• 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

1/217

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;

217 Cards in this Set

  • Front
  • Back
what are the two parts of the CNS?
afferent- sensory
efferent - motor
what is the efferent system divided up into?
somatic- skeletal muscle innervation (voluntary)
autonomic- smooth muscle, cardiac muscle, and exocrine glands (reflex, homeostasis)
what are the two divisions of the autonomic nervous system?
sympathetic- fight or flight
parasympathetic- non stress time.
what neurotransmitter is primariy stored in vescicles?
Acetylcholine
what do you call neurons that release acetylcholine?
cholinergic
what do you call drugs that mimic acetylcholine?
cholinomimetics
what do you call drugs that oppose the action of acetylcholine?
cholinoceptor antagonists
what are the two types of cholinrgeic receptors?
muscarinic- nerves, heart, smooth muscle, glands, endothelium
nicotinic- skeletal muscle, neuromusclular junctions
what neurotransmitter is released at most sympathetic postganglionic neuroeffector junctions?
norepinephrine
how is norepinephrine formed?
tyrosine -> dopamine -> norepinephrine (last step is in vesicles)
How is Norepinephrine removed after released?
1. reuptake
2. diffuse into circulation.
3. transported into effector cells (MAO and COMT)
where is norepinephrine carried a step further and converted to epinephrine?
adrenal medualla
what do you call drugs that mimic norepinephrine?
adrenergic, or sympathomimetics
what do you call drugs that oppose NE?
adrenergic antagonists, or blockers
what are 2 types of cholinomimetic agents?
direct acting - bind directly
indirect - inhibits the AChE, increasing ACh.
whate are two types of direct cholinomimetic agents?
choline esters- longer duration than ACh. resist destruction from AChE.
alkaloids- well absorbed, kidney excreted, faster with acidification.
what are the 4 cholinomimetic esters?
acetylcholine- used for miosis during cataract surgery
metacholine- longer DOA, no nicotinic activity
Carbachol- more nicotinic activity, used to treat glaucoma
bethanechol- muscarinic activity, used for urine retention, has many adverse effects
what are the cholinomimetc alkaloids?
muscarine- no nicotinic agent, (mushroom poisoning)
pilocarpine- miosis, increases gatric secretion, can be to much if diarrhea and vomiting happen.
what are the two types of AChE inhobitors?
mono and bis quaternary amine alcohols
carbamates
what are the three AChE amine inhibitors?
Ambenonium- oral, used for myastenia gravis. long DOA, more ADR's
Demecarium- topical, used for glaucoma
Edrophonium- NMB reversal, a diagnostic for myastenia gravis
what are the 3 AChE carbamate inhibitors that are reversable?
Neostigmine- treats myastenia gravis, NMB reversal, post-op non obstructive urinary retention
physostigmine- antidote to muscarinic blockers, easily absorbed
Pyridostigmine- myasthenia gravis, NMB reversal, longer DOA than neo.
what are the AChE carbamate inhibitors that are irreversable?
organophosphates- cause miosis, salivation, sweating, bronchisl constriction, NVD. Treat by maintaining ventilation and atropine. (Echothiophte)
what is glaucoma? How is it treated?
increased intraocular pressurethat can dmage the optic nerve.
miotics can imporve the aqueous flow(pilocarpine, methacholine, carbacho), cholinesterase inhibitors enhace the ACh to the iris sphincters(physostigmine, demecarium, echothiophate, isoflurophate)
what is strabismus?
turning of one or both eyes from the normal position, treated with echothiophate, demecarium, isoflurophate.
what is myasthenia gravis?
impaired neuromuscular transmission at skeletal muscles.
what do cholinomimetics do to the GI and urinary tracts?
increase lower esophogus tone, urinary retention, don't use if there is a GI blockage
how are cholinomimetics used for poisonings?
good for atropine and tricyclic antidepressant poisoning. If poisoned with them can cause serious behavior disturbances.
what are cholinergic blocking agents?
compounds that selectively antagonize the muscarinic responses to ACh. reversible. Bella Donna alkaloids, atropine, and scopoline
what is Menieres disease?
recurrent vertigo, ringing in the ears, deafness, nausea and vomiting. Use Scopolamine.
