You are on page 1of 15

Pharmacokinetics Drug Movement or Transfer

[Ma. Fidelis Espiritu-Quiza, MD, FPCP]  Paracellular diffusion through intercellular gaps
 Transmembrane transfer
2 branches of Pharmacology  Passive membrane transport
 Pharmacokinetics  Carrier-mediated membrane transport
- “what the body does to the drug”
 Pharmacodynamics Physiochemical Factors in Drug Transfer Across
- “what the drug does to the body” Membranes
 All drug movement with the body requires
Pharmacokinetics: The Dynamics of Drug Absorption, passage of drugs cell across membranes
Distribution, Metabolism and Elimination  Determinants to drug transfer:
 Physiochemical factors in transfer of drugs across - Physiochemical properties of drugs
membranes - Physiochemical properties of the
 Drug absorption, bioavailability and routes of membranes
administration
 Extent and rate of bioavailability Drug Characteristics that Determine Drug Transfer and
 Distribution of drugs Availability at Sites of Actions
 Metabolism of drugs  Molecular size
 Excretion of drugs  Molecular shape
 Degree of ionization
 Clinical pharmacokinetics  Relative lipid solubility of ionized and unionized
 Design and optimization of drug regimens forms
 Therapeutic drug monitoring  Binding to serum and tissue proteins

Pharmacokinetics Overview Barriers to Drug Movement


 Routes of drug administration  Single layer of cells
 Absorption of drugs  Several layers of cells with extracellular protein
 Drug distribution  Plasma membrane
 Drug clearance through metabolism - Common barrier to drug distribution
 Drug clearance by the kidney
 Clearance by other routes
 Design and optimization of dosage regimen
Cell Membrane Characteristics that Affect Drug Transfer
 Fluidity
 Flexibility
 Highly organized
 High electrical resistance
 Impermeable to highly polar molecules
 Relatively permeable to water
- by diffusion
- by flow resulting from hydrostatic or osmotic
differences across the membrane pressure
- Bulk flow of water can carry drug molecules
- Most large lipophilic drugs pass through cell
membrane
 Most large lipophilic drugs pass through cell
membrane
 Transmembrane movement is limited to
unbound drug (free drug)
 Proteins with bound drug are too polar and too
large for simple diffusion
 Presence of membrane proteins
 May provide selective targets for drug
actions
- Receptors Passive Membrane Transport
- Ion channels  Ion Trapping
- Transporters - An acidic drug will accumulate on the
more basic side of the membrane
- A basic drug will accumulate on the more
acidic side of the membrane
- Clinical implication: enhance absorption
and excretion of drugs

Passive Membrane Transport


 Drug molecules penetrate by diffusion along a
concentration gradient due to solubility in the
lipid bilayer (Fick’s Law of Diffusion)
 Directly proportional to:
- Magnitude of concentration gradient across
the membrane
- Lipid-water partition coefficient of the drug
- Membrane surface are exposed to the drug
 For ionic substances depend on:
- Electrochemical gradient for the ion
- Differences in pH across the membrane
 Transmembrane distribution of weak electrolytes
determined by:
- pKa of substance
- pH gradient across the membrane
Carrier- Mediated Membrane Transport
 Facilitated diffusion
- Carrier-mediated transport not requiring
energy
- Moves along electrochemical gradient
- Highly selective for specific
conformational structure
- Mediate either drug uptake or drug efflux
(P-glycoprotein)
- Ex. Glucose entry across muscle
membrane facilitated by presence of
insulin-sensitive glucose transporter
protein

 Active transport
- Direct requirement for energy
- Movement against an electrochemical
gradient
- Saturability
- Selectivity
- Competitive inhibition by co-transported
compounds
- Subtypes:
o Primary active transport
 One type of substance is
transported
o Secondary active transport (co-
transporter)
 more than one type of
substance id transported
 Same direction (symport)
 Opposite direction
(antiport)
- Examples: Na , K +, -ATPase
+

