Вы находитесь на странице: 1из 18

PHARMACOLOGY QUIZLET

CHAPTER 1 INTRO TO PHARMACOLOGY the mechanism by which a dug interacts with and is
processed by the body
Drug
chemical that interacts with an organism to alter its Elimination
function, show biological processes and provide the mechanism by which a drug leaves the body
methods of diagnosis, treatment, and prevention of
disease Selectivity
the degree to which a drug acts at one specific site or
Pharmacology receptor
the study of the interaction of drugs with an organism
Lock and Key Receptor Theory
Pharmacodynamics a visual analogy that is used to describe interactions
the study of drugs modes of action on an organism between drugs or receptors

Pharmacy Racemic Drug


the practice of compounding and dispensing medicinal drugs that contain two isomers a part of a drug's
preparations chemical structure

Therapeutics Half-Life
the branch of medicine concerned with the application the amount of time it takes for a drug's concentration in
of remedies and the treatment of disease the body to decrease by half once administered

Hypersensitivity Loading dose


an exaggerated adverse reaction to drug administration of a higher dose than that used for
maintenance in order to rapidly increase drug
Toxicology concentration
the study of drugs' toxic effects on an organism
Maintenance dose
Entral administration of a drug at dosage levels that maintain
absorbed through the gastrointestinal tract a therapeutic drug concentration

Parenteral Potency
injected directly into the body relative drug strength

Transdermal Efficacy
absorbed through the skin using timed-release the degree to which a drug exerts its intended effect

Topical Idiosyncrasy
absorbed through the skin an unexpected reaction to a drug caused by unusual
susceptibility to the drug
Inhalation
absorbed through the respiratory tract or alveoli Agonist
drug action describing a drug that has an affinity for a
Pharmacokinetic receptor and activates a response
movement of the drug
Antagonist
Pharmacodynamic a drug action describing a drug that has an affinity for a
the drug has its effect receptor and does not activate a response

Absorption Additive
the mechanism by which a drug enters the body a drug action describing two drugs' effects as being
equal to the summation of their individual effects
Distribution
the mechanism by which the drug is transported to the Syergism
site of action drug action describing two drugs effects as being
greater than the summation of their individual effects
Metabolism with both drugs being active

1
PHARMACOLOGY QUIZLET

Potentiation Prescription
a drug action describing two drugs' effects as being includes name, strength, dosage, route of
greater than the summation of their individual effects, administration, frequency and length
with one drug being active and one drug being inactive

Cumulation Preventing Medication errors


a condition that exists when a drug's rate of elimination 5 Rights: drug, dose, pt, time, and route
or inactivation is slower than its rate of administration
QD
Tolerance Each Day
refer to the need for an ever increasing amount of a
drug to continue to produce a given effect QID
Four times daily
Tachyphylaxis
is a rapidly occurring form of tolerance Q2H
Teratogenicity Every 2 hours
a drug's potential to cause fetal damage in utero when
administered to a pregnant woman
Drugs
Lethal Dose50 Chemicals that act on livings systems at the molecular
the dose of a drug that is lethal in 50% of a test group level.
of animals
Receptors
Effective Dose50 Specific molecules in a biological system that mediate
the dose of a drug that is therapeutically effective in drug effects.
50% of a test group of animals
Pharmacology
Therapeutic Index Study of the action of drugs and effects on a living
the ratio of Lethal Dose50/Effective Dose50 system.

Chemical Name Medical Pharmacology


this is the first name a potential drug receives Study of drugs used for the diagnosis, prevention, and
treatment of disease.
Code Name
this is a name assigned to the chemical by its Toxicology
manufacturer during the testing process Branch of pharmacology that deals with the untoward
effects of drugs on living systems.
Generic Name
this is the name assigned to the chemical by the USAN History of Pharmacology
when it appears to have therapeutic value and the Prehistoric methods were ineffective/toxic.
manufacturer wants to market it Relied on "serendipity" -- the idea that if it worked we
do it. Based only on effectiveness.
Official Name Eventually replaced by observation and experiment,
the Generic name becomes the Official name when the however, molecular level of knowledge was lacking.
drug receives approval from the Food & Drug Chemistry and Physiology lead us to know how drugs
Administration work on the organ level.

Trade Name History of Pharmacology (Continued)


name given to a drug by its manufacturer when the In last 50 years: Advent of the controlled clinical trial.
drug is marketed -Accurate evaluation of therapeutic claims.
Research allowed us to:
Official Source of Information -Gain info of drug action and receptor
Government issued -Improve understanding of molecular basis of drug
action
Unofficial Source of Information
Non-Government issued Pharmacogenetics
The relation of a person's specific genetic makeup to
his or her response to specific drugs.

2
PHARMACOLOGY QUIZLET

Enantiomers
Therapeutic effects Dextro and Levo. D and S.
Effects of a drug that help cure or prevent the disease. E.G. Prilosec Vs. Nexium
Prilosec: Omeprazole, racemic, S has activity.
Toxic effects Nexium: Esomeprazole, S isomer, absorption/ half life
Effects of a drug that cause secondary toxic effects. +,
Extended patent life = + money for company.
ADME
Absorption, Distribution, Metabolism, Elimination Most start racemic but eventually produced as single
isomer
Drug properties
Gas, solid, liquid, gel, suspension, solution. Pharmacodynamics
Actions of drug on body:
Factors in Aqueous Solution -Determines classification
Ionization, solubility, stability. -Determines appropriate therapy

Factors for Acting on Membranes Pharmacokinetics Definition


Lipid solubility, Diffusion, Receptor binding, Partitioning, Actions of body on drug:
Blood flow. -ADME
-Choice and administration of drug to specific patient
Drug Size
Commonly between 100-1000MW. PharmacoD. Conc./Resp.
-100MW to achieve selectivity (sufficient binding). Pharmacodynamics studies the relationship between
-1000MW to allow for proper distribution. concentration and response. The concentration of the
-If greater than 1000MW, needs to be administered at drug at the site of action, and the beneficial and toxic
site. effects.
-Depends on: CONCENTRATION
Drug Receptor Interactions
Dependent on: Concentration-response curve
-Size, shape, charge, atomic makeup Plots concentration of drug versus % of maximum
-Structure + specificity effect whether therapeutic or toxic.
-3 point attachment
-Specific, mimic endogenous Drug receptors
Most are proteins.
Affinity -Selective, and change function upon binding.
Strength of the bond that a drug has to receptor. Affects -Drugs typically look like endogenous molecule.
potency. High affinity = high potency.
Agonist
Selectivity Activate receptor
Also known as specificity. Determined by type of bond.
High specificity = few effects in body. Low specificity = Antagonist
side effects all over. Prevents action at a receptor

Covalent Bonds Pharmacokinetics Imp. stuff


Covalent / Strong bond = less selectivity (but more ADME.
reactivity) Bile = feces
-commonly irreversible Kidneys = urine
Noncovalent = most common = more selectivity (less
reactivity) Absorption
-Hydrogen, Van der Waals', ionic. The measure of how quickly a drug gets into the
-Commonly reversible bloodstream