What drug is used for seasickness?
scopolamine
what are cholinergic blocking agent effects on the eye?
mydriasis (pupil enlargment), cycloplegia (can't contract ciliary muscles), tear reduction.
what are the cholinergic blocking agents that are normally used in the eyes?
atropine, scopolamine, cyclopentolate, tropicainamide. Can cause acute glaucoma. Apply presure to lacrimal sac after administration
what are the effects of cholinergic bocking agents on the cardiovascular system?
little effect on blood pressure, slight tachycardia. prevents effects of direct acting muscarinic stimulants. low dose = initial bradycardia. larger dose = tachycardia. Only use atropine when hypotension or ventricular arrthymia
what are the effects of cholinergic blocking agents on the respiratory system?
bronchodilation, reduction of secretion. Scopolamine also causes amnesia. Glycopyrrolate is useful to reduce secretion in intubated patients.
What is the best cholinergic blocking agent to use for asthma?
ipratopium- decreased systemic effects when compared with atropine.
what effects do cholingeric blocking agents have on the GI tract?
slows motility, decreases salivation. used for diarrhea, irratable bowel, spasms
which cholinergic blocking agent is typically used for stomach spasm and pain?
propantheline, oxybutnin.
what cholinergic blocking agent is typically used for smooth muscle relaxation?
oxybutnin
what effect do cholinergic blocking agents have on the genitourinacry tract?
slows it down. good to treat urinary frequency, urgency, incontinence
which cholinergic blocking agents are best to treat overactive bladder
oxybutnin, and tolterodine (the latter has less adverse affects, and a longer DOA), and solifnacin, and Darifenacin (the last two have the greatest affinity for muscarinic receptors.
What effect does cholinergic blocking agents have on the sweat glands?
suppression.
what is used for cholinesterase inhibitor poisoning?
cholinergic blocking agents (atropine, need multiple doses becuase DOA is shorter than the poison's) 2-PAM and DAM regenerate active enzyme from the organophosphate cholinesterase complex.
What are the adverse effects of too much cholinergic blocking agent?
tachycardia, fever, delirium.
Under what conditions should you not use Cholinergic blocking agents?
glaucoma, ulcers, prostatic hyperplasia, urinary retention.
what is the difference between direct and indirecr acting adrenergic drugs?
indirect depend on the release of endogenous catecholamines.
which drugs displace the stored catacholamines from the adrenergic nerve endings?
amphetamines
which adrenergic drugs inhibit catacholamine reuptake?
cocaine and tricyclic antidepressents
which adrenergic drug is most selective for Alpha receptors? For Beta receptors?
Which does both?
Alpha- Epinephrine
Beta- Ioproterenol
Both- Norepinephrine
which drugs are adrenergic alpha blockers?
phentolamine and phenoxybenzamine
what are the 2 types of adrenergic beta receptors?
beta 1- in heart. (equal affinity)
beta 2- in lungs. (higer affinity for EPI)
What is receptor sequestration?
receptors made temporarily unavailable for activation by agonists
what is down-regulation?
disappearance of the receptors from the cell.
what is the major mechanism of desensitization?
phosphorylationof the receptor
what is homologous desensitization?
loss of receptors sensitivity exposed to a drug
what is heteroologous desensiitization?
loss of some cell surface receptor sensitivity that have not been activated by the drug.
What does epinephrine do?
acts on both alpha and beta receptors. Relaxes bronchospasms, reduces congestion and edema.
which drug is the best to use with anaphylaxis?
epinephrine
what are the side effects of using epinephrine?
tachycardia, hypertension, increased myocardial 02 demand, anxiety, decreased renal and splanchnic flow
what does norepinephrine do?
stimulant for alpha and beta in the heart, but not in the bronchi or peripheral system. Used in shock.
what are the adverse effects of norepinephrine?
arrythmias, palpitation, bradycardia, hypertension, chest pain, head ache
What doe isoproterenol do?
synthetic, acts on beta receptors. used to treat bronchospasm, shock, cardiac arrest
what are the adverse effects for isoproterenol?
premature ventricular contractions, hypotension, chest pain,
what does dopamine do?
precursor to NE. both beta and alpha receptors. indirect effect of releasing norepinephirine from storage sites.
how does the dose of dopamine effect the outcome?
low dose- D1 receptor, increases urine output.
med dose- B receptors cardiac stimulation
high dose- alpha receptors vasoconstriction
what are the adverse effects of dopamine?
tachycardia, vasoconstriction, hypertension, ventricular arrythmias, nausea and vomiting
What does Dobutamine do?
