• 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

1/1427

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;

1427 Cards in this Set

  • Front
  • Back
antiarrhythmic with negative inotropic effects
ca channel blockers
b blockers
class 1a drugs (sodium channel blockers)
what two things increase gastric acid secretion? How?
-increased levels of ca++ ( gastrin + Ach increase levels of Ca++)
-increased levels of camp (histamine increases camp, pgi2 and pge2 decrease camp)
- all act on the protein pump inhibitor
to lower renin release, give
clonidine (centrally acting)
beta blocker or an angiotensin converting enzyme (ACEI)
what type of arrhythmias (ventricular) do you get with reentry?
-extra single systoles
-sustained types of tachycardia
treatment of lepra reactions (erythema nodosum & leprosum reactions)
-steroids
-thalidomides
-clofazimine
What drug do you give to treat cutaneous manifestations of moderate to severe erythema nodosum leprosum (ENL)?
Thalidomide
Thalidomide MOA
-immunomodulatory agent
-down-regulation of both TNF-alpha and adhesion molecules involved in leukocytes migration
Which bile acid binding drug causes increased hepatic uptake of LDL?
colestipol
What's the difference between bile acids & niacin?
Bile binding acids = for isolated increases in LDL. Bile acid binding in lumen and prevent reabsorption

Niacin - for familal hypercholestemia, inhibits VLDL secretion
High TG, what drugs to treat?
Gemfibrozil
What's the difference between colestyramine and colestipol?
different indications: CTYRAMINE is used to treat pts with an isolated increase in LDL, COLESTIPOL treats pts with elevated serum total and LDL
treatment of lepra reactions
reversal reactions
-steroids
-clofazimine
a side effect of this antimycobacterium is optic neurotis (a decrease in visual activity)
Ethambutol
drugs used for MAC
-Clarithromycin
-Azithromycin
-Ethambutol, clofazimide, ciprofloxacin, or rifampin
phentolamine for CHF
- predominant effect on arteriorlar vascular bed

- H failure, decrease afterload, increase CO
active tuberculosis (possibly resistnat organisms & HIV patients)
Isoniazid, Rifampin, pyrazinamide, ethambutol or streptomycin
Diuretic used for glaucoma
Acetazolamide
Name two drug combos used for angina
- nitrates and b-blockers - can prevent a reflex tachycardia, reduce each others undesired SE

- CCB + B blockers + nitrates - all
This diuretic is indicated for treating edema used to CHF, centercephalic epilepsies, glaucoma, acute mountain sickness
Acetazolamide
When do you use captopril?
-useful in cases where elevated renin release causes
-produces hypotension plus reduced sodium retention
Name long-acting renin-angiotensin inhibitors
Enalapril & lisinopril
To maintain sinus rhythum in chronic atrial fibrillation
-class IC or Ia
-amiodarone
-sotalol
Kinins - effect on renal glands
-potent vasodilation
- increase prostaglandin synthesis
-no2 release
-increase cGMP and increased Camp
Dipyridamole MOA
- vasodilator -dilates resistance vessels in coronary beds to increase flow without increasing O2 demand

- thromboembolism - lengthens abnormally shortened platelet survival time
What drug is used for the detection of renin -dependent hypertension
saralasin
What effects do organic nitrates have on the redistribution of coronary blood flow?
-organic nitrates can selectively increase blood flow to ischemic areas (note: total coronary flow is not reduced)
Bile acid binding resin not effective in pts with homozygous familial hypercholesterolemia?
cholestyramine
antimycobacterium causes severe birth defects
thalidomide
hypolipidemic may impair carbohydrate tolerance
niacin
diazoxide MOA
- a long acting arteriolar dilator
-prevents smooth muscle contraction by opening potassium channels + stabilizing the membran potential at a more hyperpolarized voltage
- lowers tpr and arterial blood pressure
I have a life threatening ventricular arrhythmia. Give me a drug
Treatment = Procainamide

To prevent the reoccurance of life-threatening ventricular arrhythmias: amiodarone
What regulates GFR
GFR is cappillaries is regulated by the amount of tone contractivity in arterioles
What's the difference between abciximab and Eptifibatide and Tirofiban?
-abciximab - monoclomal antibody that binds IIb/iiia complex on platelets

-Eptifibatide - a fibrinogen sequence analog that binds GP iib/iiia receptors prevents aggregtion
I'm pale (pallor), what might I have?
Iron deficiency (comon presentation)
What's the difference between Thrombopotetin and GM-CSF?
-Thrombopoietin - increases size + number of megakarocytes

GM-CSF (w/ IL3 increases activity of mature granulocytes)
What drug is reserved for pts with no significant/structural heart disease but with atrial fibrillation or atrial flutter?
-quinidine
a broad spectrum - antiarrhythmics
name it which channels does it effect.
-amiodarone
-predominantly K channel blockade
-B-blockers, ca channels blocker, and Na channel block
What is the difference between Vit B12 and Folic acid?
FA: essential cofactor for synthesis of aa, purines, & DNA

VB12: a cofactor for several essential biochemical rxns

FA: treat FA deficiency
VB12: treat VB12 deficiency

Notes: VB12 deficiency causes neurological abnormalities
Urokinase and streptokinase MOA
urokinase : human enzyme
streptokinase: bacterial protein (not enzyme)

- combines with plasminogen and increases its conversion to active plasmin. Plasmin formed inside a thrombus allows them to lyse a thrombis from within
t-PA MOA
t-PA binds fibrin in a thrombus, which convers the entrapped plasminogen --> plasmin. This initiates fibrinolysis and some systemic proteolysis
what's the difference in mechanism between urokinase and t-PA
Urokinase combines with plasminogen to activate conversion to plasmin

t-PA combines with fibrin to activate plasminogen --> plasmin
SE causes hypothroidism or hyperthyroidism?
amidarone
What is the difference in Rx between atrial fibrillation, atrial flutter, and atrial tachycardia
They are all essentially the same, only use adenosisne top choice for atrial flutter in addition to regular therapy
Thrombolytic that is more fibrin-specific that t-PA
Tenecteplase
Less fibrin-specific that t-PA?
Reteplase
Less expensive than t-PA (also a thrombolytic
Reteplase
Ventricular tachycardia after MI, what is it?
Reentry near the rim of healed infarction
What factors stimulate renin release
-hypotension
-decreased blood volume
-NA depletion
-increased sympathetic outflow
Pharmacokinetics of Abciximab?
Must be given parenteraly not orally effective
antiplatelet drug used in percutaneous coroncary intervention (e.g. angioplasty, placement of coronary stints) & in acute coronary syndromes
Abciximab
Active tuberculois (susceptible)
-isoniazid and rifampin for 6 months
add pyrazinamide after 2 months
Why do you not give sotalol with class 1a antiarrhythmics? Or Class III aa?
B/c of the potential to prolong refractoriness (QT)
Ventricular tachycardia after MI
acute treatment?
chronic treatment?
acute - lidocaine, procainamide, DC cardioversion

chronic - amiodarone, class III, ICD
What is a partial Thromboplastin time (PTT) for?
PTT monitors the intrinsic coagulation system and is abnormal in deficiencies of factor i, ii, v, ix, x, xi, xii.
What is the difference between a PT test and a PTT test
PT measures deficienceis in factors I, ii, V, Vii, X
(more warfarin like)
PTT measures deficiencies in factors v, viii, ix, x, xi, xii (more heparin like)
NSAIDS on plasma renin activity?
NSAIDS decrease plasma renin activity, decrease renin
SE include weak ganglionic blocking, reduction in vagal tone
procainamide
Acute Rx for AV node reentry? PSVT?
-adenosine

-increase vagal tone with carotid sinus pressure
-digitalis
-edrophonium
-phenylephedrine
Use short term to treat erosive esophagitis, active duodenal ulcers, and GERD
proton pump inhibitors
antacids MOA
They are weak bases that react with gastric acid to form water and a salt and in the process neutralize the acid
nitrogen major effects
decrease preload
decrease venous rtn
decrease SV
decrease cardiac work
decrease O2 demand
How are atrial flutter and atrial tachycardia different from atrial fibrillation
atrial fibrillation: disorganized atrial reentry

atrial flutter: stable reentrant circuit in right atrium

atrial tachycardia: enhanced automaticity, DAD related automaticity or reentry
what drug is used for control of ventricular rate + prophylaxis for repetitive PSVT
-angina
-hypertension
Verapamil
Diltiazim also - only less good for angina and hypertension
name plasminogen activators
t-PA
Reteplase
Tenecteplase
To inhibit acid secretion, give ...
- histamine H2 antagonists
- prostaglandins
-proton pump inhibitors
name: platelet GP IIa/IIIa receptors blockers
Abciximab
Eptifibtide
Tirofiban
What's the difference between reteplase, t-PA, and tenecteplase?
They are all plasminogen activates (bind to fibrin & activate plasminogen --> plasmin)

Reteplase is a recombinant human t-PA with some aa's deleted (it is less fibrin specific that t-PA and cheaper to make)

Tenecteplase is a mutant form of t-PA with that is more fibrin specific
What's the difference in indications between

(1) histamine H2 antagonists
(2) Proton pump inhibitors
(1) Histamine H2 antagonists for prophylaxis of peptic ulcers, acute stress ulcers, and GERD

(2) proton pump inhibitors = short term treatment of erosive esophagitis, ulcers, and GERD
This antimycobacterium drug may cause peripheral neuropathy
Isoniazid
Name three drugs used to accelerate food through through the GI tract. Explain the difference between the three types
(1) bulk laxatives
(2) irritants/stimulants
(3) stool softeners

Bls are hydrophylic collids that form gels and cause water retention and intestinal distention. Whereas, stool softeners become emulsified with stool to make feces soft. Stimulates produce nicinoleic acid
What's the difference between urokinase and fibronolysis?
Both thrombolytics, but urokinase/streptokinase works on old clots

Fibrinolysins only works on young clots (< 3 days)
What is streptokinase used for?
2nd line of defense
used in pts with myocardial infarctions in cases where coronary catherization is not readily available (coronary catherization is better reduces mortality)
name a fibrinolytic
the thrombolytics
Urokinase + streptokinases
Which drugs are used to treat edema caused by CHF?
Loop diuretics - Ethacynic acid + torsemide
What type of diuretic is used to treat pulmonary edema or other edematous conditions?
Furosemide
Angiotensin II - vasoconstriction or vasodilate renal
renal vasoconstriction
major side effects of all loop diuretics
hypokalemia
catecholamines - vasodilators or vasoconstricors renal
vasoconstrictors
Which loop diuretics inhibits the Na + 2CL K+ carrier system?
torsemide
which loop diuretic reduces pulmonary congestion and left ventricular filling pressure in heart failure?
furosemide
What is a toxicity of folate definiciency?
folate deficiency in pregnant woman can cause congential malformations in newborns

Folate deficiency may also not prevent the development of vascular disease (ischemic heart disease & stroke)
major SE of potassium sparing diuretics
-abnormal elevation of serum potassium levels
-hyperkalemia may be fatal
name K sparing diuretics
triamterene
amiloride
spironolactone
alpha agonists on plasma renin activity?
decrease renin activity
what do B-blockers do to plasma renin activity
b-blockers lower plasma renin activity decrease renin activity
Folic acid MOA
essential cofactor for the synthesis of amino acids, purines, and DNA
Which type of diuretics causes NaCl to be excreted and Ca++ to be reabsorbed?
Thiazides
drugs used for short term anticoagulent therapy associated with acute primary embolism and disseminated intravascular coagulation
heparin
what are the most efficacious diuretics?
loop diuretics
(furosemide, ethacrynic acid, torsemide)
What diuretics cause impaired carbohydrate intolerance and hyperkalemia?
Thiazides - chlorothiazides and hydrochlorothiazides
What drug can be given to neutralize the action of heparin?
protamine sulfate
this antiplatelet drug increase intracellular camp inhibiting phosphodiesterase activity and by blocking adenosine uptake
Dipyridamote MOA
In the prothrombin time test, you test for deficiencies in factors ----
i, ii, v, vii, and x
To restore sinus rhythum for an acute atrial fibrillation
DC cardioversion
ibutilide
What is a toxicity of vit b12 deficiency?
leads to neurological abnormalities including degeneration of myelin sheaths in axons of the spinal cord and peripheral nerves
what are cardiac toxicities of digoxin
1) 2nd and 3rd degree AVN block
2) various arrhythmias (premature beats, bigeminy)
3) changes in ECG ( increased PR, decreased ST, and decreased QT)
PSVT acute conversion to sinus rhythum use --- which drugs are used for prophylaxis of PSVT (avn reentry)?
1) use adenosine
2) class IV - calcium channel blockers (verapamil & diltiazem)
side effects of iron dextran, how administered too?
hypersensitivty reactions given by deep IM or IV
vasoconstriction or vasodilate TXA2
vasoconstrict
1st drug for control of ventricular rate in pts with atria flutter or fibrillation?
-verapamil, diltiazim
Which antiarrhythmic is not associated with an increase in mortality in pts with coronary artery disease or heart failure?
amiodarone
what drug can : acute sinus conversion to sinus rhythum of PSVT? including when its associated with Wolff-Parkinson White syndrome?
Adenosine
Common treatments for atrial fibrillation(acute rxn) to control ventricular rate
verapamil or diltiazem
b bockers
digoxin
what is the difference between atenolol and metoprolol
indications: atenolol has more indications (management of hemodynamically stable pts iwth definite of suspected myocardial infacrction) in addiiton to hypertension + angina?
main use of sotalol
Ventricular arrhythmias?
Which type of drug is used for the healing of duodenal ulcers and peptic ulcers? how?
mucosal protective agents

MOA binds + charged groups on proteins and glycoproteins of abnormal and necrotic tissue
acts as a barrier to HCL acid and pepsin
Which drugs complicate the effects of PGs on renal glands
NSAIDS
non-steroidal anti-inflammatory drugs
which prostaglandins, increase renal blood flow, promote diuresis, and natriuresis
PGE2
PGI2
for immediate reduction of stomach pain (cheap alternative) use
antacids
abciximab MOA
a humanizeed monoclonal antibody directly against IIb/IIIa complex. it binds to the receptor complex on platelet and prevents platelet aggregation
difference between 1st generation and 2nd generation sulfonylureas?
-fewer side effects & drug interactions, more commonly prescribed

contraindicated in hepatically impaired pts
What does Erythropoietin do? Where does it act?
stimulates maturation of erythrocyte precursors

It has two sites of action
(1) BFU-E --> CFU-E
(2) CFU-GEMM --> BFU-E (acts synergistically with IL3 and GIN-CSF
4 mechanisms of vasodilation
(1) block L-type calcium channels
(2) increased cAMP
(3) increased cGMP
(4) hyperpolarized cell membrane
Which loop diuretic is effective in patients with renal insufficiency
The loop diuretic Ethacrynic acid
What's the difference between loop diuretics and thiazides?
Place they act: Loop diuretics: loop of Henle
Thiazides - distal convoluted tubule

Transport they block
Thiazides: block NaCl reabsorption, increase Ca++ reabsorption

Loop diuretis - inhibit NaCl reabsorption (some act on the Na2Cl-K+ pump)
What type of diuretics is not used in patients with renal insufficiency?
Thiazides
Kinins - renal vasoconstrictor or vasodilators
renal vasodilators
This angina occurs during exercise (climbing stairs, etc)
External angina
Name renal vasoconstrictors
-Thromboxane A2, TXA2
-Catecholamines
-Angiotensin II
Name loop diuretics
Furosemide
Ethacrynic acid
Torsemide
SE fatal pulmonary fibrosis
Amiodarone
Congenital long QT MOA :
EAD related triggered activity
Class II MOA (antiarrhythmics)
-B-blockers terminate tachyarrhythmias caused by increased sympathetic tone, increase catecholamines, tissue supervision to catecholamines

- reduces pacemaker automaticity (blocks catecholamine stimulation of Ih and Ica)
-reduces delayed after depolarization (DADs) by antagonizing B-adrenergic increase in Ca flux
Drug of first choice for treating pts with AVN reentry (PSVT)
-Adenosine
first drug choice for acute treatment
Antidote + treatment for warfarin overdose
-stop giving warfarin
-give fresh plasma
-give Vitamin K
Aspirin MOA
prevents production of thromboxane A2, Irreversible
Indications for spironolactone
-hyperaldosterone
-hypokalemia
-reduced edema & mortality in patients with CHF
Most renal vasodilators promote ----
diuresis
mild CHF treatment (mild peripheral edema on exertion) normal GFR
thiazides, ACEI
Treatment of Heparin overdose:
(1) stop giving heparin
(2) antidote: protamine sulfate
Deficiency of what compound can make warfarin toxicity worse?
a vitamin K deficiency
Transport that occurs at collecting duct
sodium is exchanged for potassium + hydrogen ions
Where in the nephron is the fluid isotonic?
hypertonic?
hypotonic?
isotonic - proximal tubule
hypertonic - descending limb loop of henle
hypotonic - ascending limb, loop of henle
Where does ADH act>
on the distal convoluted tubule and collecting duct

ADH increases water reabsorption
Which effects predominate at low doses of nitrates? nitroglycerin
venous effects predominate at low doses, but nitroglycerin does cause dilation of both arterial and venous beds
which coagulation drug should you never give IV?
Thrombin
What % of NaCl is reabsorped in the proximal tubule?
in the loop of Henle?
in the distal convoluted tubule?
-proximal tubule - 60% -70%
-loop of henle --> 15-25%
-distal convoluted tubule --> 8% - 5 %
diuretics are ...
natriuresis is ...
- agents that impair the reabsorption of sodium from tubular contents

-natriuresis is the excretion of sodium in the urine
what is the undesirable side effects of all vasodilator agents especially over administrated by intravenous infusion
-extreme hypotension
What's the difference between typical (exertional angina and variant angina)?
-typical (exertional)
-coronary insufficiency due to vessel occlusion (atherosclerosis)
-attacks usually occur during exercise (climbing stairs, etc.)
-variant prinzmetal's rest angina
-coronary insufficiency due to vasospasm?
-attacks occur during rest unstable
name 3 calcium channel blockers
-nifedipine
-verapamil
-diltiazam
which anticoagulent do you never give IV?
Thrombin
Thiazide MOA
thiazide increase NaCl excretion in the distal convoluted tubule. Loss of potassium and bicarbonate can result
anticoagulent test used to diagnosis deficiency in factors I,ii, v, vii, and x
prothrombin time test
Name one type of antianginal that can eb used for both vasopastic & effort - associated angina
-ca channel blockers
verapamil
diltiazem
nifedipine
Where do carbonic anhydrase inhibitors work?
proximal tubule
Treatment for warfarin overdose
(1) stop giving warfarin
(2) give fresh plasma
(3) give Vitamin K
What's the difference between cotting time and bleeding time?
clotting time - the time it takes blood in vitro to clot sufficiently so that the table containg the sample can be inverted

bleeding time- after you cut the ear, the length of time you cna collect blood
what;s the difference between thromus and embolism?
-thrombus -an intravascular blood clot obstructing a blood vessel or cavity of the heart

-embolism - obstruction of a blood vessel by a foreign substance or a blood clot that has been carried by the blood to a site distant from its point of origin
What's the difference between anticoagulents and thrombolytic agents?
-anticoagulents prevent the formation of fibrin clots

-thrombolytic agents remove and break fibrin deposits in blood vessels
hair growth
minoxidil
NO mechanism
No increase gCMP, decrease ca++ (becasue it goes into the SR
overdose of coumarin/ warfarin, what do you give me? why?
Vitamin K2 - cofactor required for the synthesis of factors Viii, IX, and x
major SE of heparin
SE include hemorrhage (especially ovarian hemorrhage can be potentially fatal
Dispersion of refractoriness may occur during ...
(1) an unequal ischemic lesion
(2) an extra conduction pathway
what is the effect of dopamine on glomular filtration rate, renal blood flow, and Na+
-stimulates alpha, beta, and dopamine receptors

-increase renal blood flow
-increase glomerular filtration rate
-increase Na+ excretion
Treat my hypokalemia
1. Kive K+Cl-
2. give a potassium sparing agent
3. combo drug
DC valsartan
ARB, antihypertensive
major SE of heparin
hemorrage
What's the difference between chlorothiazide + hypochlorothiazide?
chlorothiazides can also be used for kidney stones and diabetes insipidus
What ending signifies and ARB?
-sartan as in

iorbesartan
losartan
valsartan
anticoagulent which causes increased neutralization of factors II, IX, X, XI, XII, and Xiii
heparin
Why do diuretics help CHF
reduce preload and afterload
also counteract sodium retention forces ( at a max CHF) by causing natruersis
diuretics class used for CHF
loop diuretic (thiazides also work)
class of diuretics chiefly associated with hypertension
thiazides
which type of diuretic blocks the Na+2CL-K+ transporter
loop diuretics
which durgs act on the NaCl cotransporter? Where is this int he nephron?
thiazides in the early distal convoluted tubule
which drugs cause the induction of LDL receptors thereby decreasing LDL in serum?
statins - lovastatin, simvastatin
amiloride acts on?
where?

