• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/199

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

199 Cards in this Set

  • Front
  • Back
pharmacokinetics
describes the absorption, distribution, metabolism, and exretion of inhaled or injected drugs (ie. what the body does to the drug)
pharmacodynamics
describes the responsiveness of receptors to drugs, and the mechanism by which these effects occur (ie. what the drug does to the body)
what are receptors?
the components of the cell that interact with the drug to initiate a sequence of events leading to pharamcologic effects
what determines selectivity of drug action?
receptors that recognize specific drugs
how does termination of a drugs effect occur?
with metabolism, excretion, or redistribution to inactive sites
what is the definition of a drug?
a chemical compound that produces pharmacologic effects as determined by its pharmacokinetics and pharmacodynamics
differing pharmacokinetics
the pharmacokinetic characteristics of drugs in healthy and ambulatory adults may differ from those with chronic diseases (esp. renal or hepatic dysfunction), and at extremes of age, hydration, nutrition, and skeletal muscle mass
what does the pharmacokinetic profile of a drug highly depend on?
the characteristics of the nonionized and ionized fraction of that drug
characteristics of the nonionized drug fraction
they tend to be pharmacologically active and lipid soluble
and the ionized fraction?
is inactive and water soluble
what is the ionization of a drug a function of?
the pK of the drug and the pH of the surrounding fluid
what happens when the pK and pH are identical?
50% of the drug exists in the ionized form
what happens with small changes in pH?
can result in large changes in the degree of ionization, esp if the pH and pK are similar
what is an example of an acidic drug?
barbiturates
and a basic drug?
opioids and local anesthetics
when are acidic drugs, such as barbiturates, highly ionized?
at an alkaline pH
and basic drugs, such as opioids and local anesthetics?
highly ionized at an acidic pH
what happens with ionized and nonionized drugs at lipid barriers?
nonionized drugs cross lipid barriers, such as renal tubules, GI tract, placenta, blood-brain barrier); ionized drugs dont cross
which is renally excreted, and which are metabolized hepatically?
ionized drugs tend to be renally excreted, and nonionized undergo hepatic metabolism
what else does lipid or water solubility determine?
absorption and elimination characteistics of drugs
definition of absorption
the rate at which a drug leaves its site of administration
membrane transport of drugs
occurs when drugs cross cell membranes by simple diffusion, by facilitated or carrier-mediated transport, and by active (energy-dependent) transport
simple diffusion
a bidirectional process in which the rate of transfer of a drug is proportional to the concentration gradient
what do highly lipid soluble and low-molecular weight drugs do at cell membranes?
diffuse passively across cell membranes
and large molecular weight drugs?
are transported across cell membranes by a carrier-mediated process, or an active transport system requiring energy
factors affecting drug absorption
particle size, lipid solubility, drug concentration, type of drug (crystalloid vs. colloid), area of absorption and local blood supply
absorption of tablets with a large particle size
may not disintegrate easily, and will have poorer absorption
absorption of drugs with high lipid solubility
will easily pass through membranes
absorption and concentration of drugs
a higher concentration of drug will favor its absorption
absorption of crystalloids vs. colloids
liquids or crystalloids are usually better absorbed, (ie. better solubility) than solids or colloids
absorption and local blood supply
a larger area of absorption and better local blood supply favor absorption
what do heat and vasodilation do to absorption, vs. shock and vasoconstriction?
heat and vasodilation increases absorption, whereas shock and constriction decrease it
ionization and absorption
the degree of ionization of a drug affects its absorption so that nonionized drugs, which are often lipid soluble, are absorbed rapidly, whereas ionized drugs tend to be water-soluble and are absorbed poorly
site of absorption of drugs
acidic drugs tend to be absorbed readily from the stomach, whereas basic drugs tend to be readily absorbed from the alkaline environment of the intestine
and finally, what is the last factor that determines absorption of drugs?