- Na+- Ca2+ exchange protein


- Na+ dependent glucose transporters
(SGLT1 and SGLT2 in GIT and renal
epithelium)
 First-pass effect
- Reduction in bioavailability due to
inactivation or diversion of a fraction of
the administered and absorbed dose of
drug before it reaches the general
circulation and be distributed to its sites
of action
 A function of the anatomical site from which
absorption takes place
Picture
 Sites of first-pass metabolism:
- Intestinal microbial flora
- Gastrointestinal epithelium
- Liver
- Lungs Kidneys
 Pharmaceutical Equivalence
- Products contain the same active
ingredients and are identical in strength
or concentration, dosage form and route
of administration
 Bioequivalence
- Rate and extent of bioavailability of the
active ingredient of 2 products are not
much different under stable conditions
 Factors that affect bioavailability
- Differences in crystal form ad particle
size
- Conditions that alter the rate of
dissolution or disintegration of the
dosage form
- Different excipients or vehicles that may
alter pharmacokinetic properties of the
active ingredient

Pharmacokinetic Processes
 Drug Absorption
 Drug Distribution
 Drug Metabolism (Biotransformtation)
 Drug Elimination (Excretion)

Drug Absorption and Bioavailability


 Absorption
- The movement of a drug from its site of
administration into the central
Measurable site
compartment and the extent to which
this occurs
 Bioavailability Preferred is 1 or 100 percent
Iv administration

- The fractional extent to which a dose of


a drug reaches its site of action or a
biological fluid from which the drug has
access to its site of action

Picture
 Enteric coating of drugs:
- To prevent destruction or rapid
dissolution by gastric secretions
- To minimize local gastric irritation

Controlled-Release Preparations
 Controlled-release (CR), sustained- release (SR),
extended-release (XR), prolonged-action
 Potential advantages:
- Reduced losing frequency (favors
compliance)
- Sustained maintenance of therapeutic
effect over time
- Elimination of peaks and troughs of
plasma levels of the drug
 “Dose-dumping”
- Sudden release of all contents at one
time due to destruction of controlled-
release mechanism
- Resulting in toxicity due to higher dose
content than regular formulations
Picture
Major Routes of Drug Administration Major disadvantages of Oral route of Administration
 Oral (enteral) - Limited absorption due to physical
- Most common, safest, economical, and characteristics of the drug
convenient method - Emesis as a result of irritation to the GIT
 Parenteral mucosa
- More rapid availability to site of action - Destruction by digestive enzymes or low
- Utilized for emergency therapy, gastric pH
unconscious and/or uncooperative - Irregularities in absorption or propulsion
patients or those patients that cannot due to food or other drugs
retain anything by mouth - Need for patient cooperation
(compliance)
Oral Route of Drug Administration
 Absorption from GIT governed by:
- Surface area for absorption
- Blood flow to site of absorption
- Physical state of the drug (solid,
solution, suspension)
- Water solubility
- Drug concentration at site of absorption
 Most drug absorption from GIT is by passive
diffusion
 Absorption preferentially favored when drug is
non-ionized and more lipophilic
 Major site of drug absorption is in the small
intestine due to greater surface area regardless
of degree of ionization of the drug
 Factors that alter gastric emptying time will
affect rate of absorption in the SI:
- Presence of food
- Certain drugs with prokinetic activity of
the GIT
- Physiological state due to hormonal
effect (estrogen slowing of GIT transit
Sublingual Route of Administration - Avoidance of hepatic first-pass loss
 Venous drainage (plexuses of veins under the - Local application of the drug at the site of
tongue) from mouth direct to superior vena cava action especially for pulmonary diseases
 Circumvents first - pass metabolism in the GIT  Examples:
and the liver - Beta-agonists inhalation for treatment of
 Rapid drug absorption with rapid onset of action asthma
 For non-ionic and highly lipid soluble substances - Anesthetic gases
 Examples: - Corticosteroids as anti-inflammatory
- Nitroglycerin for angina or chest pain of agents for bronchial hyper-reactivity
cardiac origin
- Nifedipine for rapid decrease in BP Topical Route of Administration
 For local effects with ideally minimal systemic
Transdermal Route of Administration distribution
 Dependent on surface area and lipid solubility  Applied to mucous membranes of the following:
 Epidermis behaves as a lipid barrier - Conjunctiva
 Dermis is freely permeable - Nasopharynx
 Systemic absorption occurs more readily through - Oropharynx
abraded, burned, inflamed or denuded skin - Vagina
 Rate and extent of absorption is affected by rate - Colon
of subcutaneous blood flow or perfusion - Urethra
 Absorption may be enhanced by: - Urinary bladder
- Occlusive dressing  Sometimes used intentionally for systemic
- Hydration of skin absorption such as intranasal- insulin, ADH
- Suspending drug in oily vehicle  Sometimes associated with systemic toxicity due
 Controlled release preparations: to site characteristics which favor enhanced
- Nicotine for tobacco smoking withdrawal absorption
- Scopolamine for motion sickness  For the eyes, the site of absorption for local
- Hormonal replacement therapy effects is the cornea.
- Birth control preparations
Novel Routes of Administration
 Inserts or implants
- Ocular inserts
- Estrogen implants for prolonged release
 Drug
- Eluting stents in cardiac grafts
 Pumps
- Insulin pump