name a drug like amiloride
-amiloride blocks sodium channels in the distal convoluted tubule

triamterene
What type of drug acts on mineralocorticoid receptors?
Where? name 2
-aldosterone antagonists bind to mineralocorticoid receptors in the distal convoluted tubule (spironolactone epelerone)
MOA of drugs that work and hte loop of Henle
interferes with Na+2Cl-K+ cotransporter
why are proximal active agents only mildly potent
Na+ ions not absorbed here have multiple opportunities downstream to be absorbed
How do carbonic anhydrase inhibitors work> MOA?
-inhibits reabsorption of sodium bicarbonate

-reduce availability of cellular protons ions for exchange with luminal sodium
Where in the nephron does bulk reabsorption occur?
proximal tubule (60-70%
why are loop diuretics so powerful
-large amounts (15-25%) of NaCl are reabsorbed at this site

-nephron transport sites distal to the loop of henle have limited capabilities to reabsorb NaCl ions rejected in the loop
injudicious results of potassium sparing diuretics -->
-hyperkalemia
-metabolic acidosis

-b/c diuretic inhibits K+ and H+ secretion
which part of the nephron is under control of the mineralcorticoid systems?
How much sodium reabsorption occurs here?
-the late distal convoluted tubule and collected duct
-about 2-3% of the filtered load is absorbed here (sodium)
what types of transport occurs in the late distal convoluted tubule?
Na+ is reabsorbed in exchane for hydrogen and potassium ions
What type of ion transport is found in the loop of henle? ascending limb
an active electroneutral sodium 2 chloride potassium
how is bicarbonate reabsorbed in the proximal tubule
1. an Na/H causes secretion of H ions
2. H ions + bicarbonate = carbonic acid
3. dehydration of carbonic acid - H20 + CO2
4. CO2 diffuses into cell
5. in cell, co2 combines with H20 --> H2CO3
6. net movement. H2CO3 is moved from lumen ot the cells of the proximal tubule
where is most bicarbonate reabsorbed in the nephron?
in the proximal tubule
what can reduce the action of diuretics?
a reduction in GFR (glomerular filtration rate) rduces the supply of ions and water to the tubular system

- it also can decrease renal plasma flow
drug used to treat CHF acts on aldosterone
spironolactone
Don't use this hypolipidemic in pregnant women
statins
this type of angina is unstable - attacks oftern occur during rest
variant angina
DC losartan
ARB, antihypertensive
drugs used for combined dyslipedemias
niacin decreases TG, decreases VLDL and decreases LDL
These HMG-Coa reductase inhibitors have the SE of ...
hepatotoxicity and myopathy
what's the difference in effect between ACEI and ARBS?
(1) ARBS are more specific. They only block Ang II and ATI receptors
(2) ARBS have no effect on Bradykinin, whereas ACEI increase bradykinin
(3) ARBS more completely inhibit ang II action (ACEI only blocks some ang II production becasue there are other ways to make it
What's the difference in mechanism between drugs acting in the early distal convolution and the distal convoluted tubule?
-early distal convolution- sodium chloride cotransporter (thiazides block)

- distal convoluted tubule - exchange Na for K+ and H+ aldosterone antagnonist block)
What is the consequence of adminsitering amiloride or triamtere
mild natriuresis (increase Na+ in urine) inhibit potassium and hydrogen secretion
main use of anticoagulents
pulmonary embolism
coronary thrombosis
with myocardial infarction
peripheral vascular thrombosis
the side effects - hepatotoxicity and myopathy are characteristic of what type of hyperlipidemic?
hmg-coa inhibitors
pharmacokinetics of heparin
should be given by intermittent intravenous injections, intravenous infusion or deep subcutaneous injection
how do all vasodilators affect sodium?
All vasodilators cause sodium retention
why is hydralazine a bad antihypertensive?
-must be used with b-blockers (b/c of reflex tachycardia)
-must be used with diuretics for sodium retnetion
name 2 centrally acting drgus used to treat hypertension
clonidine & methyldopa
What are the best type of diuretics to use for hypertension?
-thiazide diuretics
-potassium sparing diuretics
-loop diuretics (high celeing) -->
can be used for edema reduction decreases volume and decreases bp
drug given to patients undergoing placement of a coronary stint
clopidogrel main indication
protamine sulfate MOA
PS forms a stable salt with acidic heparin. It neutralizes its affect. The anticoagulent activity of both drugs is lost
how should vitamin K be administered?
not by IV

b/c anaphylaxis and fatalitis have occured when it is given iv
what is the advantage of enoxaparin over heparin?
better bioavailabiiltya dn longer half life (SC injection instead of intravenous
what shoudl be monitored when giving heparin?
monitor platelet counts - perform frequently
name renal vasodilators
-dopamine
-pgs, pge2, pgi2
-kinins
what do b-blockers do to treat angina?
b-blockers block cardiac b1 receptors. thereby decreasing hr, decreasing contractility (negative inotropic effet) and decrease myocardial o2 requirements at rest and during exercise
-reduce silent ischemic
severe CHF, peripheral edema, dyspnea, and pulmonary congestion
combination diuretic therapy
loop diuretics
ACEI
What drugs can you use to treat primary triglyceruridemias?
niacin and fibric acid
heparin MOA
-anticoagulent
-straight chained anlonic mucopolysacchards called glycosaminoglycans have anticoagulent properties
-acts at multiple sites in the coagulation system
-heparin interacts with antithrombin (a cofactor) it enhances its ability to inhibit thrombosis
-inactivates clotting factors - iia, ixa, and xa by complexing with them
ca++ antagonists
-selective effects on blood vessels but not on the heart
-reduces vasospasm, reduces preload + afterload
side effects and complications of diuretics
-volume depletion (loop diuretics
-hypokalemia
-lipid elevation
-glucose intolerance
-metabolic alkalosis
-hyponatremia
-hyperuricemia
-hypomagnesia
major drug interactions with warfarin
increase warfarins effect (inhibit P450 cimetidine and amiodarone)

- decrease warfarin's effect (induce p450)
warfarin is contraindicated in
pregnancy
heparin blocks which factors

warfarin blocks which factors
heparin II, IX, X, XI, XII, XiII

warfarin II, Vii, IX, X
NSAID effects on ACEI effects?
-NSAIDS block PG synthesis
-NSAIDS also block bradykinin mediated vasodilation, therefore they impair the hypotensive effects of ACEI
how do diuretics treat hypertension?
1. decrease blood volume (decrease Bp and pressure )

2. with continued use, diuretics decrease TPR even when blood volume returns to normal
What does the PT test mean?
Test the extrinsic thromboplastin system, tests for deficiencies in factor I,II, V, VII, and X
When treating hyperlipedemia in diabetes, which drug should you be careful about giving and why?
niacin b/c one of the side effects is glucose intolerance
which factors does enoxaparin block?
inhibits clotting factors IIa (prothrombin) and Xa
With atorvastatin, you must monitor levels of ...
alanine and aspartate
dipyridamole drug class
DC: vasodilatior
platelet adhesion inhibitor
Name the major groups of drugs acting at the proximal tubule

give an example
carbonic anhydrase inhibitors (acetazolamide)
treat dyastolic dysfunction with ...
B-blockers
calcium channel blockers
therapeutic uses of coumadin/warfarin
-prophyalaxysis + treatment of venous thromboses + pulmonary embolism
-treatment of thromboembolic complication sto reduce the risk of death, recurrent myocardial infarction + thromboembolic events
heparin effects
-has little or no effect except on blood coagulation + blood lipid metabolism

- increases activity of antithrombin III (antithrombin III) neutralizes activated clotting factors II,IX, X, XI, XII, and XIII
What are the three main types of drugs used to treat angina>
(1) organic nitrates compounds
(2) propranolol and other B-blockers
(3) calcium antagonists
What's the difference between thrombolytic agents and antiplatelet agents
-thromboembolytic agents = remove and break fibrin deposits which occur in bood vessels

whereas
-antiplatelet agents are used to reduce atherosclerosis
used for pulmonary embolism, coronary thrombosis associated with myocardial infarction, peripheral vascular thrombosis, severe injury to the extremities and rheumatic heart disease
anticoagulent agents
aldosterone MOA
works in the distal parts
-genomic mechanism (increased expression of ENOC channels

old antagonists increases Na+ and decreases K+ (wasting)
which drugs blocks PDE5?
original purpose of drugs
-sibenafil
-vardenafil
-tadalofil

-pulmonary hypertension
What's the difference between heparin indications and warfarin indications
heparin is for short term anticoagulent therapy, while warfarin is more long term

both for prophylaxis treatment of venous thrombosis and thromboembolics associated with atrial fibrilation

pharm: warfarin: 100% bioavailability
Heparin: intermitt intravenous injections
ACEI MOA for CHF
ACEI reduce angiotensin II and aldosterone levels, decrease Left ventricular filling pressure, decrease TPR, increase CO
What is a side effect of guanethidine, reserpine, MAOI, and ganglionic blockers
postural hypertension
which antihypertensive can be used as monotherapy?
calcium entry blocking agents + ACEI
prostaglandin effects on renal excretion?
-increase renal blood flow
- promote diuresis
-promote natriuresis
What's the difference in renal effect between prostaglandin PGE2 and prostaglandin PGI2?
PGE2 - causes vasodilation blocks H20 reabsorption and increases diuresis

PGI2 - increases renin secretion, increases cAMP levels
What is the difference between Verapamil and Diltiazim?
- Verapamil is better for angina & hypertension.
-has a longer half life (7.4 hours max)
-different side effects - constipation + asystole in addition to bradycardia + AV node conduction block
stepwise approach versus monotherapy
- mild hypertension
1. weight reduction
2. sodium intake reduction
3. thiazide diuretics or B-adrenergic blockers
4. ACEI
5. Calcium channel blocker
6. angiotensin receptor blocker
clinical indications for B-blockers (antiarrhythmics
-prevent re-infarction & sudden death following an MI
-treat exercised induced arrhythmias
-prevent reoccurance of AVN reentry (pts with PSVT)
-control ventricular rate, increase AVN ERP
What type of drug class do you give to treat deep vein thrombosis, leading to pulmonary embolism - and in pts undergoing surgery?
think anticoagulents
(warfarin, heparin, enoxaparin)
2 indications for warfarin
1) prophylaxis and treatment of venous thrombosis
2) for the treatment of thromboembolic complications associated with atrial fibrilation and cardiac value replacement
which antianginal not only dilates resistance vessels in the coronary beds but also inhibits platelet aggregation?
Dipyridamole
pharmacokinetics of warfarin
-100% bioavailability
99% becomes bound to plasma albumin

- there is an 8-12 hour delay in warfarin action

-small Vd
what's the difference in indications between ACEI and ARBS?
-both work for hypertension, ACEI can also be used for CHF and to prevent glomerular daage in diabetes
difference between heparin's indication and warfarin's indication?
-heparin - for short term with acute pulmonary embolim

vs. warfarin is for pulmonary embolism + thromboembolic complications
what can you combine vasodilators with to improve treatment of CHF?
-vasodilators + inotropic agents
What tests should be monitored when going an anticoagulent?
-check prothrombin time and bleeding time measurements
Moderate hypertension therapy
1. thiazide diuretics
2. b-blockers + centrally-acting agent
3. ACE inhibitors
4. calcium entry blockers
5. angiotension receptor blocker
sodium nitroprusside MOA
short acting vasodilators
-decreases arterial impedence
-decreased venous pooling
which cofactor must be activated in both the intrinsinic and extrinsic coagulation pathways what is the role of this factor?
factor x -
(in intrinsic pathways - factors IX and XI are also activated)
Factor X cleaves two peptide bonds in prothrombin to form thrombin
name 2 antiplatelet drugs ---
1) aspirin
2) clopidogrel
what are the indications for nifedipine?
prophylactic treatment
1) typical (effort associated) stable (angina)
2) Prinzmetal's (variant) angina pectoris
3) hypertension
what drugs should you avoid while taking propranolol (b/c of drug interactions)
-reserpenine
-calcium channel blockers
-combination with haloperidol causes hypotension & cardiac arrest
-phenytoin& & rifampin increases propanolol's clearance
what's the effect of warfarin on vitamin K?
-warfarin prevents the reductive metabolism of the inactive form of vitamin K back to its active form by Vitamin K epoxide reductase
which antiplatelet drug irreversibly blocks the ADP receptor on platelets, thereby reducing platelet aggregation
clopidogrel MOA
drugs used to treat familal dyslipidemia
use niacin and or atorvastatin
what d b-blockers do to angina vs. arrhythmias
angina - decrease myocardial oxygen requirements by decreaseing heart rate, contractility, and bp

arrhythmia - reduce mortality and sudden death after MI
which type of angina is b-blockers not used for
contraindicated
b-blockers are not used for variant angina
what antianginal is used for acute attacks?
nitroglycerin (nitric oxide) sublingual
this anticoagulent is used for prophylaxis and treatment of thromboembolic complications associated with atrial fibrillation and cardiac valve replacement
warfarin
verapamil can be used for angina, but if the pt has --- don't use it!
don't use in patients with systolic CHF
b-blockers are often combined with --- to treat angina
organic nitrates
what's the difference between sublingual nitroglycerin tablets and nitroglycerin tablets?
nitroglycerin tablets (sublingual) are used for acute attacks

nitroglycerin patches - are used for prophylaxis against typical angina
tell me about digoxins TI
relatively low TI ( approx. 2 )
do organic nitrates have a bigger effect on the reduction of preload or the reduction of afterload?
have a greater effect on the reduction of preload
what's the difference between hydralazine and sodium nitroprusside
MOA: hydralazine - dilates arterioles not veins
sodium nitroprusside --> dilates arterioles +/- vessels

Indications -
hydralazine - essential hypertension
sodium nitroprusside - hypertensive emergencies and severe heart failure
blocks the carboxylation of several glutamate residues in prothrombin & factors VII, IX, X
warfarin
fatigue, anorexia, nausea, vomiting, diarrhea, dreams, muscle weakness, 2nd or 3rd degree AV conduction block, and various arrhythmias
digoxins famous side effects
what is the primary indication for digoxin?
What drugs do you use to treat A fib (pts have stretched atria Afib) (atrial fibrilation) and systolic CHF)
What deficiencies are seen in pregnant women?

How do you treat these conditions
(1) megalopblastic anemia due to a deficiency of folic acid --> use folic acid to treat

(2) iron deficiency due to blood loss during pregnancy --> use ferrous sulfate
AA classification

sotalol (DC & MOA)
DV: DC III
MOA: prolong action potential duration (K channel blockers)
atenolol DC + MOA
DC: DC II
MOA: B1 adrenergic receptor antagonist
quinidine MOA
Mixed Na + K channel blocker
lidocaine DC + MOA
DC: 1b
MOA: Na channel blocker
what drugs are used to treat pernicious anemia ?
what type of anemia is PA?
-folic acid and vitamin B12 are used to treat pernicious anemia (megaloblastic anemias due to folic acid deficieny)
-PA is a type of megaloblastic anemia
when would you use iron dextran instead of ferrous sulfate?
-pts with iron deficiency in whom oral administration is unsatisfactory or impossible (malabsorptions syndrome, dialysis pt)
warfarin MOA
-block carboxylation of many glutamate residues in prothrombin factors VII, IX, and X
-since these factors are not carboxylated, they are inactive coagulation
how does sodium nitroprusside reduces severe heart failure?
-in patients with heart failure & low CO, the CO often increases due to a reduction in afterload
What antimycobacterium do you give for resistant organisms?
pyrazinamide
antimycobacteria used as a preventive therapy (true chemoprophylaxis
isoniazid for 3 months if skin test + or subclinical infection isoniazid for 12 months
what is a potentially life threatening siutation resulting from treatment of atrial flutter
-if you treat w a class ia drug you can get a potentially life-threatening acceleration of ventricular rate b/c drug knock out AVN filtration ability
angina caused by coronary insufficiency due to vasospasm is called ...
variant rest angina
pulmonary fibrosis
major side effect of amiodarone
which drug interactions with digoxin? What is the effect?
-quinidine
-decrease cl and increases vd of digoxin, resulting in a doubling of p digoxin

- amiodarone
short half life 1-4 minutes
tolerance can develop
nitroglycerin pharmacokinetics
give folic acid (pts with Will's disease have a deficiency of folic acid)
i have wil's disease. Give me a drug. Tell me about my disease.
common treatments for CHF? the improtant one
-Ace INHIBITORS
-b-blockers
-diuretics
-vasodilators
-digoxin(only for systolic effects)
ventricular cardiac arrhythmias man (especially post MI) b/c class 1b, not effective against STVA
what do you use lidocaine for?
What's the difference between hydralazing vs. minoxidil
-both dilate only arteries but
hydralazine is used for essential hypertension and minoxidil is used only for treating hypertension that is sympatomatic or associated with target organ damage
what is the difference between ferrous sulfate and iron dextran
1) name an oral (absorable) iron preparation

2) name a parental iron preparation
antiarrhythmics that interact with digoxin
-quinidine
-amiodarone
atrial/ventricular tachycardia MOA
-DAD related
atrial enhanced automaticity
ventricular --> DADs triggered by sympathetic tone
difference betewen adenosine & amiodarone
amiodarone -broad spectrum antiarrhythmic

adenosine - endogenous nucleotide antiarrhythmic
where is ih most commonly found, If you block ih, what happens?
In plays a critical role in regulating automaticity in purkinje fibers, but play only a minor role in the SA node. Blocking Ih reduces automaticity in ectopic pacemaker
ACEI are contraindication
pregnancy
nitroglycerin indications
(1) effort-associated (classic) angina
(2) variant angina
(3) unstable forms of angina pectoris
causes antihypertensive causes excessive hiresutism (hair growth)
minoxidil
drug sometimes used to treat antihypertensive emergencies (after sodium nitroprusside )
diazoxide
minoxidil MOA
dilates arterioles but not veins
-opens potassium channels in smooth muscle membranes --> makes contractions less likely
hydralazine
which drug is used to treat essential hypertension
what are the primary effects of orgnaic nitrates on angina?
- dilation of arterial (resistance) and venous (capitance) vessels

-increased exercise tolerance
which antianginals are used as prophylaxis
for both typcial + variant angina :
--> nitroglycerin patches (not sublingual)
--> isosorbide dinitrate

For typical angina only
-b-blockers (propranolol)
what are b-blockers used for? in angina
B-blockers are only used for prophylaxis of typical angina
what;s the difference between nitroglycerin and isosorbide dinitrate
The 1/2 life + route of administration

nitroglycerin - sublingual half life 1-4 minutes

isosorbide dinitrate - oral dosing half life 1 hour
Irbesartan
ARB
This antihypertensives shoUdl not be given to patients with coronary artery disease
hydralazine
hydralazine MOA
-dilates arterioles, not veins
-results in decreased arterial blood pressure
-decreased peripheral vascular resistance
-causes a reflex increase in HR, SV and CO
name antihypertensives
1. minoxidil
2. sodium nitroprusside
3. diazoxide
4. hydralazine
5. ACEI
This antihypertensive is subject to polymorphic acetylation. Slow acetylaters metabolize this drugs at a different rate than fast acetylators
hydralazine
diuretics for edema associated with CHF
-all loop diuretics (Furosemide less than ethacrynic acid + Torsemide)

- Triamterene (K sparing diuretics)
-acetazolamide (carbonic anhydra. inhibitors
which antihypertensive causes a reflex increase in heart rate
hydralazine
which antihypertensive agent is given for hypertensive emergencies & severe heart failure
sodium nitroprusside
which antihypertensive agent dilates both arterial vessels and venous vessels
sodium nitroprusside
which antihypertensive is a long acting arteriolar dilatior?
diazoxide
which antihypertensive dilates arterioles but not veins?
hydralazine
minoxidil
Side effects of this antihypertensive include a reversible lupus-like syndrome
hydralazine
sildenafil
-for erectile dysfunction
-a selective inhibitor of phosphodiesterase type 5 (PDE5) results in increased cGMP, blood flow into penis
digoxin immune fab MOA + DC
DC: digoxin antidote

MOA: antibodies that bind to digoxin. The complex is then excreted through the kidneys

indications: used for life-threatening digoxin toxicity
Therapy for vasoplastic angina
-nitrates
-calcium channels blockers
what's the difference between papaverine + sildenafil
Papaverine: vasodilarot
sildenafil: erectile dysfunction drug

Papaverine: inhibitors phosphodiesterases
sildenafil: inhibitors phosphodiesterase type 5
name that carbonic anhydrase inhibitors
Acetazolamide
aspirin MOA
-aspirin irreversibly inhibits both isoforms of COX
-reduces formation of TXA2

-also interferes with chemical modulators of the kallikren system inhibits granulocyte adherence to damage vascular
why is niacid useful for treating familal dyslipidemia
it is useful b/c it reduces VLDL levels
What are saralasin and losartan?