Drug Shape Common routes of absorption


Should complement receptor site. PO/PR (oral/anal)
Many exist as chiral molecules, thus have enantiomers. Inhaled, topical, transdermal,sublingual
Dextro = Right = D Intramuscular, intravenous, subcutaneous
Levo = Left = S

3
PHARMACOLOGY QUIZLET

Drug formulations FDA Modernization Act


Affect absorption. 1997: Recognized changes needed to be made for 21st
Depends on: century.
-Barriers drug is capable of passing (B/b. B/t)
-Setting in which drug will be used (Hospital vs. home) Pediatric Research Equity Act
-Urgency of situation 2003: Requires tests in pediatrics for certain drugs for
-Drug stability certain circumstances
-First pass effect
Dietary Supplement and Non-Script Drug Consumer
Distribution Protection Act
Movement of drug from site of administration to other 2006: Requires serious adverse event reporting for
parts of the body. dietary or nutrition supplements. Non regulated so far.
Depends on:
-Vascularity. Higher = higher uptake (EG Brain/kidneys) FDA
-Drug solubility in certain tissue Responsible for approval of new drugs, and oversight
-Binding of drug to macromolecules in blood IE. of marketing/sale of drugs on market.
Albumin Prescription and OTC included
-Ability to cross special barriers IE Blood brain Blood
testes DEA
Drug enforcement agency:
Elimination Classifies drug schedules on basis of abuse potential.
2 major routes.
Metabolism: Enzymatic conversion of drugs to inactive Who can write scipts?
derivatives, more polar, more water soluble, enzymes in Depends on states.
GI and liver.
Excretion: Follows metabolism or unchanged Stages of Drug Approval
Kidneys = urine 0-4 years: Preclinical: In-vitro animal testing
Liver/bile = feces 4-8 years: Human clinical trials
8-20 years: Exclusive marketing rights
Drug legislation prior to 1906 20+ years: generics form
No regulation on sale of drugs.
-Outrageous claims of benefit and safety Human Clinical Testing Phase 1
-Unregulated use of addictive substances 25-50 HEALTHY volunteers, look for toleration, check
for safe clinical dosing range. Evaluate
Federal Pure Food and Drug Act pharmacokinetics
1906: Active ingredients placed on label, purity levels
maintained. Human Clinical Testing Phase 2
-not for efficacy, just purity. 100-200 SICK volunteers are tested. Assess dose-
1912: Amended to stop false advertising claims efficacy relationship.
Dose to pharmacological/therapeutic effect.
Food Drug and Cosmetic Act
1937: Diethylene gloycol in sulfanilamide elixir kills Human Clinical Testing Phase 3
hundreds 1000s of people. Randomized controls in actual
1938: FDCA: Label with direction for safe use / conditions.
Preapproval for safety Establish safety in patients with disease, and efficacy
compared to other treatments.
Durham-Humphrey Act
1952: Prescription vs. OTC defined and federally Human Clinical Testing Phase 4
regulated (FDA) Post marketing surveillance. Safety under actual life
conditions.
Kefauver-Harris Amendment
1962: Thalidomide babies in non USA. Proprietary Drug
Required efficacy in addition to safety. Drug with trade name, protected under patent.
Patent life = 20 years. Drugs are frequently patented 5
Controlled Substances Act years before marketing. After patents expire other
1970: Scheduled drugs companies can create bioequivalent generic drugs.
More familiar to outpatient settings.

4
PHARMACOLOGY QUIZLET

Generic Drug If drug is D and R is receptor, then what is proportional


Drug made from a once-patented formula. Cheaper, to the response?
less "development" cost. Must have same RD (drug receptor interaction)
pharmacokinetic properties as original. Promoted in
hospitals, cheaper. What are the axis (x and y) on a dose response curve?
y = e/Emax
Chemical name
Not used in practice, but in lab. Long ass. Ridic. What is the equation that models the dose response
curve?
Drug Naming Example e/Emax=([D]/[D]+ED50)
Nexium.
Esomeprazole magnesium What does the dose response curve level off at high
(S)-5-methoxy-2-[(4-methoxy-3,5 dimethylpyridin-2-yl) drug concentrations?
methylsulfinyl]-3H-benzoimidazole response saturates

CHAPTER 2 DRUG RECEPTOR AND What does the term potency refer to?
PHARMACODYNAMICS concentration (EC50 or ED50) required to produce 50%
of the drug's individual maximal effect
What are effectors?
molecules that accomplish a biological effect after Broadly, what are the factors that impact potency?
being activated by a receptor/e.g. translate drug- affinity of receptors for binding the drug, and the
receptor interaction into a change in cellular activity efficiency of the drug receptor complex to generate a
repsonse
What are major receptor mechanisms (4)
ligand gated channels, GPCRs, Kinase linked What is the most important determinant of a drug's
receptros, hormonal receptors clinical utility?
it's efficacy
What are the three main classes of receptor
molecules? What term is used interchangeably with efficacy?
proteins, nucleic acids, membrane lipids power

Broadly, what are five protein based receptor What is an agonist (broadly)?
molecules? a drug that activates a receptor upon binding, bringing
hormones, receptor or voltage gated ion channels, about a characteristic tissue response
enzymes, transport proteins, structural proteins
What is potency? (definition)
What is drug receptor theory (broadly)? Dose or concentration (EC50, ED50) required to
The size, shape, and electrical charge of a drug produce 50% of that drug's individual maximal effect
determine its binding affinity to a particular receptor,
relative to other possible binding ssites What are the two factors that affect the relative potency
of a drug?
What is the name of a drug that activates a receptor? affinity (Kd) of the receptors for binding the drug,
agonist efficiency of this drug-receptor complex for creating a
drug response

What does an antagonist do? What is a full agonist?


block the action of an agonist (so nothing happens) drugs that occupy receptors and bring about a full or
maximal response
In the absence of an agonist, what happens if you give
an antagonist? What is a partial agonist?
nothing. nothing happened before, so nothing will a drug that occupies the same receptor, but bring about
continue to happen. much like my social life. a less than maximal response

What is the broad use of a dose response curve? In clinical terms an efficacy refers to what?
Determine quantitative relationship between dose or the relative clinical effect of a given drug
concentration and its pharmacologic effect
What are the two categories of antagonists?
receptor antagonists, non receptor antagonists

