• 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

1/148

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;

148 Cards in this Set

  • Front
  • Back

What is the Federal Food, Drug, and Cosmetic Act?

Proposed in 1933 to protect consumers




1937- Public outcry due to >100 deaths from improperly prepared sulfanilamide




Passed by Congress in 1938




Created the FDA (Food and Drug Administration)




Establishes classes of drugs




Prescription vs non-prescription

What is an IND?

Investigational New Drug Application




Based on preliminary testing completed on animals




Basis of proposing human research

The IND Appraisal by the FDA is done in conjunction with what?

A local institutional Review Board (IRB) Appraisal

What does an IRB examine?

The specifics of a clinical trial, consent issues, patient protection

What is the FDA drug approval process?

Drug developed and animals tested > IND application > Clinical trials (phase I, II, and III)

What is the impact of the FDA?

Frances Oldham Kelsey (FDA scientist) kept thalidomide off the U.S. market

What happens in Phase I trials?

Involves 20-80 healthy subjects




Focus on identifying frequent side effects




Drug metabolization and excretion is studied

What happens in phase II trials?

24 to 300 subjects with the disease




Focus on effectiveness of new compound




Comparison of drug vs placebo




Gathers data on safety and side effects

What happens in phase III trials?

FDA and drug sponsor agree on structure of phase III trials




100 to 3,000 subjects




Different dosages and drug interactions studied




Safety and effectiveness data continues to be collected and evaluated

What is an NDA?

New drug application




Submitted with data from completed studies




Takes 6 to 10 months




Package Insert (PI) submitted for approval and may require revision

What happens after clinical trials?

Review meeting > NDA > Application reviewed > Drug labeling > Facility inspection > Drug approval > Post-marketing (FDA's Post-approval risk assessment systems)

What is the Controlled Substances Act (CSA)?

Passed by Congress 1971




"Scheduled" drugs- DEA and DOJ jurisdiction




Categorizes drugs into schedules or controlled classes (CI, CII, CIII, CIV, CV)




Use and distribution is controlled base on potential for abuse, addiction, and widely accepted medical use

What are Schedule I drugs?

High potential of abuse or risk




No safe or accepted medical uses




Heroin, LSD, PCP, crack cocaine, marijuana, mescaline

What are schedule II drugs?

High risk of abuse




Safe and acceptable medical uses




Severe potential for psychological and physical dependence




Hydrocodone, morphine, cocaine, oxycodone, methyphenidate, dextroamphetamine

What are schedules III-V drugs?

Low risk of abuse




Safe and acceptable medical uses




Contain smaller amounts of narcotics, anti-anxiety, tranquilizers, sedatives, stimulants

How is TX PA prescriptive authority delegated?

Delegated by supervising physician to the PA through formal agreement

Can controlled substances be prescribed by Pas?

Yes, with specific limitations

What must Pas who prescribe controlled substances have?

DPS registration number




DEA registration number

Can PAs prescribe schedule II drugs?

Only under physician delegation in the following circumstances:




In a hospital to a patient who has been admitted to the for an intended stay of >24 hours




To a patient who is receiving services in the ED of a hospital




Part of a plan of care for treatment of a patient with a terminal illness or in hospice

How often is physician consultation required when prescribing controlled substances?

Consult for refills after initial 90 day supply




When prescribing controlled substances for children <2

What is a pregnancy category A drug?

Studies in pregnant women indicate no risk to fetus

Wha is a pregnancy category B drug?

Animal studies indicate no risk to fetus

What is a pregnancy category C drug?

Animal studies report adverse affects on fetus




Potential benefits may indicate use despite risks





What is a pregnancy category D drug?

Positive human fetal risk




Benefit may outweigh risk in some select cases

What is a pregnancy category X drug?

Fetal abnormalities reported




Positive human fetal risk




Risks outweigh benefits




Drug should not be used during pregnancy

What pregnancy drug categories can be prescribed during pregnancy?

Category A and B

What are the written prescription components?

Clinician Identification




Patient identifiers




Inscript- Drug name and strength




Subscript- Dosage from and number of units to dispense




Signa/Sig- Words on bottle (directions for patients)




Number of refills




Date of Rx written




Prescriber's DEA number




Generic vs Trade

What is pharmacokinetics?