Which is better?
They are both angiotensin II inhibitors
only Losartan has few side effects - therefore its better
(sarlasin may cause hypertension )
What do vasoconstrictors do to GFR?
-vasoconstrictors
-decrease hydrostatic pressure
-decreases GFR decreases glomerular filtration rate
what do vasodilators do to GFR in capillaries
-vasodilators decrease pressure in the afferent arterioles

-increases raise GFR
-increases raises hydrostatic pressure in capalliaries
3 major factors in peptic ulcer disease and treatment
1. infection with H. Pylori
2. Increased HCL acid secretion
3. Inadequate mucosal defenses against acid

Treatment
1. antimycobacterials
2. 2 week course of triple therapy, with a proton pump inhibitors and antibiotics
name an antidote used to reverse digoxin toxicity
digoxin immune Fab
major drug interactions of quinidine
hepatic eliminartion is increased by drygs that induce P450 (phenobarbital, phenytoin & rifampin)
What's the international normalized ratio?
INR = a more sensitive test of prothrombin time using human thromboplastia measures relative PTT

INR = (PT test/ PT normal)
Pharmacokinetics of sodium nitroprusside
given IV drugs
Effects of ACE on bradykin and on ang II
ACE --] inhibits bradykinin
ACE increases Ang II levels
ACEI MOA -->
effect on bradykinin
ACEI =
-inhibits peptidyl dipeptidase (ACE)
-prevents conversion of ang I --> ang II
-elevates levels of bradykinin
-lowers ang II levels
if it ends in pril it's a ...
an ACEI
Name ACE inhibitors
-Benazepril
-Captopril
-Enalapril
-Fosinopril
-Lisinopril
-quinapril
-ramipril
sodium nitroprusside MOA
-dialates both venous & arterial vessels
- results in reduced TPR & venous return
-releases NO = incresed intracellular cGMP, decrease Ca++ and relaxes vasulars smooth muscles
what is the advantage of the international normalized ration over the PT test?
INR is more sensitive
- it also is relative (between labs)
Why are other vasodilators agents (other than nitroglycerin and dipyridamole, etc is not sed to treat angina?
because they increase blood flow, but also increses O2 demand on heart
antimycobacterial used for both tuberculosis and leprosy
Rifampin
Tuberculosis - MDR
use combination of 3 drugs to which the organisms is susceptible
this anticoagulent is used to treat venous thrombosis (it is also used for prophylaxis purposes)
Warfarin
name the 2 types of angina and tell me the difference
-exertional angina - attacks occur during exercise

-variant rest angina --> attacks occur during rest
what drug metabolism sidenafil?
metabolized by cyt p450
which part of the nephron has NaCl Cotransporters (major transport mechanism here
NaCl cotransporters @ early distal convoluted tubule
how does nitroglycerin effects myocardial o2 consumption and delivery
myocardial O2 consumption demand is decreased bc of the arterial + venous effect of nitroglycerin
nitrals may increase mo2 delivery in variant angina
What is the only current use of verapamil
-not used for antihypertensive events
-used only for PSVT
-blocks Ca++ entry into the AV node
How do thiazide diuretics help hypertension
appear to alter arteriolar tone
2 drugs used to treat familal dyslipidemia
niacin + atorvastatin
What;s the difference between traimterene and amiloride?
place of action

triamtere - collecting tubule
amiloride - distal convoluted tubule & collecting duct

action - same action but amiloride also reduces H+ secretion

indications - triamtere is used mainly for edema associated with heart failure
which diuretics block na entry through na selective ion channels
k sparing diuretics - triamtere + amiloride
what's different about ethacrynic acid compared to other loop diuretics
-acts on the ascending loop of henle + the proximal and distal tubules
torsemide + Furosemide only work on the ascending limb of the loop of henle
What compound promotes prostaglandin synthesis ( a potent vasodilator)
kinins
how do organic nitrates (nitroglycerin) help angina
-these drugs release nitic oxide
-these drugs result in decreased O2 demand by
-reducing preload and afterload
1. how do organic nitrates reduce preload

2. How do they reduce afterload
1. They reduce preload by causing venous dilation (big effect)

2. They reduce afterload by dilating the arteries (small effect
which calcium antagonists is used for both typical + variant angina
diltiazem
name to obsolete antianginal drugs illicitly used to prolong erections
sodium nitrate + amyl nitrites
inhibits RNA synthesis by inhibiting DNA dependient RNA polymerase
Rifampin MOA
Isoniazid MOA
antimycobacterium that inhibits biosynthesis of mycolic acids which are important for mycobacterial cell wall synthesis
Rifampin
antimycobacterium that makes you piss red orange
warfarin/coumadin, coumarins
anticoagulent that blocks synthesis of factors II, VII, IX, X
The effects of prostaglandins on renin excretion
prostaglandins increses renin, thereofre COX2 inhibitors decrese renin
severe hypertension therapy
1. thiazide diuretics
2. direct vasodilators
3. B-blocker to block reflex tachy
4. guanethidine or clonidine
5. calicum entry blocker
6. calcium channel blocker
7. alpha receptor blocker
8. angiotensin receptor blocker
SE = lupus erythematosus syndrome (antihypertensive)
hydralazine
if i have chronic renal failure, what drug is safe to give me (antianemic drug) and which is not safe?
-you can give me Epoetin alpha

-do not give me deferoxamine (an iron chelator)
don't take --- with sildenafil
nitrates - b/c can cause profound hypotension if injected together
coumarin activity MOA
-antagonizes vit Ks role
-blocks synthesis of factors II, VII, IX, X
hypolipidemic should be avoided in pregnancy
statins
use of this hypolipidemic increses the risk of gout
niacin
through what factors does heparin influence factors II, IX< X, XI, XII, XIII
Antithromin III
Competitive inhibitors of HMG-CoA reductase are called ....
statins (lovastatin & simvastatin)
what type of transport occurs at the distal convoluted tubule?
sodium reabsorption is active, cl- follows passively
Name 2 thiazides
-hydrochlorothiazide
-chlorothiazide
class 1b antiarrhythmics are effective against ...

class 1b antiarrhythmics are not effective against
-think, b only works on the bottom half the heart effective only against ventricular arrhythmias, not effective against supraventricular arrhythmias
acetazolamide MOA
-carbonic anhydrase inhibitors
-acts at the proximal tubule
-inhibits proton source (Na/H exchanger) necessary for absorption of sodium bicarbonate can K+ indicrectly
which drug used in the treatment of CHF must be given by continuous infusion intravenous infusion
sodium nitroprusside
niacin MOA
decrease TG
decrease VLD esp LDL
what are the two important things about digitoxin
-hepatic metabolism
-half life is approx 1 week
is used for the treatment of normal & high renin hypertension, CHF, and may prevent glomerular damage in diabetes
ACEI
what transport processes maintain the volume and composition of body fluid in the kidney?
-glomerular filtration
-tubular transport (anionic + cationic transport systems
-passive transports
This drug is competitively antagonized by caffine, theophylline, and dipyridamol
adenosine
how do you block 2nd and 3rd degree AVN block caused as a SE of digoxin , Why?
atropine - a muscarinic antagonists, blocks parasympathetic b/c AVN block is due to excessive increase in vagal tone
to control ventricular rate in chronic atrial fibrillation
-verapamil or diltiazem
-b-blockers
-digoxin
-warfarin
(don't use in pts with evidence of myocardial damage
what is the difference between nifedipine + verapamil
does the opposite to the heart?
nifedipine reduces afterload and preload via peripheral vasodilation
diuretics used for symptoms of a cute mountain sickness
acetazolamide
how do digoxin effects the heart
- increase vagal tone (through barorreceptors mediated - central vagal stimulation

-positive inotropic effect (due to inhibition of the Na/K pump - enhance ica, increse SR release
digoxin MOA
-binds to Na/K ATPases
-increases vagal tone, therefore increase AVN ERP and decrease automaticity

esp in SAN + atria (vagal inervation is big here)
what drugs should you not give with digoxin? Why
-quinidine
-verapamil
-amidarone (antiarrhythmics)
they inhibit digoxin secretion in urine through p-glycoprotein pump competition
side effects include teratogenicity and eripheral neuropathy
-thalomide
when do you use a PT
drug tests used:

-useful in measuring Vit K
-useful in liver disease
-monitor oral anticoagulation therapy to ensure a decrease in factors II, VII, X
therapy for unstable angina
-aspirin, herparin
-anticoagulents and antiplatelet drgus
clofazimine MOA
builds to DNA and inhibits template function
competitively inhibits the enzyme dihydropteroate synthase - thus blocking folic acid synthesis
dapsone MOA
papaverine
vasodilator inhibits phosphodiesterase increases cAMP
used for cerebal + peripheral ischemia with arterial spasm
side effect in acute renal failure in pts with bilateral renal artery stenosis
ACEI
which calcium channel antagonists is used mainly for variant angina because of its antispasmotic effects
-nifedipine
therapy for chronic stable angina of effort
-long acting nitrates
-b-blockers
-calcium channel blocerks
reflex tachycardia results from hypotension caused by which drug used to treat CHF
sodium nitroprusside
what two drugs shoudl digoxin be given with if possible
-use with a diuretic and an ACEI
bile acid binding resin useful in treating pts with isolated increases in LDL
cholestyramine
name binding acid binding resins (hypolipidemics)
cholestyramine + colestipol
prodrug hypolipedimics
the statins - lovastatins and simvastatins
these drugs decrease TP and increase HDL
fibric acid derivates gemfibrixil and fenofibrate
used to treat conditions with hypertriglyceridemias _ dysbetalipoprotenima
fibric acid derivatives
indapamide
drug interactions include
-additive effects with other antihypertensives
-enhanced lithium toxicity
-may decrese response to catecholamines
diuretics only indication is antihypertension
indapamide
congenital long QT acute Rx and chronic Rx
acute - pacing, magnesium, isoproteronol

chronic - b-blockers, pacers
ventricular fibrillation acute treatment chronic treatment
acute: DC caridoversion
Lidocaine
procainamide

chronic:
ICD4
amiodarone
don't give during pregnancy
1. acei - angiotensin converting enxyme inhibitors

2. indapamide
3. ANG II receptors blockers
4. warfarin
acetazolamide
diuretics used for centripencephalic epilepsies
petit mal
-unlocalized seizures
name and MOA of diuretics that act at the early distal convolution
block NaCl cotransporters

chlorothiazide
metolazone
indapamide
which drugs bin bile acids in the intestines and cause and upregulation of LDL receptors
-chloestyramine
-colestipol
cholestyramine
-hypolipedimic increases TG and increase VLDL most
diuretic class primarily associated with edema from CHF
-potssium sparing diuretics
-loop diuretics also work
this bild acid binding resin may delay or reduce absorption of concoitant oral medication
cholestyrarmine
4 main uses of diuretics
A) nephrotic syndrome
B) CHF
C) advanced liver disease
D) hypertension
most common important complication of diuretics usage
hypokalemia
metabolic disturbances (alkalosis, acidosis
glucose intolerance
hyperuricemia
name two other drugs act at the same site as indapamide
chlorothiazide
metolazone (early distal convoluted tubule)
side effects include acute renal failure, hyperkalemia, and drug cough
ACEI
what is one of the most potent vasoconstrictors
one of the most potent vasoconstrictors in ang II
pyrazinamide MOA
inhibits ETC in mycobacterial (Not sure
name drugs used for leprosy
-dapsone
-clofazimine
-rifampin
-thalidomide
drugs used for prophylaxis for prophylaxis of peptic ulcers acute stress ulcers and GERD
cimetidine, rantidine, famotidine, and nizatidine
how do you control gastric acid secretion in patients with peptic ulcer
histamine is the major factors controlling gastric acid release

can give a histamine H2 antagonist to block gastric acid secretion
what drug is ineffective in treating familal dyslipideMia
binding resins don't work
what diseases cause a reduction in GFR
-organic diseases
-congestive heart failure
- antihypertensive
drug interactions with ACEI
-potassium sparing diuretics - b/c may cause hyperkalemia

-NSAIDs b.c they block the hypotensive effects of ACEI by blocking bradykinin - mediated vasodilation
nephrotic syndrome
excrete greater than 3-3.5 g of protein/ 24 hrs

--> hypoalbuminemia, hyperlipidemia
ACEI indications
-normal L high resin hypertension
-CHF
-may prevent glomerular damage in diabetes
cholestyramine MOA
-binds bile acids in the intestinal lumen & prevents reabsorption

-upregulation of LDL receptors
best for chronic asthma
-corticosteroids - beclamethasone, bidesonide, fluticasone
i am a patient who requires more than occasional inhalations of b2 agonists for symptoms relief
use corticosteroids aersol
cromolyn sodium MOA nedocromil sodium
- inhibiory effect on mast cell + eosinophils

-prevents release of cell mediators
Respiratory drug

-can be affected by liver disease, cigarette smoking _ diet changes

-narrow therapeutic index
theophylline
theophylline side effects
high dose
(1) convulsions
(2) cardiac arrhythmias
(3) death

lots of drug interactions
hansen's disease
use WHO short course antibacterial regiments
who treatment of for leprosy
rifampin, dapsone, clofazimine (X2 year minimum
drugs used in reversing the lepra reaction and erythema nodossum leprossum
lepra rxn - steroids, clofazimine

ENL- steroids, thalidomide, clofazimine
antimycobacterial used for H. pylori infections
bismuths + metronidazole , tetracycline, amox, clarithromycin
theophylline MOA
-inhibit cell surface receptors for adenosine
-causes direct bronchodilator and has anti-inflammatory acts
-adenosine receptor modulate - adenylate cyclase activity
belcamethasone MOA -->
which drugs share the same MOA
-budesonide + fluticasone
-inhibits phosphotipase A2, thereby inhibiting production of inflammatory cytokinesis
-inhibit lymphocytic + eosinophilic mediated airway inflammation in asthmatics not curative
prophylaxis of chronic asthmas
- 1 degranulating inhibitors cromolyn sodium _ nedocromil sodium

-leukotrienes (monteleukast _ zileuton)
ipratroprium bromide MOA
-degree of muscarinic involvemtn in bronchomotor responses varies among patients
pharmacokinetcs of ipratropium bromide
can be given in high doses b/c is poorly absorbed
a bronchodilators used for maintaines treatment of bronchospasm associated with chronic obstructive pulmonary disease
ipratropium bromide antimuscarinic bronchodilator
long term treatment - along actign B2 selective sympathetic
salmeterol
asthma & pulmonary disease medications can reduce fluid congestion b/c of alpha effects
epinephrine/adrenaline
acute asthma
-use short acting b2 agonists
-epinephrine (unwanted side effects)

-albuterol (B2 selective)
what's the difference between monteleukast and zileuton
-different pats of the pathway -
monteleukast and leukotriene inhibitor

- zileuton - leukotriene pathway inhibitor

different mechanism - monte - LTD4 receptor antagonists
zileuton - 5-lipoxygenasese inhibitor
an LTD4 receptor antagonists
monteleukast
name a

1. leukotriene inhibitors
2. leukotriene pathway inhibitors
1. monteleukast
2. zileuton
what is the advantage of azithromycin over clarithromycin
-less likely to efffect the QT interval than clarithromycin

-only taken once a day
which antimycobacterial cannot be administered pimozide
clarithromycin b/c prolongs the QT interval
What's the difference between dapsone + clofazimine?
-dapsone --> first line of defense
clofazimine --> 2nd defense

different MOA

Dapsone - competitively inhibits enzyme dihydropteroate synthetase block folic acid synthesas

clofazimine --> binds to DNA + inhibits template function
red colored urine
rifampin
clofazimine
dapsone indications
-mycobacterium leprae
-pneumocystis pneumonia in AIDS
leprosy drugs
clofazimine
rifampin
dapsone
what is an alternative to dapsone for treating resistant leprosy
clofazimine
This type of hypolipidemic drug activates lipoprotein lipase
fibric acid derivatives
gemfibrozil and fenotibrate
-antimycobacterial used as a sterilizing agent against residual intercellular organisms for tuberculosis bacilli
-pyrazinamide
side effects of isoniazid
-hepatotoxicity
-peripheral neuropathy
-may indice hemolytic anemia in pts iwth G6P dehydrogenase deficiency
ethambutol MOA
-inhibits the synthesis of arabinogalactan (a component of mycobacterial cell walls) bacteirostatic
side effects of this antimycobacterial include
hepatoxicity, peripheral neuropathy, can ay indice hemolytic anemia in pts iwth G6P dehydrogenase deficiency
isoniazid
which antimycobacterials are bacteriocidal
-cidal

-isoniazid in dividing cells
-rifampin
antimycobacterials that are bacteriostatic
-isoniazid in resting cells
-ethambutol
-dapsone
inhibits ETC in mycobacteria
pyrazinamide
different acetylators - fast acetylators and slow acetylators metabolize at different speeds
isoniazid
dopamine MOA
-reduce afterload combined with inotropic stimulation
-acts on D, a, b receptors
primary action of this vasodilator for CHF is reduced preload
torsemide
foresemide and torsemide drug interactions
NSAIDs interfere with prostaglandin synthesis
carbohydrate tolerance --> hypolipidemic, vasodilators for CHF?
niacin
hydrochlorotriazide
if worried about cyt p450 drug interactions, which statin can you give?
pravastatin
side effects of this hypolipidemic is elevations of serum aminotransferase
statins- HMG-Coa reductase inhibitors
lovastatin MOA
-specific inhibitors of HMG-Coa reductase which catalyzes conversion - HMG-CoA --> mevalonate
-mevalonates is a precursor of cholesterol
-decreases cholesterol synthesis + upregulation of LDl
-decreased oxidative stress + vascular influentia
which hypolipidemic do you use after MI?
irrespective of lipid levels?
statins
hypolipidemic - decreases cholesterol synthesis and upregulates LDl
-HMG-CoA reductase inhibitors
-statins
which of the statins is not a prodrug
pravastatin
for high ldl to treat use
isolated: cholestyramine
what's the difference bteween niacin and gemfibrozil
both decrease triglycerides, decrease LDl, and decrease VLDL
but ...

niacin - familial hyperlipidemia
gemfibrozil - hypertriglyceridemas and dyslipoproteienma
used for hypertriglyceridemia snad dysbetalipoproteinemia
gemfibrozil
gemfibrozil 0 ligand for nuclear Tx receptor PPAR alpha
PPAR - alpha is asociated with which hypolipidemic?
gemfibroxil MOA
-functions as a ligand for nuclear transcription receptors - peroxisome proliferator- activated receptor alpha (PPAR-alpha)
name fibric acid derivatives
gemfibrozil
niacin MOA
-inhibit VLDL secretions in hepatocytes
-inhibits intracellular lipase of adipose tissue via receptor mediated signaling
-decrease catabolic rate for HDL
-reduce VLDL by decreaseing influx of free fatty acids into the liver
what is the most effective hypolipidemic for increasing HDL levels
niacin
used for familal hypercholesterolemia in combination with a resin or reductase inhibitor
niacin
side effects of hypolipidemic - cutaneous vasodilation
niacin
ethambutol MOA
not well understood - but inhibits syntehsis of arabinogalacan, an essential component of mycobacterial cell walls. Enhance activity of lipophilic drugs such as rifampin
tuberculosis resistant isoniazid
rifampin and ethambutol
atrial fibrillaiton and atrial flutter can be treated by ...
ca channel blockers
b-blockers
-digoxin

b/c these drugs increase AV node ERP
Ca channel blockers MOA
slow AVN conduction
increas AVN ERP

this prevents reoccurances of AVN reentry (increase AVN ERP)
PSVT

control ventricular rate in pts with atrial tachyarrhythmias (increase AVN ERP)
vitamin K
drug used to treat hypoprothrombinemia secondary to factors limiting absorption of synthesis of vitamin K
moderate CHF treatment, decrease GFR, moderate peripheral edema vascular congestion
-digitalis/digoxin
-loop diuretics (thiazides)
-ACEI
which antihypertensives have SE - interferes with glucose metabolism & risk of diabetes and uricoic acid, increase plasma lipid levels
thiazide diuretics
antiseizure med nystagmus, gingival hyperplasia and hirustrism
phenytoin
--- receptors mediate long term potentiation when stimulated by glutamate
NMDA receptors
I have acute pain, pulmonary edema, dyspnea, and I need a preanesthetic medication
morphine
of maximal analgesia is 12-15% of morphine - used for moderate pain
codeine
fentanyl
80X more potent than morphine, used for general anesthesia
2 most important excitatory transmitters used for the transmission of pain
-glutamate
-substance p
GABA MOA
1. GABAa -= binding opens CL ion channesl caused hyperpolarization and inhibiton of neuronal firing
2. GABAb - metabotropic receptors decrease spasticity
3. GABA c -inotropic
testosterone DEA schedule
Schedule III
Antipsychotic drugs for which extrapyramidal symptoms are a side effect
-haloperidol
-risperidone
name 3 atypical antipsychotics and the main receptor they inhibit
1. rieperidone - blocks D2 and 5HT2 - schizo and dementia

