prolonged elimination   dosing interval
• Hepatic Disease: same renal elimination,
  same/increased volume of distribution,
  slower rate of enzyme metabolism  
  dosage,  dosing interval
• Cystic Fibrosis Patients:          increased
  metabolism/ elimination, and larger volume
  of distribution   dosage,  dosage interval
Pharmacologist role
      Clinical Pharmacokinetics
• Application of pharmacokinetic methods in drug
  therapy
• Optimized dosing strategies based on
   – Patient disease state
   – Patient specific consideration
• Influence of disease on drug disposition
   – Not adequately studied
• Age, Gender, genetic & ethnic factors
   – Pharmacokinetic difference
• Population approach
• TDM
Pharmacokinetics in drug development
Stage of development
Initial PK studies in Humans (decision Phase)
   – emphasis on safety and tolerance
      • Number of subjects limited but intensive
      • Descriptive evaluation of pharmacokinetics
      • Look for first evidence of concentration-effect
        relationships
         Goal: First-Time Knowledge About PK of the Drug
Pharmacokinetics in drug development
• Later PK Studies in Humans (Registration Phase)
   – Emphases on Expansion and Depth of Knowledge
      • Use number of subjects necessary to be definitive
      • Define concentration-effect relationships
      • Expand studies to wider population (gender, age, ethnic
        origin)
      • Link data to target population (population PK)
            Goal: Broaden Understanding, Special Populations
• Therapeutic drug monitoring (Commercialization Phase)
Key Pharmacokinetic Descriptive Variables
Half-Life, T½
                        CL = V X 0.693 / T½
Clearance, CL
Volume of Distribution, V
Primary Pharmacokinetic Measurements
   – Concentration (mass per volume), Cp
   – Rate constants (time-1), ka ke k12 λ β
   – Amount of Drug (mass), A Ae Dose
   – Area Under the Curve (integration of time and
   mass per volume), AUC
    Why estimate pharmacokinetics
• "Need to know" versus "Nice to know“
•FDA and other regulatory hurdles
•Absolute Bioavailability
  - Dosage form design
  - Bioavailability problems (F=5% or 95%)
  - Intersubject Variability (absorption vs DME)
•Estimate Rate Processes
   – Distinguish rate process from rate constant
Why estimate pharmacokinetics
 • Characterize drug exposure
       – time duration
       – degree of exposure
 • Predict dosage requirements
       – how much, how often
 • Assess changes in dosage requirement
       – special populations
       – drug interactions
      Why estimate pharmacokinetics
• Pharmacokinetic – Pharmacodymamic Relationships
      – Concentration effect relationships
      – Use PK to provide concentration when PD
        measurement is performed
      – Establish safety margins and efficacy
        characteristics
• Efficient and safe drug utilization
     Interrelationship
Key Pharmacokinetic factors
  Biopharmaceutics & Pharmacokinetics
                   Dynamic relationship
       Drug, Drug Product & pharmacologic effect
Drug release and         Drug in systemic
                                             Drug in Tissue
   dissolution              circulation
                          Excretion and     Pharmacologic or
                           metabolism         clinical effect
     Biopharmaceutical factors –
            Dosage form
• Protection of activity of drug within the
  drug product
• Release of drug
• Rate of dissolution
• Systemic absorption
Drug Disposition- drug interaction in body
                                                               Volatile
                                   BLOOD
                                                               drug in
    Oral        Adipose           Effector                     expired air
 Ingestion       Tissue            tissue
                Storage        drug receptor         Lung
                                  binding
                                                Peripheral
   DRUG         DRUG                             tissues
                                                metabolism
                                     Liver          Kidney
              Drug - Plasma          Drug
             Protein Complex       Metabolism                  Drugs and
                                                               metabolites
                                      Bile                     in urine
INTESTINE       Intestinal                       Drugs and
                reabsorption                     metabolites
                                                 in stools
              Hard drugs
• Nonmetabolizable drugs.
• Not only does it solve the problem of
  toxicity due to reactive intermediates or
  active      metabolites,        but   the
  pharmacokinetics also are simplified
  because the drugs are excreted primarily
  through either the bile or kidney.
• Eg. ACE inhibitors and bisphosphonates
                 Soft drugs
• A soft drug is pharmacologically active as such,
  and it undergoes a predictable and controllable
  metabolism to nontoxic and inactive metabolites.
• The main concept of soft drug design is to avoid
   oxidative metabolism as much as possible and
  to use hydrolytic enzymes to achieve predictable
  and controllable drug metabolism.
  Eg. Atracurium
           Drug Disposition
• BCS – system
• BDDS
  – Metabolism
  – Solubility
  – Permeability
          BCS & its Application
• The Biopharmaceutical Classification System
  (BCS) is based on solubility tests, correlating for
  drugs with their bioavailability in human body.
• It is widely used in design and development of
  innovation drugs
• New dosage forms (permeability amplifiers)
• In clinical pharmacology (drug-drug, drug-food
  interaction)
• Regulation agencies of several countries as the
  scientific approach, for testing of waiver on
  bioavailability.