What the body does to the drug

What is pharmacodynamics?

What the drug does to the body

What are the properties of pharmacokinetics

Absorption




Distribution




Metabolism




Elimination

What is absorption?

Entry of the drug to the plasma via the administration site

What is distribution?

Drug may reversibly leave the blood stream and distribute into interstitial and intracellular fluids

What is metabolism?

Drug is biotransformed by metabolism in the liver or other tissue

What is elimination?

Drugs and metabolites are excreted via urine, bile, or feces

How are routes of administration determined?

Drug properties (water or lipid soluble, ionization)




Therapeutic objectives (rapid onset needed, long term treatment, restriction of drug to a local site)

What is the enteral route?

By mouth

What is an enteric coated drug?

Coated to protect from stomach acid




Coating dissolves in small intestine

What is an extended release drug?

Coatings or ingredients delay drug release (slow absorption/prolonged duration)




Improves patient compliance




Less peaks and troughs

What are the sublingual/buccal routes?

Under tongue/in cheek




Drug diffuses into capillary network and goes directly to systemic circulation




Rapidly absorbed




Avoids harsh gastric pH and 1st pass metabolism

What are some advantages of the enteral route?

Safe




Convenient




Economical

What are some disadvantages of the enteral route?

Limited absorption




Affected by food




Patient compliance concerns




1st pass metabolism effects

What is the parenteral route?

Drug goes directly into systemic circulation via blood stream




Highest degree of bioavailability




Irreversible




May cause pain, irritation, tissue damage, and infection

When is the parenteral route ideal?

Drugs are unstable in the GI tract




Drugs are poorly absorbed from the GI tract




Patient not able to take oral medications




Rapid onset of effect is needed

What is the intravenous route?

Directly into vein




Rapid onset




Maximum control over amount of drug delivered




Bolus dosing vs Continuous infusion (Bolus is for quick effect and continuous infusion is for maintenance effect)

What is the intramuscular route?

Directly into the muscle




Prepared in aqueous solutions (rapid absorption) or in a specialized depot injection (slow absorption)

What is the subcutaneous route?

Injection into the subcutaneous tissue




Minimizes risk of hemolysis or thrombosis




Can cause pain and tissue necrosis

What are the advantages of the parenteral route?

Immediate effects




Large volumes can be easily administered




Helpful in emergencies




Easy to titrate to effect




Used for high molecular weight proteins and peptide drugs

What are the disadvantage of the parenteral route?

Unsuitable for oily or poorly absorbed substances




Bolus injections can cause adverse effects




Need for prolonged injection times




Aseptic technique required

What are the oral and nasal inhalation routes?

Rapid administration through mucous membranes




Beneficial in patients with respiratory disorders (drug can go directly to lungs)

What are the intrathecal/intraventricular routes?

Introduces drugs directly to the cerebrospinal fluid

What is the topical route?

Applied to local area of skin




Local effects

What is the transdermal route?

Skin patches often used for sustained drug delivery




Thickness of skin and lipid solubility influence absorption

What is the rectal route?

Drug administered into rectum




Useful when patients are unconscious or vomiting




Absorption is often erratic and incomplete




Bypasses the liver and GI

What absorbs better, solutions, suspensions, or tablets?

Solutions absorb better than suspensions or tablets

What factors influence absorption?

Factors related to the drug-




Degree of ionization (highly ionized drugs are poorly absorbed)




Degree of solubility (high lipid/water partition coefficient increases absorption)

What is the amount of drug that reaches systemic circulation?

Bioavailability

What is bioavailability highly dependent on?

Apsorption

How are most drugs absorbed into systemic circulation?

Passive diffusion

What are other mechanisms that drugs can be absorbed by?

Active transport




Facilitated diffusion

What is passive diffusion?

Drugs move from high to low concentrations across a membrane




No carrier




Not saturable




Low structural specificity




Water soluble drugs diffuse through pores




Lipid soluble drugs diffuse across membranes

What is facilitated diffusion?