B1 receptor. caridac stimulation increases introphy and cardiac output. Has less arrythmia issues than all the others
what does phenylephrine do?
pure alpha agonist. decongestant for the eyes.
What does Ephedrine do?
releases the stores of catecholamines. nonselective. Nasal decongestant. Ma Huang and herbal teas have it.
what does pseudoephedrine do?
ephedrine enantiomer. OTC decongestant
What are the topical decongestants?
Xylometazoline and Oxymetazoline, may cause hypotension
What are the three characteristics of ADD and ADHD?
impulsiveness, heightened distractability, short attention span
what are the goals of ADHD medications?
improve core symtoms, associated symptoms, functional outcomes, allow the child to exert control instead of have the drug control the child
what do stimulant medications do in regards to ADHD?
dopamine and norepinephrine reuptake inhibitor. (MOA)
what is Methylphenidate
patch worn up to 9 hours a day. Has many adverse effects
What is Amphetamines?
Adderall- mix of dextroammphetamie and lisdexamphetamine. Lasts up to 12 hours. CNS stimulation can lead to abuse. Was a study aid.
Which stimulant medication is used to treat narcolepsy?
modafinil
what are SSRI's?
selective serotonin reuptake inhibitors
what are the two functions of writing prescriptions?
1- tells pharmasist specific drug, dosage, concentration, and amount issued
2- directions to patient
why prescribe generics?
flexibility for the pharmasist and savings for the patient
are generics and brand names always of equal satisfaction?
no
how many grams in an ounce? how many ml in an ounce?
30 gms, 30 mls
how many ml in a pint?
500 ml
how many ml in a teaspoon and a table sppon
5/15ml
what are legend drugs?
require a prescription
1906?
interstate regulation
1914?
Narcotic drug federal regulation
1938?
interstate regulation of drugs that haven't been proven safe
1952
prescriptions introduced
Federal warning introduced
1962
drug must demonstrate effectiveness and ingredients
1965
accounting of abuseful drugs. Refill limitations
1970
controlled substances divided into 5 classes
1983
orphan drugs
what is the law for a controlled drug?
DEA # required.
I -not prescribed.
II -IV -require prescription
III-IV can be telephoned and can have up to 5 refills
II must be written and signed in ink.
What are Schedule I drugs?
drugs w/ high potential for abuse with no accepted medical use.
Schedule II?
high potential for abuse with accepted medical use
Schedule III?
lower abuse potential
Schedule IV?
lower abuse rate than III
Schedule V?
exempt narcotics, very low
BID?
GM?
QD?
QID?
Q4h?
STAT?
TID?
twice a day
gram
as required
4 times a day
every 4 hours
immediately
3 times a day
What does the IRB do?
requires federal funds, reviews studies, protects the rights of those involved in trials
What is informed consent?
study subjects have the right to know what will happen to them if they participate
what are the phases of clinical investigation?
I - establish safety
II - establish efficacy and dose
III - verify efficacy and detect adverse effects
IV- obtain additional data following approval
Phase I?
dose level where toxicity first appears.
Phase II?
find optimal dose range, patients in test should have no medical problems other than the condition for which the new drug is being administered
phase III?
detect adverse effects, over 2 million serious adverse effect reactions a year. 100,000 deaths a year many adverse effects are not discovered until after phase 3
What does parenteral mean?
injection - given to uncooperative , good for poorly absorbed
painful
what is the difference between solutions, suspensions, emulsions, and dry powders?
suspensions - decreased water solubility
emulsions- water insoluble
dry powders- drugs unstable for long periods
Bioavailability?
fraction of unchanged drug reaching the systemic circulation following administration by any route
pharmicodynamics?
what the drug does to the body.
what do drugs do?
enhance, discontinue or regulate something the body already does
what is the pH of most drugs?
weak acids and bases.