2. clozapine - blocks D4 and a1 -used for resistant schizo

3. olanzapine - block 5-HT2 > DI - D4 - used for bipolar disorder
lithium MOA
-antimanic drugs
-prevents mood swings - mood stability in pts with manic depressive orders

-resembles sodium, enters the cell using Na + channels --> accumulates intracellulary b/c not pumped out by Na/K atpase
-causes partial depolarization of cells
-reduces hormone ADH induced cAMP production
-inhibis depolarization evolved NT release
-inhibits several enzymes involved in normal recycling of membrane phosophoinositol decreas PIP2, decrease IP3, decrease DAG
Benzodiazepine is used for insomina
flurazepam (a hypnotic)
drugs used to treat heroin withdrawal
methadone (agonist)
naltrexone (antagonist)
buprenophine - more potent long acting
what are the three opioid receptor types
what's the difference between them>?
u, k,s

u - most important responsible for analgesic effects + major side effects
k = responsible for some analgesic effects
which type of pain do opioids work for? WHich type of pain are they ineffective against?
opiods are for acute pain, not chronic pain
used for insomnia
-sedating
-benzodiazepine like drugs (zolpidem & zaleplon
1st line of defence for OCD
SSDRI (fluoxetine (Prozac))
if you have a family history of hypothermai, don't give this skeletal muscle relaxant
succinylcholine
diazepam (valium) is a DEA schedule --- what is it>?
scheudle IV
diazepam is an anxiolytic and an antiepileptic
primary NTS at the dorsal horn fo the spinal cord
glutamate + substance p
endogenous compounds with u >> dk selectivity
endomorphins
teat seizures associated with alcohol withdrawal with ...
short acting lorazepam
for anxiety (long term)

for anxiety (short term)
- diazepam (long term)

- alprazolam (short term)
DOC muscular disorders especially cerebral palsy and MS
diazepam
detoxification druring withdrawal from physiological dependence of barbituates, BZs, and ethanol - use -->
long acting sedative hypnotics with dose tapering

- chlordiazepoxide
-diazepam
idosyncratic blood dyscrasias, aplastic anemia
carbamazepine
anesthetic but not analgesic
thiopental sodium
local amide with higher incidence of cardiac arrhythmias
bupivacatine
DOC - tonic clonic & grand mal
phenytoin and carbamezipine
DOC partial seizure
phenytoin and carbamezipine
DOC status epilepticus q
phenytoin and diazepam
DOC myoclonic seizures
clonazepam + valproic acid
DOC absence seizures
Ethosuzimide (alternative clonazepam and valproic acid)
buprenorphine
long duration opioids
don't give with meperdine
MAOI probably shoudl avodi TCAS and antipsychotics
facilitates surgical amnesia
diazepam
name an antidepressant that inhibits cyt p450
fluxoetine
-slower onset of action than BZ
-no muscle relaxation or anticonvulsant activity
busprione --> for chronic anxiety with symptoms of irritabilitya dn hostility especially in ex-drug abusers
treatment of generalized anxiety disorders --> acts on the 5-h-t receptors
buspirone
1. epilepsy DOC
2. DOC for grand mal
3. status epilepticus
1. clonazepam
2. diazepam
3. diazepam
DOC for alcohol withdrawal
-chlordiazepoxide
-diazepam
-oxazepam
SE of THC

SE of LSD
THC - amotivational syndrome

LSD- alters users sense of time and self may change
Blocks NMDA-type glutamate receptors

Hallucinogens
-PCP - dangerous hallucinogen
-ketamine - anesthetic agent used for diagnostics & surgical procedures
endogenous compounds with d> uk selectivity
enkephalins
major side effects of morphien
decreased respiration

also: miosis, block of cough reflex, emesis, GI distress, cardiovascular effects, constipation, nausea, depressed renal function, pruitis and tolerance
a centrally acting analgesic (not chemically related to opiates
tramadol
difference between hydrocodone and tramadol?
hydrocodone is an antitussive and a narcotic analgesic, while tramadol is a centraly acting analgesic

hydrocodone shoudl eb used with caution in pts iwth head injuries

tramadol should not be used with naloxone
inhalation GA MOA
may activate GABA-A receptors _ depress spontaneous evoked activity of neurons in the brain
amide local anesthetics associated with high incidences of cardiac arrhythmias
bupivacaine
name amide local anesthetics
-remember the i before caine rule

-lidocaine
-bupivacaine
-ropivacaine
hallucingoen (angel dust, heroin) are DEA shedule
one
antipsychotic contraindicated in pts with long QT
haloeridol
tranylcypromiae pharm
fast recovery of MAO due to weak bond to enzyme
antipsychotic used to treat dementia
Risperidone
name 2 drugs under DEA schdule 1
PCP & heroin
contraindicated with SSRI fluoxetine
-drugs metabolized by cyt p450
-MAOI presumably also
antiepileptic drug - highly bound to plasma protein
phenytoin
-valproate
-carboanzapene also strongly bound
the most common most dangerous hallucinogen is
PCP
antiepileptic used only for partial seizures
GABA pentin
opioid administration
-oral administration of opioids
-huge first pass effect but the amount that can get into the blood is still sufficient
what should not be administered with meperidine?
tricyclics
lithium MOA
-alters sodium transport in nerve and muscle cells
-inhibits the recycling of neuronal membrane phosphoinositides involved in the generation of secondary messengers
antidepressants that shoudl not be given to the elderly
TCAs becasue of the severe side effects
DOC for atypical depression
phenelzine
amide local anesthetic s
-medium duration of action
-long duration of action
medium - mepivacaine
long- ropivacaine
Which inhaled GA is an incomplete anesthetic
nitrous oxide
MAC > 100%
what type of patients can thiopental sodium and methohexital not be used for ?
-contraindicated in pts with porphyria
hypnotic used for short term treatment of insomnia
zolpidem
has a long half life and a longer duration of action than morphine
methadone
what's the difference between glycine and gaba>
they are both inhibitory NTS but glycine increases Cl permeability, while GABA works on GABA A and GABA B
DEA schedule I
heroin
acute opioid poisioning
signs and symptoms
treatment
1. comatose, miosis, cyanosis
2. treatment - artificial respiration and give naloxone
what's the difference between inotropic receptrs and metabotropic receptors
inotropic: ligand gated ion channels, fast

metabotropic: g protein coupled receptors, slow
name 2 areas of the brain, regions, where opioids work
PAG and RVM - regions of the brain with lots of opioid receptors
skeletal muscle relaxant used for allievation of severe spasticity resulting from multiple sclerosis & in patients with spinal cord injuries
baclofen
durham - humphrey admendment
define what drugs are prescripiton and which are over the counter
kefauver-Harris amendment
establishes stronger drug-saftey regulation establishes clinical testing phase i-IV
1. DEA schedule III drug
2. DEA schedule IV drug
3. DEA schedule V drug
1. testosterone
2. diazepam
3. codeine
name a narcotic antagonist
-naltrexone
-naloxone
opioid MOA - where does it block?
1. presynaptic SP receptors
2. opioids can also effect calcium release in the presynaptic terminal, so that the SP NTS are not released
used for ADD in children greater than 6 years old? Other uses of this drug?
methamphetamine

other uses - short term treatment of obesity
can be used for post-traumatic stress & premenstrual dysphoric disorder + social anxiety disorder
sertraline (zoloft)
blocks 5-HT2 > D1- D4 etc. Uses?
olNZAPINE
USED FOR bipolar disorder
antagonizes action of benzodiazepines - with what result?
flumazenil - reverses the sedative effects of benzodiazepines
phenobarbital MOA
antiepileptic
-potentiates GABAergic stimulus
-inhibits Ca++ channels
-blocks AMPA receptors
used for partial seizures and absence seizures
lamotrigine
pharmacological action of morphine
1. analgesia
2. euphoria
4 uses of diazepam
1. anxiolytic
2. alcohol withdrawl
3. muscle spasm
4. seizure
what are tricyclic antidepressants used for now?
-as an alternative for enuresis and chronic pain
epinephrines effect on LA
-vasoconstriction
-decrease systemic toxicity
-decrease bleeding (local)
-increase duration of action of LA
cocaine MOA
Mechanism of Action: blocks monoamine reuptake transport into nerve terminals; Na channel blocker
cocaine indications
Indications: topical anesthesia of the upper respiratory tract (due to its combined vasoconstrictor & local anesthetic properties); use in EM as an ingredient in TAC (tetracaine, adrenaline & cocaine) prior to wound cleaning & suturing.
tell me about cocaine mixed with alcohol
Major drug Interactions: MAOI inhibitors would be expected to increase cocaine's effects & toxicity. Ethanol consumption will convert cocaine to cocaethylene, a derivative that has a half life of 3-4 hours and shares a similar pharmacology as cocaine. Most cocaine abusers consume ethanol to prolong their high. This may also increase cocaine's cardiotoxicity.
for ADD or narcolepsy
-ritalin methylphenidate
-dextroamphetamine

ADD - also methamphetamien & pemoline
drug interactions with MAO inhibitors
diuretics
NSAIDS
ACEI
pregnant and head injury, what do you not give?
morphine
THC acts on (MOA)
cannabinoid receptors CB1 and CB2
antiepileptic used for all seizure types
valproate
most important side effect that can kill is decreased respiration
opioids
NSAID used for short term management of acute pain requiring analgesia at the opiate level
ketorolac (can be used in place of opioids in patients whom nausea and vomiting is a problem
used in hospital for severe and chronic pain
fentanyl
narcotic analgesic morphine types (mu receptor agonists)
what compound is a benzodiazepine receptor antagonist
flumazenil
used for both partial seizures and tonic clonic, grand mal seizures
-phenytoin
-carbamazepine
-phenobarbital
antiepileptic given IV or rectally to stop continous seizure activity
diazepam
valproate MOA
antiepileptic enhanves GABA mediate inhibitors
-blocks Na channels
-blocks K currents
-blocks T types Ca++ channels
partial agonist @ alpha adrenergic receptors in blood vessels resulting in vasoconstriction

SE = Saint Anthony;s fire
Ergotamine MOA
possibly live threatenting hepatic failure - why its used as a 2nd line of defense for ADD
pemoline major toxicity
chemical structures unrelated to benzodiazepines and barbituates
tell me about the chemical structure of zolopidem
for which antidepressant is it especially important to avoid an abrupt withdrawal
paroxetine
name SSRIs
- fluoxetine
- sertraline
- citalopram
- paroxetine
newer designer antidepressants that target more than one NT
multiple mechanism:

- venlafaxime
-nefazodone

i.e. inhibit neuronal uptake or serotoin & norepinephrine + antagonizes central 5-Ht 2 receptors + alpha 1 adrenergic receptors
local anesthetic effects
1. what increses duration of action
2. what decreases LA effects
1. increase duration of action - hydrophobicity
2. decrease LA effects
inflammation (extracellular acidosis)
fluoxetines (norfluoxetine) metabolite is active and has an 8 day half life
pharmacokinetics of fluoxetines - length of action
This inhaled anesthetic sensitizes the heart to epi-induced arrhythmias
-rare but can induce hepatitis
halothane
which drugs cause an increase in CO2
what does it mean>
what drug can you give to treat
opioids can cause an increase in Co2
give Dantrolene to treat
may eb headed for malignant hypothermia
cause less sexual dysfunction than selective serotonin reuptake inhibitors
-nifazodone
-bupropion
MAOI MOA
- increased amine levels by interfereing wtih metabolism
- increased vesicle stores of NE+ serotonin
-irreversible + non-selective
Antidepressant used for bulima nervosa (list drug type/classification )
-fluoxetine
-SSRI
I have bipolar disorder (mania -depression)
I need drugs now.
- lithium
-carbamezepine
-valproic acid
olanzapine
manic depression drugs
0lithium
-carbamazepines
-valproic acid
fluoxetine (prozac)
#1 drug choice for depression, OCD, panic disorder
MAOI - contraindication don't give with ---
-sympathetic drugs
- SSRIs
-tyramine - rich foods
#1 treat the mania of bipolar disorder
lithium
typical antipsychotic drugs
-blocks D2> D1
-typical drus include chlorpromazine & Haloperidol
Bupropion (contraindicated in patients suffering from seizure disorders)
used in pts trying to break their addiction to smoking cigarettes
this drugs has direct depressants effects on monosynaptic reflex pathways in the spinal cord - thus it can produce skeletal relaxation without sedation
diazepam
benzodiazepines used for panic + phobia
-alprazolam
-clonazepam

buspirone can also be used - the advantages are less likely to get addicted.
increased risk of seizures if nalaxone is used to treat --- overdose
tramadol
only local anesthetic that causes vasoconstriction
cocaine
bulima - 1st line of defense
SSRI (Fluoxetine)
more lipid soluble + crosses the BBB better than morphine
Heroin
drugs that block neuromuscular transmission are either
-competitive (non-depolarizing)
-cholinomimetics (depolarizing)
migraine (mild) pharmacological strategy
1. NSAIDS
2. Acetaminophen
3. Caffeine
4. sumatriptan
Two series of alkaloids in opium?
Phenanthrenes + Benzylisoquinolines
which opioid receptor is responsible for most of the opioids analgesic effects?
u receptors (also responsible for side effects)
Lamotrigine MOA
(Carbamazepine too)
antiepileptic blocks Na channels
Phenytoin MOA
-blocks Na channels
-increases GABA-mediated
-decreases Ca++ influx
for generalized tonic-clonic (grand-mal) seizures
-phenytoin
-carbamazepine
-phenobarbital
antiseizure drug has SE

Nystagmus, gingival hyperplasia, hirsutism
Phenytoin
carbamazepine MOA
antiseizure
blocks sodium channels
antiseizure drug has SE idiosyncratic blood descrasis, aplastic anemia
carbamazepine SE
antiseizure drug blocks t-type ca++ channels
Ethosuximide MOA
for absence (petit mal) seizures
-Ethosuximide
-Lamotrigine
partial seizures
-phenytoin
-carbamazepine
-phenobarbital
-valproate
-GABApentin
-Lamotrigine
for status epilepticus
-diazepam
-lorazepam both benzodiazepam/ antiepileptic
name 2 benzodiazepines that are antiepileptic
diazepam + lorazepam
which inhaled antiesthetic may cause megaloblastic anemia?
nitrous oxide

-from prolonged exposure due to a decrease in methionine synthase activity
what's the difference between methanol and ethanol?
methanol and its metabolites are much more potent toxins than ethanol
inhibitory NTs
glycine + GABA
difference between glutamate and GABA?
glutamate is a primary excitatory NT, while GABA is the primary inhibitory NTs.
serotonin and catecholamines are ...
monoamines
SE of propofol
-marked hypotensions (greater than that causes by barbs
this drug is used primarily in patients with limited cardiac or respiratory reserves
Etomidate: short duration of action little effect on CV or respiration
a dissociative anesthetic used for shock states (hypotensive, pts at risk for bronchospasm) and children + young adults for short procedures
ketamine
benzodiazepines vs. barbituates (clinical uses)
benzodiazepines = anxiety, panic attacks, insomnia, muscle spasm

barbituates = anesthesia + seizures
buprenorphine
for the relief of moderate to severe pain
tramadol
+ analgesic effects of this drug is only potentially antagonized by naloxone
meperidine
this opioids has no antitussive effects
anti-histamine and anti-serotonergic agent

SE= sedative & weight gain
prophylaxis of severe migraine (cyproheptadine MOA)
side effects
treatment of alcohol withdrawal
1. without hepatic dx
2. with hepatic dx
w/o hepatic dx
1. diazepam + chlorodiazepoxide

w/ hepatic dx

1. oxazepam + lorazepam
name a sedative -- hypnotic & antiepileptic
phenobarbital
have selective anticonvulsant effect without marked sedation
phenobarbital & clonazepam
methylphenidate MOA
compared to Dextroamphetamine MOA
Methylphenidate: blocks reuptake of dopaminergic neurons (mild effects)

Dextroamphetamines: causes releases of monoamine and competes for reuptake with monoamine (stronger CNS effects)
What are the advantages of codeine over morphine?
-causes less respiratory depression
-less addiction
-less euphoria
-slower developement of tolerance
a longer duration of action compared to naloxone & therefore more appropriate drugs for long-term treament of addiction to heroin r other opioids
naltrexone
most widely used antitussive
dextromethorphan
what is a problem of giving morphine? (not respiratory distress)
tolerance
psychological + physical dependence
give an example of a full agonist + partial agonists + weak agonists antagonists
full - morphine
partial - oxycodone
weak - codeine
antagonist - naloxone
antagonist that blocks all receptors (u, k, d)
naloxone
a non-narcotic antitussive
dextromethorphan
only for a short term relief of muscle spasms associated with acute, painful, musculoskeletal conditions
cyclobenzaprine
dantrolene MOA
-produces skeletal muscle relaxation by interfering with relaxation by interfereing with release of calcium from the sarcoplasmic reticulum through SR calcium channel complex
succinylcholine MOA
what type of skeletal muscle relaxant
- a depolarizing NMJ blocker

MOA - 1. phase I : excites skeletal muscle by binding nicotinic receptors

2. phase II = then prevents connection by prolonging the time that receptors at the NMJ cannot respond to Ach
Skeletal muscle relaxant that's contraindicated if you have a genetic disorder of plasma pseudocholinisterases
succinylcholine
selectively depresses the cough center in the medulla
dexomethorphan
opioid that makes your pupils dilate
meperidine
a narcotic antagonist used as an antidote against resporatory depression resulting from overdosage or sensitivity to narcotics
naloxone hydrochloride
side effects include constipation, nausea, vomiting (emesis), dizziness, sedation, respiratory distress, sedation, respiratory distress , miosis, circulatory depression, shock apnea
morphine
used for morphine or fentanyl dose
naloxone: short duration of action
chronic ethanol consumption can ---
induce cyt p450. This can increase hepatoxicity of acetaminophen
side effects include blurred vision, retinal damage, and metabolic acidosis with an evaluated anion gap
methanol
what are endomorphins, b-endomorphines, enkephalins,and dynomorphins, and what's the difference?
they are all endogenous opioids

endomorphins = u> dk
b-endomorphins - u> dk
enkephalins = d> uk
dynomorphins = k> ud
in acute pain, which pain are opioids effective at blocking? Which fibers mediate this pain?
opoids block dull second pain mediated by c-fibers
it is less effective at blocking sharp, first pain mediated by alpha delta fibers
opioid antagonists used to treat respiratory depression induced by natural + synthetic narcotics like butorphanol, methadone, nalbuphine, pentazocine + propoxyphene. DOC when nature of depressant drug is unknown
naloxone
what drugs are used with oxycodone for a synergistic effects
synergistic effects
oxycodone + acetaminophen = percoset

oxycodone + aspirin = percodan
arrange codeine, morphine, oxycodone in order of potency
morphine> oxycodone > codeine
a CNS depressants
ethanol
this opioid gets into the CNS fast and then is converted to morphine, why?
heroin
b/c its more lipid solubule and crosses the BBB better
what is the one type of chronic pain that opioids have some action agonists?
-opioids are sometimes useful against cancer pain
dangerous pharmacodynamic interactions between MAO inhibitors and
SSRIs (selective serotonin reuptake inhibitors)
what's the difference between

