Block 1 Flashcards
pharmacology definition
biomedical science concerned w/study of drugs and their effects on life processes
goal of pharmacology
understand mechanisms by which drugs interact w/biologic systems to enable rational use of effective agents in diagnosis & treatment of disease
2 subdivisions of pharmacology
- pharmacokinetics
- pharmacodynamics
Pharmacokinetics definition
study of actions of body on drug (ADME)
Pharmacodynamics definition
Study of actions of drug on body
4 domains of pharmacokinetics
ADME:
- Absorption
- Distribution
- Metabolism (biotransformation)
- Excretion
dose-response relationship
relationship btwn concentration of drug in tissue and magnitude of tissue’s response to drug
Most drugs produce their effects by
bind to protein receptors in target tissues → activate signal transduction cascade
Toxicology definition
study of poisons and organ toxicity; focuses on harmful effects of drugs & mechanisms by which these agents produce pathologic changes, disease, and death
Pharmacotherapeutics definition
medical science concerned with the use of drugs in the treatment of disease
clinical trials
Human studies used to determine efficacy & safety of drug therapy in human subjects
Pharmacy definition
science & profession concerned w/preparation, storage, dispensation, proper use of drug products
Pharmacognosy definition
study of drugs isolated from natural sources, including plants, microbes, animal tissues, and minerals
Medicinal chemistry definition
branch of organic chemistry that specializes in design & chemical synthesis of drugs
Pharmaceutical chemistry / pharmaceutics definition
concerned w/formulation & chemical properties of pharmaceutical products, such as tablets, liquid solutions and suspensions, and aerosols
drug definition
natural product, chemical substance, or pharmaceutical preparation intended for administration to human or animal to diagnose or treat a disease
types of drugs produced by body
hormones, neurotransmitters, or peptides
xenobiotic
synthetic or natural drug produced outside the body
poison
a drug that can kill
toxin
a drug that can kill and is produced by a living organism
alkaloid definition
contain nitrogen groups and produce an alkaline reaction in aqueous solution
alkaloid examples
morphine, cocaine, atropine, quinine
Antibiotics
drugs targeted to bacteria; isolated from numerous microorganisms, including Penicillium and Streptomyces species
structure-activity relationship
relationship btwn drug molecule, its target receptor, and resulting pharmacologic activity
crude drug preparations
obtained from natural sources; can be made by drying or pulverizing plant or animal tissue (ex: opium), or extracting substances from a natural product (ex: coffee)
pure drug compounds
isolated from natural sources or synthesized in lab (ex: morphine extract)
pharmaceutical preparations
intended for administration to patients (ex: morphine solution)
routes of drug administration
- enteral
- parenteral
- systemic
types of drugs
natural, semi-synthetic, or synthetic
drugs regulated by
the FDA’s Center for Drug Evaluation and Research (CDER) – except biologics
CDER
FDA entity that regulates drugs (except biologics)
Prescription drug
aka “legend drug;” considered potentially harmful if not used under supervision of licensed health care practitioner
biologics
complex mixtures that are not easily identified or characterized; incl. vaccines, recombinant therapeutic proteins, and gene therapy
biologics regulated by
FDA’s Center for Biologics Evaluation and Research
CBER
FDA entity that regulates biologics
Controlled or scheduled drug
prescription drug whose use and distribution is tightly controlled b/c of abuse potential or risk
Classification of controlled drugs
Schedules CI, CII, CIII, CIV, and CV
Over-the-counter drugs
Do not require a prescription but still require FDA drug approval process
Behind-the-counter drugs
OTC drugs with restricted access (ex: sudafed)
regulation of dietary supplements
FDA’s Center for Food Safety and Applied Nutrition
What to consider when Choosing a Drug for Your Patient
benefit vs risk
STEPS Approach to Evaluating and Comparing Drugs
- S – Safety
- T – Tolerance
- E – Efficacy
- P – Price
- S – Simplicity
medication nonadherence
At least 50% of patients do this
percentage of adults > 65 years old who take more than 5 drugs
42%
top 3 therapeutic classes of prescriptions
- antihypertensives
- mental health
- pain
top 3 prescriptions (number prescribed)
- levothyroxine
- acetominophen/hydrocodone
- lisinopril
how many phases of clinical testing
4
Clinical Testing Phase 1
safety, PK, dose range, 20-80 subjects
Clinical Testing Phase 2