5
PHARMACOLOGY QUIZLET

What are the two types nonreceptor antagonists? Enzymes (ex: Dihydrofolate reductase is the receptor
chemical, physiological for the antineoplastic drug Methotrexate); Transport
Proteins (ex; Na+/K+ ATPase is the membrane receptor
What is the action of chemical antagonists? for cardioactive Digitalis Glycosides); Structural
binds the agonist directly Proteins (Ex: Tubulin is the receptor for Colchicine, an
anti-inflammatory agent)
What is the action of physiological antagonists? What are examples of how Enzymes, Transport
bind to a differen receptor Proteins, & Structural Proteins serve as drug
receptors?
What are the two types of active site bind modes and
their relative classifications?
reversible (competitive), irreversible (non competitive) E = (Emax x C) / (C + EC50), where E is the effect
observed at concentration C, Emax is the maximal
How can a competitive, reversible antagonist be response that can be produced by the drug, and EC50
overcome? What is its effect of potency, and ED50? is the concentration of the drug that produces 50% of
increase concentration of agonist, potency is reduced, maximal effect
but ED50 remains the same The relation b/t drug concentration & effect is described
by a Hyperbolic Curve according to which equation?
What is the effect of an irreversible, competitive agonist
on the dose response curve? B = (Bmax x C) / (C + Kd), where B = total conc. of
ED50 unchanged (e.g. potency is the same) but Emax receptor sites, Kd = equilibrium dissociation constant
will be reduced (you are essentially removing which represents the conc. of free drug at which half-
receptors) maximal binding occurs, & C = conc. of free unbound
drug. The LOWER the Kd, the HIGHER the binding
affinity.
The receptor's affinity for binding a drug determines the What is the equation that resembles the Mass Action
concentration of drug required to form a significant # of Law, which describes the association b/t 2 molecules of
drug-receptor complexes, and the total # of receptors a given affinity?
may limit the maximal effect a drug may produce.
How do receptors largely determine the quantitative The transduction process that links drug occupancy of
relations b/t dose or concentration of drug & receptors & pharmacologic response. Occurs after a
pharmacologic effects? receptor is occupied by an agonist, the resulting
conformational change is only the 1st of many steps
The molecular size, shape, & electrical charge usually required to produce a pharmacologic response.
determine whether, and with what affinity, it will bind to What is Coupling?
a particular receptor among the different binding sites
available in a cell, tissue, or patient. Antagonists that reduce receptor activity below basal
How are receptors responsible for selectivity of drug levels observed in the absence of bound ligand
action? What are Inverse Agonists?

Agonists: Some drugs & many natural ligands Higher concentrations of Agonist are required to
(hormones, NTs) activate the receptor to signal as a produce a given effect (agonist conc. is shifted to the
direct result of binding to it; Antagonists: Other drugs RIGHT, EC50 is changed); High Agonist concentrations
act by binding to receptors and interfere w/ the ability of can OVERCOME inhibition by a Competitive Antagonist
an agonist to activate the receptor How do Competitive Antagonists affect the Agonist
How do receptors mediate the actions of both effect?
pharmacologic agonists & antagonists?
Reduces the maximal effect the Agonist can achieve by
Receptors which ligands are yet still unknown irreversibly binding to a receptor, usually covalently,
What are "Orphan" Receptors? preventing agonists from binding. (Curve does not shift,
EC50 stays the same usually)
The best characterized drug receptors which mediate How do Noncompetitive Antagonists affect the Agonist
the actions of endogenous chemical signals such as effect?
NTs, autacoids, & hormones. These mediate the effects
of many agents. Drugs that bind to a separate site on the receptor
What are Regulatory Proteins? protein & alter receptor function w/out inactivating the
receptor; Ex: Benzodiazepines bind noncompetitively to

6
PHARMACOLOGY QUIZLET

ion channels activated by the NT GABA, enhancing the What are the 2 Therapeutically Important
net activating effect of GABA on channel conductance Consequences of using Lipid-Soluble Agents such as
What are Allosteric Modulators? Example? Steroid & Thyroid Hormones?

Partial Agonists produce a Lower Response, at Full Insulin, EGF (epidermal), PDGF, ANP (atrial natriuretic
Receptor Occupancy than do Full Agonists when ALL peptide), TGF-beta, & other trophic hormones
receptors are occupied; Partial Agonists do NOT What are examples of Ligands used in Ligand-
produce a Maximal Response b/c they competitively Regulated Transmembrane Enzymes including
inhibit the responses produced by Full Agonists Receptor Tyrosine Kinases?
What are Partial Agonists?
Begins w/ binding of ligand, typically a polypeptide
One drug (positively charged Protamine) acts as a hormone or growth factor, to the receptor's extracellular
chemical antagonist of the other (negatively charged domain -> change in receptor conformation -> receptor
Heparin) simply by Ionic Binding that makes the other molecules bind to 1 another -> tyrosine kinase domains
drug Unavailable for interactions w/ proteins involved in brought together & activated -> PHOSPHORYLATE
Blood Clotting each other & downstream signaling proteins w/ tyrosine
What is a Chemical Antagonist? residues
How do Ligand-Regulated Transmembrane Enzymes
Glucocorticoid hormones lead to Increased Blood Including Receptor Tyrosine Kinases work?
Sugar, an effect opposed by Insulin. Insulin therefore is
administered to oppose the Hyperglycemic effects of a When ligand binding induces accelerated Endocytosis
Glucocorticoid hormone; Physiological Antagonist of receptors from the cell surface, followed by the
produces less specific & less easy to control effects degradation of those receptors (& their ligands); Cell's
than a receptor-specific antagonist responsiveness to the Ligand is diminished
What is an example of Physiologic Antagonism? What is Down-regulation?

1) A Lipid-Soluble Ligand that cross the membrane acts Growth Hormone, Erythropoietin, Interferons (several
on an Intracellular Receptor; 2) A Transmembrane types), & other regulators of Growth & Differentiation
receptor protein whose intracellular enzymatic activity is Cytokine Receptors respond to which peptide ligands?
allosterically regulated by a ligand that binds to a site
on the protein's extracellular domain; 3) A Unlike Tyrosine Kinase receptors, the protein tyrosine
Transmembrane receptor that binds & stimulates a kinase activity is not intrinsic to the receptor molecule.
Protein Tyrosine Kinase; 4) A Ligand-Gated Instead, a separate protein tyrosine kinase from the
Transmembrane Ion Channel that can be induced to JAK family binds Noncovalently to the receptor ->
open or close by the binding of a ligand; 5) A receptors Dimerize -> JAKs activated to Phosphorylate
Transmembrane receptor protein that stimulates a tyrosine residues on the receptor -> STATs bind ->
GTP-binding signal transducer G protein, which in turn STATS phosphorylated by JAKs -> 2 STAT molecules
modulates production of an intracellular 2nd messenger dimerize -> STAT/STAT dimer dissociates from
What are 5 different ways that TransMEMBRANE receptor, travels to nucleus, & REGULATES
Signaling occurs? transcription of specific genes (STAT = signal
transducer & activators of transcription)
Thyroid Hormones & Steroid Hormones How do Cytokine Receptors work?
(Corticosteroids, Mineralocorticoids, Sex Steroids,
Vitamin D) ACh, Serotonin, GABA, & Glutamate (all are synaptic
What are examples of Lipid-Soluble Ligands that when transmitters)
binding to receptors, stimulate the transcription of What ligands are involved in Ligand-Gated Channels?
genes by binding to specific DNA sequences (response
elements) near the gene whose expression is to be Each NT receptor transmits its signals across the
regulated? plasma membrane by increasing transmembrane
conductance of the relevant ion & thereby altering the
1) The effect is produced 30 mins to several hours b/c electrical potential across the membrane. Ex: ACh
this is the time required for the synthesis of new binds to AChR -> Na+ flows down concentration
proteins; 2) Once the effects take place, they can gradient into cells producing a Depolarization
persist for Hours or Days after the Agonist How do Ligand-Gated Channels work? Example?
Concentration is at 0; This is due to the slow turnover of
most enzymes & proteins which remain active in cells cAMP, Ca2+ ion, & the Phosphoinositides
for hours to days after being synthesized What are 3 examples of 2nd Messengers?

