Shared Flashcard Set

Details

PDA EXAM II
Flashcards
117
Health Care
Professional
03/08/2008

Additional Health Care Flashcards

 


 

Cards

Term
What is a Xenobiotic?
Definition
A drug or chemical that is foreign to the body. It can't be something that is made in the body such as insulin.
Term
Explain Additive Drug Effects
Definition
It is the sum of the effect of 2 drugs w/ similar pharmacological activity.
Term
Explain Potentiation
Definition
This occurs when the effect of a combo of 2 drugs is greater than the sum of their individual effects.
Term
What is an Idiosyncracy
Definition
An unusual side effect such as a barbituate causing pain or excitement.
Term
Explain the Therapeutic Index
Definition

TI = LD50/ED50

LD50 - lethal dose that would kill 50% of people.

ED50 - effective dose that would cure 50% people.

The larger the TI, the safer the drug.

So, for a larger TI, the LD50 must be larger, too. If an LD50 is 100mg, this doesn't mean that you're goint to take 100 mg. It means that you can take up to 100 mg before it's lethal. So, you want the LD50 to be big and the ED50to be small.

Term

What is Tolerance?

 

What are the 2 types of tolerance? 

 

T or F: Tolerance requires chronic use of a drug over a period of time.

Definition

Tolerance occurs when a larger dose is needed to produce the same biologic effect.

 

Drug disposition tolerance & pharmacodynamic tolerance.

 

True

Term

What is drug disposition tolerance?

 

What is pharmacodynamic tolerance?

Definition

Drug disposition tolerance occurs when drugs induce more enzymes. Enzymes metabolize the drug faster & the drug is excreted faster. As a result, a larger dose is required to keep up w/ all of the enzymes.

 

Pharmacodynamic tolerance is an adaption that occurs in the CNS. FYI: It is possibly due to down-regulation of receptors to reduce sensitivity to large amounts of a drug. The body is trying to tell you that it has had enough.

Term
Tachyphylaxis
Definition
This occurs when it only takes one or two doses to develp tolerance.
Term

What is antagonism?

 

What are the three types of antagonism?

Definition

Antagonism is where one drug (the antagonist) depresses or inhibits the action of another drug (the agonist).

 

 1) receptor (aka pharmacological) 2) physiologic 3) chemical

Term

What is receptor antagonism?

Definition
It's where the antagonist is competing with the agonist for the receptor site. The antagonist can also bind to the allosteric site, which can alter the shape of the active site so the agonist can't bind.
Term

What is chemical antagonism?

What is an example?

 

What is physiological antagonism?

Definition

Chemical antagonism is where one drug (the antagonist) reacts w/ another drug (the agonist) and changes its chemical structure so that it cannot bind to a receptor.

 

An example is when protamine sulfate (a weak base) binds to warfarin (a weak acid) to prevent it from binding to its receptor.

 

It's a functional antagonist that's actually an agonist, b/c it produces an effect (through a different receptor) opposite to that of the original agonist.

Term
What is pharmacodynamics?
Definition

Factors involved in absorption, distribution, & elimination of drug in the body.

Term

What are the 5 major factors involved in drug absorption?

Definition

Think ACCDS (ACC DUKE SUCKS)

1)Absorbing surface area - the greater the surface area, the greater the absorption.

2) Concentration - Many drugs are absorbed via passive diffusion. Therefore, the greater the amount of drug, the higher the [gradient] and more drug is absorbed.

3) Circulation to the site - The greater the vasculature, the more the absorption.

4) Dissolution rate - especially solid dosage forms

5) Solubility - especially solid dosage forms

 

 

Term
What are the advantages and disadvantages of the oral route of administration?
Definition

Advantages:

1) its the easiest, safest, & most convenient ROA.

2) Since absorption takes 15-30 minutes, it can be removed via charcoal or vomiting if the wrong drug is given or if negative side effects occr.

Disadvantages:

1) Many drugs are poorly absorbed, some as low as 20-30%.