Transmembrane carrier proteins facilitate the passage of large molecules




Conformational changes allow passage of drugs into cells




Move from high to low concentration




No energy required




Saturable




Competing compounds can inhibit the process

What is active transport?

Specific carrier proteins




Carrier proteins transport specific drugs that closely resemble the naturally occurring metabolites across membranes




Energy dependent (ATP)




Drugs can go from low to high concentration




Saturable




Can be competitively inhibited

How are exceptionally large molecules transported in the body?

Endocytosis (engulfment of drug by cell membrane for transport into the cell)




Exocytosis (secretes substances out of the cell)

Are most drugs weak or strong acids and bases?

Weak acids and bases

What does a weak acid look like?

HA <=> H+ + A-




Weak acids release a proton (H+) causing a charged anion (A-) to form

What does a weak base look like?

BH+ <=> B + H+




Weak bases release a proton (H+) producing an uncharged base (B)

What type of drugs pass through membranes more readily than cations and anions?

Uncharged drugs (HA and B)

What is pKa?

The measure of the strength of interaction of a compound with a proton

What type of drug has a lower pKa?

More acidic drugs

What type of drug has a lower pKa?

More basic drugs

What happens when pH = pKa?

HA = A-




BH+ = B

When the pH is less than pKa, what forms predominate?

HA and BH+

When the pH is greater than pKa, what forms predominate?

A- and B

How does blood flow influence absorption?

More blood flow = greater absorption




Intestines receive more blood flow than stomach

How does total surface area influence absorption?

Greater surface area = greater absorption




Intestines have 1000x more brush border containing microvilli

How does contact time influence absorption?

More contact time = greater absorption




If a drug moves too quickly (diarrhea) it won't be absorbed as well




Delaying passage of drugs from the stomach to the intestines will slow absorption (take with food > slow gastric emptying)

How does the expression of P-glycoprotein influence absorption?

Increased P-glycoprotein = less absorption




P-glycoprotein pumps drugs out of cells




P-glycoprotein is found in the kidneys, placenta, intestines, and brain

How can you determine bioavailability?

Compare plasma levels after various routes of administration




IV = 100%




Oral = variable




Calculate the AUC (area under the curve) by plotting plasma concentrations over time

What does oral bioavailability depend on?

The amount absorbed and the amount metabolized before reaching systemic circulation (first pass metabolism)

What is first pass metabolism?

Metabolism of orally administered drug in the liver before it reaches systemic circulation




Significant first pass metabolism limits drug's bioavailability




Consider other routes of administration for drugs with high first pass metabolism

How does solubility influence bioavailability?

Extremely hydrophilic or lipophilic drugs are poorly absorbed




Ideal- largely lipophilic with some solubility in aqueous solution

How does chemical instability influence bioavailability?

Certain drugs can unstable or destroyed in the GI tract

How does the nature of the formulation influence bioavailability?

Drug absorption may be altered by factors unrelated to the chemistry of the drug




Ex. Particle size, salt form, crystal polymorphism, enteric coating and excipients

What is bioequivalence?

Two drugs that have comparable bioavailability and similar times to achieve peak blood concentrations




Brand vs. generic

What is therapeutic equivalence?

Pharmaceutically equivalent




Same dosage form, active ingredient and route




Similar clinical and safety profiles

Does bioequivalence = therapeutic equivalence?

No

What is distribution of a drug from systemic circulation to tissues dependent on?

Lipid solubility




Ionization




Molecular size




Binding to plasma proteins




Rate of blood flow

What does it mean if a drug has an uncommon distribution parameter?

Ex. VD, protein binding, storage in fat depots




Have to be dosed carefully to avoid toxicity while ensuring therapeutic efficacy

How are drugs present in the blood?

Free form- Active, diffusible, available for metabolism and excretion




Bound form- Inert, not available for metabolism and excretion (acts as a reservoir for drug)

Is binding of a drug to albumin reversible?

Yes

What is the significance of a drug binding to albumin?

Drugs may have affinity for protein binding sites




Drugs can compete and cause drug interactions




Drugs highly bound to plasma proteins are in general expected to persist in the body longer than those less bound and are expected to have lower therapeutic activity

Drug molecules in the blood are in what two forms?