7
PHARMACOLOGY QUIZLET

Extracellular Ligand is detected by receptor -> receptor Why does Desensitization occur w/ G Protein mediated
activates a G protein on cytoplasmic surface of responses to drugs & hormonal agonists over time?
membrane -> activated G protein changes activity of an
effector element (an enzyme or ion channel) -> Mobilization of stored energy (the breakdown of carbs
changes conc. of intracellular 2nd messenger in liver or TGs in fat cells via beta-adrenomimetic
(Hormone & NTs activate Gs coupled receptors which catecholamines), Conservation of water by the kidney
stimulates Adenylyl Cyclase, a membrane protein that (via vasopressin), Ca2+ Homeostasis (via PTH),
converts ATP to CAMP) Increased Rate & Contractile Force of Heart Muscle
How do G Proteins work? Example? (via beta-adrenomimetic catecholamines), Production
of Adrenal/Sex Steroids (via corticotropin or FSH),
Receptors: Beta-adrenergic amines, Glucagon, Relaxation of Smooth muscle, etc
Histamine, & Serotonin -> Inc. Adenylyl Cyclase -> What are the functions of cAMP?
INCREASED cAMP
What Receptors & Effect does Gs protein have? cAMP stimulates cAMP-dependent protein kinases
(composed of 2 catalytic chains (C) & 1 cAMP-binding
Receptors: Alpha2-Adrenergic Amines, ACh, Opioids, & regulatory dimer (R)) -> When cAMP binds to the R
Serotonin; Effects: Dec. Adenylyl Cyclase -> Dec. dimer, active C chains are released to diffuse thru the
cAMP; Open Cardiac K+ channels -> DECREASED cytoplasm & nucleus, where they transfer phosphate
Heart Rate from ATP to substrate proteins (specificity of cAMPs
What Receptors & Effect do Gi1, Gi2, & Gi3 proteins regulatory effects depends on the protein substrates of
have? the kinases) Ex: Liver is rich in Phosphorylase Kinase &
Glycogen Synthase, enzymes which are regulated
Receptor: Odorants (olfactory epithelium); Effects: Inc. reciprocally by cAMP-dependent phosphorylation ->
Adenylyl Cyclase -> INCREASED cAMP Carb Storage & Release
What Receptor & Effect do Golf Protein have? How does cAMP exert most of its effects
biochemically?
Receptors: NTs in Brain; EFFECT UNKNOWN
What Receptor & Effect do Go Proteins have? 1) cAMP-stimulated phosphorylation of Enzyme
Substrates are rapidly reversed by PHOSPHATASES;
Receptors: ACh, Bombesin, & Serotonin; Effect: Inc. 2) cAMP itself is degraded to 5'-AMP via Cyclic
Phospholipase C -> INCREASED IP3, DAG, Nucleotide Phosphodiesterases (PDE); 3) Caffeine,
Cytoplasmic Ca2+ Theophylline, & other Methylxanthines Competitively
What Receptor & Effect do Gq Proteins have? Inhibit & Degrade cAMP
What are the different ways cAMP & its effects are
Receptors: Photons (Rhodopsin & Opsins in retinal rod/ terminated?
cone cells); Effect: Inc. cGMP Phosphodiesterase ->
DECREASED cGMP (phototransduction) Receptor Activates G Protein -> PLC (Phospholipase
What Receptor & Effect do Gt1 & Gt2 Proteins have? C) -> (1) DAG & (2) IP3 -> DAG activates PK-C
(Protein Kinase C) -> Phosphorylation -> Response; ->
7-transmembrane (7-TM) aka "serpentine" receptors; IP3 (water soluble) diffuses across membrane ->
Agonist binds to hydrophobic transmembrane regions Triggers release of Ca2+ from internal vesicles -> Ca2+
of 7-TM receptor -> G Proteins interact w/ cytosolic binds to CaM (Calmodulin) -> CaM-E (Calmodulin-
loop part of 5th & 6th domain -> Receptor's binding enzymes) -> Response
Cytoplasmic Terminal Tail contains Serine & Threonine How does Ca2+ & Phosphoinositide work as a 2nd
resides whose OH groups can be phosphorylated -> Messenger System?
Phosphorylation -> Diminished receptor-G protein
interaction IP3 is inactivated by Dephosphorylation; DAG is
What type of Receptors are coupled to G Proteins in phosphorylated to yield Phosphatidic Acid ->
general? How do these receptors work w/ G Proteins? Phospholipids, or it yields Arachidonic Acid; Ca2+ is
Actively removed from the cytoplasm by Ca2+ pumps
After reaching an initial high level, the response (cAMP What are the different ways the IP3 & DAG pathway
accumulation, Na+ Influx, Contractility) diminishes over are terminated?
seconds or minutes, even in the continued presence of
the agonist. This "desensitization" is rapidly reversible; cGMP is more rare than cAMP and is found in Intestinal
a 2nd Exposure of the agonist to the receptor when Mucosa & Vascular Smooth Muscle tissue; It's
provided a few minutes after termination of the 1st mechanism is similar to cAMP but instead involves
exposure results in a response similar to the initial stimulating Guanylyl Cyclase to produce cGMP which
response activates cGMP-dependent Protein Kinase;