Pulmonary Route of Administration


 Inhalational for gaseous or volatile substances or
aerosolized particles
 Rapid access to circulation due to large surface
area of the lung
 Minimizes systemic distribution when in the right Parenteral Route of Administration
dose  Major routes
 Potential toxicity due to inhalation of toxic - Intravenous (IV)
chemical fumes - Subcutaneous (SC)
 May also result to local irritation of the - Intramuscular (IM)
respiratory tract - Intradermal (ID)
 Advantages include the following: - Intra-arterial
- Almost instantaneous absorption of drug - Intra-thecal
into the blood
 May be subject to first-pass elimination in the
lungs (except intra-arterial) Intra-arterial and Intrathecal Route
 Subcutaneous, intramuscular and intradermal  Intra-arterial
rate of absorption limited by: - For direct access to a specific organ
- Area of absorbing capillaries - Localized effects
- Solubility of substance in the interstitial - Treatment of organ tumors
fluid - Bypass first-pass effect of lungs
Intravenous (IV) Route of Administration  Intrathecal
 100% bioavailability (complete and rapid) - Into the meninges of the CNS
- Drug delivery is immediate and controlled - For direct access to the CNS especially
and achieved with accuracy since the CNS has tight junctions
- Ideal for irritating substances due to
dilution by blood Major disadvantages of Parenteral Route of
- High risk for acute adverse effects of the Administration
drug due to rapidity of onset  Need for maintenance of asepsis
- May be given as bolus or drip by “piggy-  Pain in injection site
back”  Difficulty of self-administration by the patient
 Drug characteristics that contraindicate use of IV  High risk for acute adverse effects of the drug
route  Less chances of drug removal once absorbed
- Oily vehicle
- Can precipitate blood constituents
- Can hemolyze blood elements
 Once administered, can no longer be withdrawn
 Warrants close monitoring during administration
and some period immediately after
administration

Subcutaneous (SC) Route of Administration


 For drugs not irritating to the tissue
 May cause pain, necrosis and sloughing off
 Constant and sustained release
 May be given as pellets under the skin
 May be given with vasoconstrictor agent to
retard absorption (epinephrine with lidocaine)

Intramuscular (IM) Route of Administration


 Drugs in aqueous solution
 Rate and extent of absorption may be influenced
by the following:
- Local heat applied to the area
- Activities that increase blood supply
(exercise/massage)
- Amount of fat tissue in the surrounding
area
 May be used for irritant substances
Drug Distribution
 Factors that affect the rate and extent of drug
distribution in the body:
- Physiochemical properties of the drug
- Physiological and pathological conditions
of the body
o Cardiac output
o Regional blood flow
o Capillary permeability
o Tissue volume
 Two phases of drug distribution  Potential for alteration in intensity of drug effects
1) First (rapid) phase due to changes in plasma protein binding and/or
 Well-perfused organs (liver, competition of binding sites
kidney, brain and heart)  Increase plasma protein may result in decreased
2) Second (slower) phase therapeutic efficacy due to increased binding
 Muscles, viscera, skin, fat  Decrease plasma protein (liver disease, renal
losses of albumin) may result in increased
therapeutic efficacy due to decreased binding
 Tissue binding of drugs
- Generally reversible
- Binding to cellular components
 Proteins
 Phospholipids
 Nuclear proteins