2) Many drugs, such as protein drugs (eg, insulin), destroy the GI.

3)First pass metabolism - blood first goes directly through the liver. Some drugs are metabolized before they ever reach their destination.

 

Term

What are two ways that drugs are administered through the oral mucosa?

 

Name the advantage & disadvantage of this ROA.

 

Definition

Sublingual and buccal (in the cheek)

 

Advantage:

Avoids first pass metabolism

 Disadvantage:

There is very little surface area for absorption.

 

Term

What type of ROA includes retention enemas?

 

What are retention enemas?

 

Besides retention enemas, what is another dosage form included in this type of ROA? 

 

What are advantages and disadvantages of this type of ROA?

Definition

Rectal

 

Fluids injected rectally to treat constipation. The fact that it is retentive means it is supposed to stay in the bowel long enough to soften the stool

 

Suppository

 

Advantages:

1) Good for pediatrics when swallowing an oral pill is not feasible.

2) No vomiting

3)Avoidance of portal circulation by 50%

Disadvantages:

1)Absorption is incomplete due to feces.

Term

T or F:

IV is the most common parenteral route.

Definition
False, its IM.
Term

T or F:

IM has good absorption

Why?

Definition
True, because it is surrounded by blood vessels.
Term

Which type of parenteral administration is injected directly into the spinal fluid?

 

Give an example of when this method is often used

Definition

Intrathecal

 

during labor

Term

Which is easier to do: intravenous or intrathecal?

 

What type of drugs are normally administered intrathecally?

Definition

Intravenous

 

Toxic drugs

 

 

Term
What is the pathway of  entero-hepatic recirculation? What does this do for the half life?
Definition

Blood - liver - bile - intestinte - blood

It increases the half life.

Term

T or F:

Rapid absorption occurs when a drug is given nasally due to a large surface area.

Why?

 

T or F:

Drugs that are taken nasally can cross the BBB.

Definition

False

Rapid absorption does occur, but the surface area is actually small.

 

True

Term

T or F:

The absorptive surface area of the large intestine is the size of a tennis court.

Definition
False, the absorptive surface area of the small intestine is the size of a tennis court.
Term
What three features does the small intestine have that increase its absorption potential.
Definition

1) mucosal folds

2) villi

3) microvilli

Term

T or F:

Drugs that are poorly ionized display good absorption.

Definition
True
Term

T or F:

Absorption most often occurs via passive diffusion.

Definition
True
Term

Which will have more favorable absorption in the small intestine: weak acids or weak bases?

Definition
Weak bases (pg 36 Pento)
Term

What is passive filtration? 

 

T or F:

The brain and placental capillaries are the only tissues that use passive filtration instead of passive diffusion?

Definition

It's where drugs that are small, water-soluble ions move through small membrane channels or small para-cellular spaces.

 

False, it's actually the peripheral capillaries that use passive filtration (they also use passive diffusion). The brain and placental capillaries use passive diffusion only.

Term
If a weak acid drug is in a basic environment, will it absorb? Why or why not?
Definition
It will not absorb, b/c it will ionize in the basic environment. Ionized molecules have great difficulty being absorbed through cell's lipid membranes.
Term

T or F:

Facilitated diffusion moves with a concentration gradient.

 

What is facilitated diffusion? Is it selective? Is it saturable?

 

Definition

True

 

It's where a drug uses a carrier

(eg, transport protein) to get across the membrane. Carrier types include antiporters, symporters, and uniporters. It is selective and saturable.

Term

How is active transport similar to facilitated diffusion?

Definition

They both require selective and saturable carriers.

Term

Multi-Drug efflux is a type of               transport in which a selective/non-selective transport protein, such as a                    transporter, moves drugs and other                in/out of the cell.

 

T or F:

Multi-drug efflux functions in cancer cells and pathogens to produce drug resistance.