test hypothesis of effectiveness, controlled trial, 100-300 subjects
Clinical Testing Phase 3
randomized, blinded, placebo controlled trials for specific indications
Clinical Testing Phase 4
post-marketing surveillance
FDA Drug Development and Approval Process
- Pre-clinical research (animals)
- Investigational New Drug Application (IND)
- Phases 1-3 of clinical testing New Drug Application (NDA) filed
- FDA subcommittee review and approval
- Phase 4 of clinical testing
drug patent length
20 years; can be extended 5 years but total life of patent cannot go beyond 14 years after NDA approval
chemical drug name
based on chemical structure
generic drug name
public nonproprietary name; United States Adopted Names (USAN); Standards set by US Pharmacopeia (ex: sildenafil citrate)
trade drug name
exclusively owned by manufacturer (ex: Viagra)
most common formulation of drugs is for what kind of administration
oral
most common preparations for oral administration
tablets and capsules
benefits of tablets and capsules
suitable for mass production; stable and convenient to use; can be formulated to release drug immediately after ingestion or over a period of hours
Variations in rate and extent of tablet disintegration and drug dissolution can give rise to
differences in the oral bioavailability of drugs from different tablet formulations
drug in tablet must do what to reach circulation
dissolve in gastrointestinal fluids
Enteric coatings
- don’t disintegrate in gastric acid
- break down in basic pH of intestines
- protect drugs that would otherwise be destroyed by gastric acid
- slow release & absorption when large dose given at one time (ex: fluoxetine)
two methods used to extend the release of a drug
- controlled diffusion
- controlled dissolution
controlled diffusion
regulated by a rate-controlling membrane
controlled dissolution
caused by inert polymers that gradually break down in body fluids
osmotic pressure can be used for
sustained release – formulations contain osmotic agent that attracts gastrointestinal fluid at a constant rate, which then forces drug out of tablet thru small laser-drilled hole
benefits / downsides of solutions and suspensions
- convenient method for administering drugs to pts who can’t easily swallow pills or tablets
- less convenient than solid dosage b/c liquid must be measured each time dose is given
sterile solutions and suspensions be administered in these ways
parenterally: needle & syringe or IV infusion pump
Transdermal administration
- drug slowly released for absorption thru skin into circulation
- most suitable for potent drugs w/lipid solubility (ex: fentanyl patch)
Aerosols
- inhalation thru nose or mouth
- particularly useful for respiratory disorders b/c of direct delivery
Nasal sprays
type of aerosol used either for drugs that have a localized effect on the nasal mucosa or that are absorbed through the mucosa and exert an effect on another organ (ex: nasal butorphanol for pain)
types of enteral administration
sublingual, buccal, oral, rectal
enteral administration definition
drug is absorbed from GI tract
benefits of sublingual and buccal administration
- enable rapid absorption of certain drugs
- not affected by first-pass drug metabolism in liver (ex: nitroglycerin; hyosciamine)
oral administration definition
medication is swallowed, and drug is absorbed from stomach and small intestine
Per Os (PO)
oral administration
benefits / disadvantages of oral administration
- convenient, relatively safe, most economical
- absorption can vary widely
- some drugs inactivated by first-pass metabolism
- not suitable for pts who are sedated, comatose, or experiencing nausea and vomiting
benefits / disadvantages of rectal administration
- useful when pts can’t take medications by mouth (nausea, vomiting)
- can be administered for localized conditions like hemorrhoids
- undergo relatively little first-pass metabolism
Parenteral administration definition
administration w/needle and syringe or IV infusion pump
most commonly used parenteral routes
- intravenous
- intramuscular
- subcutaneous
benefits / disadvantages of IV administration
- bypasses absorption
- greatest reliability and control over dose reaching systemic circulation
- 100% bioavailability
- preferred for drugs w/short T1/2
- good for drugs that have to be carefully calibrated to physiologic response
- dangerous b/c potential for serious toxicity from rapid administration
intramuscular and subcutaneous administration suitable for these types of drug preparations
solutions and particle suspensions
intramuscular or subcutaneous absorbed faster?
intramuscular – greater circulation of blood to muscle
solutions or suspensions absorbed faster?