Drug Distribution in the CNS


 Drug penetration is transcellular rather than
paracellular due to presence of “tight junctions”
 Utilizes membrane transport proteins (influx and
efflux transporters)
 Blood-brain barrier
- Brain capillary endothelial cells and
pericapillary glial cells
 Lipid solubility of non-ionized and unbound drug-
clinical implication:
- Modification of ability to penetrate CNS
to limit adverse side effects of certain
drugs
 Meningeal and encephalic inflammation increase
local permeability
Drug Distribution
 Other clinically significant sites of drug
 Determinants of tissue distribution:
distribution
- Relative binding of drug to plasma
- Fat
proteins and tissue macromolecules -
- Bones and teeth
more important determinant
- Placenta
- Partitioning of drug between blood and
 Fetal plasma is slightly more acidic
tissue
than that of the mother – ion
- Lipid solubility
trapping of basic drugs
- Transmembrane pH gradients
- Milk
 Plasma protein binding is usually reversible
- Hair
 Occasional covalent binding
- Saliva, sweat
 Plasma proteins with which drugs bind:
o Albumin
Drug Metabolism (Biotransformation)
 For acidic drugs
 Major site of drug metabolism: LIVER
o ꭤ1- acid glycoprotein
 Other sites of drug metabolism
 For basic drugs
- GIT epithelium
o Specific proteins especially for hormones
- Lung endothelium
 Sex hormone-binding globulin
- Renal tubular cells
 Thyroid hormone-binding
 Lipophilic characteristics of drugs that promote
globulin
access to sites of action hinder their excretion
 Only unbound drug crosses membranes, are from the body
metabolized and excreted
 Main purpose: conversion of hydrophobic
 Only unbound drug binds to receptors substances to more hydrophilic substances in
 Fraction bound is a function of number of binding preparation for elimination from the body and
sites and drug concentration
 Plasma protein binding is nonlinear and saturable
termination of biological and pharmacological
activity
 In general: biotransformation reactions generate
more polar, inactive metabolites