Bound to plasma proteins (albumin)




Unbound or free

What type of drug can distribute into tissues and exert its action?

Free drug

Affinity of protein binding site for a drug determines what?

Bound/unbound concentrations

Saturation of binding sites may result in what?

A large increase in unbound drug concentration




Toxicity may be a problem

Competitive protein binding can cause what?

Significant drug interaction




In general, only drugs that are highly protein bound (90%) are involved in drug interactions due to competitive binding

What can hypoalbuminemia lead to?

Increases in free drug

What is volume of distribution (Vd)?

The hypothetical volume of blood and body tissue into which the drug distributes




No physiologic or physical basis




Amount of drug in the body/Concentration at time zero

What can Vd be altered by?

Tissue binding (Increase tissue affinity = increased Vd)




Tissue perfusion (Decrease perfusion = decreased Vd)




Plasma protein binding (Increased protein binding = decreased Vd)

What is the primary location of metabolism?


Hepatic (phase I and II)

What are other locations (extrahepatic) of metabolism?

Kidneys




Lungs




Blood

What happens in Hepatic Phase I metabolism?

Oxidation, reduction, and/or hydrolysis




Following Phase I, the drug may be activated, unchanged, or, most often, inactivated

What happens in Hepatic Phase II metabolism?

Results in conjugation products




Conjugated drug is usually inactive

What is cytochrome P450?

Super-family of cytochrome P450 enzymes has a crucial role in the metabolism of drugs




Almost every drugs is processed by some of these enzymes, which causes a reduction in bioavailability




Responsible for drug interactions

What impact can genetic variability have on a person's reaction to a drug?

May alter drug efficacy and increase risk of adverse events




Ex. 2D6 mutations result in low capacity to metabolize substrates which causes little analgesic effect with opioids like codeine




Ex. Poor metabolizers of 2C19 have a higher risk of cardiovascular events when taking the prodrug clopidogrel

What are the type of cytochrome P450 drug interactions?

Inhibition




Induction

What results from CYP450 inhibition?

Leads to decrease rates of metabolism of other drugs metabolized by the same enzyme, resulting in higher drug levels and increased potential for toxicity




Usually reversible




Irreversible inhibition can occur, requiring new CYP450 enzyme to be synthesized




Tends to occur quickly with maximal effect occurring when highest concentration of the inhibitor are reached

What results from CYP450 induction?

Increased clearance of concomitant medications metabolized by the same enzyme




The body responds by increasing the production of specific enzymes of the CYP450 system




Increased enzyme production can lead to increased metabolism and decreased drug concentrations

What factors affect drug metabolism?

Enzyme induction increases drug clearance




Aging




Diseases (hyperthyroidism)




Drugs




Conditions (smoking, alcoholism)




Higher doses of drugs may be required or other options for drug therapy may be needed

What are the effects of aging on drug metabolism?

Decreased hepatic blood flow is often associated with decreased first pass effect and reduced phase I metabolism




Phase II reactions are well preserved with age and severe liver disease

What is the Beers criteria for the elderly?

Clinical tool developed to assist healthcare providers in improving medication safety in older adults




Purpose is to inform clinical decision-making concerning the prescribing of medications for older adults in order to improve safety and quality of care

What is the principle of elimination?

Dosing of drugs needs to be adjusted according to the elimination characteristics of the patient (ex. renal impairment) in order to avoid toxicity from or metabolite accumulation

What is the main organ of drug excretion?

Kidneys

How are drugs eliminated from the kidneys?

Passive glomerular filtration




Active proximal tubular secretion




Passive distal tubular reabsorption

What occurs during passive glomerular filtration?

Free/unbound drug flows through capillary slits

What happens during active proximal tubular secretion?

Transport systems for acids and bases




Low specificity (can see competition between drugs)

What happens during passive distal tubular reabsorption?

Solute and water are removed from the tubular fluid and transported into the blood

What are other routes of elimination?

GI tract (salivary glands, stomach, large intestines, liver)




Sweat




Lungs




Milk

What is linear (first order) elimination?