Definition

Multi-Drug efflux is a type of active transport in which a non-selective transport protein, such as a MDR (multi-drug resistance) transporter, moves drugs and other toxins out of the cell

 

True

Term
What is the mechanism of transport for macromolecules?
Definition
Transcytosis (vesicular transport). It involves endocytosis and exocytosis.
Term

Choose one: Plasma proteins can increase/decrease a drugs half life by binding to it. Why?

 

Which protein binds to weak acids? Which protein binds to weak bases?

 

What type of bonds are involved in protein binding?

Definition

It increases its half life, b/c it makes it more difficult for the drug to cross cell membranes and be metabolized.

 

Serum albumin - a1-glycoprotein

 

Weak bonds such as H-bonds, VDW's, etc. - not covalent or ionic bonds.

 

 

Term
What type of drugs will saturate fatty tissues?
Definition
Lipophilic (non-ionized) drugs.
Term
Name three major types of drug storage.
Definition

1)Protein binding

2)Adipose tissue

3)Bones and/or teeth

Term
What are 6 major routes of excretion?
Definition

Martin Luther King Healed Forlorn Spirits 

1) Milk 

2) Lungs

3) Kidneys

4) Hair and skin

5) Feces

6) Sweat, saliva, tears

Term

What is the primary route of excretion?

Definition
The kidney
Term

T or F:

Drugs that are non-ionized are readily excreted.

Definition

False. As a general rule, they are usually reabsorbed so they can be metabolized to a more polar metabolite.

Term
What are the two mechanisms for excretion via the kidney?
Definition
Glomerular filtration and tubular secretion
Term

T or F:

All molecules of less than 80k MW are filtered by the glomerulus.

 

Definition

False, all molecules of less than 70k MW are filtered by the glomerulus.

 

Term
How can drug interactions affect drug absorption?
Definition

pH changes - drug A may increase pH while drug B requires a lower pH

altered absorption - drug A may extend gastric emptying time. Then, drug B ends up in the stomach for an extended period of time. Since drug B is slow in getting to the small intestine, this delays absorption.

altered peristalsis - drug A may speed up motility, causing drug B to go through the small intestine faster. The faster it moves through, the less it is absorbed.

Term
What are the 4 major mechanisms of drug interactions?
Definition

Absorption

protein binding

Metabolism

Excretion

Term
How is protein binding involved with drug interactions?
Definition

Drugs may compete for a protein. Drug displacement is where drug A kicks drug B off of a protein and drug B is absorbed (maybe faster than it should have been)

Term
How can drug interactions affect drug metabolism?
Definition

nonspecific enzyme induction - drug A may induce more of an enzyme that will degrade drug B.

Metabolic competition - self-explanatory

Term
How can drug interactions effect drug excretion?
Definition

1) pH changes - if drug A lowered the pH, a weak base would ionize and it would be excreted faster.

2) Impairment of renal secretion.

Term
Identify levels of measurement associated w/ defining a "mechanism of action"
Definition

The level of measurement that we use depends on the amount of info that is available. 

An anesthetic will be used to describe the levels of measurement.

Symptomatic - If we know very little, the action is described on a symptomatic level. Fe, the pt feels no pain.

Physiological - If we know a little more, we can describe the effects on a physiological level. Fe, we may know that nerve impulses are not being conducted.

Cellular - If we know more, we may be able to describe actions on a cellular level. Fe, we may be able to tell that a membrane potential is stabilized.

Molecular - The most specific level measurement. Fe, there may be a decreased capacity of Na channels.

Term
What is the difference b/w specific and non-specific drugs? Give examples.
Definition

Specific drugs are aimed directly at a disease. Non-specific drugs ameliorate symptoms. They are used for syndromes in which no clear cause is known.

Examples of specific drugs are most chemotherapeutic drugs. Examples of non-specific drugs are APAP and hypertension drugs.

Term

T or F:

Chemotherapeutic agents have to be lethal to an invader to be effective. 

 

 

 

Definition

False, chemotherapeutic agents can be suppressive and still be effective.