the route of administration
route of administration and absorption
IV administration of drugs allows for a predictable plasma concentration, whereas oral and IM injection of drugs often results in unpredictable absorption and is dependent of local blood flow
absorption of drugs from the GI tract
drugs absorbed from the GI tract, principally the small intestine, enter the portal venous blood, and pass through the liver before entering the systemic circulation and delivery to tissue receptors, known as first-pass hepatic metabolism
first-pass metabolism
for drugs that undergo extensive hepatic metabolism (eg. propranolol, lidocaine) this is the reason for such a large difference between effective oral and IV doses (ie. drug gets delivered to the receptors without first passing through the liver)
in addition to the liver, what other organ may play an important role in pharmacokinetics?
the lungs
pulmonary metabolism
the effect of the lungs on pharmacokinetics is reflected by basic lipophillic amines (eg. lidocaine, propranolol, and fentanyl) into lung tissue; the first-pass pulmonary effect may influence the peak arterial concentration of these drugs, and the lungs may subsequently serve as a reservoir to release drug back into systemic circulation
what does the passage of a drug through cell membranes depend on?
the pH of the drugs environment, the degree of ionization, the dissociation constant of the drug (pKa), protein binding, molecular weight, and lipid solubility
what is the pKa of a drug?
the pH at which the nonionized and ionized drug concentrations are equal
example of pKa and its effect on drug distribution
alfentanil has a more rapid onset of action than fentanyl because its pKa is close to physiologic pH, so most of the drug exists in the lipid-soluble, nonionized form at physiologic pH
what determines the concentration of a drug in the plasma and other tissues?
the binding of a drug to proteins
circulation of drugs after absorption
after absorption, drugs circulate in plasma in the unbound (free) form and in the form bound to protein
what proteins are drugs usually bound to?
acidic drugs to albumin, basic drugs to a1-acid glycoprotein
what does protein binding of drugs act to do?
act as a temporary reservoir of drug, and prevent large fluctuations of the unbound or free drug
what else can it do though?
protein binding can reduce a drugs metabolism and renal excretion by preventing its transport across renal tubular cell membranes
what things influence the binding of drugs to proteins?
increasing age, hepatic and renal disease, trauma, and surgery
what phenomenon can limit drug distribution and in whom does it occur?
ion trapping, in the fetus
ion trapping
fetal blood has a lower pH than normal adult blood, which means that drugs, such as local anesthetics and opioids which cross the placenta in the nonionized form, willl become ionized in the fetal ciculation, and because ionized drugs cant readily cross the placenta they accumulate in the fetal blood in a concentration gradient
what happens after systemic absorption of drugs?
the highly perfused tissues, eg. brain, heart, kidneys, and liver; recieve a proportionally larger amount of the total dose

for example, 75% of CO in a normal adult is delivered to the vessel-rich group of organs, which make up only 10% of body mass
so due to this, what happens after IV administration of lipid-soluble drugs (eg. barbiturates, opioids)?
there will be rapid onset of CNS effects
redistribution
as the concentration of drugs in the plasma decreases below those in the highly-perfused tissues, drugs leave the highly-perfused tissues and are delivered to less well-perfused tissue beds, such as skeletal muscle and fat
thiopental redistribution
redistribution from the brain to inactive tissues is primarily whats responsible for awakening after a single dose of the drug; giving repeated doses can saturate inactive tissue sites decreasing the amount of sites available for redistribution, delaying awakening until metabolism can decrease plasma concentrations
fentanyl redistribution
the duration of action of fentanyl is normally short resulting from redistribution, but its effect can be prolonged when single, large doses or continuous infusions saturate inactive tissue sites available for redistribution
drug metabolism
done primarily in the liver, but to a lesser extent in the kidneys, lungs, and GI tract, its the conversion of pharmacologically active, lipid-soluble drugs to water-soluble, often inactive metabolites
what does increased water solubility do to a drug?
decreases the volume of distribution of that drug (Vd) and enhances its renal excretion
renal excretion of lipid-soluble drugs
they undergo minimal renal excretion because of the ease of which they are reabsorbed from the renal tubules into the pericapillary fluid
what else can metabolism do to a drug?
convert a prodrug into its active form, or cause toxic metabolites
what exactly metabolizes most drugs, and where are they located?
microsomal enzymes, and they are located principally in the hepatic smooth endoplasmic reticulum
where does the term microsomal enzymes come from?