8
PHARMACOLOGY QUIZLET

How does the 2nd messenger cGMP work and where is


it found? aka Maximum Efficacy or Emax; is the greatest effect
(Emax) an agonist can produce if the dose is taken to
It causes RELAXATION of Vascular SM by a kinase- very high levels; determined mainly by the Nature of the
mediated mechanism that results in Drug & its associated effector system; It can be
DEPHOSPHORYLATION of Myosin Light Chains; measured w/ a graded dose-response curve but not w/
cGMP synthesis is elevated in these SM cells via ANP a quantal dose-response curve
(Atrial natriuretic peptide) & NO (Nitric Oxide) to help in What is Efficacy? What is it determined mainly by?
treating Cardiac Ischemia & Acute HTN Which curve can measure efficacy and which cannot?
What is the function of cGMP?
aka as the EC50 or ED50, Potency is the amount of
1) Amplification & 2) Flexible Regulation drug needed to produce a given effect; Determined
All 2nd messenger signaling involves Reversible mainly by the Affinity of the Receptor for the drug and
Phosphorylation which performs which 2 functions in the # of Receptors available; Both Graded & Quantal
Signaling? curves can determine Potency
What is Potency? What is it determined by? Which
GTP Bound to a G Protein leads to the attachment of a curves measure Potency?
Phosphoryl group to a Serine, Threonine, or Tyrosine
Residue which powerfully amplifies the initial regulatory If the maximal drug response (Emax) is obtained at less
signal by recording molecular memory that the pathway than maximal occupation of the receptors (Bmax); If the
has been Activated EC50 < Kd; 1) The Duration of the activation of the
How does Amplification occur? effector may be much greater than the duration of the
drug-receptor interaction; 2) Actual # of receptors may
Different substrates of the multiple protein kinases exceed the # of effector molecules available
regulated by 2nd messengers provide signaling When are Spare Receptors known to exist? What are
pathways that can be independently regulated by the 2 mechanisms that lead to spare receptors?
cAMP, Ca2+ or other 2nd messengers to produce
Different Effects in Different Cell Types Ratio of TD50 or LD50 to the ED50; determined from
How does Flexible Regulation work? QUANTAL response curves; Represent an estimate of
the SAFETY of a drug (b/c a very safe drug might be
The GRAPH of the Response vs. Drug Concentration/ expected to have a very large toxic dose & a much
Dose when the response of a particular receptor- smaller effective dose) The SMALLER the #, the Safer
effector system is measured against increasing the drug
concentrations of a drug; Efficacy (Emax) & Potency What is Therapeutic Index (TI)? What is it determined
(EC50 or ED50) are derived; The smaller the EC50 or by? What does it represent an estimate of?
ED50, the GREATER the Potency of the drug
What is a Graded Dose-Response Curve? What Actually more clinically relevant than TI b/c it describes
parameters are derived from this graph? the DOSAGE RANGE b/t the minimum Effective
therapeutic concentration or dose & the minimum Toxic
Potency concentration or dose
The smaller the EC50 or ED50, the GREATER the What is Therapeutic Window (TW)?
______ of the drug
1) Intracellular proteins may block access of the G
Affinity protein to the activated receptor molecule (Beta-
The smaller the Kd, the GREATER the _______ of the Arrestin prevents the access of Gs coupling protein &
drug thus desensitizes the tissue to further beta agonist
activation); 2) Receptors are Internalized via
Graded: The concentration/dose that causes 50% of Endocytosis and are later either reinserted into the
the Maximum Effect or Toxicity; Quantal: The membrane (morphine receptors) or degraded (beta
concentration/dose that causes a specified response in adrenoceptors, EGF receptors0
50% of the Population under study What are 2 Ways Receptor Response is diminished
What is EC50, ED50, TD50, etc in GRADED Dose- after Frequent or Continuous Agonist Exposure?
Response Curves? In QUANTAL Dose-Reponse Examples?
Curves?
CHAPTER 3 PHARMACOKINETICS AND
EC50/ED50 is the median Effective dose; TD50 is the PHARMACODYNAMICS
median Toxic dose; LD50 is the median Lethal dose
What is EC50/ED50? TD50? LD50? Define Pharmacokinetics and Pharmacodynamics

9
PHARMACOLOGY QUIZLET

Pharmacokinetics: what the body does to the drug Remember: ions are water-soluble, and non-ionic/
neutrally-charged atoms are lipid-soluble.
Pharmacodynamics: what the drug does to the body
What is the log of a number?
In what ways can some drugs have direct effects
without a receptor? What are the logs of 10, 100 and 0.1?
- Interact directly The power that the base has to be raised to in order to
- Interact with water molecules (e.g. Mannitol) make the number.

Define Bioavailability 10 = 1
The fraction of unchanged drug reaching the systemic 100 = 2
circulation following administration by any route 0.1 = -1
What is the log of 0.2, then?
What is the general bioavailability for drugs given via
the following routes: IV, Inhalation and Oral? State the Henderson-Hasselbach Equation and define
IV = 100% the relationship between the parts.
Inhalation = 5 to <100% Log Unprotonated/Protonated = pH - pK
Oral = 5 to < 100%
As pH goes down, the amount of protonated drug
First-pass effect increases. As pH goes up, the amount of UNprotonated
Hepatic elimination can reduce bioavailability. Drugs drug increases.
can be absorbed by the gut wall (can be metabolized in
gut wall), portal blood (can be metabolized in portal SCM's
blood) delivers drug to the liver, can be excreted into
bile, metabolized in liver, and a portion eventually - What are they
enters systemic circulation. - What do they do
- How do they work
What are two routes that can be used to avoid the first- Special Carrier Molecule
pass effect?
Sublingual and transdermal, as they bypass the portal - Transports substances that are too large or too
veins insoluble, such as peptides, amino acids, glucose.
- Occurs via active transport or facilitated diffusion

Fick's Law How do S.C.M.'s work?


Speed of diffusion = (C1-C2) x Area x k / Thickness Give an example.
- SCM's are saturable and inhibitable
Barriers to aqueous diffusion (2) - Less selective SCM can also eliminate foreign
- Binding to large plasma proteins molecules from the cell, such as multi-drug-resistance
- "Charged" molecules - the electrical field may slow type 1 [MDR1] transporter.
diffusion
Name four types of plasma protein
What can a lipid:aqueous partition coefficient tell us?
Indicates how well or poorly drugs move between lipid What do they have affinity for?
and aqueous compartments - Albumin = most abundant, affinity to acidic but can
bind to all drugs
What is pH? - a-1 acid glycoprotein = favors basic
- Measure of activity of H+ -B-globulin = favors basic
- Closely related to H+ concentration - Lipoproteins, transcortin = less important
- Acidic vs. Basic
Protein Binding Formula
Fill in the blank: Lipophilicity __________ with each [free drug] + k on
additional carbon atom (R) added. [unbound protein binding sites] ---> [bound drug]
Increases <---
k off
Protonated forms for weak bases and weak acids
Weak bases = ionic or "charged", water-soluble Define volume of distribution (Vd)
Weak acids = non-ionic or "neutral", lipid-soluble The apparent volume of plasma that a given dose of
drug is dissolved in.

10
PHARMACOLOGY QUIZLET

Cytochrome P450 nomenclature


Vd formula All start with CYP
Vd = amount of drug given / plasma concentration of
drug First number = genetic family
First letter = genetic subfamily
Vd is INVERSELY proportional to plasma concentration Last number = specific gene

Plasma concentration refers to both free fraction and Elimination vs clearance


protein bound. Elimination: all processes which result in removal of
drug from the body
Vd formula for the mathematically inclined - considered eliminated if changed to new form or
Vd = Vp + Vt (Fp/Ft) excreted
- units are fraction of drug eliminated / time
Vp = volume of plasma
Vt = volume of tissue Clearance: measures the rate at which drug is
Fp = fraction of unbound drug in plasma completely removed from the body, typically plasma
Ft = fraction of unbound drug in tissue - Units are volume/time

Two phases of drug decline after IV injection Two orders of elimination, give an example of a process
- Alpha or "distribution" phase that involves both
- Has rapid and slow components Zero order: constant rate
- Beta or "elimination" phase First order: constant percentage, this is the most
-Represents terminal elimination common