Term

T or F: 

Chemotherapeutic agents are always selective for organs

 

 

What is the hallmark of chemotherapy?

Definition

False, Abx are often selective for invaders.

 

 

Selective toxicity b/w host and invader

Term

T or F:

Pharmacotherapeutic agents are always reversible.

 

Definition

False. It's rare, but they are sometimes irreversible. When they are , they are associated w/ toxicity.

 

Term

T or F:

Anti-growth cancer drugs are very effective for slow growing tumors.

Definition

False

Anti-growth cancer drugs are NOT very effective for slow growing tumors

Term

T or F:

Pharmacotherapeutic agents will increase or decrease activity of the organ function when producing its therapeutic effect.

Definition
True
Term

T or F:

Any Abx that doesn't inhibit cell wall synthesis will only suppress bacterial growth.

Definition
True, Abx that only suppress bacterial growth must have assistance from the immune system to destroy invaders.
Term
List 3 examples of how drugs act as pharmacotherapeutic agents.
Definition

1) They can replace naturally occurring substances in your body that are either gone or not working sufficiently. These drugs would be mostly hormones, but some neuromediators.

 2)There are drugs that change regulation of body functions. Examples would include the treatment of symptoms, antihypertensives,diuretics, antivonvulsants, etc.

3)Some pharmacotherapeutic drugs alter mood.

 

Term

Methotrexate and Trimethoprim

are inhibitors of                 .

Definition

folic acid reductase

Term
T or F:
Chemotherapeutic agents never target organs, only invaders.
Definition
False, inhibition of growth rate is the primary target for cancer drugs. Therefore, cancer drugs are usually specific for organs that continually reproduce cells (eg, hair, skin, mouth, GI, blood, and immune-cell forming processes).
Term
Darvon & Novrad are an example of                .
Definition
Enantiomers
Term

T or F: 

Receptors for pharmacotherapeutic drugs are often binding sites for a variety of endogenous, normally occurring, compounds including hormones and neurotransmitters

Definition
True
Term
Whether or not a drug binds to a receptor depends on structural complementation, or orientation in space, or whether it's cis or trans. Explain.
Definition

Structural complementation - Receptors are very specific. Fe, vasopressin and Qytocin are both 8 aa proteins that vary by 2 aa's. They act w/ different receptors.

Orientation in space - (stereoselectivity) chiral isomers are mirror images and  have identical properties, but differ in the receptors they bind to. Quite often, one isomer is active and the other is inactive.

cis-trans isomers - they differ in their properties and in their ability to bind to receptors.

 

Term

T or F:

Antagonists must be identical to agonists.

Definition

False

Most often, antagonists hardly resemble the endogenous molecule they are trying to inhibit.

Term

T or F:

With pharmacotherapeutic agents, toxicity is often an extension of therapeutic effect.

Definition
True
Term
Why is the FDA concerned about racemic mixtures?
Definition
B/c one isomer may not be active enough or it may be overly active resulting in toxicity. An example is labetalol.
Term

T or F:

Unlike geometric isomers, stereoisomers are easily isolated as pure substances, b/c they are racemic.

Definition

False

Geometric isomers are easily isolated as pure substances. Stereoisomers are not.

Term
What is sarin?
Definition
It's a poisonous nerve gas that binds covalently (irreversibly) to acetylcholinesterase. Consequently, your muscles are unable to relax.
Term

T or F:

An allosteric effect can either increase or decrease the binding of a ligand.

Definition
True
Term

What is positive cooperativity?

 

 

What is negative cooperativity?

Definition

Cooperativity involves identical ligands.

Positive cooperativity is where one ligands binds to a receptor and causes another receptor on the same molecule to fit with an identical ligand. An example involves Hb's affinity for oxygen.

 

Negative cooperativity is where one of the ligands binds to the allosteric site and consequently changes the shape of the active site so that the other ligand cannot bind.  

Term

What are membrane transporters?

 

What is Cystic Fibrosis?

 

What are the 2 types of transporters?