from the fact that centrigugation of homogenized hepatocytes concentrates fragments of the smooth endoplasmic reticulum in whats called the microsomal fraction
what does the microsomal fraction contain?
the cytochrome P450 system, which likely contains many enzymes responsible for metabolism of many foreign compounds
what is enzyme induction?
stimulation of microsomal enzyme activity by drugs (classically phenobarbitol) which leads to accelerated metabolism of other drugs
what is the principle determinant of microsomal enzyme activity?
likely is genetic, which illustrates the large individual variations in the rate of metabolism of drugs among patients
what types of metabolic reactions can drugs undergo?
phase 1 or phase 2 reactions
phase 1 reactions
include oxidation, reduction, and hydrolysis
phase 2 reactions
includes conjugation
what does oxidation?
cytochrome p450 system
reduction?
by halogenated compounds
hydrolysis?
by procaine and amides
what do oxidation, reduction, and hydrolysis do to drugs?
they make them more water soluble by introducing polar groups like hydroxyl, amino, sulfhydryl, and carboxyl groups
conjugation
involves coupling a drug with an endogenous substrate, like glucuronate, acetate, or an amino acid so it can be excreted
how can drugs be excreted?
either unchanged or as metabolites
sites of drug excretion
primarily in the kidneys, but also the lungs, skin, bile, intestines, breast milk, saliva, and sweat
by what processes do the kidneys excrete drugs?
by passive glomerular filtration, active tubular secretion, and passive diffusion
nonionized drug excretion in the kidneys
they are easily filtered through the glomerulus, but they can also diffuse back across renal tubular cell membranes, leaving only a small amount of drug in the urine
ionized drug excretion
are not reabsorbed after filtration, and appear unchanged in the urine
how are many organic acids, like penicillin, excreted?
they are transported across renal tubules by systems that secrete naturally occuring substances, such as uric acid
passive diffusion of drugs
a bidirectional system where drugs diffuse across renal tubules according to a concentration gradient, lipid solubility, and pH
renal damage and excretion
renal damage impaires the ability of the kidneys to excrete drugs, which can result in high plasma concentrations and prologed duration of action of drugs
excretion and protein
protein binding can significantly alter the renal excretion of drugs, and hypoprotenemia can increase the amount of drug available for filtration
excretion and pH of urine
weak acids will be ionized in alkaline urine, and will therefore not be reabsorbed, while weak bases will be ionized in acidic urine and not be reabsorbed
how are volatile anesthetics excreted?
in the lungs
what happens to drugs excreted in the bile, and what are good examples of these?
they are reabsorbed repeatedly from the intestine (enterohepatic circulation), which plays a significant role in the excretion of drugs such as vecuronium and erythromycin
what are the pharmacokinetics of IV drugs influenced by?
the Vd of the drug and the clearance
what is the elimination half-time and what determines it?
its the rate at which the plasma concentration of a drug decreases with time, and it is also determined by Vd and clearance
what measures are more useful in characterizing the clinical response to a drug than elimination half-time?
context-sensitive half-time, and effect-site equilibrium
plot of drug concentration after an IV bolus of drug
there will be 2 phases, the first is the distribution phase, the 2nd is the elimination phase

the distribution phase is the initial rapid decrease in plasma concentration, and corresponds to the distribution of drug from the circulation to the peripheral tissues

the elimination phase is the gradual decrease in drug concentration and reflects its elimination from the central vascular compartment (ie. clearance) by the kidneys and liver
distribution and elimination half-lives
are the time necessary to decrease the drug concentration 50% during the distribution or elimination phase
what is one of the most important pharmacokinetic variables to consider when setting a constant rate of IV drug infusion to maintain a steady-state plasma concentration?
clearance
clearance
the volume of plasma (ie. of the central compartment) cleared of drug in mL/min by renal or hepatic excretion, metabolism, or both
what happens when the rate of drug infusion exceeds clearance?
the plasma concentration of the drug will increase progressively, and cumulative drug effects occur
total clearance
is additive, and is the summation of the clearance rates for the liver, kidneys, and other routes
what are important factors determining clearance of a drug?
its concentration, and blood flow to the organ of clearance
what organ is most important for clearance of unchanged drugs or their metabolites?