Define distribution half-life and elimination half-life Saturable kinetics: first order until a certain point, then
Distribution half-life zero order. Alcohol is an example. This is also known
- Amount of time it takes for 1/2 of drug administered to as Michaelis-Menten elimination.
move from central compartment to rapid equilibrating
peripheral compartment Clearance formula
Clearance = rate of elimination / C
Elimination half-life, or what we generally refer to as
"half-life" C = concentration of drug in the blood, plasma, or
unbound drug in water
- Amount of time it takes to eliminate 1/2 the drug from
the body Clearance is inversely proportional to drug
- Indicator of amount of time it takes to attain 50% of concentration.
steady state
-INdicator of amount of time it takes to decay 50% from Also: rate of elimination = clearance x concentration
steady state after a change in the rate of drug
administration So elimination is DIRECTLY proportional to drug
concentration
Describe the processes involved in the two phases of
drug metabolism. Second clearance formula
Phase 1 - oxidation, hydroxylation, reduction, hydrolysis Cl = KeVd
Phase 2 - conjugation
Ke = elimination rate constant
Aim of both is to create more polar, water-soluble Vd = volume of distribution
molecules.
Half life formula
What enzyme is primarily involved in Phase I Half life (T 1/2) = log of 2 / Ke = 0.693/Ke
reactions? Ke = elimination rate constant
Cytochrome P450
Formula demonstrating half-life relation to clearance
What happens in conjugation reactions? t 1/2 = 0.7 x Vd/Cl. Half-life is inversely proportional to
Enzymes link one chemical to another to create more clearance.
water-soluble molecules to facilitate elimination from
the body. Rule of thumb for half-lives and full effects of drugs

11
PHARMACOLOGY QUIZLET

Four half-lives must elapse before full effects will be make. Reflects percentage of people exhibiting desired
seen. effect, not individual responses.

Extraction Ratio fomula Isomer


ER = liver clearance (CL) / Q two or more different chemical compounds that have
the same molecular formula; divided into two broad
Q = hepatic blood flow Approximately 90 L/hr. classes:
- structural isomerism
Context-Sensitive Half-Time - stereoisomerism
The time required for the drug concentration in the
central compartment to decrease by 50% after Structural Isomers
discontinuation of drug administration. Same molecular formula only. Linkages between atoms
can vary.
For most drugs, the half-time increases markedly as the
infusion duration lengthens. However, a drug with a Stereoisomers, entantiomers and diastereomers
long elimination half-life CAN have a brief CSHT. • Isomers that have the same structure (linkages
between atoms) but different spacial arrangements.
What is an inert receptor? What problem can this Further divided into entantiomers which are
potentially cause? molecules that are mirror images of each other but
A receptor that does not cause a direct biologic effect. are not superimposable upon one another, or
An example is albumin, which binds with a ligand, but diastereomers, which are not mirror images of each
causes no subsequent biological activity. other.

An effect would be that it may cause problems by Chirality


taking an important drug out of the plasma. The property of handedness, of not being
superimposable on a mirror image.
Potency
Concentration (EC50) or dose (ED50) of drug required Racemic
to produce 50% of maximum effect. A drug can have Made up of two entantiomorphic isomers as an
less effect but be more potent. equimolar mixture and therefore is optically inactive (+)

Efficacy Dextrorotary
Maximum response possible with any dose. Polarized light rotated to right

ED50
Median effective dose - that dose at which 50% of Levorotary
subjects exhibit the specified effect Polarized light rotated to left

EC50 2 other systems of naming isomers


Concentration at which 50% of subject exhibit the D/L = relationship to glyceraldehyde molecule
specified effect R/S = for hardcore chemistry majors only

TD50 Racemic Mixtures


Median toxic dose - dose required to produce a Most drugs are racemic mixtures
particular toxic effect in 50% of subjects Consequence: patients are receiving doses of which
50% or more has a different or no effect
LD50
Median lethal dose. 5 types of receptor mechanisms
• Ligand gated ion channels
Therapeutic index • Metaphobores
Therapeutic index is the ratio of TD50 to ED50. • G protein coupled
TD50:ED50 (or LD50:ED50) • Intracellular receptors
Larger = safer • Transporters

Quantal dose-effect curves Ligand Gated Ion Channel


A curve constructed if a response to a drug is not Definition and examples
graduated and a dose-response curve is difficult to • Bonding of ligand results in opening of ion channel
• Examples: GABA or acetylcholine receptor

12
PHARMACOLOGY QUIZLET

• Drugs that use this mechanism: muscle relaxants (as • Phenytoin


antagonist), propofol, benzodiazepines, inhaled • Carbamazepine
agents • Other drugs:
• Rifampin
Voltage-gated Ion Channels • Glutethimide
Definition and examples • Phenylbutazone
• Spironolactone
• Some ion channels are opened by changes in • Other factors:
voltage, which can be affected by drugs. • Cigarette smoking
• E.g. verapamil effects calcium (Ca++) channels • Charcoal broiled beef
altering sensitivity of the cell to depolarization. • Dietary protein/carbohydrate ratio
• Chronic alcohol abuse
Metaphobores
Definition and examples CHAPTER 4 DRUG BIOTRANSFORMATION
• Receptor complex is trans-membranous.
• Ligand binding extracellularly results in directly Biotransformation
enzymatic conversion of intracellular substance that Chemical alteration of the drug molecule by the cells of
has further physiologic effects. the animal
• Ex: insulin, many trophic hormones such as
epidermal growth factor, atrial natriuretic peptide, One of the results of biotransformation (a)
work this way. Change in the physiochemical properties of the drug
-->The metabolite is more water soluble, polar and
G Protein coupled ionized
Definition and examples
• Like metaphobores, except the intracellular One of the results of Biotransformation (b)
substance release is not the effector Change in pharmacological activity
• Characteristic 7 transmembrane loop protein with --> Bioinactivation or detoxification (default)
intra and extra cellular components --> Bioactivation or lethal synthesis (few drugs)
• Bonding of (extracellular) ligand results in release of
(intracellular) G protein that has effect Examples of biotransformation
• Example: B adrenergic receptors Active drug to active metabolite
(aspirin --> salicylic acid)
G Protein receptor processes
• Release of G-protein complex results in activation of Inactive drug to active metabolite
adenylyl cyclase which in turn results in increased (chloral hydrate --> trichloroethanol)
cAMP.
• After binding GTP substituted for GDP. Nontoxic drug to toxic metabolite (this is lethal
• Hydrolysis of G-Protein leads to GDP and synthesis; we don't want this)
recombination with the receptor complex. (parathion --> paroxon)
-Antifreeze is an example of this
Intracellular receptors
• Ligand is lipid soluble so crosses membrane easily, Sites of Biotransformation
then acts on cellular receptor. • Liver (most important)
• E.g. steroids. • Nervous tissue
• Mechanism of action results in slow onset and • Kidney
prolonged effects. • GI tract
• Lungs
Transporters • Skin
• Some substances are too large to cross cell • Plasma
membrane after release, such as serotonin.
• Normally, transporter proteins bring these substances Characteristics of Hepatic metabolism: Enzymes lack
back into cell. _____.
• Drugs can be used to block this function, such as Specificity
fluoxetine.
Characteristics of Hepatic metabolism: Enzymatic
Inducers of drug metabolism in humans reactions are _______ reactions
• Most important to remember are: saturable
• Phenobarbital and other barbiturates