1. neurotransmitters
2. neuromodulators
3. neurohormones
1. NTs exert effects within a single synapse (fast)

2. neuromodulators act on multiple synapses (slower onset)

3. neurohormones - released into blood, act globaly (slowest onset)
what is the primary excitatory NT in the CNS?
glutamate
considerable crossover pharmacology, specific NTs
monoamines
difference between morphine + codeine?
morphine : full agonists PAIN RELIEF?

codeine : partial agonists mild to moderate pain relief

tolerance can also develops with morphine use, much less likely with codeine
cocaine is a DEA schedule ---
Two
DEA schedule 2
chronic pain is a --- disease of the CNS
pathological
competitive antagonist of skeletal muslce nicotinic receptors
tubocurarine MOA
difference between tubocurarine & succinylcholine
tubocurarine - non-depolarizing succinylcholine - depolarizing NMJ blocker

MOA
T -competitive antagonist of nicotinic antagonists of nicotonic receptors in skeletal muscle

S - D excites skeletal muscle prevents contraction by prolonging NMJ responses time to Ach
skeletal muscle relaxant used to treat malignant hypothermia
dantrolene
MOA of analgesia?
basically the spinal cord mediates transmission of pain signals to the brain --> if you block anywhere in this pathway you get an analgesic effet
Name a DEA schedule II drug its drug class + utility
cocaine = CNS stimulant

uses: topical anesthetic for the upper respiratory tract
and int he TAC prior to wound cleaning and suturing
1. short acting opoid antagonist
2. long acting opoid antagonist
1. naloxone
2. naltrexene
succinylcholine contraindications
-genetic disorders of plasmacholinesterases

-family hisotry of malignant hypothermia
at high blood contraceptives, the rale of oxidation (of ethanol) follows ---
zero order kinetics for ethanol
Name a depolarizing neuromuscular blocking drug
tubocurarine
for mild to moderate pain. major effects on CNS and bowel
codeine
give an example of a DEA schedule III drug + its uses
testosterone (Androgen)
-abuses use this drug to increase muscle mass & strength
produces opioid only mild euphoria
methadone that's why they're used to treat heroin withdrawal
weak acting opioid agonist with actions like codeine shoudl be used with caution in patient wtih head injuries
hydrocodone
vd of this drug is similar to body water content
ethanol vd
lithium
irreversible MAO Inhibitors
selective for MAO-B
selegiline
social anxiety disorder used
-zoloft (sertraline)
-paroxetine
pharm of lithium
-narrow therapeutic window
-distribution space of lithium approximates that of total body water
phenobarbital indications
1. sedation
2. hypnosis
what do you use for
-surgical relaxation
-control of ventillation
-treatment of convulsion
NMJ blocking drugs
controlled substances act
legal foundation of gov't fight against drug abuse
establishes DEA schedules
controlled substances act
prophylaxis of severe migraine
1. b-blockers (propranolol, timolol)
2. calcium channel blockers (verapamil)
3. TCA 5-HT2 receptor antagonists
4. H1 antagonists
5. anticonvulsants (valproate)
1. skeletal muscle relaxant used during surgery to set fractures + dislocations

2. skeletal muscle relaxants used as an adjunct to facilitates ET tubation
1. d-tubocurarine
2. succinylcholine
a partial agonist of morphine, it will precipitates an abstinence syndrome in morphine abusers
pentazocine
2 mechanisms by which opioids work
1. presynaptically - decrease opioids decrease calcium influx, decrease NT release

2. postsynaptic - opioids block receptors in spinal cord, by increasing k+ conductance --> IPSP
indicated only for opiate tolerant clients
oxycodone
if you have malignant hypothermia - don't use these drugs
-succinylcholine
-inhaled general anesthetics
narcotic analgesic used for indication, maintenacne and an analgesic of postoperative care
Remifentanil
--- receptors exhibit molecular heterogeneity
GABAa receptors
most common therapeutic uses of morphine
adjuvant to anesthesia
-analgesia
opoid act centrally bind to receptors and inhibit reuptake.

May be used for chronic pain
Tramadol
Percodan vs. Vicodin
Percodan - oxycodone + aspirin
vicodin - hydrocodone + acetaminophen
a depolarizing NMJ blocking drug
succinylcholine
don't give this skeletal muscle relaxant to clients in whom histamine release is hazardous
tubocurarine
acute pain is what type of a signal?
a physiological signal
pure food and drug act
prohibits mislabeling and adulteration of food, establishes official agencies that monitor food and drug manufacturing
harrison-narcotic act
identifies what a narcotic is and establishes classes of abusable drugs
NT only acts on metabotropic receptors
monoamines
short acting (24 hours) duration reversible Inhibits MOA-a
mocolbemide
NT excites both inotropic and metabotropic
glutamate
ingestion is potentially fatal, call poision control immediately, treat with activated charcol, gastric lavage, breathing difficulties
isopropyl alcohol (rubbing alcohol)
morphine contraindications
1. head injury or craniotomy
2. pregnancy
3. hepatic failure
inotropic receptors
are ligand gated ion channels when open allow movement of specific ions down their electrochemical gradient
what's the difference between cyclobenzaprine + diazepram + baclofen?
cyclobenzaprine : for short term relief of muscle spasm

diazepram: for relief of skeletal muscle spasm caused by local pathology (inflammation, trauma, or cerebral palsy)
baclofen: for severe spaticity from MS or spinal cord injury
+ baclofen?
panic disoorder - 1st line of defense
MAOI & SSRIs (benzodiazepines might also work)
Name monoamines
histamine, norepinephrine, dopamine, and serotonin
name TCAs
-Amitryptyline
-imipramine
monoamines type of receptors
all receptors are metabotropic
nicotonic, 5-ht3, glycine, + GABA-A receptors are ...
ionotropic receptors
baclofen MOA
- a GABA- B receptor agonist. Activation of receptors increases K conductance (hyperpolarization) that produces a presynaptic inhibitory effect to reduce the release of excitatory NTs by decreasing Ca influx
Phenobarbital MOA
-potentiates GABAergic stimulates by incresing duration of GAB-gated Cl channel opening
-compared to benzodiazepines, barbs are less selective
-also suppresses exulatory NTs (glutamate) + exert non-synaptic membrane effects

-lowers margin of safety compared to BZs
morphine metabolism
hepatic conjugation to glucuronides
1. short opoid duration of action

2. long duration of action compared to morphine (full agonists)
fentanyl - short acting
methadone - long acting
A GABA-B receptors antagonists ( a skeletal muscle relexant
baclofen
sumatriptan/triptans

1. contraindications
2. side effects
1. contraindications : cardiac ischema, cerebrovascular or peripheral vascular disease

2. side effects: coronary artery spasm
benzodiazepine anxiolytic - sedatives
lorazepam
2nd line antidepressive with incidence of sometimes fatal hypertensive crisis
tranylcypromide
endogenous compound with k > ud
dynomorphin
used for tetanus
diazepam
name MAOI
phenlzine, selegiline, tranylcyromine, mocolbemide
caffine MOA and indications
MOA: adenosine receptor antagonists (increase intracellular camp)

indications: sleep retardant, migraine, apnea of prematurity
antidepressants - SE is prolong QRS
TCA amitryptyline can lead to potentially fatal cardiac arrhythmias
name hypnotics with chemical structures unrelated to BZ and barbituates
zolpidem & hydroxyzine
what's the difference between selegiline and mocolbemide?
selegiline: irreversibel MAO inhibitor selective for MAO-B (less likely to cause cheese reaction)
mocolbemide - short acting reversible inhibitors of MAO-A
Name a glycine antagonist
strychnine
what's the difference between hydrocodone and oxycodone
strength
hydocodone - weak aagonist
oxycodone - full agonist

MOA - hydrocodone = narcotic analgesic + antitussive

oxycodone - narcotic analgesic only
what's the difference between the two intravenous anesthetics
thiopental sodium - repeated intravenous doses lead to prolonged anesthetics becasue fatty tissues act as a reservoir

methohexital - duration of action 1/2 as long as thiopental
doesn't concentrate in fatty tissue
what's the difference between the two intravenous anesthetics
thiopental sodium - repeated intravenous doses lead to prolonged anesthetics becasue fatty tissues act as a reservoir

methohexital - duration of action 1/2 as long as thiopental. doesn't concentrate in fatty tissues
a potent, long acting partial agonist. can be used for heroin addiction
buprenorphine - naltrexone can also be used for heroin addiction (long acting) but its an antagonist
name classes of NTs
-peptides (neuromodulators, neuropeptide y)
-Excitatory amino acids (glutamate)
-inhibitory amino acids (GABA, glycine)
-Ach
-purines
-monoamines
NMDA receptors involved in learning & memory
glutamate
glutamate interacts with ...
AMPA, Kainate, + NMDA
NT involved in regulation of pain, asthma, psoriasis, inflammatory bowel disease into CNS, emesis, migraine, schizophrenia, depression & anxiety
substance p
where is substance p found?
in the periphery and in the CNS
treatment for methanol poisioning
1. suppress metabolism by alcohol dehydrogenase to toxic products by giving ethanol

2. hemodialysis
3. treat acidosis with bicarbonate
used for the relief of skeletal muscle spasm due to reflex spasm caused by local pathology (inflammation or trauma or upper motor neuron disorders - (cerebral palsy) tetanus
diazepam
ester local anesthetic
- esters
-tetracaine (long duration)
-2-chloroprocaine (short onset, short dur)
-procaine (short duration)
naloxone may not be effective in reversing respiratory depression by this drug
buprenorphine
cyclobenzaprine vs. baclofen
baclofen can be used in patients with spinal cord injuries, cyclobenzaprine is ineffective in patients with spinal cord injuries
more potent + addicting than codeine
oxycodone (Percodan)
food, drug, and cosmetic act
no drugs can be marketed until proven safe
name spasmolytics
-dantrolene
-baclofen
-diazepam
-cyclobenzaprine
nausea management
1. metoclopromide (dopamine antagonists)
2. diphenydramine + dimenhydrinate (H1 antagonists)
3. Ketoralac
Lithium toxicity
-narrow therapeutic windows
-SE includes polydypsia, polyuria, + diabetes insipidus
-highly toxic levels include seizure, circulartory collapse and coma
-don't be dehydrated or you can get severe toxicity
benzodiazepine used as an antiseizure med?
clonazepam
diazepam
lorazepam
benzodiazepine used for anxiety disorder or the treatment of panic disorders
-Alprazolam (an anxiolytic)
Benzodiazepine used for mild anxiety
chlordiazepoxide (an anxiolytic)
ondanseteron MOA
-antiemetic
-serotonin receptor 5-HT3 antagonists serotonin binding to 5-HT3 receptors stimulates vagal effererent nerves, stimulating vomiting
barbiutates MOA
-increase duration of GABA mediated chloride channel opening
-block glutamate (excitatory)
-block sodium channels
-exert GABA-mimetic effects
triptans (e.g. sumatriptans) MOA
-activation of the serotonin receptors 5HT1 B-D inhibits activation of the trigeminal nerve & mennigal vasodilation
-produce vasoconstriction
treat acute attack of moderate or severe migraine headache
1. ergotamines (alpha-receptor blockers)
2. MAOI (phenylzine)
3. anticonvulsants (valproate)
4. 5HT antagonists (methylsergide)
5. opioids (reserved)
6. Triptans
ethanol MOA
-CNS depressants
-ethanol effects a # of membrane proteins involved involved in neurotransmissions, includes enhancement of GABA + GABA-A receptors + inhibiton of glutamate in opening NMDA receptor channels
narcotic analgesic + an antitussive agent
hydrocodone
TCA side effects
-antimuscarinic effects = dry mouth, constipation, urinary retention, aggravation of glaucoma
-antihistamine: sedation
-NA channel blockers increase QRS arrhythmics
-alpha blockade : orthostatic hypertension
-tachycardia
-erectile dysfunction
TCA MOA
-blocks 5-HT (serotonin) and NE reuptake
-antimuscarinic
-antihistamine effects
-NA channel block
- SE orthostatic hypertension + weight gain
benzodiazepines for anxiety
-chlordiazepoxide (mild anxiety)
- alprazolam (management of anxiety disorder)
buspirone MOA
a partial agonist of the 5-HT a serotonin receptor
diazepam MOA
skeletal muscle relaxant
-acts on the limbic system, the thalamus and the hypothalamus
-induces a calming effect
LSD MOA
- agonism & antagonism (partial agonist) at 5-HT receptor subtypes
short acting benzodiazepine sedative; used as an adjunct to general anesthesia
what are it's 3 clinical indications>
1. preoperative sedation
2. amnesia
3. anxiolysis

midazolam (BZ)
anxiolytic drug + 5-HT1a partial agonist
buspirone
social phobia
SSRI
angel dust side effects
phenyclidine - most dangerous hallucinogen

1. psychosis
2. dissociation
3. disorientation
4. loss of pronociception
5. catatonic posturing
6. aggressive behavior
adverse effects of depolarizing blockade
-hyperkalemia
-increased ocular pressure
-increased intagastric pressure
-muscle pain
opioid used to treat alcohol dependence
naltrexone
drug used for severe pain and detoxification + maintenance of narcotic dependence
methadone
one tenth as potent as morphine - used in minor surgeries
meperidine
what are NMJ blocking drugs used for?
-surgical relaxation
-control of ventilation
-treatment of convulsion
monoamine MOA
-receptors are metabotropic
-receptors are G-protein coupled (when stimulated, they effect CA or K channels)
-considerable cross over pharmacology
1. what tye of receptors does morphine act on
2. where does morphine act?
1. morphine acts on all receptors subtypes
2. morphine acts
a. in the spinal cord to inhibit NT release and neuron activity
b. at opioid receptors in the brain (PAG + RVM)
c. at site of injury to inhibit nociceptors
glutamate MOA
excites inotropic receptors (ligand gated ion channels - NMDA, AMPA, & kainate) + metabotropic (G protein coupled) receptors - basically does both
SSRI side effects
-sexual dysfunction
-weight gain
Metabotropic receptors
- g protein coupled
-leads to ion channel activation
transduction of phospholipases, guanyl cyclase, and protein kinase activation
-slow response
MDMA (exstasy) MOA
CNS stimulant and hallucinogen
MOA: release of serotonin, dopamine, and NE
-distorts perception of time, facilitates intrapersonal communication
-acts as a sexual enhancer
metoclopramide MOA vs. diphenhydramine MOA
metoclopramide
-antiemetic: D2 receptor antagonist in the chemoreceptor trigger zone to reduce

diphenydramine
-antihistamine + antiserotonergic agent
-H1 receptor inhibitor, muscarinic, alpha adrenoceptor
-decrease GI hyperactivity
acute alcohol use may ...
metabolism of drugs due to decreased metabolism & decreased liver blood flow (tricyclic antidepressants, phenothiazines, sed-hypnotic drugs)
cyclobenzaprine MOA
-not clear but
-acts within the CNS and the brain stem to reduce tonic somatic motor activity
-metabotropic receptor subfamily
muscarinic Ach receptors monoamine receptors histamine
anesthesia (MOA)
1. enhance GABA effects
2. block nicotinic receptors (analgesia)
3. activate K+ channels hyperpolarize
4. block NMDA glutamate)
5. inhibit synaptic proteins
6. enhance glycine effects
drug interactions with morphine
-sedative hypnotics
-antipsychotic drugs
-MAO inhibitors
benzodiazepine hypnotics
-tenezepam (short term managemetn of insomnia)
-flurazepam (insomnia)
name an anticonvulsant used for prophylaxis of severe migraines
-valproic acid
-blocks neuronal Na channels and reduces the repetive firing of neurons
hypnotics - actions are due to suppression of activity in certain key regions of the subcortical area of the CNS
hydroxyzine
pharmacokinetics naloxone vs. naltrexone
naloxone: t1/2 1-4 hours
iv use only

naltrexone : t1/2 = 10 hours
oral use only
used for moderate pain - acts on k receptorsa and u receptors
pentazocine
benign, reversible thyroid enlargemetn is a SE
lithium b/c decrease tyrosine iodination
possible life-threatening hepatotoxicity is a side effects
MAOI
tranylcypromide
doctor treat my hunnington's disease
haloperidol
used to treat chronic generalized anxiety
buspirone
short term treatment of anxiety
also has hypnotic activity
zolpidem uses
#1 seizure med?
alternative meds?
diazepam
alternatives - lorazepam
also clonozepam
contraindicated in pts with porphyria
phenobarbital
levodopa MOA
MOA - symptoms of parkinsons related to depletion of scriatal dopamine
-dopamine is a metabolic precursor of dopamine but crosses the BBB
- carbidopa
- a dopa decarboxylase inhibitor in the periphery
-an aromatic amino acid decarbozylation inhibitor
carbidopa reduces peripheral dopamine formation
what is sinement?
-levodopa + carbidopa
- don't give to pts with history of melanoma
drug of first choice for parkinsons disease
bromocriptine (sinemet also works)
bromocriptine MOA
- D2 agonist
ergot derivative activates D2 receptors
-does not require enzymatic conversion ot an active metabolite have no potential toxic metabolites
atropine MOA
1. improves rigidity and tremor in parkinsons by decreasing Ach activation
2. preanesthetic med , decrease respiration and secretion
3. restor cardiac rate when vagal stimulation is produced by intra-abdominal surgical traction
4. decrease AV block produced by digitalis
5. overcome severe bradycardia
6. antidote for CV collapse
drug like atropine may improve tremor and rigidity of parkinsonism, no effect on bradykinesia
benzotropine
effects of selegiline on dopamine? why?
effect on amatadine of dopamine
selegilline : retards breakdown of dopamine prolongs antiparkinson effect, allows reduction of dose of L-dopa needed
MAO-B inhibitor

amantidine - potentiates dopaminergic function by influencing synthesis/ release of reuptake of dopamine
major side effect of levodopa limitating factor in therapy
dyskinesias (repetive involuntary gross movemetn of face + limbs , treat by decreaseing levodopa dose
don't use levodopa with ....
non selective MAOI like tranylcypromine b/c it can cause a severe hypertensive crisis
half life of carbamazepine is ---
12-18 hours
antiepileptic used to treat trigeminal neuralgia _ other pain syndromes
carbamazepine
the lower the MAC the ---

the higher the partition oil-gas coefficient the ----
more potent the anesthetic agent
how can you increase the rate of anesthetic induction?
- increase anesthetic concentration
-increase rate and depth of ventillation
-change pulmonary blood flow
which inhaled anesthetics can cause cardiovascular system depression?
halothane isoflurane too
what is neuroleptic malignant syndromes - treat me
results from too rapid block of dopamine in pts with highly sensitive to extrapyramidal effects

treat with dantrolene or diazepam
1. prostatic hypertropy also avoid in pts with angle-closure glaucoma
benztropine shoudl eb used in caution with patient swith ....
verapamil DC and MOA
class IV
MOA: calcium channel blockers
iron poisoning due to multiple transformations
deferoxamine is the answer
CHF with EF < 40 %
what type?
what drug?
systolic type of CHF
(weakened force of ejection)
can used digoxin
which drugs should not be given as monotherapy becasue they can increase ventricular rate?
class Ia agents including procainamide and quinidine
what is the difference between the class 1a antiarrhythmics quinidine and procainamide
quinidine : for atrial flutter and fibrillation

procainamide: for life threatening ventricular arrhythmias (is also effective against atrial arrhythmias
metroprolol DC and MOA
dc: DC II
MOA: B1 selective adrenergic receptor antagonists
what is Ih
ih is a nonselective Na/K pacemaker current
what is ferritin?
ferritin is the stored form of iron - stores iron in the liver and the heart
if a pregnant woan has a folic acid deficiency, the baby might have a ...
what drug would you use to treat this?
ans: a neural tube defect
in megaloblastic anemia
treat with IL-11
i overdosed in ferrous sulfate. what's my antidote?

what's its drug class and MOA
1. deferoxamine - what's it used for?