the kidneys
clearance in the kidneys
water soluble compounds not bound to protein are the easiest to excrete, while protein-bound and lipid-spluble ones are harder, emphasizing how important metabolism is to convert lipid-soluble drugs to water-soluble metabolites
what are the most useful clinical indicators of the ability of the kidneys to eliminate drugs?
creatinine clearance, or serum creatinine concentration; the magnitude of increase of these will give a good estimate of the downward adjustment of drug dose required to prevent accumulation of drug in the plasma
volume of distribution (Vd)
is a calculated number which reflects the apparent volumes of the compartments that constitute the compartmental model for that drug:

dose of drug administered IV/plasma concentration
what factors will lead to a small Vd?
binding of drug to plasma proteins, a high degree of ionization, and low lipid solubility will all limit passage of drug to the tissues
examples of drugs with a small Vd
neuromuscular blockers
examples of drugs with a large Vd
thiopental and diazepam: they are nonionized lipid soluble drugs that readily pass into peripheral tissues from the circulation, and thus have low plasma concentrations
elimination half-time
the time necessary for the plasma concntration of drug to decrease 50% during the elimination phase
how many elimination half-times are required to completely eliminate a drug?
5
What will repeated doses of drugs equivalent to the initial dose at intervals more frequent than five elimination half times result in?
Cumulative drug affects
What will drug accumulation continue until?
The rate of drug elimination equals the rate of drug administered
What is the time necessary for drug to achieve steady-state plasma concentration (Cps) with intermittent doses?
Five elimination half-time
What is a common practice for achieving therapeutic concentration rapidly?
To administer a large initial IV dose i.e. loading dose, of drug to achieve a therapeutic concentration rapidly, and then give continuous or intermittent IV injections of decreased doses of drug to match the rate of elimination and maintain an optimal and unchanging plasma concentration
What must occur to the maintenance dose in the presence of renal or hepatic dysfunction?
It must be adjusted downward to prevent drug accumulation due to a prolonged elimination half-time
What is the descriptor most often used to characterize a drug's pharmacokinetic behavior?
Elimination half-time
However, what is the problem of elimination half-time?
It is useful only in the computation of central compartment drug concentration in a one compartment model, and it is of little value in describing the pharmacokinetics of drugs and multi-compartmental models
What is the anesthesiologist more interested in?
How long it will take the plasma concentration to decrease to a level that allows the patient to awaken, rather than the slope of the plasma drug concentration curve. Elimination half times alone provide virtually no insight into the rate of decrease in the plasma concentration after discontinuation of IV drug administration
So what then might the anesthesiologist wish to focus on?
Context-sensitive halftime
Context-sensitive halftime
The time necessary for the drug concentration to decrease to a predetermined percentage, for example 50%, 60%, 80%; after discontinuation of a continuous IV infusion of a specific duration; context refers to the duration of infusion
How are context-sensitive half times calculated?
Using computer simulation of multi-compartmental pharmacokinetic models drug disposition for drugs administered as continuous infusions during anesthesia
What is the context-sensitive halftime shown to be related to?
It increases in parallel with the duration of continuous IV administration, depending largely on the drugs lipid solubility and the efficiency of its clearing mechanisms: each drug will increase at a different rate based on the above factors
What is the relationship between the context-sensitive halftime and the drugs elimination half-time?
There is none
What does the time to recovery depend on?
How far the plasma concentration must decrease to reach levels compatible with awakening
Why is there a delay between the IV administration of a drug and the onset of its clinical effect?
This reflects the time necessary for the circulation to deliver the drug to its site of action, for example brain tissues. This delay reflects the fact that the plasma is not usually the site of drug action the circulation is merely the route by which the drug reaches its effect site
Effect site equilibrium time
The time for equilibrium between drug concentration in the plasma and the drug effect, for example time to produce a specific effect on the EEG
What drugs have short effect site equilibrium time?
remifentanil, alfentanil, thiopental, propofol
What drugs have a longer effect site equilibrium time?
fentanyl, sufentanil, midazolam
What is knowledge of the effect site equilibrium time important for?
Determining dosing intervals, especially when titrating IV drugs to a given clinical effect
What can failure to appreciate the importance of effect site equilibrium time result in?