13
PHARMACOLOGY QUIZLET

Characteristics of Hepatic metabolism: Drugs can Examples of Non-microsomal hydrolases (For


compete on _______ enzyme hydrolysis in Phase I)
Same • Esterases
• Amidases
Characteristics of Hepatic metabolism: Enzymes can • Peptidases
be _______ or _______ by drugs
Induced or inhibited Examples of Esterases
• Acetylcholine
Characteristics of Hepatic metabolism: Microsomal • Succinylcholine
enzymes cause ________, _________,__________ • Procaine
and ________ to glucuronic acid
• Oxidation Example of Amidase
• Reduction Procainamide
• Hydrolysis Example of Peptidase
• Conjugation Proinsulin
Metabolites from phase I reactions that are more
Phase I reactions (non-synthetic) involve what reactive (toxic) are further handled by _______.
processes? Phase II reactions aka Conjugation
• Oxidation
• Reduction What is the most common conjugation reaction?
• Hydrolysis Conjugation to glucuronic acid
Phase II reactions (synthetic) involves what process?
Conjugation Conjugation is __________.
Combination of a drug or its metabolite (after Phase I)
Processes involved in microsomal (Smooth ER) with an endogenous substance
oxidation (Phase I)
• Hydroxylation Products of conjugation
• Deamination • Glucuronic acid
• Dealkylation • Sulfuric acid
• Desulfuration • Acetyl group
• Sulfoxide formation • Methyl group
• Glycine
Sites involved in non-microsomal oxidation (Phase I) • Methionine
Cytosol or mitochondria • Glutathione

Enzymes involved in oxidation Glucuronidation is ________.


Alcohol and aldehyde dehydrogenase Microsomal
(ethanol -->acetaldehyde)
All other conjugations are nonmicrosomal
Xanthine oxidase
(xanthine --> uric acid) Conjugation is _______ in the neonate
Deficient
Tyrosine hydroxylase
(Tyrosine --> dopa) Metabolite conjugates are usually ______ and
_______.
Monoamine oxidase Inactive and water-soluble
(Metabolism of catecholamines and serotonin)
Can conjugation reactions result in formation of
What is the most common biotransformation reaction? metabolites?
Oxidation Yes

Process involved in microsomal reduction Example of conjugation that forms metabolites


Nitro reduction of chloramphenciol Acyl glucuronidation of NSAID which is toxic to the liver

Process involved in non-microsomal reduction Factors altering biotransformation


Aldehyde reduction of chloral hydrate • Species
• Individual
• Route of Administration

14
PHARMACOLOGY QUIZLET

• Enzyme inducers How does route of administration affect


• Enzyme inhibitors biotransformation?
• Liver disease • There is a first pass effect for oral and intraperitoneal
• Hepatic blood flow routes (and drugs administered through the skin)
• Plasma protein binding
• Distribution and storage Examples of drugs that have extensive first pass effect
• Age Lidocaine
• Sex
• Diet and nutrition Propranolol
• Body Temperature
• Environmental Factors Morphine

Cats are deficient in ________. What are enzyme inducers


Glucuronyl transferases for phenols and aromatic Up regulators
amines
Drugs that stimulate the liver to produce more
(GT needed to break down aspirin) metabolic enzymes

Cats are deficient in ______ but may be able to Only _____ enzymes are inducible by drugs
conjugate endogenous substrates such as ______, Microsomal
_______ and _______. • Examples are phenobarbital, phenylbutazone,
Glucuronyl transferases griseofulvin, rifampin (and chlorinated hydrocarbon
insecticides)
Steroids, thyroxine, and bilirubin
Enzyme induction is _______.
Cats are also deficient in _______ and ________. Reversible
Hydroxylation and dealkylation
Enzyme induction may lead to ______ or _________.
Dogs lack _______ enzymes • Tolerance
acetylating • Drug-drug interactions

Ruminants have ______ plasma _______ than horses, What are enzyme inhibitors?
dogs and cats Down regulators (slow down)
Less
Pseudocholinesterase Drugs that inhibit the liver to produce metabolic
enzymes
Ruminants and horses have _____ levels of _______
enzymes Examples of enzyme inhibitors
High • Chloramphenicol
Oxidative • Cimetidine
• Ketoconazole
Pigs are deficient in ________ enzymes
Sulfate conjugating Enzyme inhibition may lead to _________.
Drug-drug interactions
Birds lack _______ enzymes
Oxidative Distribution and storage _______ biotransformation.
Decrease
Fish have _____ levels of _______ enzymes
Low Newborns and geriatric patients may have ________
Drug metabolizing biotransformation than adults
Less
How does an individual effect biotransformation?
• Certain individuals may be deficient or lack certain Examples of sex-dependent variations in drug
enzymes due to genetic differences metabolism in humans are ______, ________,
________ and _______.
Ex: 50% of dogs do not have CYP2D15 which Ethanol, propranolol, benzodiazepines, salicylates
metabolizes celecoxib
Don't recognize this factor much in animals

15
PHARMACOLOGY QUIZLET

Synthetic reactions that involve addition (Conjugation)


Charcoal broiled foods are _______ of subgroups to -OH, -NH2, & -SH functions on the
Enzyme inducers (CYP1A) drug molecule. Subgroups added are Glucuronate
(Glucuronidation), Acetate (Acetylation), Glutathione
Grape fruit juice is an ________. (Glutathione Conjugation), Glycine (Glycine
Enzyme inhibitor (CYP3A) Conjugation), Sulfate (Sulfation), & Methyl
(Methylation)Groups; These groups are POLAR so they
Malnutrition and under-nutrition may _______ make it less lipid soluble; Reactions that increase water
biotransformation solubility by conjugation of the drug molecule w/ a polar
Decrease moiety such as glucuronate, acetate, or sulfate
What are Phase II Reactions? What are the subgroup
Nutrition plays an important role in the regulation of processes involved? How do these groups make the
_______ product less lipid soluble?
drug conjugates
Mostly LIVER, Sometimes Kidneys, and a few drugs
Liver disease may not have an effect on function liver. T are metabolized in many tissues (ex: liver, blood,
or F? intestinal wall)
True What are the Sites of Drug Metabolism?