2. drug class - iron chelator
MOA> binds iron avidly (loosely bound iron)
it can't compete with already bound iron (biologically chelated iron
drug used for life threating ventricular arrhythmias
procainamide
i have microcytic anemia - what drug should you give me and why
give me ferrous sulfate
iron deficiency causes microcytic anemia due to the formation of small erythrocytes with insufficient hemoglobin, By giving ferrous sulfate, you give a iron source so that erythrocytes will be normal sized (with porphyrin ring)
what is the only class II and class III antiarrhythmic we've studied?
-sotalol - blocks K channels and b-adrenergic receptors
ventricular fibrillation MOA
disorganized reentry
what is the difference between

epotein a
ferrous sulfate and iron dextran
vitamin B12 and folic acid
epotein a - stimulates RBC production in normocytic anemia pts

2. ferrous sulfate - iron source in pts with microcytic anemia
3. drugs used to treat megalopblastic anemia (vitamin B12 treats pernicious anemia, folic acid should only be given to pts with pernicious anemia
what is atrial fibrillation? the MOA
disorganized atrial reentry
verapamila dn diltizim DC and MOA
DC: calcium channel blockers
mOA: blocks L-type calcium channels
permature atrial nodal or ventricular beat
don't treat don't do anything
how do you promote GI motility
wich receptors, which action
-antidopanergic (D2)
-activates serotonin receptors (5 HT3
i have a left main coronoary artery leion, what should I do ?
get bypass surgery
if you don't have serious lesions in the left main coronary artery you shoudl use drug treatments
can we use NO to treat calcified flow limiting lesions of the coronary artery?
no - cannot dilate a plaque or plaque area
which side does nitroglycerin act on
effects arterial side
major SE of nitroglycerin
hypotension
variant angina treatment
-give nitroglycerin first
-give ca++ blockers to maintain

don't give B-blockers
ventricular tachycardia without structural heart dx what is it?
DAD's triggered by increase in sympathetic tone
quinidine DC and MOA
class ia , na + channel blocker
which clas sof antiarrhythmias has anticholinergic SE b/c they block m2 receptors and block autonomic ganglia
major side effect of class 1a drugs?
what is unusual about the pharmacokinets of amiodarone?

the pharmacokinetics of adenosine?
-amiodarone has a very long half-life (10-50 days)

adenosine has a t1/2 of ten seconds
which drugs reduce mortality and sudden death after MI
b-blockers propranolol
atrial/ ventricular fibrillation
disorganized reentry
in what type of patients can deferozamine not be used to treat iron poisioning
in pts with severe renal disease (iron chelators are excreted by the kidney)
SE includes lupus erythromatosus-like syndrome after prolonged use
procainamide
propafenone DC + MOA
DC: 1c
MOA: Na channel blocker
CHF with Ef > 45%
diastolic type of CHF
weakened cells replaced by fibrous tissue)

do not use digoxin
which antiarrhythmic is administred in a stenle solution for rapid bolus injection and has a half life of less than 10 seconds
adenosine
propranolol DC+ MOA
DC: class II
MOAL b1 receptors antagonists (non-selective)
prostaglandins PGE2 and PGI2
renal vasodilator or vasoconstritors
renal vasodilators
prevents reoccurance life threating ventricular arrhythmias
amiodarone
which test measures extrinsive thromboplastin and
which test measures intrisic thromboplastin?
which test is used to monitor coumarin/ warfarin or heparin
extrinsic thromb = Pt coumarin/ warfarin
intrinsic thromb = PTT heparin
deficiency that cuases macrocytic anemia and peripheral neuropathy
vit b12 deficiency
in addition to being used for CHF what type of arrhythmia is digoxin also used for?
atrial fibrillation and atrial flutter
procainamide DC + MOA
DC: 1a
MOA = Na channel blockers
list the major drug categories. Tell me their MOA give me an example
1 - sodium channel blockers
II - b blockers
III - potassium channel blockers prolonged APD
IV - calcium channel blockers
IV - multiple MOA
another name for AVN reentry
PSVT Paroxysmal supra ventricular tachycardia
Amiodarone DC + MOA
class I - IV - multiple MOA (sometimes listed as class III
what do diuretics do to digoxins effects ? why ?
diuretics cause hypokalemia (a SE) which oeduces the activity of ATPase. which can increase digoxin's effects
what's the difference between treating a pt with atrial fibrillation or flutter with ca channels blockers or b blockers compared to digoxin?
ca channel blockers + B-blockers also have a negative iontropic effects and harmful to pts with systolic CHF )low EF < 45

digoxin has a postiive inotropic and can be used in pts with low EF
I have hypertension
what antianemic drug should not give me
Epoetin a
which drugs has SE cinchonism, torsades de pointes , syncope, antimuscarinic effects, diarrhea?
quinidine
why do we not give folic acid alone to treat pernicious anemia
b/c you need to determine if the patients also has a vit b12 deficiency. folic acid supplements mask the signs of vitamine B12 deficiency but not prevent the development of irreversible neurological disease due to vitamine b12 deficiency
fatigue, visual complaints, muscular weakness, nausea, anorexia, psychic complaints, adominal painjs, dizziness, dreams, headache, diarrhea, vomiting
signs of digitalis toxicity
1. name a glycoprotein that stimulates red blood cell production

2. normocytic anemia - erythrocytes are normal, there just aren't enough of them
how does epoetin alpha help anemia?

what type of anemia does epoetin a cure?
what drug is used mainly to treat
digoxin -induced arrhythmias
phenytoin
what type of angina is due to coronary insufficiency due to vessel occlusion
exertional (typical angina)
amiodarone drug interactions
many review careful amiodarone is a p450 substrate
am inhibits most cytochrome enzymes can elevate digoxin plasma levels
class III drugs MOA
-prolong APD by blocking K channels
-indirectly results in increase ERP b.c of increase APD
- increase in ERP abolishes reentry b/c ERP > CT
what does transferrin do?
transferrin transports iron around the body
name 1b drus what are not effective agains?
-lidocaine
-phenytoin
-tocainide
-mexiletine

No good for SVTA
which class of antiarrhythmics can have nerologic side effects (stimulation of depression and convulsions
class 1b drugs (lidocaine, phenytoin, and mexiletine)
what do you use to treat methotrexate overdose?
what is the advantage of this drugs over other available drugs?
-citrovorum factor (leukovorin)
to replinish endogenous folic acid bec methotrxate inhibits DHFR
what's the difference betwene quinidine, procainamide, and disopyramide
the side effects

quinidine --> syncope, torsades de pointes, cinchonism (tinnitus, visual disturbances, hearing loss, GI upset)

procainamide - much weaker vagal effect than quinidine - SE - systemic lupus erythematosus-like syndrome in 1/5 patients on drug for > 1 year

disopyramid - anticholinergic effects
pharmacokinetics of lidocaine
DC - 1b

administered iv or im due to extensive 1st pass hepatic metabolism and short half life adminsitered in the ICU generally
these drugs cause irreversible inhibitor of the H+ K+ and ATPase proton pump in the parietal cells
proton pump inhibits
how are subclasses of drug class 1 determined
1a, 1b, 1c are distinguished based upon the magnitude of their effect on conduction (decrease upstroke velocity - QRS) and repolarixation (APD in vitro or increased QT
antimycobacterial that induces p450
rifampin
difference between heparin _ warfin ?
herparin is a direct acting drug
warfarin is an indirect acting drug?

heparin is given iv, warfarin is oral
ang II effects on vasculature
causes vasoconstriciton, increase rise in BP
renins effect on bp, why?
end results increases bp and increase blood volume
b-agonists on plasma renin activity
increased renin activity
symptoms include paresthesias, and eweakness in peripheral nerves, progressing to spasticity, ataxia and CNS dysfunction

What drug to treat?
neurological abnormalities from vit b12 deficiency

treat with vitamin B12
adenosine drug class
endogenous nucleotide antiarrhythmics
i have normocytic anemia, what type of patient am I likely to be?
what drug would you give me?
-give me epoetin a b/c it stimulates red blood cell production

I am a
1. cancer pt - chemotherapy pts
2. AZTs treatment AIDS pt (HIV pts)
3. chronic renal failure
4. anemia b/c I didn't get a bood transfusion following surgery for whatever reason
drugs (antiarrhythmics contraindicated in asthma
sotalol and propranolol
when are b1 selective blockers contraindicated?
-sinus bradycardia
-heart block
-cardiogenic shock
-overt cardiac failure
3 antidiarrheals types
1. antimotility agents - inhibits parasympathetics activation. block ach release

2. adsorbents - absorbs intestinal toxins + microorganisms

3. agents modifying fluid + e transports decreases fluid secretion
name trmbolytics
urokinase
streptokinase
t-pa
reteplase
name an angiotensin II
antagonist
losartan - oral competitive ang II antagonist
difference between v. tachycardia after MI, v. tacycardia without structural heart dx ventricular fibrillation
v. tachycardia after MI - reentry near rim of healed infarction

v. tachycardia without structural heart dx.
DAD triggered by increase sympathetic tone

ventricular fibrillation disorganized reentry
dopamine renal vasodilator or constrictor
renal vasodilator
aspirin indications
what drug is used for

1. antiplatelet effects
2. antiinflammatory effects
3. MI prophylaxis
4. transients ischemic attacks
class I antiarrhythymics MOA used for?
4. selective block of early extrasystoles effects

1. increse ERP
2. class 1a prolongs APD by blockign K+ channels
3. selective depression of conduction in depolarized tissure - bidirectional block

used for multifocal premature beats
-sustained tachycardia
-sustained flutter and fibrillation
side effects include hyperkalemia
ACEI
potassium sparing diuretics
indapamide
this diuretic is contraindicated in pregnancy
ventricular tachycardia without structural heart dc
acute - b-blocker
verapamil
adenosine

chronic
verapamil and b-blockers
why are calcium antagonists used to treat angina?
-mechanism blockage of calcium influx via l-type channels into cardiac and vascular smooth muscle --->

produces decreased contractility, vasodilation, etc ...
chronic Rx
AV nodal reentry treatment (PSVT)
-verapamil or diltiazem
-b-blockers
-digoxin
-ablation
renal excretion of digoxin is proportional to glomerular filtration rate
pharmacokinetcs of digoxin
digoxins famous drug interactions
-potassium depleting diuretics
-calcium
-quinidine
-verapamil
-amiodarone
-erythromycin, clarithromycin
-tetracycline
how does nifedipine effect chronic (effort associated) stable angina differently than prinzmetal's variant on vasospastic angina (think MOA)
effort associated - nifedipine decreases arterial pressure at rest by dilating peripheral arterioles and decreasing afterload (TPR). THis reduces myocardial O2 consumption.

variant associated - dilates the main coronary arteries + inhibits coronary artery spasm
name prodrugs
-mercaptopurine
-fluorouracil
-thioguanine
-cytarabine

cycloplos
methotrexate
interactions with cyt p450
-metabolized by cyt p450
imatinib

p450 biactivation
-dacarbazine
-tamoxifen
actinomycin D indications
-Wilm's tumor, choriocarcinoma
Topotecan indications
advanced ovariaran
cancer + small cell lung cancer
burkitts lymphoma
cyclophosphamide
vincristine
methotrexate
treat my acute lymphocytic leukemia (ALL)
induction: Vincristine
Prednisone
doxorubicin

maintenance: methotrexate mercaptopurine
which drug is M phase specific?
paceitaxel (the spiderman poision, spiderman like Mary Jane)

Vinblastine is also m-phase specific
which drug is s phase specific
cytarabine
which drug blocks cell dividion in the late s-G2 phase of the cell cycles
etoposide
treatment for small cell lung cancer?
1. etoposide + PEB
2. Topotecan
3. Paclitaxel
what drugs are used to treat choriocarcinoma?
1. methotrexate
2. Dactinomycin or Actinomycin D
What does procarbazine interact with? (2 drug) (drug interaction)?
interacts with ethanol for anautabuse rxn

procarbazine is a MAO inhibitors and potentiates drug effects that are MAO substrates
drugs used for osteogenic sarcoma?
1. doxorubicin
2. cisplatin
3. high doses of methotrexate

(use leucovorin to balance against high doses of methotrexate
primary indications of mercaptopurine?
acute lymphocytic leukimia (ALL)
+
acue myelocytic leukemia (AML)
when is mercaptopurine contraindicated?
-contraindicated in renal failure
what compound can be found in high concentrations in smokers urine?
radioactive polonium-210
what compounds are present in second hand smoke?
dimethylnitrosamide (DMNA) and nornitrosinicotine carcinogen
which two drugs need to be activated by cyt p450 to be active (alkylating agents)
1. cyclophosphamide
2. dacarbazine
what do thiolated purine analogs do? name two? what type are they?
-inhibit glutamine dependent synthesis
-inhibit xanthine oxidase

agents - mercaptopurine
thioguanine --> guanine analogs
lethal synthesis type I mechanisms?
1. type 1: (purine) mercaptopurine + thioguanine
lethal synthesis type II mechanism?
type 2: pyrimidine
5-fluorouracil
hydroxyurea
which drugs used in testicular cancer are cell cycle specific
-bleomycin - G2
-Vincristine
-Etoposide
what is high dose-intermittent therapy called?

what diseases is it especially good for?
1. pulse therapy - for acute leukemia testicular cancer + Wilm's tumor

2. recruitment therapy
1. burkitts?
1. methotrexate + cyclophosphamide
2. ALL?
cyclophosphamide
3. AML?
danuorubicin
4. Breast cancer?
tramoxifen doxorubicin
5. ovarian?
paclitaxel + cisplatin
6. lung?
cisplatin + paclitaxel + docltaxel
which drug helps shorter neutropenia?
1. growth factor G-CSF
2. which drug(s) affect platelets more than white blood cells for toxicity
2. nitrosoureas (cisplatin affects red blood cells more)

most drugs decreased white BC > decreases platelets > decreases RB
treatment options for advanced ovarian cancer
1. cisplatin
2. topotecan
dacarbazine side effects
hepatic necrosis
methotrexate indications

1. mercaptopurine indications
methotrexate ALL, chlorocarcinoma, burkitts, lymphoma, breast cancer

2. ALL + AML
trastuzumab side effects

tamoxifen is metabolized by?
1. cardiomyopathy
2. tamoxifen is met by cyt p450
side effects st. cisplatin
-nephrotoxicity + ototoxicity
side effects of imatinib
imatinib = hepatotoxicity + edema
which drugs causes cardiomyopathy as a side effects?
trastuzumab (a monoclonal antibody to EGF receptors)

doxorubicin --> causes cardiotoxicity leading to CSF
name 4 plant alkaloids
1. Irinotecan
2. topotecan
3. paclitaxel
4. etoposide
which antitumor agents have broad spectrum aplications of solid tumors
1. doxorubicin --> solid tumors
2. paclitaxel --> breast, ovarian, bladder, head, neck, small + non-small cell lung cancer
indications for methotrexate?
ALL (acute lymphoeytic leukemia
-chlorocarcinoma
-burkitt's lymphoma
-breast cancer
what's the difference between complete response to chemotherapy, partial respons and muinor response?
1. complete --> disappearance of tumor by physiological examination + radiography studies
2. partial --> greater than 50% decrease
3. minor --> greater than 25% but less than 50% decrease
what scales are used to measure performance status in cancer pts?
what do the numbers mean?

for what numbers are chemotherapy likely to work?
ECOG and karnofsky
0 = normal
1= symptomatic
2 = out of bed
3 = in bed > 50% of the time
4 = bedridden

Pts with ECOG > 2 have very little chance of responding to therapy
name alkylating agents
1. busulfan
2. cyclophosphamide
3. dacarbazine
4. mechlorethamine
5. melphalan
6. nitrosoureas
1. thioguanine MOA?

2. thioguanine can be combined with this drug to give a synergistic effect against /// ?
1. MOA = inhibits several enzymes in the purine nucleotide pathway, decrease guanine nucleotides interfere with DNA & RNA synthesis

2. combine with cytarabine to treat adult acute leukemia
what is the primary indications for 5-fluorouracil
-colorectal cancer
-solid tumor
what is the mechanism of 5-fluorouracil
Mechanism of Action: a prodrug that undergoes a complex series of biotransformations to ribosyl and dexoxyribosyl nucleotide metabolites. One of these (FdUMP) forms a covalently bound ternary complex with thymidylate synthase and the reduced folate N5-N10-methylenetetrahydrofolate, a reaction critical for the synthesis of thymidylate. This results in an inhibition of DNA synthesis through a "thymineless death". 5-FU is also converted to FUTP, which is incorporated into RNA, where it interferes with RNA processing & mRNA translation. In addition, 5-FU is converted FdUTP, which can be incorporated into DNA, resulting in inhibition of DNA synthesis & function. Its cytotoxicity is therefore due to effects on both DNA- and RNA-mediated events.
what is methotrexat activated to
Must be bioactivated intracellularly to polyglutamate derivatives, which are selectively retained within cancer cells & have increased inhibitory effects on enzymes involved in folate metabolism, making them important determinants of the duration of action of methotrexate.
what is the mechanism of drug resistance to methotrexate
Drug Resistance: tumor cell resistance has been attributed to: 1) decreased cellular uptake (which may be related to incresed activity of P-glycoprotein), 2) decreased polyglutamate formation, 3) synthesis of increased levels of DHFR through gene amplification, and 4) altered DHFR with reduced affinity for methotrexate.
what is the similarity and the difference between thioguanine and mercaptopurine MOA
thioguanine and mercaptopurine both inhibit DNA and RNA synthesis by inhibiting DNA and RNA synthesis by inhibiting enzymes in the purine pathway

thioguanine - purine nucleotide pathway, decrease GMP

mercaptopurine - purien nucleotide interconversion, decrease AMP
the most likely mechanism of drug resistance to cytarabine?
increased levels of non-specific phosphatases
name 2 anthracycline antibiotics
1. daunorubicin
2. doxorubicin
3 MO drug resistance to alkylating agents
Drug Resistance: Cancer cells can acquire a resistance to alkylating agents by at least 3 mechanisms including: 1) increased ability to repair DNA lesions; 2) decreased permeability to the alkylating drug; 3) increased production of glutathione or increased glutathione S-transferase which catalyzes the conjugation of the drug to gluathione.
name 2 natrual products used as antitumor drugs
1. bleomycin
2. actinomycin D or dactinomycin
what is the difference between leukemia and lymphoma?
leukemia: progressive disease of blood forming organs, characterized by distorted proliferation of leukocytes in blood and bone marrow

lymphoma: any neoplastic dissorder of lymphoid tissues
What is the difference between daunorubicin and doxorubicin?
danuorubicin: Indications: acute myelocytic leukemia (AML) "only"

Drug: Doxorubicin (Indications: broad spectrum of clinical indications including solid tumors (e.g. breast cancer) and hematoligic malignancies.

side effects
daunorubicin: Side Effects: dose-limiting myelosuppression & cardiotoxicity

doxorubicin: Side Effects: myelosupression and cardiotoxicity including acute transient ECG changes (reversible) and chronic cardiomyopathy leading to the development of congestive heart failure. The development of CHF is related to the cumulative dose taken over time: ~1% at 450-500 mg/m2; ~11% at 500-600 mg/mm2; ~33% at >600 mg/mm2.
name 5 antimetabolites (cancer drugs)
1. cytarabine
2. 5-FU
3. Methotrexate
4. Thioguanine
5. Mercaptopurine
what is the difference between vinblastin and vincristine?
diff side effects: vincristine : allopecia and peripheral neuropathy

vincristine has a wider spectrum of clinical activity than vinblastine

vincristine is used for acute lymphoblastic leukemia (ALL)

vincristine is also used to treat hematological malignancies (hodgkin, non-hodkin lymphoma, multiple lymphoma)
what are the two break down products of cyclophosphamide?
1. acrolein - break down of aldophosphamide
2. an inactive metabolite (carboxyphosphamide)
what enzyme is responsible for bioactivation
--> the enzyme responsible for bioactivation is cyt p450
what drug is used against kaposis sarcoma?
(tumor in connective tissue)

vinblastine
what is a pitfall of using actinomycin D or Dactinomycin to treat wilms tumor choriocarcinoma

what is it's MOA
/1. a highly toxic drug

2. inhibits transcription resulting in cell death by intercolating into DNA
what alkylating agent is used to treat brian tumors and metastatses?

what other alkylating agent crosses the BBB?
1. nitrosoureas - penetrate BBB

Busulfan also penetrates the BBB can lead to seizures as a side effect.
5-fluorouracil MOA

what enzyme does 5-fluorouracil inhibit majro SE?
Mechanism of Action: a prodrug that undergoes a complex series of biotransformations to ribosyl and dexoxyribosyl nucleotide metabolites. One of these (FdUMP) forms a covalently bound ternary complex with thymidylate synthase and the reduced folate N5-N10-methylenetetrahydrofolate, a reaction critical for the synthesis of thymidylate. This results in an inhibition of DNA synthesis through a "thymineless death". 5-FU is also converted to FUTP, which is incorporated into RNA, where it interferes with RNA processing & mRNA translation. In addition, 5-FU is converted FdUTP, which can be incorporated into DNA, resulting in inhibition of DNA synthesis & function. Its cytotoxicity is therefore due to effects on both DNA- and RNA-mediated events.

inhibits target enzyme thymidylate synthetase

major SE : myelosuppression
Describe the chenucak structural requirements for carcinogenicity of polycyclic aromatic hydrocarbons (PA)
1. compound must be coplanar
2. methyl substitution can enhance or detract from cardiogenicity
3. a plenanthrene nucleus must be present is a tricyclic aromatic hydrocarbon occuring in coal tar
4. Requires bioactivation by cyt p450 to the epoxide form
which one of the following agents can produce hearing loss and removal impairment

a. cisplatin
b. nitrogen mustard
c. melphalan
d. mercaptopurine
e. vinblastine
a. cisplatin
which of the following cancer chemotherapeutic agents is least likely to exhibit cross resistance to other agents listed?
a. actinomycin D
b. doxorubicin
c. 6-mercaptopurine
d. vincristine
e. bleomycin
c. mercaptopurine is a small molecule. All the others listed are bulky natural products transported out of the cells, adn multidrug resistance phenyotype can confer cross resistance
which of the following anitcancer drugs inhibit DNA dependate RNA synthesis marks as a result of intercalculation between adjacent guanine cytosine base pain in double standardred DNA?
a. dactinomycin
b. flutamide
c. methotrexate
d. paclitaxel
e. vinblastine
a. dactinomycin binds tightly to double stranded DNA interferes with RNA synthesis
Which of the following classes of anticancer drugs is used in the treatmetn of this pateint is cell cycle specific and is used in MOPP + ABVD
Bleomycin and V+O (plant alkaloids) are cell cycle specific
What are the side effects of most alkylating agents
1. myleosuppression (Busulfan has the most myleosuppresion)
2. GI side effects
3. Nausea and Vomiting
Which cells are susceptibles to alkylating agents
1. alkylating agents are not cell cycle specific

2. rapidly dividing cells are most susceptible to alkylating agents
name (2) vinca alkaloids
1. Vinblastine
2. Vincristine
EWhat is the only indication of daunorubicin
AML only acute myelocytic leukemia
what type of anti tumor agent is it?