Solutions are absorbed more rapidly than particle suspensions, so suspensions are often used to extend the duration of action of a drug over many hours or days.
Intrathecal administration
- injection of drug thru thecal covering of spinal cord and into subarachnoid space
- useful in administering antibiotics that don’t cross blood-brain barrier
Epidural administration
targets analgesics into space above dural membranes of spinal cord
intraarticular administration
parenteral route used for arthritis drugs
intradermal administration
parenteral route used for allergy tests
insufflation / intranasal administration
parenteral route used for sinus medications
transdermal patches
- most use rate-controlling membrane to regulate diffusion
- drug intended to reach circulation
benefits / disadvantages of transdermal administration
- bypasses first-pass metabolism
- reliable route for drugs effective at relatively low dosage and highly soluble in lipid membranes
- controlled release possible
Topical administration
- application of drugs to the surface of the body to produce a localized effect
- when applied over inflamed skin, can reach circulation
This route of drug administration is used with potent and lipophilic drugs in patch formulation and avoids first-pass metabolism
transdermal
This route of administration does not have an absorption phase
intravenous
Which parenteral route is used to administer drug suspensions that are slowly absorbed?
intramuscular
An elderly patient has problems remembering to take her medication three times a day. Which one of the drug formulations might be particularly useful in this case?
extended-release
Which form of a drug name is most likely known by patients from exposure to drug advertisements?
trade name
These administration routes bypass first-pass metabolism / the hepatic portal vein
- sublingual
- buccal
- rectal
- transdermal
- parenteral
These administration routes bypass GI tract
- intravenous
- intramuscular
- subcutaneous
- transdermal
mechanisms of absorption
- Passive diffusion
- Active transport
passive diffusion
- drug enters cell until intracellular conc = extracellular conc
- rate depends on drug conc gradient
- Most drugs absorbed by this method
Most drugs absorbed by this method
passive diffusion
active transport
drugs can enter cells against concentration gradient by linking to transport proteins
function of Transport Proteins
Allow efficient transport of molecules across epithelial membranes in the intestine
OATP
- organic anion transporting proteins
- transporters that facilitate uptake
P-glycoprotein (Pgp)
- Efflux transporter that actively removes drugs from epithelial cells and prevents absorption
- Essential mechanism to prevent toxin absorption
- Can also lead to chemotherapeutic drug resistance
essential mechanism to prevent toxin absorption
efflux transporters like Pgp
form of drug that can cross lipid membranes
Only non-ionized form
pKa value of a drug tells you this
pH value at which ½ of drug is in ionic form (i.e. ½ ionized & ½ non-ionized)
pKa and diffusion capability of weak acids
- low pKa
- diffuse across membranes at low pH
pKa and diffusion capability of weak bases
- high pKa
- diffuse across membranes at high pH
gastric acid pH
1.4
plasma pH
7.4
weak acids do what with protons?
donate them to form anions (A-)
weak bases do what with protons?
accept them to form cations (HB+)
non-ionized form of weak acid protonated or nonprotonated?
protonated (HA)
non-ionized form of weak base protonated or nonprotonated?