What is the difference between hormones and xenobiotics?
xenobiotics are synthesized outside of the body
what is the difference between a toxin and a poison?
toxins are organics or biological only. Poisons can be synthetic. Both are of no benefit to the body.
do all drugs produce their effect by an interaction with a receptor?>
no
what determines the concentration of drug required for drug action?
number of receptors available and affinity for binding
what is more important? specific nature of a particular drug receptor bond, or drugs with low affinity to their receptors are more selective than drugs that have a high affinity?
the latter
what are enantiomers?
asymetric centers with same chemical makeup
What is structure activity relationships? (SAR)
activity is related to chemical structure and affinity for receptors
what is an agonist?
drugs that have a direct stimulatory effect on a receptor.
what is the difference between a full agonist and a partial agonist?
full- envokes the max response
partial- doesn't elicit full response, even when occupying all the receptors.
what is an antagonist
agents that interfere with the activities of an agonist
what effect does an antagoinst binding to a receptor site have?
none, remains inactive
What is the difference between competitive and noncompetitive antagoinsts?
competitive- is reversible, can be overcome by increasing the concentration of the agonist
noncompetitive- irreversible, increase in agonist concentration will have no effect on the receptors.
what are chemical antagonists?
drugs that antagonize by binding to and inactivating the agonist
what are the 2 main functions of a receptor-substrate interaction?
direct cemical effect on the receptor
involvement of a second messenger
what is the dose-response relationship curve?
relationship where the maximum effect is reached at a dose, or percentage of receptors occupied, lower than what would be expected on a linear graph.
what is a quantal dose/effect?
the effect will be either/or. It will either have the desired effect, or it won't.
what is the ED50 and the LD50 mean?
ED50- median effective dose, 50% of subjects demonstrate quantal effect
LD50- median lethal dose, 50% die
what is the therapeutic index?
range between the ED50 and LD50 (before the LD50)
what is a graded response?
small response to a low dose of drug, and the response increases as the dose increases. It is directly related to the number of receptors with which the drugs effectively interact
how can the efficacy of the drug be determined?
it is seen where the drug plateaus on the dose/response curve.
how can you determine the potency of a drug?
the concentration at which the drug produces 50% of the maximal effect.
Can a drug be more potent and less efficient than another?
yes, it can reach 1/2 maximum effect at a lower dose, but not reach the same efficacy level as the other drug
does greater potency or greater efficacy determine superiority?
no, each patient and circumstance is different.
what are spare receptors?
receptors still unfilled once the maximal effect has been reached by the drug.
what do spare recpetors do?
increase the sensitivityto drugs since lower concentrations of antagonist can be blocking receptors and drug responses can still occur.
what is pharmicokinetics?
absorption, distribuition, metabolism, and excretion of the drug. Compares these to the therapeutic effects of the drug.
how does drug concentration effect absorption?
rate of diffusion is directly proportional to the concentration.
how does the physical state of the drug effect absorption?
the hardness and the interactions among various ingredients plays a role.
how does the surface area effect absorption?
smaller surface area = slower absorption
how does vascularity and blood flow effect absorption?
more rapid from tissues that are more vascular and inflamed.
where are most drugs absorbed?
small intestine
what is drug distribution?
the partitioning of a drug among the numerous locations where a drug may be found in the body.
what kind of drug would be found in the total body water?
small hydrophilic compounds
what kind of drug would be found only in the extracellular water?
large hydrophilic compounds
what kind of drug would be found in blood plasma?
strongly plasma bound and charged compounds
why is the blood brain barrier important to consider before administering a drug?
it allow diffusion certain chemical to pass along into the CNS. Lipid soluble
what is the only route of administration that does not require the drug to transport from the sight of administration to the the area desired?
intravenously
how can drug binding to protein effect the drug distribution?
only the free drug that is not bound th protein is available to exert it's biological effect. If a competitor dislodges the drug from the protein, icreased effect or even toxicity results.
how are drugs effected by metabolism?
most drugs are acted on by metabolisms and turned into metabolites, which can be excreted ot turn into toxic products.
what are active metabolites?
metabolites that still have therapeutic effects
what are prodrugs?
drugs that have no therapeutic benefit until it has indergone metabolism first.
what is first pass metabolism?
after absorption, portal vein delivers drug to liver before body. Significant metabolism can take place there first.
when discussing drug metabolism, what are phase 1 reactions?