 Maybe altered due to age, gender, genetic


variability, disease and xenobiotics
 Phases of drug metabolism
- Phase 1 (functionalization phase)
- Phase 2 (biosynthetic phase)
 PHASE 1 (functionalization phase)
- Introduce or expose functional group on
parent compound
- Generally, results in loss of
pharmacologic activity Drug Metabolism (Biotransformation)
- Enzyme systems primarily involved  Result of drug biotransformation
reside in the endoplasmic reticulum - Altered pharmacologic activity
- Enzymes groups  Active substance to inactive moiety
 Cytochrome P450 isozymes  Inactive drug (prodrug) to active
 Transferases molecule
 PHASE II (biosynthetic phase)  Toxic product to non-toxic
- Conjugation reactions metabolite
- Lead to formation of covalent linkages  Conversion of non-toxic substance
between a functional group on parent to toxic metabolite
compound or on results of phase I
reactions
- Glucuronic acid, sulfate, glutathione,
amino acids, or acetate
- Results to highly polar substances
- Enzyme systems primarily located in the
cytosol
 PHASE I (functionalization phase)
- Activity may be enhanced or inhibited
- Enzyme inducers
 Anticonvulsants (phenytoin,
barbiturates, diazepam)
 Rifampicin
- Enzyme inhibitors
 Metronidazole, erythromycin,
ketoconazole
 Cimetidine
Termination of Drug Effect  ABC transporters
 Metabolism - Organic cationic drugs (organic bases)
 Excretion  Tubular reabsorption – drug is passively
 Redistribution of drug from its site of action into reabsorbed back into the blood
other tissues or sites - Mostly by non-ionic diffusion
Excretion of Drugs - Some through action of membrane
 Drugs are eliminated either as unchanged form transporters
or as metabolites  Ion trapping
 Except from lungs, excretion of polar compounds
from excretory organs is more efficient than for Biliary and Fecal Excretion
substances with high lipid solubility  Active transporter secretion into the bile
 Most lipid soluble drugs are not readily excreted canaliculi
 Excretory organs:  Active transporter secretion from enterocytes of
 Kidney: major organ of excretion systemic circulation directly into the intestinal
 Lungs: importantly mainly for anesthetic lumen
gases  Enterohepatic recirculation
 GIT: unabsorbed orally ingested drugs or - Reabsorption of substances from the GIT
drug metabolites excreted either in bile lumen into the systemic circulation
or secreted directly into GIT and not - Responsible for prolonging duration of
reabsorbed action of a drug
 Skin - Ezitimibe:
 Breast milk  Reduces intestinal absorption of
cholesterol by interfering with the
Renal Excretion sterol transporter system in the
 3 major processes: intestinal epithelial cell- prolong
- Glomerular filtration duration of action
- Active tubular secretion - Toxicity potential:
 Predominantly in the  Reabsorption of toxic substances
proximal tubule already excreted into the GIT
- Passive tubular reabsorption lumen resulting in prolonged or
 Non-ionized forms of repeated manifestations of
weak acid and bases in toxicity
proximal and distal - Ex. Organophosphates (insecticide)
tubules  Enterohepatic recirculation
 Determinants to amount of drug entering renal responsible for intermittent
tubular lumen: recurrence of cholinergic toxicity
- Glomerular filtration rate  Repeat dose activated charcoal
- Extent of plasma binding of the drug; for adsorption with eventual
only unbound drug is filtered excretion into the feces
 Tubular secretion Excretion by Other Routes
- Drug is actively secreted mostly in the  Sweat, Saliva and Tears
proximal tubule  Depends mainly on:
 Active carrier-mediated process - Diffusion of the non-ionized lipid soluble
 Transporters: form of drugs through the epithelial cells
 P glycoprotein (P-gp) - pH
 Multi-Drug-Resistance-  Milk is more acidic than plasma
Associated Protein Type - Traps basic drugs, resulting to increased
2 (MRP2) delivery of basic drugs to infants
 ATB-binding cassette  Some excretion in hair and skin
(ABC) transporters
 P-gp and MRP2 Transporters
- Amphiphatic cations
- Conjugated metabolites (glucuronides,
sulfates and glutathione adducts
Clinical Pharmacokinetics
 Fundamental tenet:
- Relationship exists between the
pharmacological effects of a drug and an
accessible concentration of the drug
(blood or plasma)
 Attempts to provide both a quantitative
relationship between dose and effect and a
framework within which to interpret
measurements of concentrations of drugs in
biological fluids and adjustment through changes
in dosing for the benefit of the patient
 Importance in patient care: Clearance
- Improvement in therapeutic efficacy  Most important concept in designing a regimen
- Avoidance of unwanted effects can be for long term administration
attained by application of principles
 Refers to elimination of drug from systemic
when dosage regimens are chosen and
circulation
modified
 Important in assuring maintenance of drug levels
 Dose adjustment for individualization of patient
within steady-state within the therapeutic
therapy is dependent on physiological and window during the entire course of treatment
pathophysiological variables which affect
 Steady-state concentration:
pharmacokinetic parameters
- rate of drug elimination equals rate of
 4 most important parameters that govern drug
drug absorption
disposition:
 The amount of plasma cleared of 100% of the
 Clearance
drug per given unit of time (ml/min)
 Measure of body’s efficiency
 Indicates the volume of biological fluid (blood or
eliminating the drug
plasma) from which drug is completely removed
 Volume of distribution (Vd)
 Measure of apparent space
 Total systemic clearance:
in the body available to
CL= CL renal + CL hepatic + CL other
contain the drug
 Dosing Rate (maintenance):
 Elimination half-life (T 1/2)
Dosing rate = (CL * Target C ss)/ F
 Measure of rate of removal
 C ss refers to drug concentration in the body at
of the drug from the body
steady state
 Bioavailability
 First - order kinetics:
 Fraction of the drug
- Constant fraction of drug in the body is
absorbed in the systemic
eliminated per unit time
circulation
o Governs elimination of drugs
through non-saturable
mechanisms (enzymes and
transporters involved in
elimination are not saturated)
 Zero – order kinetics:
- Constant amount of drug is eliminated
per unit time
o Occurs when mechanisms of drug
elimination become saturated
FIRST ORDER ELIMINATION - Affects rate at which drugs can be
 The decline in Cp varies with the concentration transported in the blood to the liver
 The term “first order” reflects the fact that the - Affected by physiologic and disease
rate of elimination is dependent on conditions
concentration
 The concentration declines ‘exponentially’ with Faster flow  decreased rate of removal
time Slower flow  increased rate of removal
ZERO ORDER ELIMINATION
 Rate of elimination is constant with time Volume Distribution
 Constant amount of drug is eliminated with time  Relates the amount of drug in the body to the
concentration of the drug in the blood or plasma
 Does not refer to a real physiologic space
 Merely an apparent space or volume of fluid that
would be required to contain all the drug in the
body

Vd= amount of drug in the body (dose)/ Co


Initial dose = C * Vd
 Often provided by the drug developer
- Often provided as volume per kg body
weight (L/kg)
Clearance
 Rate of drug elimination of a particular organ is
the product of blood flow and the difference
between the arterial drug concentration entering
the organ and the venous drug concentration
exiting from the organ
 Extraction ratio:
- An expression of clearance of the drug
by a specific organ
(CA-CV)/ CA
- CA = arterial concentration of the drug
- CV = venous concentration of the drug