nonprotonated (B + H+)
Henderson-Hasselbach eqn
- used to determine ionized:nonionized ratio
- log (prot/nonprot) = pKa - pH
Factors Affecting Oral Absorption
- dosage formulations (coatings, controlled-release designs)
- blood flow
- gastric motility
- first-pass effect
Bioavailability (F)
fraction of drug that actually enters the systemic circulation in active form
bioavailability = 100% for this kind of administration
parenteral
First pass effect
drugs absorbed from GI tract are metabolized by liver before reaching systemic circulation
Drugs subject to first-pass effect have this kind of extraction rate and bioavailability
- high hepatic extraction ratios
- low oral bioavailability
examples of drugs w/high (hepatic) extraction ratios
- oral propanalol
- morphine
- meperidine
- nitroglycerin
- verapamil
- lidocaine
Factors Affecting Drug Distribution
- Organ blood flow
- plasma protein binding
- lipid solubility
- molecular size
highly perfused organs that get faster drug distribution
- Liver
- kidney
- heart
- lungs
less-perfused organs that get slower drug distribution
- Skin
- fat
- bone
drug bound to albumin is active or inactive?
inactive – only free drug is active and able to cross cell membranes
Lipid-soluble drugs distribute to a greater or lesser extent than polar and ionized molecules?
lipid-soluble have higher volume of distribution (Vd) and cross BBB
apparent volume of distribution (Vd)
volume of fluid in which a drug would need to be dissolved to have the same concentration as in plasma
can amount of drug in the body be directly measured?
no – usually measure the conc of drug in plasma
Plasma concentration of drug depends on
- dose of drug
- extent of distribution into tissues
Vd
Vd (L) = [amount in body or dose administered (mg)] / [plasma drug concentration after administration (mg/L)]
L = mg / (mg/L)
Plasma volume for 70 kg Individual
- 2.8 L
- 0.04 L/kg
Extracellular fluid volume for 70 kg Individual
(ECF = Plasma + interstitial fluid)
- 17.5 L
- 0.25 L/kg
Intracellular fluid volume for 70 kg Individual
- 24.5 L
- 0.35 L/kg
Total body water volume for 70 kg Individual
- 42 L
- 0.6 L/kg
High Vd indicates…
Drug highly distributed into tissues and fat
Low Vd indicates…
Drug primarily in plasma
Importance of Vd
- Fundamental pharmacokinetic parameter for all drugs
- Useful for calculating loading dose
- Essential for determining elimination rate constant and T1/2
loading dose
- 1st dose of drug
- necessary to reach therapeutic serum levels quickly for drugs w/long T½
Calculate digoxin loading dose if
- T½ = 2 days
- Steady state plasma concentration desired (Cp) = 1.5 mcg/L
- Vd = 7.3 L/kg
- Vd = Dose / Cp
- Dose = Vd x Cp
- Dose = 7.3 L/kg x 70 kg x 1.5 mcg/L = 766.5 mcg or 0.75 mg
Vd formula
Vd = Dose / Cp
or
Dose = Vd x Cp
Drug Metabolism
- activate OR inactivate compounds
- transform compounds into easily excretable metabolites
- 2 phases
Drug Metabolism 1o locations
- liver
- intestinal cell lining
Phase I of Drug Metabolism
Addition of small polar groups to drug structure by oxidation, reduction, or hydrolysis converts lipid-soluble drugs to more polar and water-soluble metabolites (active or inactive)
Phase II of Drug Metabolism
Formation of highly water-soluble conjugates to create inactive and easily-eliminated compound
Cytochrome P450 Enzymes general characteristics & location
- Responsible for most Phase I metabolism reactions
- in all living organisms
- primarily in liver, but also in intestine, lung, brain, and placenta
Cytochrome P450 Enzymes structure
- bound to membranes within a cell (cyto)
- contain heme pigment (chrome and P)
- absorb light at 450 nm when exposed to CO
Cytochrome P450 Enzymes function
- Essential for production of endogenous compounds like cholesterol, steroids, and prostacyclins
- Necessary for detoxification of exogenous compounds like foreign chemicals and drugs
Cytochrome P450 Enzyme Family
- Over 50 distinct P450 enzymes in humans
- Five (1A2, 2C9, 2C19, 2D6, 3A4) metabolize 90% of drugs
Most drug interactions caused by this
changes in Cyt P450 metabolism
Pro-drug
- needs to be metabolized to become active
- parent compound usually inactive
When could pro-drug have little or no clinical effect?