drug is made into a more polar metabolite by either masking or unmasking a functional group.
when discussing drug metabolism, what is enzyme induction?
acceleration of the metabolism leads to a decrease in drug therapeutic action.
in discussing drug metabolism, what is enzyme inhibition?
inhibition of the drug metabolism leads to an increase in drug therapeutic effect, increasing the half life.
when discussing drug metabolism, what are phase 2 reactions?
conjugating the drug or metabolite with an endogenous compound more water soluble to be eliminated by the kidney.
what are the three processes that lead to kidney excretion of drugs?
glomerular filtration rate, tubular secretion, and reabsorption.
What is Therapeutic Drug Monitoring? (TDM)
safe and effective therapeutic management of drugs in an individual patient
It is used to improve efficacy and decrease toxicity.
what can be extrapolated about drug concetrations in blood plasma?
whatever trends are seen in the blood plasma, should be similar in the target tissues as well.
what is the therapeutic range?
aid to individualize medications, it is derived from population kinetics, and it provides a guideline for safe and effective medeication use.
are the boundry concentrations of therapeutic ranges the sane for everyone?
no
what factors can cause variability in serum concentration?
difference in absorption, metabolize, and eliminate ability
disease
age
drug interactions
what is compartmental monitoring?
estimates an actual physiologic circumstance.
=dose/Vd
(amount/volume it is found)
what is the difference between one and two compartment modeling?
one- organs and tissues are grouped into one. Instant distribution is assumed, and elimination does not affect concentration.
second- closer to reality becuase it accounts for quick distribution to the plasma, and slow distribution to the tissues.
what is volume of dsitribution?
the extent into which the drug is distributed. THE Vd DOES NOT CHANGE WITH DOSE.
what is clearance?
removal of drug from plasma, and removal from the body. Doesn't say how much, just how fast.
what is the difference between first order and Zero order elimination?
1st- fraction remains constant. Amount changes.most drugs exhibit 1st order.
0- amount does not change. Fraction does.
What is the elimination rate constant?
calculated form the first order elimination. amount of drug removed from the body over a given period of time.
what is half-life?
time it takes for drug concentrationin the measured compartment to be reduced by half.
What is steady state?
when the amount eliminated is equal to the amount taken in. concentration of peak times are consistent. Concentrations of trough times are consistent.
= 5 half lives
What is a loading dose?
amount of drug required for a patient to achieve therapeutic concentration of the drug. It does not reach the steady state faster, but reaches the range fater.
what is pharmacotherapeutics?
use of drugs in the diagnosis, prevention, and treatment of a disease.
what are empircal results?
results that may occur because the prescribed drug covers a vat range of symptoms beyond the just the one that you are trying to alleve.
How is the optimal dose determined?
diagnosis, severity, stage of disease, concurrent disease and drug therapy.
what are the steps of rational prescribing?
1- make diagnosis
2- consider pathophysiology of diagnosis
3- select therapeutic objective
4- select drug
5- determin dose
6- plan for observation
7-plan for patient instruction
what is pharmacogenetics?
different drug responses due to heredity
what is genetic polymorphism?
variation in structure of a gene or an allele
what are the important genetic factors to consider?
1- metabolism rates among different ethnic groups
2- cellular resistence
3- alteration of protein synthesis (in microorganisms and cell lines)
4- transfer of genes that encode a resistence to the drug (in microorganisms and cell lines)
5- excess gene amplification that leads to excess enzyme production that inactivates drugs
how is the weight of the patient used to calculate dose?
get actual weight for obese to use for their loading dose, then use the ideal body weight for their maintenence dose.
Use actual for normal and underweight patients.
what type of drugs do most pregnant women on medication take?
nonprescription
what is teratogenesis?
malfunction of fetal organs either structurally or functionally
do most drugs cross the placenta into the baby?
yes
when is the greatest teratogenic susceptability for the fetus?
1st trimester
what kind of drugs pass into the fetus most easily and how do they do it?
lipid soluble drugs- passive diffusion, facilitated transport, and active transport.
what are the FDA pregnancy safety categories?