Definition

They are transporters in the membranes that help maintain the plasma concentrations of drugs. They have nothing to do with mediating drug effects.

 

CF is due to a genetic defect in ABC's. The defective ABC's can't efflux the chloride ions.

 

ATP Binding Cassettes (ABC's) and Solute Carrier Transporters (SLC's)

Term

What are ABC's

 

 

What are SLC's

Definition

ATP binding cassettes (ABCs) are primary active transporters that are responsible for causing an efflux from cells in the liver, kidney, intestine, and BBB. The most important ABC is the MDR.

 

SLC's are solute carrier transporters. They use facilitated transporters or ion-coupled secondary active transporters. They are involved in the reuptake of nt's, transport of organic anions (OATs) and organic cations (OCTs)

Term

Organic anion transporters are important in what organ?

 

 

What popular class of drugs uses these OATs?

Definition

Liver

 

 

Statins

Term
Organic cation transporters are important in what organ.
Definition
OCTs are important in the kidney.
Term

T or F:

All drug actions require that a drug bind with a receptor.

Definition

False

Chelating agents, osmotic cathartics, & anesthetic agents act through physical or chemical processes.

Cholinesterase inhibitors and MAOIs act through enzymatic mechanisms.

Term
Penicillin used to be administered parenterally. Why is penicillin now administered orally?
Definition

Since penicillin is a water soluble drug, it doesn't absorb well. If it's given orally, it will have more time to be absorbed.

Term

Why is drug metabolism necessary?

Definition

Most drugs are lipid soluble, meaning they would stay in the body for a long time and become very toxic. Metabolism converts the drugs into water soluble metabolites that can be excreted.

Term
What is a prodrug and why is it given?
Definition
A prodrug  is administered as an inactive drug to enhance absorption, avoid destruction, or to enhance delivery to a certain site. Once in vivo, the drug is metabolized to its active form. Their main purpose is to improve ADME.
Term
Prodrugs are often designed to improve bioavailability for oral drugs. Why?
Definition
because oral drugs are sometimes poorly absorbed in the GI tract.
Term
Prodrugs are used in many cancer treatments to limit adverse effects. Explain how they work.
Definition
These prodrugs are not toxic until the reductase enzymes found in hypoxic cancer cells convert them to their cytotoxic form.
Term

What is the main difference b/w the Phase I and Phase II proportions of metabolism?

 

Which one is synthetic (or conjugated) and which one is nonsynthetic?

Definition

Phase I involves small chemical changes occurring in one or more functional grps of a drug. Phase II is where a molecule provided by the body is added to the drug. 

 

 Phase II is synthetic (or conjugated) b/c there are many changes. Phase I is non-synthetic b/c there are minor changes.

Term

T or F:

Phase I rxns always precede PhaseII rxns

Definition

False

The purpose of phase I rxns is to prepare drugs for phase II rxns by making them more reactive. Some drugs already have plenty of reactive grps and can be directly metabolized by phase II conjugation.

Term

What are the main types of metabolic

phase 1 rxns?  What types of groups are commonly added b/c of these rxns?

Definition

oxidation, reduction, and hydrolysis

 

Think SHAC

Sulfhydryl (SH), hydroxyl, amino, & carboxyl.

Term

T or F:

Only metabolic phase II rxns involve conjugation.

Definition
False
Term

T or F:

Any drug that is modified by a metabolic phase I rxn will lose its biological activity.

Definition
False, a prodrug will become active.
Term
What is the most common type of biotransformation?
Definition

Oxidation

 

 

FYI: Biotransformation = metabolism

Term

T or F:

Most oxidations take place in the SER of cells in metabolizing organs such as the liver and kidneys.

Definition
False, most oxidations take place in the SER of cells in metabolizing organs such as the liver and intestines.
Term
Name three types of enzymes that carry out oxidations. Which are the most important?
Definition
flavin monoxygenases, various dehydrogenases, and CYP450. CYP450 are the most important.
Term
What are microsomal enzymes?
Definition

They are enzymes found in the SER

Term
What is a common pathway of biotransformation for esters and amides?
Definition
hydrolysis (a phase I rxn)
Term

What are the products after an ester undergoes hydrolysis? What enzyme catalyzes this rxn? 