Unnecessary or premature administration of drug
Bioavailability
The amount of active drug that is absorbed and reaches the systemic circulation; for example a drug that is absorbed from the intestine passes through the liver, first pass effect, before it reaches the circulation, and if the liver metabolizes the drug extensively the amount of active drug reaching the circulation and its site of action will be limited
What is an example of a drug degraded by the liver?
Propofol
What is the most important mechanism by which drugs exert pharmacologic effects?
By the interaction with a specific protein molecule in the lipid bilayer cell membranes, called a receptor
Examples of membrane receptors
Membrane or G protein coupled receptors, ligand gated ion channels, voltage sensitive ion channels, and enzymes
Drug interaction with receptors
A drug administered as an endogenous substance in contrast to endogenous hormones or neurotransmitters is an incidental passenger for these receptors; a drug – receptor interaction alters the function of a specific cellular component, which initiates or prevents a series of changes that characterize the pharmacologic effects of the drug
Receptor definition
Excitable transmembrane proteins that are responsible for transduction of biologic signals
Voltage sensitive ion channels
Depend on cell membrane voltage to open and close, for example sodium, chloride, potassium, calcium channels
Ligand gated ion channels
Function as receptor ion channel complexes in which the ion channel is an integral part of a larger and more complex transmembrane protein, for example nicotinic cholinergic receptors and amino acid receptors, e.g. GABA
What does activation of GABA – chloride channels, i.e. receptors, result in?
Cell hyperpolarization or an increase in ion conductance that prevents depolarization, thereby inhibiting neuronal activity; such activation enhances endogenous GABA mediated inhibition in the central nervous system, providing neurobiologic basis for the hypnotic and sedative effects of drugs such as benzodiazepines, barbiturates, and propofol
Prevalence of GABA receptors
Approximately 1/3 of all synapses in the CNS are responsive to GABA
What two parts contribute to the anesthetized state?
1. Sedation and loss of memory and consciousness – brain?

2. Analgesia and loss of pain sensation – spinal cord?
How might opioids and alpha two agonists contribute to the anesthetized state?
Through inhibition of calcium channels and potassium channel activation – presynaptic inhibition
How do sedative – hypnotics, such as benzodiazepines propofol, etomidate, steroid anesthetics, contribute to the anesthetized state?
By activation of GABA a receptor chloride channels – leading to neuronal inhibition
How does ketamine work?
By inhibition of glutamate receptors
Guanine, G, proteins
Receptors that are important intermediaries in cell communication that reflect the molecular mechanisms of action of multiple classes of drugs, including opioids, sympathomimetic's, and anti-cholinergic's
G protein mechanism of action
An exogenously administered drug is recognized by its specific receptor, and the receptor – ligand interaction induces conformational changes, enabling the receptor to activate a specific gene protein which mediates the final cascade of biologic steps within the cell that leads ultimately to the pharmacologic and physiologic response characteristic of the administered drug
Examples of clinically important G protein coupled receptor systems
Include adrenergic, opioid, muscarinic, cholinergic, dopamine, and histamine receptors
Subtypes of G protein receptors
Multiple subtypes of receptors exist, including alpha 1 and two, beta one and two, muscarinic one and two, and histamine one and two receptors
Types of ion channels
Characterized as either voltage – sensitive or ligand – gated ion channels
Ligand – gated ion channels
Function as receptor – ion channel complexes in which the ion channel is an integral part of the larger and more complex transmembrane protein, for example glutamate activated-N-methyl-D-aspartate (NMDA) receptors, GABA receptors, nicotinic cholinergic receptors
What may be the principal molecular target for ketamine?
NMDA receptor
How do GABA – activated ion channels mediate the response to GABA?