A- sufficent studies with no adverse effects
B- adverse in animals, but not in humans, or no animal risks and not many human studies
C- adverse effects, not many human studies, benefits outweigh the risk
D- eveidence of fetal risk, but effect still outweighs the risk
X- Risk outweighs the benefit
where do most drugs taken during pregnancy fall into?
B and C
How are drugs passed on to the infant in breatfeeding from the mother?
bidirectional diffusion. Milk proteins can bind to drug, but bind less well than the proteins in plasma. Emulsified fat in milk can cause drug concentrating
what is the milk/plasma ratio?
drug in milk/drug in plasma. are directly porportional. The lower (0.1) the better.
what is the relative infant dose and absolute infant dose
absolute- drug concentration in milk X volume of milk per day
relative- infant exposure due to maternal dose. absolute dose/maternal dose X 100
what are the methods to minimize drug exposure to infants because of maternal dose?
1- withhold or delay feeding
2- change drug route for mother
3- monitor infant adverse effects
are adults or children at a greater risk for medication errors?
children
how is the body composition different in babies compared to adults
more water(Vd) and less fat(Vd) than adults
what are the primary organs for metabolism?
LIVER, kidneys, intestine, lugs, skin
what are the two categories of metabolic processes seen in children?
phase I- decreased P450 enzyme activity in neonates, reaches normal levels around 6 months
phase II- conjugation of drug with endogenous protein to be excreted.
what are the types of conjugation reactions seen in infants?
sulfation- around week
acetylation- around a month
glucaron- around 2 months
AA conjugation- around 3 months
how is elimination rate effects in children?
lower glomerular filtration rate b/c of fewer nephrons, or fewer functional nephrons.
Tubular function is decreased b/c of secretion and reabsorption low levels
what are some good ways to mask medicines?
chill, popsicle, cold liquid, hold nose, mix with something else
how does absorption change in elderly patients?
decreased saliva, teeth, active transport, stomach acid production, GI blood flow, esophogus action.
increased gastric pH, ulcers, diverticulum in colon
how is body composition different in elderly patients?
increased fat, decreased water and plasma. Decreased albumin, increasing free fraction of drugs.
how is metabolism different for elderly patients?
decreased liver size, decreased first pass metabolism. Phase I has most change
How is excretion different for elderly patients?
less glomeruli, less GFR, less renal blood flow
what is physiologic reserve in elderly pateints?
functional reserve capacity and the ability to maintain homeostasis
what is the functional reserve capacity in elderly?
ability to perform activities under abnormal sistuations
what is homeostasis in the elderly?
ability to maintain a constant enviroment while adjusting to change.
what patients get the majority of prescriptions in the US?
elderly (50%)
what is polypharmacy?
use of several medications by one patient
what are 8 risky drugs to use with the elderly and why?
1- amitriptyline and doxepin- used for neuropathic pain- it dires them out
2- Diazepam and chlordiazepoxide- long half lives, cause sedation hangover anf can injure them selves
3- propoxyphene- causes CNS disfunction due to a toxic metabolite
4- Dipyridamole- used to decrease platelet aggregation, causes standing hypotension
5- Meperidine- creates a toxic metabolite
6- cyclobenzaprine and carisoprodol- has poor efficacy, is a skeletal muscle relaxant
7- COX2 inhibitors- GI ulcers
8- Fluoxetine- long half life
what should be done to patients with lver disease that have a prescription for a drug that is either highly or intermediately extracted?
reduced because the liver diseae will porlong the half life otherwise.
what is azotemia?
elevated blood urine nitrogen (BUN) and creatine in urine (can cause vomiting)
what is uremia?
metabolic abnormalities and symptoms resulting from the accumulation of uremic toxins
what is creatinine clearance?
men= (140-age)(weight)/(72 x serum creatinine)
women= men x .85
how does the loading dose change in patients with organ dysfunction?
stays the same. The subsequent doses change to account for the degree of organ dysfunction. You can change either the dose or the interval
what is hyporeactivity and hypereactivity?
hypo- needs bigger dose
hyper- needs smaller dose
what is idiosyncracy?
unpredicted reaction indepeendent of dose, not an allergic reaction
What is tachyphylaxis?
tolerance that quickly develops over only a few doses
what are 4 types of noncompliance?
failure to obtain or access medicine
2- failure to take as prescribed
3- failure to start and stop as directed
4- medication sharing