Hepatic Drug Clearance:


 Determinants of hepatic drug clearance:  Factors that determine the volume of distribution
1) The intrinsic clearance (Cl int.) of any given drug
2) Hepatic blood flow - Degree of plasma protein binding vs.
 Extraction Ratio: tissue protein binding
- Refers to the combined effect of the - Degree of binding to high-affinity
intrinsic clearance and rate of hepatic receptor sites
blood flow – proportion of administered - Extent or rate of blood flow or perfusion
drug that is removed by the liver to storage site
 Intrinsic Clearance (Cl int.) - Lipid solubility
- Refers to the liver’s inherent ability to - Body mass: fat content vs. body water
metabolize the drug - Amount of drug in other body storage
- Dependent on enzymatic activity compartment
- Activity may be decreased or increased - Age, gender
by the use of either the following: - Disease
o CYP 450 enzyme inhibitors HALF-LIFE (T1/2)
(cimetidine, metronidazole,  The time it takes for the plasma concentration or
erythromycin) the amount of the drug in the body to be
o enzyme inducers reduced by half (50%)
(phenobarbital, rifampicin)  Half-life of absorption
 Hepatic blood flow  Half-life of elimination
T1/2 = 0.693* (Vss/CL) THERAPEUTIC DRUG MONITORING
 Major use of measured concentrations of drugs
 Often provided by the drug developer for normal at steady state is to refine estimate of CL/ F for
physiologic conditions the patient being treated
 Affected by volume of distribution and clearance  Sampling time is important issue
parameters  If measurement is to determine attainment of
peak plasma concentrations within the
therapeutic window- soon after administration of
drug
----------------------------------------------------------------------------
 Loading dose: (oral) Target Cp * Vss/F
 Loading dose: (IV) Target Cp * Vss

 Dosing rate (oral): Target Cp * CL/F


 Dosing rate (IV): Target Cp * CL/F
----------------------------------------------------------------------------
 It takes four (4) half-lives of absorption for near
attainment of steady state  A 40-year-old male patient (70 kg) was recently
 It takes five (5) half-lives of elimination for near diagnosed with infection. He received 2,000 mg
100% removal of the drug from the body vancomycin as IV loading dose. The peak plasma
 Provides basis for dosing interval in the concentration of vancomycin was reported to be
administration of maintenance doses 28.5mg/L. What is the apparent V4?
T1/2 = 0.693 * (Vss/ CL)
 Directly proportional to volume of distribution Vd = dose/C
and inversely proportional to clearance  2,000 mg / 28.5 mg/L
 Half-lives constitute the basis for dosing interval  70.1 L
 Half-lives constitute the basis for time to achieve
steady-state  Because the patient is 70 kg, the apparent
 Half-lives constitute the basis for time for volume of distribution in L/kg will be
elimination of drug from the body approximately 1L/kg (70.1 L/ 70kg).

CHARACTERISTICS OF DRUGS ASSOCIATED WITH TDM


 Narrow therapeutic range
 Unpredictable dose/response relationship
 Significant consequences from toxicity
 Correlation between SDCs and efficacy or toxicity
 Readily available assays

INFUSION LENGTH
 The length of infusion has a significant effect
upon the peak serum level.
 If we were to administer the same dose to the
same patient at different infusion rates, the peak
levels would differ significantly

Infusion Dose Interval Peak level


(mins) (mg) (hrs) (mg/dl)
30 500 8 26.8
60 500 8 25.3
120 500 8 22.5
APPLIED PHARMACOKINETICS: Practical Considerations

 Choose practical, convenient doses and


administration schedules.
- Odd doses and schedules may lead to
errors in administration
 Consider a loading dose
- Some drugs, which are usually
administered without a loading dose,
may require a loading dose in order to
quickly attain therapeutic levels

Some reasons for variation of CL


Low CL High CL
Normal variation Normal variation
Renal impairment Increased renal blood
Genetic poor metabolism flow
Liver impairment Genetic
Enzyme inhibition hypermetabolism
Old age/ neonate Enzyme induction
Drugs with pH-dependent renal elimination

ACIDS BASES
 Elimination  Elimination
enhanced by enhanced by acid
alkaline diuresis diuresis
Amphetamines
Methadone
Phenobarbitone Mexiletine
Phencyclidine
Phenylpropanolamines,
Salicylates e.g. ephedrine,
pseudoephedrine
Quinidine

You might also like