when it depends on a P450 enzyme that isn’t functioning b/c:
- in short supply (poor metabolizer)
- inhibited by something else
Examples of pro-drugs
- Hydrocodone → hydromorphone (pain relief)
- Tramadol (Ultram) → metabolite 33x more active (pain relief)
- Enalapril → enalaprilat (HTN)
- Vyvanse (lisdexamfetamine) → dextroamphetamine in gut (prevent intravenous abuse)
Examples of Active Metabolites of Old Drugs Developed as New Drugs
- Zyrtec (cetirizine) from Atarax (hydroxyzine)
- Allegra (fexofenadine) from Seldane (terfenadine)
- Clarinex (desloratidine) from Claritin (loratidine)
- Invega (paliperidone) from Risperdal (risperidone)
- Pristiq (desvenlafaxine) from Effexor (venlafaxine)
- Trilipix (fenofibric acid) from fenofibrate
Biliary excretion
- Conjugated drug metabolites and large molecular weight compounds
- Drug may be reabsorbed by enterohepatic cycling
- requires active center
Renal excretion
Most drugs eliminated this way, either as parent compound or as inactive metabolite formed in liver
Most drugs eliminated this way
renal excretion
Glomerular filtration depends on
extent of protein binding
3 methods of renal excretion
- Glomerular filtration
- Active tubular secretion
- Passive tubular reabsorption
Passive tubular reabsorption depends on
- lipid solubility
- ionization
Clearance (Cl) definition
- Volume of plasma from which drug is eliminated per unit time (L/hr or ml/min)
- Summation of clearance of drug metabolized by liver and excreted by kidney
- Does not indicate how much drug is removed
Clearance calculation
Elimination rate (mg/hr) = Clearance [Cl] (L/hr) x Plasma conc [Css] (mg/L)
Creatinine Clearance (ClCr)
- allows estimation of GFR
- need accurate estimate of renal function to dose drugs eliminated by kidney
- most dosage adjustments for renally excreted drugs based on estimate of ClCr
Creatinine
- metabolic byproduct of muscle
- constant rate of formation
- elimination almost exclusively by GFR
Formula name for estimating Creatinine Clearance
- Cockroft and Gault formula widely used
- Need to know sex, age, ideal body weight, and Scr
Cockroft and Gault formula
- ClCr = [(140 - age) x IBW in kg] / (Scr in mg/dl x 72)
- multiply ClCr by 0.85 for women
Zero-Order Elimination
- aka capacity-limited or nonlinear elimination
- Elimination rate constant and independent of plasma concentration
- Cl inversely proportional to drug concentration (toxic levels can be reached quickly)
- Very few drugs eliminated by this method
Drugs eliminated by Zero-Order Elimination
- phenytoin (Dilantin)
- aspirin at high doses
- ethanol
First-Order Elimination
- At normal doses, rate of drug elimination is proportional to plasma drug concentration
- if drug concentration increases, so does clearance
- most drugs eliminated by this method
most drugs eliminated by this method
First Order Elimination
Elimination Rate Constant (Ke)
- a fraction of drug eliminated over a set period of time
- ex: Ke = 0.25/hr → 25% of drug remaining in body is removed each hour
Calculating Ke
Ke = Cl (L/hr) / Vd (L)
Mathematical Relationship btwn K and T½
T½ = 0.693 / k
Relationship btwn Ke and Half-Life
T½ = 0.7 / Ke
Relationship btwn Vd, Cl, and Half-Life
T½ = 0.7Vd / Cl
Steady state
drug administration rate = elimination rate
time it takes for a drug to reach steady state is dependent on…
T½ of drug
It takes about how many half-lives to reach steady state after starting drug (no loading dose)?
5
It takes about how many half-lives for elimination of drug after last dose?