By selectively allowing chloride ions to enter and thereby hyper polarizing neurons
Ion pumps
Are examples of excitable membrane proteins, such as the sodium/potassium – ATPase
Sodium potassium ATPase action
Action potentials activate sodium ion channels, allowing sodium the pass from outside to inside the cell. Sodium potassium ATPase then pump sodium out of the cell in exchange for potassium, returning the cell to its original cation composition
Digitalis mechanism of action
Inhibits the energy dependent sodium potassium ATPase ion pump, improving myocardial contractility
Agonist
A drug that initiates pharmacologic effects after combining with the receptor, therefore an agonist has a high efficacy and high affinity for the receptor; agonists induce stimulatory or inhibitory effects that mimic endogenous hormones and neurotransmitters
Antagonist drugs
Drugs that prevent receptor – mediated agonist effects by occupying agonist receptor sites, therefore they have the same affinity as the agonist for the receptor but their efficacy is poor
Mechanisms of action of antagonists
Can inhibit agonist effects by competitive inhibition, e.g. neuromuscular blocking drugs, or noncompetitive inhibition
What is a drug with an affinity equal to or less than that of the agonist but with lesser efficacy called?
A partial agonist
Inverse agonists, or super antagonists
Drugs that produce a response below the baseline response measured in the absence of the drug
Number of receptors
The number of receptors in the lipid cell membranes is dynamic, increasing or decreasing (up regulation or down regulation) in response to specific stimuli: for example prolonged beta adrenergic agonists as in the treatment of asthma are associated with tachyphylaxis and a concomitant decrease in the number of beta-adrenergic receptors, and conversely chronic beta blockade may result in increased number of beta-adrenergic receptors so an exaggerated response occurs if the block is abruptly reversed such as during the preoperative period
Receptors and aging
Changes in the responsiveness of receptors with no increase or decrease in the number of receptors may occur with aging, for example more isoproterenol is necessary to increase heart rate in the elderly compared with younger patients despite an unchanged number of receptors with aging
Dose – response curves
Depict the relationship between the dose of drug administered, or the resulting plasma concentration, and the resulting pharmacologic effect
How was the dose or concentration commonly graphed?
As the logarithmic transformation of dosage since this permits display a large range of doses
What causes the dose response curve to shift to the right?
A competitive antagonist or desensitization, or an agonist with a lower receptor affinity or potency
What causes a shift to the left?
An agonist with higher receptor affinity or potency
Drug potency on dose response curves
Is depicted by the location along the dose axis of the dose response curve
What is the dose required to produce a specific effect called?
Effective dose, ED, just the dose necessary to produce that effect in a given percentage of patients, for example ED 50, ED 90
What does increased affinity of a drug for its receptors due to the dose response curve?
Shifts it to the left
Drug potency in clinical practice
The potency of the drug makes little difference as long as the necessary dose of drug can be administered conveniently
Calculating drug dosage in common practice
Drug dosages commonly calculated on the basis of body weight, but when total body weight exceeds ideal body weight the total body weight will increasingly overestimate lean body mass, so in adults it is rarely necessary to scale dose to body weight more than 80 kg woman or 100 kg man
What influences the slope of the dose response curve?
The number of receptors that must be occupied before drug effect occurs
If the drug must occupy most receptors before its effect occurs what will happen to the slope of the dose response curve?
It will be steep
What are examples of drugs with steep dose response curves?
Neuromuscular blocking drugs and inhaled anesthetics
Clinically what does it mean when there is a steep slope of the dose response curve?
Small increases in dose evoke large increases in drug effect, for example one Mac of a volatile anesthetic prevents skeletal muscle movement in response to a surgical skin incision in 50% of patients, ED 50, where is a further modest increase to 1.3 Mac prevents movement and at least 95%, ED 95, of patients
What is also true when the dose response curve is steep?
The difference between a therapeutic and toxic concentration may be small, which is true for volatile anesthetics where there is a small difference between doses that produce desirable central nervous system depression and undesirable cardiopulmonary depression
Drug efficacy
The maximal effect of a drug as depicted by a plateau in the dose response curve
What may limit dosage to below the concentration associated with maximal effect?
Undesirable side effects of the drug
What is the ceiling effect?
A phenomenon where the degree of effect produced by increasing doses of drug eventually reaches a steady level, and if the dose of drug exceeds the ceiling dose there is no further increase in therapeutic effect and undesirable side effects may predominate
Ceiling dose
The dose at which the ceiling effect is obtained
Are efficacy and potency related?