Amount of drug eliminated per unit of time is directly proportional to the amount in the body




Non-saturable elimination




Most drugs

What is non linear (zero order) elimination?

Amount of drug eliminated per unit of time is constant, regardless of the amount in the body




Saturable elimination




Few drugs




Small changes in dose lead to large changes in concentration




Never double the dose

What is half-life (t1/2)?

Time required for the amount of drug (or concentration) in the body to decrease by 50%

What are the two types of half-life?

Distribution t1/2- a (alpha)




Elimination t1/2- B (beta)

What does elimination t1/2 determine?

Duration of pharmacologic effect




Optimum dosage regimen




Time to reach steady state

What is steady state (Css)

Time at which the rates of drug administration and drug elimination are equivalent




Concentration of drug is constant




Attained after 3-5 t1/2 at a constant dosing rate




Ideal time for serum drug concentration measurements

What is designing and optimizing drug regimens?

Process of determining the patient plasma concentrations to optimize drug therapy




Maximize therapeutic benefits




Minimize toxic effects

What is assumed when designing and optimizing drug regimens?

Serum drug concentration reflects concentration at the receptor site




Intensity and duration of pharmacodynamic effect is correlated with the receptor site drug concentration

What is a maintenance dose?

Keeps concentrations in a therapeutic response window to minimize toxicity and side effects




Caution with drugs that have narrow therapeutic index

What is a loading dose?

Rapidly obtains desired plasma levels




Often seen with antibiotics and antiarrythmics




Increased risk of toxicity




Usually followed by maintenance dose

What are serum drug levels influenced by?

Timing of sample




Correct dosing regimen and dose




Patient's clinical state




Drug's pharmacokinetics




Renal or hepatic function




Analytical methodology used

What does pharmacodynamics focus on?

The mechanism, intensity, peak, and duration of a drug's physiological actions

What is a drug-receptor complex?

Drug binds to the receptor and elicits an effect

What are the two receptor states?

Inactive and active

What are the major receptor families?

Ligand gated ion channels (cholinergic nicotinic receptors)




G-protein coupled receptors (a & B receptors)




Enzyme liked receptors (insulin receptors)




Intracellular receptors (steroid receptors)

What occurs during transduction?

Receptors transduce their recognition of a bound agonist by initiating a series of reactions that ultimately result in a specific intracellular response

What is signal amplification?

G protein and enzyme linked receptors can amplify intensity and duration

What is desensitization and down regulation of receptors?

Protective




Repeated agonist exposure can lead to tachyphylaxis




Chronic use of antagonists can lead to up-regulation of receptors

What are agonists?

Affinity- the ability of the agonist to bind to the receptor




Efficacy- the ability to cause a response via the receptor interaction

What are antagonists?

Have affinity but no efficacy

What are full agonists?

Activate receptors to produce an effect similar to a physiologic signal molecule




Maximal efficacy

What is an inverse agonist?

Activates receptors to produce an effect in the opposite direction of the agonist

What is a partial agonist?

Activates receptors to produce sub maximal effects but antagonizes a full agonist

What is an antagonist?

Prevents the action of an agonist on a receptor




Does not have an affect of its own

What are competitive antagonists?

Compete with agonist for the receptor




Reversible (can be overcome by increasing the agonist concentration)




Displaces the agonist dose response to the right




Reduces the apparent affinity of the agonist

Wha are non competitive antagonists?

Drug binds to the receptor and stays bound




Irreversible




As more and more receptors are bound the agonist drug becomes incapable of eliciting a maximal effect

What is efficacy?

Maximum intensity of response to a drug

What is ED50 (effective dose-50%)?

The dose of a drug that gives rise to the designated response in 50% of the subjects




Easier to measure than maximum effect and is used to determine efficacy

What is potency?

A comparison of the ED50 of two or more drugs that have parallel log dose-response curves




The drug that reaches the ED50 at the lower dose is the more potent




Potency is not important if you can increase the dose of the less potent drug without causing side effects

What is therapeutic index?

The ratio of the dose that leads to toxic effects in 50% of cases, to that that leads to therapeutic effect in 50% of the cases




Safer-Large therapeutic index




More toxic- Narrow therapeutic index