MOA?
2. anthacycline antibiotic
3. MOA inhibit Topo II intercalcates into DNA, thereby blocking DNA and RNA synthesis
What drugs inhibit the proliferation of human tumor cells that overeexpress HER2
Trastuzumab
which drug(s) inhibit Topoisomerase II?

Which drugs inhibit topo I?
1. topo II = 1. daunorubicin, 2. doxorubicin 3. etoposide

2. Top I : 1. irinotecan, 2. topotecan
Which alkylating agent is frequently given orally (most are given IV
cyclophosphamide
which drug can be used to treat ALL and AML?
mercaptopurine
what is the difference in MOA between the antimetabolite 5-Fluorouracil, methotrexate, and thioguanine
1. Mechanism of Action: a prodrug that undergoes a complex series of biotransformations to ribosyl and dexoxyribosyl nucleotide metabolites. One of these (FdUMP) forms a covalently bound ternary complex with thymidylate synthase and the reduced folate N5-N10-methylenetetrahydrofolate, a reaction critical for the synthesis of thymidylate. This results in an inhibition of DNA synthesis through a "thymineless death". 5-FU is also converted to FUTP, which is incorporated into RNA, where it interferes with RNA processing & mRNA translation. In addition, 5-FU is converted FdUTP, which can be incorporated into DNA, resulting in inhibition of DNA synthesis & function. Its cytotoxicity is therefore due to effects on both DNA- and RNA-mediated events.

2. methotrexate: Mechanism of Action: a folic acid antagonist that inhibits dihydrofolate reductase (DHFR), interfering with the synthesis of thymidylate, purine nucleotides & the amino acids serine & methionine, thereby interfering with the formation of DNA, RNA and proteins. Must be bioactivated intracellularly to polyglutamate derivatives, which are selectively retained within cancer cells & have increased inhibitory effects on enzymes involved in folate metabolism, making them important determinants of the duration of action of methotrexate.

3. Thioguanine: Mechanism of Action: inhibits several enzymes in the the purine nucleotide pathway (denovo purine biosynthesis), resulting in inhibition of nucleotide interconversion (e.g. IMP --> GMP), resulting in a decrease of intracellular guanine nucleotides, interference in DNA & RNA synthesis.
Which cells are susceptible to alkylating agents?
1. alkylating agents are not cell cycle specific
2. rapidly dividing cells are most susceptible to alkylating agents
name 2 vinca alkaloids
1. vinblastine
2. vincristine
what is the only indication of daunorubicin
AML only
what type of antitumor agent is it

MOA?
anthacycline antibiotic
3. MOA inhibit TOPO II intercalates into DNA, thereby blocking DNA and RNA synthesis
What drug inhibits the proliferation of human tumor cells that overexpress HER2
Trastuzumab
which drugs inhibit ToPO II
etoposide
daunorubicin
doxorubicin
What drugs inhibit TOPO I
irinotecan
topotecan
which alkylating agetn is frequently given orally (most are given IV)
cyclophosphamide
which drug can be used to treat AML and ALL?
Mercaptopurine
What is the difference in MOA betwene the antimetabolites 5-FU, methohexate, and thioguanine
1. 5-FU - thymine less death DNA and RNA effects + Proteins

2. Methotrexate - folic acid antagonists inhibit dihydrofolate reductase DNA, RNA and proteins

3. Thioguanine inhibit enzyme in purine nucleotide pathway , decrease guanine nucleotide
which drug has side effects leading to congestive heart failure?
What are the other side effects of these drugs?
doxorubicin

other side effecs: myelosuppression
cardiotoxicity
chronic cardiomyopathy and CHF
releated to cumulative dose over tome
Alkylating agent MOA
Mechanism of Action: Aklylating agents have a chemical structure that contain a bifunctional nitrogen mustard moiety which includes two reactive alkyl groups (hence the term aklylation). These groups can cyclize in an aqueous environment to form a highly electrophilic "immonium ion" that can covalently bind to any nucleophilic compound, including the N-7 nitrogen position on guanine, which therapeutically is a major site of action. Consequently they can produce a cross-linking of DNA when both akyl groups react with pairs of guanine residues in DNA (intrastrand or interstrand). Crosslinking of DNA ultimately results in breaks of the DNA sequence, and cell death. FWIW: these agents can also react chemically with other cellular components containing nucleophilic groups (sulfhydryl, amino, hydroxyl, carboxyl & phosphate groups). Although aklylating agents are not cell cycle specific, rapidly dividing cells are most susceptible to their effects.
What is diferent about the MOA of 5-FU compared to methotrexate?
5-fu : Mechanism of Action: a prodrug that undergoes a complex series of biotransformations to ribosyl and dexoxyribosyl nucleotide metabolites. One of these (FdUMP) forms a covalently bound ternary complex with thymidylate synthase and the reduced folate N5-N10-methylenetetrahydrofolate, a reaction critical for the synthesis of thymidylate. This results in an inhibition of DNA synthesis through a "thymineless death". 5-FU is also converted to FUTP, which is incorporated into RNA, where it interferes with RNA processing & mRNA translation. In addition, 5-FU is converted FdUTP, which can be incorporated into DNA, resulting in inhibition of DNA synthesis & function. Its cytotoxicity is therefore due to effects on both DNA- and RNA-mediated events.

Methotrexate: Mechanism of Action: a folic acid antagonist that inhibits dihydrofolate reductase (DHFR), interfering with the synthesis of thymidylate, purine nucleotides & the amino acids serine & methionine, thereby interfering with the formation of DNA, RNA and proteins. Must be bioactivated intracellularly to polyglutamate derivatives, which are selectively retained within cancer cells & have increased inhibitory effects on enzymes involved in folate metabolism, making them important determinants of the duration of action of methotrexate.
What two drugs cause mitotic arrest? What is the difference in their mechanism ?
1. vinblastin and paclitaxel

2. diffference in mechanism
paclitaxel enhances tubulin polymerization
vinblastine promotes depolymerization its blastin away all MTs this interferes with chromosome segregation
what drugs can kill cells in all stages of the cell cycle?
cisplatin
what drug can eb given to protect the body from the side effects of cyclophosphamide
mensa - the Na+ salts of methylethyl sulfonate can be given to protect the bladder from the toxic effects of cyclophospamide
allopurinol
uricosic agent (adjuvant in Rx of blood dyscrasias

MOA: reduces uric acid syntehsis from purines by inhibiting xanthine oxidase

-treatment of pts with blood dyscrasias (leukemia, lymphoma) and other maligancies with cancer chemotherapy resulting from elevated levels of serum purines
G-CSF or Filgrastin
-recombinant GCSF
- regulates the production of neutrophils

indicatoins: decreases the incidence of infection especially in patients with nonmyeloid malidancies on myelosuppressive drugs

-reduce the time to neutrophil recovery
trastuzumab
-HER2 is observed in 25-30% of primary breast cancer

-monoclonal antibody to EGF receptor

-inhibits the proliferation of human tumor cells that overexpress HER2

-indications for pts with metastatic breast cancer whose tumors overexpress the HER2 protein SE cardiomyopathy
procarbazine
-inhibits the syntehsis of DNA, RNA + protein, produces chromosomal breaks

-used in MOPP for Hodgekin's
Tamoxifen
Drug Class: Antiestrogen
Mechanism of Action: a competitive partial agonist-inhibitor of estrogen and binds to the estrogen receptor of estrogen-sensitive tumors. It has a 10 fold lower affinity for ER than does estradiol, indicating the importance of ablation of endogenous estrogen for optimal antiestrogen effect.
Indications: Exteremly effective useful for: 1) treament of both early-stage and metastatic breast cancer, 2) as a chemopreventative agent in women at high risk for breast cancer; 3) treatment of endometrial cancer.
cyclophospamide
Drug Class: Antineoplastic (Alkylating Agent)

Indications: One of the most widely used alkylating agents. A broad spectrum anticancer drug that can be given alone, but is more commonly given in combination with other drugs (e.g. CHOP, CAF or CMF). Major indications include non-Hodgkin's lymphoma, leukemias, multiple myeloma, breast & ovarian cancer.

Side Effects: Hemorrhagic cystitis occurs occasionally (& common side effects listed above). Mensa (Na salt of methylethylsulfonate) can be given to protect the bladder from the toxic effects of cyclophosp
rituximab
Drug Class: Monoclonal Antibody

Mechanism of Action: a genetically engineered chimeric murine/human monoclonal IgG1 (human Fc) antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes. The CD20 antigen is expressed on > 90% of B-cell non-Hodgkin's lymphomas. The mechanism of action includes complement-mediated lysis, antibody-dependent cellular cytotoxicity, and induction of apoptosis in the malignant lymphoma cells. This drug appears to be synergistic with chemotherapy for lymphoma.

Indications: therapy of patients with relapsed or refractory B cell non-Hodgkin's lymphoma.
cisplatin
Mechanism of Action: an inorganic metal complex that kills cells in all stages of the cell cycle, inhibits DNA synthesis, and binds DNA through the formation of interstrand cross-links similar to other alkylating agents. The primary binding site is N7 of guanine, but may form covalent interactions with other nucleotides. Platinum complexes appear to synergize with certain other anticancer drugs (cisplatin is the P in PEB drug combinations).

Indications: testicular carcinoma, breast, ovarian & bladder cancer.

SE: Nephrotoxicity and Ototoxicity
Imatinib
Drug Class: Antineoplastic "designer drug"
Mechanism of Action: an inhibitor of the tyrosine kinase domain of the Bcr-Abl oncoprotein. Imatinib prevents the phosphorylation of the kinase substrate by ATP. (see Notes below).
Indications: treatment of chronic myelogenous leukemia (CML).
Side Effects: hepatotoxicity, edema, fluid retention. tyrosine kinases associated with the platelet-derived growth factor receptor, stem cell factor and c-kit are also inhibited by Imatinib.
Bleomycin
Drug Class: Antitumor Peptide

Mechanism of Action: bleomycin is a small peptide that contains a DNA binding region & an iron-binding domain at opposite ends of the molecule. It acts by intercollating into DNA & reacts with ferous ions (Fe2+) to produce free radicals that cause DNA fragmentation, resulting in single & double strand breaks, and inhibition of DNA biosynthesis. Bleomycin is a cell-cycle specific drug that causes accumulation of cells in the G2 phase of the cell cycle.

Indications: Hodgkin's disease , Non-Hodgkin's lymphoma, testicular carcinoma

Side Effects: little myelosuppression (unusual for antineoplastics), and as a result it can be combined with other drugs that do produce myelosuppression (e.g. BACOP, ABVD, PEB). Lung toxicity

drug interactions - metabolized by cyt p450
compare drug mechanism of antimetabolites vs. anticancer antibiotics

Name them
antimetabolites: compete for utilization of normal metabolites
1. mercaptopurine - exhibits feedback inhibition of all purine biosynthesi and incroporated into DNA

2. thioguanine - inhibits enzyme sin purine synsthesis pathways
3. 5- Fu - inhibits thymidylate synthase anticancer antibiotics --> inhibit nucleic acid synthesis by blocking DNA replication of transcriptions
what is the advantages of ifosfamide over cyclophosphamide?

Who contains a phenylalanine?
ifosfamide --> hemorrhagic cystitis --> cyclosphosphamide analog

--> melphalan
The m is MOPP is for ...

toxicities you forget - secondry

1. bleomycin
2. cisplatin
3. cyclophosphamide
4. Etoposide
5. Mercaptopurine
6. Paclitaxel
m is for mechlorethamine

1. alopecia
2. ototxicity, BMS
3. Alopecia
4. alopecia
5. GI disturbances
6. peripheral neuropathy + alopecia
which alkylating agent causes profound myelosuppression in all patient ?
busulfan
differences in mechanism between irinotecan and imatinib?
irnotecan - topo I inhibitor
used for colonorectal cancer

imatinib - inhibits tyrosinekinase domain of Bcr-Abl oncoprotein
-used for CML
when do you use intermittent, high dosing?
1. drugs acting on all phases fo the cell cycle

b. tumors in which a majority of cells are actively proliferating

c. toxicity effects schedule
Two reason to give a drug contious infusion or with frequent dose administration?
1. drug is rapidly metabolized or excreted

2. drugs are phase specific + act only on one potion of the cell cycle
what chemical mediator is responsible for nausea + vomiting? why? which drugs produce this effects?
serotonin interacts with there receptors in the chemoreceptors trigger zone to produce vomiting

dacarbazine, doxorubicin, high dose cyclophosphamide nitrogen mustard, dactinomycin
what are three major toxicities of bleomycin
1. acute pneumonitis
2. pulmonary fibrosis
3. oxygen toxicity
1. drugs causing nephrotoxicity
2. drugs that cause neurotoxicity
1. cisplatin - acute renal failure

2. methotrexate - acute tubular necrosis

neurotoxicity - vinca alkaloids = peripheral neuropathy
most important side effects of

1. adriamycin
2. methotrexate
3. cyclophosphamide
1. adriamycin - cardiotoxicity
2. methotrexate - cirrhosis of the liver
3. cyclophosphamide - hemorrhagic cystitis
what side toxicities (i.e. what other cells) does chemotherapy effect?
chemotherapy effects rapidly proliferating cells, therefore chemo effects gastrointestinal mucosa hair follicles and bone marrow
what type of anticancer drug most commonly causes alopecia
rapid turn over of hair follicles

alkylating agents etoposides authracyclines (doxorubicin), dactinomycin, and the vinca alkaloids
what is another name for adriamycin
doxorubicin
define doubling time

define growth fraction

tumors most susceptible to a chemotherpay curve have a high or low growth fraction
1. doubling time - time required for a neoplasm to double its cell numbers

2. growth fraction - the fraction of cells actively proliferating

3. high
explain log kill & log kill hypothesis
chemotherapeutic agents: work by first-order kinetics - the drugs kill a constant fraction of tumor cells rather than a fixed # of cells

log kill --> the killing of a fraction f cells rather than an absolute number. in adjuvant chemotherapy --> concept remove detectable tumor (109) the body removed eradicates remaining cells
why is combination chemotherapy used?

Examples
- use drugs with different mechanisms of action and or which act on different phases of the cell cycle to delay the development of resistanct cells and to maximize cell kill
-MOPP + PEB
-choose drugs with differing non-overlapping toxicities.
1. adjvant chemotherapy
2. neoadjuvant chemotherapy
1. chemotherapy used in addition to another treatmment modality, such as surgery or radiotherapy witht he goal of eridacting micrometastasis prevent relapse

2. neoadjuvant - use chemotherapy prior to surgery or radiation - organize reduce load
Why do we use bone marrow transplant in chemotherapy patients
we can't give high enough doses of chemo therapy drugs b/c of myleosuppresion

by getting a bone marrow transplant resure allows us to give a higher previously lethal doses of chemotherapy by allowing means to repopulate the bone marrow
3 type of bone marrow transplant
allogenic - bone marrow from an HLA matching donor transfer to another pts

autologous: graft individual marrow to himself collect before remission --> give after

peripheral stem cell transplants = harvest progenitor cells by plasma-pharesis
what immunotherapy can be used to help cancer pts with infections
1. BCG
2. Interferons
3. Interleukin-2
4. Differentiating agent retinoids
5. monoclonal antibodies
6. amgiogenesis inhibitors
Describe the three steps in the progession from DNA damage to cancer
initation: DNA damage

promotion: cell with DNA damage encounters (often chemical) incourage cell multiplication + cell division)

progession = developing tumor becomes malignant
list regiments

1. hodgekins
2. non-hodgekins
3. testicular carcinoma
4. AML
5. ALL
6. CML
list regiments

1. hodgekins = ABVD or MOPP
2. non-hodgekins = CHOP and BACOP
3. testicular carcinoma = PVB
4. AML = Cytarabine
5. ALL = MOPP
6. CML
A cancer cells that is resistant to the effects of both vincristine + methotrexate probably has developed that resistance

a) changes in the properties of the target enzymes
b) decreased activity of an activing enzyme
c) increase in expression of p-glycoprotein transporter
d) Increase in proteins that are involved in DNA repair
e) increase in production of drug trapping molecules
c) p-glycoprotein transporters more foreign cells (increased expression)
what drug cause significant peripheral neuropathy
vincristine
what is a complete carcinogen
drug containing both a initiator and a prometer
which drugs are safe to be given intrathecally to treat meningial + brain cancer
1. cytarabine
2. methotrexate
what are the three classes of chemotheraoy? and give examples
class 1: non-cell cycle dependent
e.g. radiation, mechlorethamine, carmustine

class 2: phase specific
e.g. cytarabine (s phase), bleomycin (G2/M)

class 3: Kill proliferating cells prefrentially to resting cells (non0cell cycle specific ) e.g. cyclophosphamide, dactinomycin, 5- FU
What do you ave to do if you are giving an s-phase specific or G2/M phase specific drugs? Why? which ones?
cytarabine + bleomycin

b/c they affect only a small proportion of the cells population at a given time they are often given at very frequent intevals or by continous infusion
treat my breast carcinoma
cyclophosphamide , methortrexate, cyclophosphamide + doxorubicin = fluorouracil or paclitaxel . doxorubicin + paclitaxel, Tamoxifen
wHAT TWO ALKYLATING AGENTS ARE used to treat multiple myeloma ?
What is the benefit of one over the other?
multiple myeloma = mechlorethamine
melphalan

The advantage of melphalan over, mechlorethamine is that it is less carcinogen
Treat my non-Hodgkins Lymphoma
BACOP (Bleomycin, Doxorubicin, cyclophosphamide, vincristine, prednisone)

CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone)
What drug is used to treat Wilm's tumor?
Actinomycin D or Dactinomycin + Vincristine
What compounds are good for cancer prevention
1. BHA + BHT in cereal promote glutathione, which destroys dangerous electrophiles

2. Antioxidants are good - destroys free radicals that might damage DNA

3. Differentiating agents promote differeentiation
what is common of many carcinogen?
1. Many needs to be bioactivated by cyp-450 to be carcinogen in the body

most are prodrugs externally

most carcinogens are activated to reactive, electrophilic compounds
Which alkylating agent is used primarily to treat Hodgkin's disease?
What are the side effects?
1. Dacarbazine in combination with other drugs (bleomycin can also be used)

2. side effectss - hepatic necrosis (MOPP + ABVD) (Bleomycin SE = lung toxicity and some myelosuppression)
treat my hodgekins
MOPP (Mechlorethamine, vincristine, procarbazine, predisone)

ABVD (Doxorubicin, bleomycin, vinblastine, dacarbazine
DOC and secondary drugs for testicular cancer
1. Etoposide + PEB (Cisplatin + etoposide + bleomycin)

2. Bleomycin
What is the primary drugs used to treat colorectal cancer? other choices
1. irinotecan with 5-FU & leucovorin

2. Irinotecan by itself if 5-FU is ineffective
Compare drug used for
1. acute myelogenous leukemia?
2. chronic myelogenous leukemia ?
1. AML --> cytarabine + daunorubicin + mercaptopurine

2. CML --> Imatinib (treatment) Busulfan + cyclophosphamide --> a conditoning regement used before cell transplantation
What is the DOC for AML?

a secondary drug choice used?
daunorubicin + cytarabine
1. cytarabine is the DOC. It inhibits DNA polymerase, & blocks DNA synthesi. Cytarabine can be incorporated into DNA and RNA and leads to chain elongation interference

2. 2nd drug: mercaptopurine
what drugs do you give to treat chronic myelogenous leukemia (CML)
What drugs do you give as a condition regimen prior to cell transplant
1. give busulfan (conditioning regiment)with cyclophosphamide

2. imatinib = treatment
a chemopreventative agent for women at high risk for breast cancer?
tamoxifen
1. paclitaxel MOA

2. what semisynthetic analog of paclitxael is approved for a second-line therapy therapy of various cancer
MOA: a mitotic spindle poision that binds microtubules & enhances tubulin polymerization. M-phase specific

uses: breast, ocarian, bladder, head and neck , small cell lung cancer.