 

What are the products after an amide undergoes hydrolysis? What enzyme catalyzes this rxn?

Definition

An esterase hydrolyzes an ester to an alcohol and a carboxylic acid. 

 

An amidase hydrolyzes an amide to an amine & a carboxylic acid.

Term

Why are phase I rxns nonsynthetic?

 

 

 

Why are phase II rxns called synthetic or conjugated?

Definition

It's b/c only small changes occur in one or more functional groups on a drug.

 

 

It's b/c the body provides a highly polar molecule or group to be attached to the drug.

Term

What are the 6 types of conjugation rxns involved in phase II of biotransformation?

 

Which is the most common?

Definition

Think GASMAG 

Glucuronidation

Amino acid conjugation

sulfation

methylation

acetylation

glutathione conjugation

 

Glucuronidation is the most common

Term

Explain the process of glucuronidation in phase II rxns.

 

Why is this the most common phase II rxn?

Definition

With the help of a microsomal enzyme, the GA attached to UDPGA is transferred to the drug, making it a glucuronide. The glucuronide is a weak acid that will ionize at physiological pH, and be excreted.

 

It's the most common rxn, b/c UDPGA is not selective and it comes from glucose (which the body has plenty of).

Term

During biotransformation, what type of cmpds are sulfated?

 

Explain sulfation.

Definition

aromatic hydroxyl cmpds (eg, phenols and catechols) and some amines

 

 

First, sulfate combines w/ ATP to convert to its active form, PAPS. Sulfotransferases then catalyze the conjugation rxn b/w PAPS & the drug (or the phase I derivative), making it more water soluble and readily excreted in the urine and bile.

Term

Concerning biotransformation, explain amino acid conjugation.

Definition
COOH's are activated by ATP and converted to CoA thioesters. Endogenous aa's (usually glycine and glutamine) then conjugate with the CoA thioesters. These aa conjugates are more water soluble and readily excreted in urine and bile.
Term
Concerning biotransformation, explain the process of acetylation.
Definition
Primary alkyl and aromatic amines conjugate with acetic acid to form an acetyl conjugate. The ionizable amine is actually converted to a nonionizable amide; nevertheless, acetylation does deactivate the drug. See pg 203 for details.
Term

T or F:

Concerning biotransformation, glutathione conjugates are rarely excreted in bile, but are excreted only in urine.

Definition

False

They're rarely excreted in urine b/c of their large MW. They can be excreted in bile.

Term
What are the two phase II rxns that result in conjugates that are less polar than the original cmpd?
Definition
Acetylation and methylation
Term
Concerning biotransformations, what drugs are readily methylated?
Definition
epinephrine and isoproterenol among others.
Term
What is the functional hepatic unit?
Definition
The liver lobule
Term

T or F:

There is absolutely no barrier in the hepatic sinusoids to limit the uptake of solutes.

Definition
True.
Term

T or F:

Polar and ionized drugs cannot enter hepatocytes.

Definition

False

they can, but they have to enter via carriers or specific transporters.

Term
Discuss 2 factors that could increase metabolism.
Definition

Environmental or therapeutic factors can cause enzyme induction. Examples of therapeutic factors would be barbituates & rifampin increasing CYP3A4.

 

Genetic mutations can also increase metabolism.

Term
Discuss factors that decrease the rate of metabolism.
Definition
"Intentional inhibition" (of an enzyme) is the goal of many drugs. Fe, allopurinal prevents gout by inhibiting xanthine oxidase from making uric acid. A decreased rate could also occur if 2 drugs are competing for the same enzyme. Other factors include age (young or old), disease states, grapefruit juice, diet, and genetic differences.  
Term
Which occurs more often: decreased rates of metabolism or increased rates of metabolism?
Definition
Decreased rates
Term

How many CYP450's are there?