How does liver disease effect biotransformation? Cytochrome P450 enzyme species (ex: CYP2D &
May decrease liver enzymes CYP3A4) that are responsible for much of drug
metabolism. Many isoforms of CYP have been
Hepatic blood flow may be _____ due to liver disease recognized.
or drugs resulting in ______ biotransformation What are CYP Isozymes?
• Decreased
• Decreased
Stimulation of drug-metabolizing capacity; usually
Plasma protein binding _______ biotransformation manifested in the liver by increased synthesis of
Decreases Smooth ER (contains high concentrations of Phase I
enzymes) Inducers selectively INCREASE the
Hypothermia, shock or anesthesia may _______ subgroups of Isozymes
biotransformation What is Enzyme Induction?
Decrease
An ATP-dependent transport molecule found in many
Animals in the tropics may have ____ biotransformation epithelial & cancer cells. The transporter expels drug
than animals in temperate climate molecules from the cytoplasm into the extracellular
• Less space. In epithelial cells, expulsion is via the external or
• Ex: Trimethoprim in goats luminal face; Expels unwanted molecules immediately
after absorption
Foreign chemical compounds in the air, water, & food. What is P-glycoprotein?
What are Xenobiotics?
1) Hydrolysis of Esters (Succinylcholine is metabolized
Lipid-Soluble; Body undergoes Drug Biotransformation by plasma cholinesterase very SLOWLY w/ genetically
to make drugs less lipid-soluble abnormal forms of the enzyme leading to an extended
What important property of most drugs makes it neuromuscular paralysis); 2) Acetylation of Amines (Ppl
favorable for absorption across membranes but also are "Slow Acetylators" if they are deficient in acetylation
results in very slow removal of the body b/c the capacity of Amine Drugs); 3) Oxidation (rate of
molecule is readily absorbed from the urine back into oxidation of particular drugs by certain P450 isozymes)
the renal tubule? What does the Body undergo to Which 3 Drug Metabolizing Systems have a Genetic
hasten the excretion of these drugs? Factor component to it?

Oxidation (cytochrome P450 enzymes), Reduction, Carbamazepine, Phenobarbital, Phenytoin, & Rifampin
Deamination, & Hydrolysis; Reactions that convert the What are the most common Strong Inducers of Drug
parent drug to a more polar (water-soluble) or more Metabolism?
reactive product by unmasking or inserting a polar
functional group such as -OH, -SH, or -NH2 Drugs: Acetaminophen, Clozapine, Haloperidol,
What are Phase I Reactions? Theophylline, Tricyclic Antidepressants, (R)-Warfarin;
Inducers: Smoking, Charcoal-broiled Foods,

16
PHARMACOLOGY QUIZLET

Cruciferous Vegetables, Omeprazole; Inhibitors: Since, P-gp expels drugs from the intestine into the
Cimetidine, Fluoroquinolones, Grapefruit Juice, lumen, Inhibitors inhibit P-gp and therefore INCREASE
Macrolides, Isoniazid, & Zileuton Bioavailability & may result in toxic plasma
What are the Inducers, Inhibitors & Drugs Metabolized concentrations of drugs given at normally nontoxic
by the CYP1A2 Family? dosage; Ex: Verapamil, Mibefradil & Furanocoumarin
(Grapefruit Juice)
Inducers: Barbiturates, Phenytoin, Primidone, Rifampin; What is the effect of Inhibitors of P-glycoprotein (P-gp)?
Inhibitors: Amiodarone, Chloramphenicol, Cimetidine,
Isoniazid, Metronidazole, SSRIs, & Zafirlukast; Drugs: Digoxin, Cyclosporine, & Saquinavir
Barbiturates, Cholarmphenicol, Doxorubicin, Ibuprofen, What are examples of Drugs normally expelled by P-
Phenytoin, Chlorpromazine, Steroids, Tolbutamide, & gp?
(S)-Warfarin
What are the Inducers, Inhibitors & Drugs Metabolized Drugs being metabolized does NOT mean they are
by the CYP2C9 Family? being Inactivated. In fact, some drugs are converted to
active products which may be Toxic. Example: A large
Inducers: Carbamazepine, Phenobarbital, Phenytoin, & overdose of Acetaminophen results in Reactive Toxic
Rifampin; Inhibitors: Fluconazole, Omeprazole, & Intermediates such as N-acetyl-p-benzoquinoneimine
SSRIs; Drugs: Tricyclic Antidepressants, Phenytoin, b/c the metabolic pathways are overwhelmed
Topiramate, & (R)-Warfarin What is Toxic Metabolism? Example?
What are the Inducers, Inhibitors & Drugs Metabolized
by the CYP2C19 Family? Amphetamines, Barbiturates, Phenytoin, Caffeine,
Morphine, Theophylline, Codeine, Acetaminophen,
Inducers: Ethanol & Isoniazid; Drugs: Acetaminophen, Nicotine, Chlorpromazine, Cimetidine, Thioridazine, &
Ethanol (minor), & Halothane Diazepam
What are the Inducers & Drugs Metabolized by the What are examples of Drug Substrates that undergo
CYP2E1 Family? P450 DEPENDENT Oxidations?

Inducers: Barbiturates, Carbamazepine, Epinephrine, Chloral Hydrate, & Ethanol


Corticosteroids, Efavirenz, Phenytoin, Rifampin, & What are examples of Drug Substrates that undergo
Troglitazone; Inhibitors: Amiodarone, Azole Antifungals, P450 INDEPENDENT Oxidations?
Cyclosporine, Erythromycin, Grapefruit Juice, HIV
Protease Inhibitors, Quinine, SSRIs, Tacrolimus; Drugs: Chloramphenicol, Clonazepam, Dantrolene, &
Antiarrhymthmics, Antidepressants, Azole Antifungals, Naloxone
Benzodiazepines, Calcium Channel Blockers What are examples of Drug Substrates that are
What are the Inducers, Inhibitors & Drugs Metabolized involved in Phase I Reductions?
by the CYP3A4 Family?
Aspirin, Clofibrate, Procaine, Succinylcholine,
Drugs: Antidepressants, Flecainide, Lidocaine, Indomethacin, Lidocaine, & Procainamide
Mexiletine, & Opioids; Inhibitors: Amiodarone, What are examples of Drug Substrates that are
Cimetidine, Quinidine, SSRIs involved in Phase I Hydrolyses?
What are the Inhibitors & Drugs Metabolized by the
CYP2D6 Family? Acetaminophen, Diazepam, Digoxin, Morphine, &
Sulfamethiazole
Drug metabolism is diminished usually by What are typical Drug Substrates for Phase II
Amiocadarone, Cimetidine, Furanocoumarins Glucuronidation?
(Grapefruit Juice), Ketoconazole, & the HIV Protease
Inhibitor Ritonavir Clonazepam, Dapsone, Isoniazid, Mescaline, &
What is Enzyme Inhibition? What are the most common Sulfonamides
inhibitors of drug metabolism? What are typical Drug Substrates for Phase II
Acetylation?
Drugs that are metabolized to products that
IRREVERSIBLY Inhibit the metabolizing enzyme; Ethacrynic Acid, Reactive Phase I metabolite of
Examples: Ethinyl Estradiol, Norethindrone, Acetaminophen
Spironolactone, Secobarbital, Allopurinol, Fluroxene, & What are typical Drug Substrates for Phase II
Propylthiouracil Glutathione Conjugation?
What are Suicide Inhibitors?
Deoxycholic Acid, Nicotinic Acid (Niacin), & Salicylic
Acid

17
PHARMACOLOGY QUIZLET

What are typical Drug Substrates for Phase II Glycine


Conjugation?

Acetaminophen, Estrone, & Methyldopa


What are typical Drug Substrates for Phase II
Sulfation?

Dopamine, Epinephrine, Histamine, Norepinephrine, &


Thiouracil
What are typical Drug Substrates for Phase II
Methylation?

Hydralazine & Procainamide


Which 2 Drugs are associated w/ SLOWER Metabolism
in White & African-Americans than in most Asians?

18

Вам также может понравиться