 PHARMACOKINETIC CHARACTERISTICS
        OF BIOAVAILABILITY
Bioavailability is based on the physiological
process of absorption, which include three stages
•    Transfer a substance through apical plasma
membrane inside cells
•    Intracellular transport of substances followed
by their possible metabolism
•    Transfer of the transported and transformed
substance from cells into blood or lymph
  Characteristics of absorption & bioavailability
                    processes
       Absorption                 Bioavailability
Strictly corresponds        Corresponds to an API dose
to API dose                 and clearance value
In some cases corresponds   Strictly corresponds
to a therapeutic effect     to therapeutic effect
Depends on permeability     Depends on both API
of corresponding bio        entrance to blood circulation
membranes                   and elimination from it
(enterocytes)
The effect of food on API absorption
    BCS      The effect of food   Action mechanism
    class    on bioavailability
                parameters
I           Reduction of rate  Decrease of GIT
            but not duration   evacuation
2           Reduction of rate  Decrease of solubility
(Bases)     but not duration   due to the increase
                               of gastric pH
2 (Acids) Increase of the rate Increase of solubility
          and possibly         due to the increase
          duration             of gastric pH
3         The effect is not
          observed
The ratio of solubility/permeability
  parameters in BCS classes
Measurement of Drug Concentration
    • Milk                  ▪ Saliva
    • Plasma                ▪ Urine
Sampling of biological specimens
• Invasive Method
  – Sampling Blood, Spinal Fluid, synovial
    fluid, Tissue biopsy
• Non invasive Method
  – Sampling urine, saliva, feces, expired air
         Pharmacokinetic Model
• Quantity study of various kinetic process
  of drug disposition in the body
• Biological nature of drug distribution and
  disposition is complex and drug events
  often happen simultaneously
           Steps in modeling
• Model development
• Model characterization, i.e. methods to describe
  how consistent the model is with biology;
  strengths and limitations of available model and
  data, such as sensitivity analyses,
• Model documentation,
• Model evaluation, i.e. independent review
  Basic pharmacokinetic model
• Various mathematical model can devised to
  simulate the rate process of drug ADE
• development of equations useful in describing
  drug concentration in the body as a function of
  time
• Predictive capability of model lies in the proper
  selection & development of mathematical
  function (s) that parameterize the essential
  factors governing the kinetic process
             Variables in model
• Key parameters in a process is commonly
  estimated by fitting the model to the
  experimental data
• Pharmacokinetic     function   relates  an
  independent variable (time) to a dependent
  variable (response)
• Types of model
   – Empirical or Physiological
      • Empirical models are practical but not very useful
        in explaining the mechanism of the actual process
        of ADME in the body is not possible
        Pharmacokinetics
     Pictorial and Graphical
      Understanding of the
  Shapes of Concentration Time
             Profiles
Mathematical Models that describe
  and track these time profiles
Concentration profile depends - On
• Route of Administration
  – Intravenous (bolus, infusion)
  – Extravascular (oral, IM, SQ)
  – Specialized
• Disposition of the drug (ADME)
  – distribution
  – metabolism
  – elimination
Pharmacokinetic Variability
Compartment models
        Compartment Models
• Well stirred model
• Based on assumption using linear
  differential equation
• Provides simple way of grouping all the
  tissues into one or more compartments
Types
• Mammilary Model and catenary model
IV Bolus One Compartment
IV Bolus Two Compartment
Oral One Compartment
Oral One Compartment
Compartmental Model
Oral Two Compartment
Oral Two Compartment
      compartment model
One compartment open model IV Injection
               Central      Ke
             compartment
   One compartment open model with
         first order absorption
      Ka        Central       K
              compartment
      Multi compartment model
     Two compartment open model IV Injection
                        K12
           Central              Tissue
         compartment          compartment
                        K21
              k
       Two compartment open model with
             first order absorption
                        K12
ka         Central              Tissue
         compartment          compartment
                        K21
              k
  Functions of Drawing Models
• To write differential equations to describe
  drug concentration changes in each
  compartment
• Visual representation of rate process
• Shows how many pharmacokinetic
  constants are necessary to describe the
  process adequately
Deficiencies of compartmental analysis
•   Lack of meaningful physiological basis for derived
    parameters
•   Lack of rigorous criteria to determine No of
    compartments necessary to describe disposition.
•   Lack of ability to elucidate organ specific elimination
•   Inability to relate derived parameters to quantifiable
    physiological parameters
•   Inability to predict impact of pathophysiology
•   Inability to provide insight into mechanism of drug-drug
    and drug-nutrient interactions
•   Highly sensitive to sampling frequency
 Physiological compartment Model
• Blood flow or perfusion model
• Describes the data with the consideration
  that blood flow is responsible for
  distributing drug to various part of body
• Uptake of drug into organs is determined
  by the binding of drug in these tissues
• Tissue volume describes the drug
  concentration
                     PBPK
• Experimentally difficult
• In spite of this limitation the PBPK model does
  describe much better insight into hoe physiologic
  factor may change drug distribution from one
  animal species to another
• No data fitting is required
• Drug concentration is predicted by organ tissue
  size, blood flow & tissue – blood ratio (partition)
• The above facts may vary due to
  Pathophysiologic condition
Physiologic pharmacokinetic model
           (Flow Model)
                 From: Rowland M, Tozer TN. Clinical
                 Pharmacokinetics – Concepts and
                 Applications, 3rd edition, Williams
                 and Wilkins, 1995, p. 12.
     Models in Toxicokinetics
• There is no single method or model that
  can extrapolate the toxicity from animals to
  humans (Boxenbaum et al., 1988), the
  species differences in toxicity often can be
  explained     by     pharmacokinetic      or
  pharmacodynamic effects of drugs.