How are they distributed?

Definition

57

15 metabolize xenobiotics

14 are involved with sterol metabolism

4 oxidize fat soluble vitamins

9 oxidize fatty acids in addition to synthesizing prostaglandins

15 have no xenobiotic substrate

 

 

Term
Which CYP is most prevalent in tissue and most efficient at metabolizing drugs?
Definition
CYP 3A4
Term

What 2 drugs induce CYP3A4 and CYP2CP/19?

 

 

What CYP is most abundant? Most polymorphic?

Definition

Rifampin and phenobarbital. This is one of the few examples of where metabolism is increased. If anything, metabolism is more often decreased

 

 

CYP3A4; CYP2D6

Term

Compare zero and first order kinetics

Definition

zero order kinetics                              first order kinetics

-there is a constant amount of drug      -there is a constant % of drug lost  

lost per unit time                                  per unit time

-It's dose dependent, b/c the half-life   -It's dose-independent, b/c the 1/2

increases as [plasma] increases.           life remains steady at any [plasma]

-The graph is non-linear, b/c as the      -The graph is linear, b/c as the  

dosage increases, the [plasma]             dosage rate increases, the

increases exponentially.                        [plasma] stays the same. 

-the saturation of an enzyme is            -There is no saturation.

associated w/ its rate of excretion,         

thus the disappearance is a constant

amount per unit time.

-The exponential increase occurs b/c     -The linear increase occurs b/c the

the [plasma] is > Km.                           [plasma] is < than the Km.

-Prediction of [plasma] is difficult          -Prediction of [plasma] is easy and

                                                          toxicity is avoided.

Term

What does steady state mean?

 

What is the steady state formula?

Definition

It means that a drug has equilibrated in all fluids.

 

1.5 X dose/time X t1/2

Vd

 

When given data, find the mass/volume and multiply it by 1.5

Term
What 6 factors determine plasma concentration? Explain each of them.
Definition

1) Rates of absorption and elimination - [peak] occurs when rates absorption and elimination are =.

2) ROA - [peak] will occur soon after an IV. For all other ROA's, the [peak] varies.

3) Target tissue

4) Apparent Vd - it's apparent, b/c it's not accurate. It's = to the dose / [plasma]. If the [plasma] is low (due to drug being stored in tissues), the Vd will be high. If the [plasma] is high, the Vd will be low.

5) Clearance - the volume of blood (ml's) from which drug is removed per unit time (minutes).

6) Half life - the time for the agent to decrease in concentration by one-half.

Term
Be able to calculate clearance, Vd, half-life, or Ke when given the appropriate info. List some formulas.
Definition

C = Ke * Vd

t1/2 = .693/ke

 

Term

Dose rate = ?

 

 

Loading dose = ?

Definition

Dose/Time = Cl (mL/min) * [plasma] (in mg/ml) 

 

 

Loading dose = [plasma] (in mg/L) * Vd (in L)

Term

For drugs that follow first order kinetics, the time to reach the final concentration is dependent on                  .

 

If a drug has a long/short (choose) half life, you can give a priming dose of the drug to reach the therapeutic concentration faster.

Definition

half-life

 

 

long

Term

T or F

A prodrug has to be administered as inactive

Definition

False

It can be significantly less active

Term
Which second messengers are mainly involved with cell growth and proliferation?
Definition
tyrosine kinases and phosphatases
Term

What is the main purpose of NO?

What are some of the other actions of NO?

Definition

Vasodilation via cGMP.

It acts as a NT in the CNS. NO alters platelet activation and has roles in inflammation and septic shock.

Term
What is the principal effector of vasodilation? Name three things that increase this effector?
Definition

cGMP

NO; increased by ACh through the production of NO; and, phosphodiesterase V inhibitors (viagra)

Term

T or F

Drugs made for hypertension are non-specific

Definition
true
Supporting users have an ad free experience!