5
Therapeutic Drug Monitoring
- Usually for drugs with narrow therapeutic index
- Drug needs to be at steady state, otherwise lab result may lead to dosage error
- Depending on drug, may need peak level, trough level, or both
Examples of Drugs Requiring Therapeutic Monitoring
- Aminoglycoside antibiotics (Gentamicin, tobramycin, amikacin)
- Clozapine (atypical antipsychotic)
- Digoxin (antiarrhythmic)
- Theophylline (bronchodilator)
- Vancomycin (antibiotic)
- Warfarin (anticoagulant)
Summarize the 5 important pharmacokinetic equations
- Vd (L) = Drug in body (mg) / Plasma drug concentration or Cp (mg/L)
- Loading Dose (mg) = Vd x Cp
- Ke = Cl (L/hr) / Vd (L)
- T½ = 0.7 / Ke
- T½ = 0.7Vd / Cl
what protein can lead to chemotherapeutic drug resistance
Pgp
examples of Pgp inhibitors
amiodarone, erythromycin, propranolol
acidic drugs generally bind to what in plasma?
albumin
basic drugs generally bind to what in plasma?
glycoproteins and β-globulins
conjugation
- attachment of polar groups
- often allows for faster excretion
microsomal P450 monooxygenase reaction requires what?
- CYP (a hemoprotein)
- NADPH-dependent CYP reductase
- membrane lipids in which the system is embedded
Most drug biotransformation is catalyzed by which three CYP families?
- CYP1
- CYP2
- CYP3
which CYP subfamily catalyzes more than half of all microsomal drug oxidations?
CYP3A
polymorphism
individual variation in the genes coding for drug-metabolizing enzymes
examples of polymorphisms that affect drug metabolism
- SA phenotype:
- slow acetylators – reach toxicity of certain drugs faster
- CYP2D6 and CYP2C19 differences:
- altered enzymatic rxn rates (codeine; omeprazole)
enterohepatic cycling
- drug excreted in bile
- bile empties into intestines
- fraction of drug may be reabsorbed into circulation and eventually return to liver
biliary excretion eliminates substances from the body only to the extent that…
some of the excreted drug is not reabsorbed from the intestine
one-compartment model
drug undergoes absorption into blood according to rate constant Ka, and elimination from blood with rate constant Ke
two-compartment model
drugs are absorbed into central compartment (blood), distributed from central compartment to peripheral compartment (the tissues), and eliminated back from central compartment
how to accelerate rate of excretion of weak acid?
- alkalinize urine via NaHCO3 administration
- particularly useful for
- aspirin overdose
- salicylate overdose
- phenobarbital overdose
- 2,4-dichlorophenoxyacetic acid (herbicide) poisoning
how to accelerate rate of excretion of weak base?
- acidify urine
- has been largely abandoned b/c does not significantly increase elimination of these drugs and poses serious risk of metabolic acidosis
when is manipulation of the urine pH worthwhile for accelerated excretion?
when drug is excreted to a large degree by kidneys
Parameters of plasma drug concentration curve
- maximum concentration (Cmax)
- time needed to reach maximum (Tmax)
- minimum effective concentration (MEC)
- duration of action
area under the curve (AUC)
measure of total amount of drug during time course
formula to determine oral bioavailability of a particular drug
dividing the AUC of an orally administered dose of the drug (AUCoral) by the AUC of an IV-administered dose of the same drug (AUCIV)
Pharmaceutical factors that can affect bioavailability
- rate and extent of tablet disintegration
- drug dissolution
Biologic factors that can affect bioavailability
- food, which can sequester or inactivate a drug
- gastric acid, which can inactivate a drug
- gut and liver enzymes, which can metabolize a drug during absorption and first pass through the liver
indication that drug has reached intracellular fluid?
Vd = total body water
drugs w/renal clearance close to ClCr…
eliminated primarily by glomerular filtration, w/little tubular secretion or reabsorption
drugs w/renal clearance higher than ClCr…
undergo tubular secretion
drugs w/renal clearance lower than ClCr…
are highly bound to plasma proteins or undergo passive reabsorption from renal tubules
hepatic clearance
usually determined by multiplying hepatic blood flow by arteriovenous drug concentration difference
why is hepatic clearance difficult to determine?
hepatic drug elimination includes biotransformation and biliary excretion of parent compounds
What does graph of first-order kinetics look like?