Not necessarily
Individual responses to drugs
Individual responses may very as reflections of differences and pharmacokinetics, e.g. renal liver or cardiac function or patient's age, or pharmacodynamics, such as enzyme activity in genetic differences. For example benzodiazepines have a more pronounced effect in the elderly than in young adults
Malignant hyperthermia
A hyper metabolic state triggered in some individuals in response to administration of succinylcholine or volatile anesthetics, which represents a genetic pharmacodynamic abnormality
Therapeutic index
The ratio between the lethal dose in 50% of patients, LD50, and the effective dose in 50% of patients, ED50
Therapeutic index and safety
The higher the therapeutic index of a drug, the safer it is for clinical administration because the LD is far above the ED
Additive effect
When the pharmacologic effect of two or more drugs administered together is equivalent to the summation of their individual effect
Synergistic response
When the pharmacologic effect of two or more drugs administered together is greater than the sum of the individual effects
Time synergism
The prolongation of action of one of the drugs when two drugs are administered together, for example combination of lidocaine and epinephrine increases the duration of action of lidocaine
Competitive antagonism
The competitive antagonist can usually be displaced from the receptor by administration of high doses of agonist, it shifts the dose response curve to the right, and it is usually reversible
Noncompetitive antagonism
When bounde to receptors noncompetitive antagonists produce a confirmational change in the receptor which results in diminished receptor response when exposed to agonist even at high doses, the dose response curve shifts to the right, the slope is reduced and the maximum pharmacologic response diminishes
Is noncompetitive antagonism reversible or irreversible?
It can be either
Drug stereospecificity
The molecular interactions which are the foundation of pharmacokinetics and pharmacodynamics are stereoselective or stereospecific, emphasizing the drugs are expected to interact with other biological components, e.g. receptors, in a geometrically specific way.
Racemic mixture
When two isomers, dextro (D) and levo (L), of opposite shape are present in equal proportions
Administration of the racemic mixture of drugs
May represent pharmacologically two different drugs with distinct pharmacokinetic and pharmacodynamic properties, the two isomers may have different rates of absorption, metabolism, and excretion and different affinities for receptor binding sites. And although only one isomer is active it is possible that the other isomer contributes to its side effects
Racemic mixture examples
D – bupivacaine remains in sodium ion channels for a longer period than the L – isomer which may result in cardio toxic effects; but ropivacaine and levobupivacaine are present only has the L – isomer and are not as likely to produce cardio toxic effects as bupivacaine

D – ketamine is predominantly hypnotic and analgesic, whereas L – ketamine is the likely source of the drugs unwanted side effects
How should the inactive isomer in a racemic mixture be regarded?
As an impurity
Drug tolerance
When a large dose of drug is required to elicit an effect that is usually produced by smaller therapeutic doses of the drug; it can be natural or acquired
Natural tolerance
Can be species or racial specific
Acquired tolerance
Developed in response to repeated administration of the drug and can be either tissue tolerance or cross – tolerance
Tissue tolerance
Is tolerance confined to certain pharmacologic effects such as tolerance to euphoric effects of morphine but not its constipating effects
Cross tolerance
Occurs when an individual develops tolerance to a specific drug, for example alcohol, and other drugs, e.g. sedative – hypnotics, inhaled anesthetics, producing similar pharmacologic effects
Tachyphylaxis
Acute tolerance to the pharmacologic effects of certain drugs, for example ephedrine and amphetamines, which may occur when they administered at short intervals. The mechanism responsible is unclear but may reflect depletion of norepinephrine stores or altered dissociation of drug from its receptor sites
Drug dependence
The psychic or physical state characterized by behavioral responses including a compulsion to take the drug on a continuous or periodic basis to experience its effects and sometimes to avoid the discomfort of its absence; tolerance may or may not occur, and a withdrawal syndrome which may be life-threatening may develop on discontinuation of drug
Drug interactions
Simultaneously administered drugs can alter each other's pharmacokinetic and pharmacodynamic behaviors, for example ranitidine or metoclopramide can alter drug absorption by changing the pH of gastric secretions and G.I. motility
Cholinesterase inhibitors
Antagonize neuromuscular block by increasing the amount of acetylcholine, which displaces non-depolarizing neuromuscular blocking drugs from nicotinic receptors