2. docetaxel
a competitive partial agonist - inhibitor of estrogen used for early-stage and metastatic breast cancer?
tamoxifen
what is another name for F-CSF?
What is another name for filgrastin?
Antibody that binds to the CD20 antigen formal on B lymphocytes
Rituximab
What phase is cytarabine most active against?
2. What is the mayor indications for cytarabine?
3. cytarabine MOA?
4. What's special about its pharmacokinetics?
1. cytarabine is s phase specific

2. acute myelogenous leukemia (AML)

3. competitively inhibits DNA polymerase incorporate into DNA strands leading to chain termination

4. metabolized rapidly, must be given by continous infusion
regulates the production of neutrophils
G-CSF or Filgrastin
CMF -->
COP -->
COP - cyclophosphamide, vincristine, prednisone

CMF - cyclophosphamide, methotrexate, fluorouracil
name the disease?

abnormal chromosomal translocation that results in the formation
--> CML

non-hodgkins --> rituximab
i have renal problems are macrolides, safe for me?
yes. b/c biliary excretion
name 4th generation cephalosporin + list incations
cefepime - better for gram negative bacteria (lower MIC values) active against enterobacter
can erythromycin be used systemically?
for meningitis?
1. non, not absorbed
2. no, doesn't cross the BBB
macrolides should not be used in ----
HIV or cancer patients, no immunocompromised pts - need better drugs
macroide MOA
inhibits translocation step
how are macrolides excreted?
biliary + fecal
name 3rd generation cephalosporins
-cefotaxime
-ceftriaxone
-ceftazidime
what is special about 3rd generations cephalosporins
-penetrate the CSF (useful for meningitis)
-also active against gram negative bacilli
carbapenem MOA
imipenem is structurally related to b-lactam antibiotics
can macrolides be used for bilary infections?
yes b/c high [] in bile
what are extended & broad spectrum penicills more effective against _ why?
-gram negative bugs b/c of an enhanced ability to penetrate the gram negative outer membrane
side effects of cephalosporins
hypersensitivity
hypersensitivity to penicillin
what don't use?
what do you use?
Don't use cephalosporins
use vancomycin
meningitis
penetrates CSF
3rd gen ceph and ceftriaxone
penicillins MOA
-bind and inactivating PBPs inhibits transpeptidation reaction and block cross-linking of the cell walls cell wall tysis results b/c of high internal osmotic pressure inactivation of the inhibitor of autolysins
penicillinase resistant penicillins MOA
same as pen G + resistant to B-lactamase
repository penicillins
penicillins G procaine -
IM + adequate serum levels for 12 hours
penicillin G benzathine
-IM adequate serum levels for 1 month
anaerobic infections in boxes + joints (B. fragilis)
clindamycin penetrates into bones and joints
what are the two major advantages of extended & broad specrum penicillins?
1. inactivated by B-lactamase
2. greater activity against gram negative bugs
benefits of fourth gen ceph
-greater activity against organisms (lower MIC)
- active against enterobacter
-more resistant to b-lactamase
clindamycin MOA
binds 50 s ribosomal subunits inhibits peptidyl transferase no more peptide
MRSA - vancomycin resistant I have staph or strep
streptogramins (combo drugs)
multi drug resistant gram + cocci
streptogramins
steptogramins MOA? USes?
-proteins synthesis inhibitor
-used for staph + strep
when vancomycin resistant _ MRSA multidrug resistant gram + cocci
are they oral? macrolides
yes
which drug side effects cardiac arrhythmias
erythromycin
what is a toxicity of erythromycin and clarithromycin but not azithromycin
interaction with cyp-p450
legionnaires
macrolide erythromcyin
streptomcyin
old, mainly used for tuberculosis
what type for meningitis?
bacteriostatic?
or bacteriocidal?
-cidal
list aminoglycosides
-gentamycin
-netilmycin
-amikacin
-tobramycin
-streptomycin
-vancomycin
toxicities
-nephrotoxicity
-phleboscelerotic (vein enlargement)
-ototoxicity (hearing loss)
two reasons to use vancomycin
-pts allergic to penicillin (especially for endocarditis)

- indications --> b-lactamases producing MRSA
why shoudl you be carful about administering vancomycin (CWI) with an aminoglycoside
hard on renal b/c renal excretion + similar toxicity
nephrotoxicity and ototoxicity
how is vancomycin excreted?
renal
side effect of fast administration of vancomycin
hypotension, red man syndrome from histamine release
2nd line of defense from staph infection + strep ?

3rd?
last?
1. penicillinase resistant penicillins
2. clindamycin
3. streptogramins
4. linozolid
lifethreatening bacterium
3rd generation ceph and aminoglycoside
klebsiella UTI
1st generation ceph, cephalexin and cefaclor
typhoid fever, samonella, and typhoid
ceftriaxone is DOC

ampicillin is used in the 3rd world (cheap)
main use of ceftriaxone
meningitis
meningicocal meningitis
ceftriaxone
meningicocal bacteriamia use ...
use 3rd generation ceph ceftriaxone
difference between 1st and 2nd generation cephalosporins
increased activity against gram negative bascillae, greater stability against b-lactamase inactivation
used for surgical prophylaxis
cefazolin
in general --> UTIs are resporatory infections -->
Use 1st generation cephalosoporins, cephalexin or cefazolin
UTI Klebsiella pneumoniae
cephalexin
To treat pneumonia Klebsiella pneumoniae respiratory infections
Ans. use piperacillin (mega penicillins) or better yet or better yet use a 1st generation cephalosporins - cephalexin or cefazolin
which penicillin are susceptibles to B-lactamases and which aren't
ans. all susceptibles
ceftriaxone
-drug choice for gonorrhea
-parentarlly
long half-life
Why is the use of CWSI with aminoglycosides synergistics
CWSI breaks down cell walls, making it easier for amino glycosides to penetrate even gram + cocci with staph infections
i have a serious infection life threatening
synergistics combination CWSI + aminoglycosides
what are cephalosporins ineffective against
enterococci --> use penicillins
can you use 3rd generation cephs to treat staph or strep
no, use penicillinase resistant penicillins
cefaclor
-oral
-h. influenza
aminoglycoside MOA
bind 30s ribosome inhibit protein synthesis

frozen @ elongation step
miscoding results if made
resistance to aminoglycosides MOA
acylation
adenylation
decreased drug uptake
why are aminoglycosides ineffective against anerobes? Which ones?
bacteroides fragilis closterium
need oxygen for oxygen dependant active transport process
which aminoglycoside is least resistant
amikacin
(b/c less ways to activate)
aminoglycoside toxicity
nephrotoxicity + irreversible + ototoxicity
what can aminoglycosides not be used for?
gram positive anaerobes strep pneumonia
aminoglycosides cross BBB?
no don't cross BBB

not useful for meningitis
where do aminoglycosides distribute
small vd

vd = 1/4 of body weight b/c vd is all extracellular fluid - doesn't distribute into tissues
pseudomonas aeruginosa
ceftazidime (can't use carbenicillin b/c of b-lactamase)
if B-lactamase is present use --
-use carbapenems (imipenem/ cilastatin)

- or cephalosporin
treat my staph aureus infection
use penicillinase resistant penicillins - oxacillin, cloxacillin, dicloxacillin
treat my strep infection
use a macrolide like erythromycin
cephalexin
-oral
-esp klebsiella pneumonia
-relatively stable against b-lactamase
-for gram + except enterotococci
UTI with pseudomonas with B-lactamases
cephedrine (3rd)
what is penicillin G active against?
inactive against?
active against gram negative cocci (gonococci + meningococcus and al gram + bacteria)

inactive against : gram negative bascilla (gram negative rods)
name gram + rods
staph aureus + streptococcus
what are cephalosporins not generally used for?
enterobacter (except cefepime) and staph aureus and strep
GI infection from bacteroides fragilis
ticarcillin
staph aureus is a ...
gram + cocci
I am a streptococcus, I am a ---- bacterian
gram + cocci
oral cephs
-cephalexin
-cefprozil
-cefaclor
whad do they have in common?
do aminoglycosides work in pelvic infections
no, use cefoxitin
pelvic infection likely bacteria?
antibiotic treatment?
b. fragilis anaerobic
use cefoxitin for abdominal + pelvic infections
1 have gonorrehea, give me an antibiotic
or cefoxitin possibly or amoxicillin + clauvanic acid possible

ceftriaxone
antibiotic used to treat anaerobic infections
use cefoxitin
tonsilitis
use amoxicillin if B-lactamase use clauvalinic acid
BAD side effect of clindamycin
can lead to superinfection overgrowth
excretion - clindamycin - okay in renally compromised
renal and biliary
yes, can be used in renally compromised
These drugs ending in -udine are this type of drug

one drug ends with -iridine, what type of drug is it? What about uridine?
-uridine - NRTI = lamvudine, stavudine, zidovudine

-iridine - delavirdine - NNRTI (non-nucleoside reverse transcriptatse inhibitor)

-uridine - thymidine analogs
what does probenecid do?
probenicid blocks the secretory pump which for instance can inhibit the rapid ecretion of Pen G by the renal acid secretory mechanism
MOA of nucleoside reverse transcriptase inhibitors NRTI
-competitively inhibits HIV-1 reverse transcriptase & can be incrorporated into the growing viral DNA chain to cause DNA chain termination. Each requires intracytoplasmic activation as a result of phosphorylation by cellular enzymes to the triphosphate form
differences between NRTIS and NNRTIS
NRTI - compete with nucleoside triphosphates require intracellular activation to be active

NNRTI - do not compete with nucleoside triphosphates, do not require phosph to be active

resistance to NNRTIs is generally rapid
non-nucleoside reverse transcriptase inhibitors (NNRTIS)
MOA: NNRTIs bind directly to a side on the HIV-1 RT, resulting in blockade of the RNA and DNA dependent DNA polymerase activities. The bidning side is near to but distinct from that of the NRTI
common side effects of NNRTI
- drug hypersensitivity
-seriou possibly life-threatening skin rashes. (Steven-Johnson syndrome can occur)
pyrophosphate analog MOA

name that pyrophosphate
MOA : an inorganic phosphate compound that inhibits

1. viral DNA polymerase
2. RNA polymerase
3. HIV reverse transcriptase
(competes for the pyrophosphate binding = "Fos"carnet is a pyrophosphate
which drugs inhibit Peptidyl transferase?
Clindamycin and chloramphenicol
herpes simplex (Acyclovir resistant)
Foscarnet
CMV DOC + alternative
ganciclovir + valganciclovir

alternatives: Foscarnet + vidarabine
Herpes simplex (encephalitis) DOC + alternative
acyclovir and vidarabine
which antifungals penetrate the CSF?
fluconazole
flucytosine

"flu" gets to your head man all the other antifungals such
dermatophyte infection DOC and second line of defense
griseofulvin then ketoconazole
antifungals that interact with cyt p450
griseofulvin - induces Cyt p450

ketoconazole - inhibits mammalian cyt p450
which drugs are fungicidal

which drugs are fungiostatic?
fungicidal - polyene antifungals - amphotericin B and nystatin (azoles can reach fungicidal effect at high concentrations)

fungistatic -
azoles - ketoconazole , itraconazole, fluconazole, fluycytosine, griseofulvin, caspofungin
systemic candida treatment
-amphotericin B + flucytosine

2nd line of defense - Fluconazole
what are the pharmacokinetics advantages of fluconazole?
1. oral bioavailability
2. good CSF penetration - cna be used for meningitis
3. has fewer hepatic interacting a wide therapeutic index and permitting aggressive dosing
which antifungal has antiandrogenic side effects? what are other side effects?
ketoconazole

1. hepatoxicity
2. hypersensitivity - anaphylaxis
3. antiandrogenic effect (lower testerone effect)
name 3 NNRTIS
1. nevirapine
2. delavirdine
3. efavirenz
name nucleoside reverse transcriptase inhibitors (NRTIS)
1. Lamivudine
2. Didanosine
3. AZT
4. Stavudine
5. Abacavir
AZT zidovudine
-antiretroviral , NRTI
-deoxythymidine analog for HIV 1 infections
-avoid giving with other myelosuppressive drugs
what drugs cause myelosuppresion> or bone marrow suppression?
1. myleo - common SE for NRTIs - zidovudine, didanosine, lamivudine, stavudine (ganciclovir, ribavirin)

2. BMS - vidarabine (purine analog) antiviral
imipenem
-used as a last resort not used for common infections
-rapidly metabolism in the kidneys
-highly resistant against B-lactamase
-a carbapenem used against staph aureus, streptococcus, gram - cocci + anaerobes
flucytosine
-converted to F-dUMP + FUTP, which inhibit DNA and RNA synthesis

-narrow spectrum
-for serious infections caused by candid and cryptococcus

-not good in patients without renal function
oral only
DOC for clostridium perfringens - type of bug?
peniciillin G
gram positive bacilli
DOC for penicilin resistant infections
vancomycin + ceftriacxone + rifampion + levofloxocin
DOC for streptococcus pneumonia (pneumococcus
penicillin G or Penicillin V
DOC for escherichia coli? type of bug?
ceftriaxime, ceftizoxime, ceftriaxone, ceftazidime, or cefepine

enteric gram negative baascilla
DOC for proteus mirabilis? type of bug?
ampicillin

enteric gram negative bascillae
DOC for proteus, indole positive (includes proteus vulgaris) type of bug?
cefotaxime, ceftizoxime, ceftriaxone, ceftazidime, or cefepine

enteric gram negative bascili
DOC for salmonella typhi

type of bug?
ceftriaxone or a fluoroquinolone
DOC for Haemophilus influenzae?

1. meningitis, epiglotitis, arthritis, and other infections

2. upper respiratory infections and bronchitis
1. cefotaximine or ceftriaxone
2. Trimethoprim -sulfamethoxazole
DOC for pseudomonas aeruginosas? UTI

Type of bug?
ciprofloxacin

enteric gram negative bascillae
DOC for pseudomonas pseucloniallel?

Type of bug?
ceftazidime

gram negative bascillae
ketoconazole
SE : causes torsades de pointes
MOA: impairs the synthesis of ergosterol (a component of cell membranes)
-poor penetration into the CSF
-causes fatalities
-contraindicated when terfenadine and astemizole are administered together.
-inhibits mammalian cytp450
pseudomonas aeruginosa
piperacillin or carbenicillin, ceftazidime
Taz like pseudo mona
common SE fo NRTIS
1. myelosuppresion, including neutropenia and severe anemia
2. lactic acidosis and severe hepatomegaly
give me a drug to treat herpes simplex keratitis
idoxuridine or trifluridine given in eye drops

vidarabine will also work
pseudomonas aeruginosa
carbenicillin (if no B-lact)
ceftazidime if B-lactamase
H. influenzae - no B-lactamase
yes B-lactamase
1. no B-lact = ampicillin

2. yes B-lact - Ceftriaxone or cefaclor
b. fragilis
ticarcillin
cefoxitin
what spectrum does ampicillin have not covered by pen g?
-covers e coli, proteus mirabilis, H. influenza
antacids + H2 antihistamines interferes wtih ___ 's bioavailability
ketoconazole
antifungals contraindicated in pregnancy
griseofulvin - because of teratogenicity
varicella-zoster (DOC + alternate)
acyclovir
alternate - foscarnet
differentiate between ganciclovir + valgancicloir in terms of therapeutic indications
ganciclovir : DOC for CMV (cytolomegalovirus)

valganciclovir: prodrug used primarily for CMV retinitis in immunocompromised pts and transplant pts
(retinitis - inflammation of the retina)
Interferons alpha MOA
- a polypeptide cytokine

MOA: activates ribonucleases, which degrade viral mRNA

- blocks protein synthesis by inhibiting the translation initiation complex, resulting in suppression of the new viral particle generation
Ribavirin MOA -
- a synthetic triazine riboside analog, has a two prong effect

- converted intracellularly to 5' triphosphate derivative which inhibits viral RNA polymerase

-also inhibits capping of viral mRNA at the 5' position
cryptococcal (fungal) meningitis DOC treatment and prophylaxis
-fluconazole - secondary prophylaxis

-flucytosine - used with amphotericin B for cryptococcal meningitis

why good? b/c both F's penetrate the BBB
DOC for oropharyngeal candida
fluconazole

2nd line of defense : nystatin or ketoconazole
hypokalemia and hypomagnesemai are side effects of which antifungal drugs?
amphotericin B
8 drug sthat work on anaerobes
1. clindamycin
2. piperacillin & mezlocillin (extended spectrum)
3. ticarcillin
4. metronidazole
5. penicillin G
6. cefoxitin
7. chlroamphenicol
8. imipenem
SE of flucytosine administration
-dose dependent bone marrow suppression

neutropenia, thrombocytopenia, and anemia
I have complete renal failure and I'm allergic to penicillin, give me a drug
macrolides
clofaximine
which antiviral does not require activation to block DNA polymerase actiivty?
foscarnet is a trisodium salt of phosphonoformate
It completes for pyrophosphate binding side on DNA polymerase and blocks cleavage of the pyrophosphate group rom nucleotides during DNA synthesis
I need a drug for respiratory syncytial virus
RSV - use ribavirin
Antivirals with CNS toxicity side effects
amantadine
some rimantadine, ganciclovir
antivirals with drug interactions with zidovudine (AZT)
-Acyclovir
-Valganciclovir
-Interferon alpha
avoid drug interactions with nephrotoxic drugs (Antivirals)
Foscarnet - avoid aminoglycosides amphotericin B)

Valganciclovir
Drug interactions with probenecid (antiviral)
-acyclovir, valacyclovir
-valganciclovir
Drug interactions with coricosteroids (antiviral)
vidarabine
oral second generation cephalosporin active against B-lactamase producing H. influenza, primarliy used for sinusitis otitis and lower respiratory tract infections
cefprozil
gram + penicillinase producing staphylococci infections
1. use cloxacillin or dioxacillin (oral)

2. if severe use parental admin of oxacillin or nafcillin
drugs to treat B fragilis
-ticarcillin
-cefoxitin
Drug used to treat pseudomembranous colitis?
What is pc?
Vancomycin

(pseudomembranous colitis associated with the use of clindamycin, due to an overgrowth of clostridium difficile
treat staphylococcal enterocolitis , streptococcal or enterococcal endocarditis
-use vancomycin
what enzyme converst valganciclovir to ganciclovir?
stomach esterases
antivirals
-iv administration only?
-subcutaneously only?
-topical only?
IV - foscarnet + vidarabine + ganciclovir

subcutaneously - interfereon alpha

topical - Idoxuridine + trifluridine (thymidine analogs)
I have Hep C , what durg combination would you give me?
ribavirin with interferons alpha-2beta
interferon alpha-2 b
-antiviral, antihepatitis virus B and C

-MOA - a recombinant cytokine with a host of antiviral properties
= inhibits RNA and DNA viral replication
=activates ribonucleases that degrade mRNA
=inhibits translation initiation complex

-DOC for Hep B and Hep C

SE flu like symptoms
acyclovir
-not effective against CMV
-antiviral, anti-herpes and varicella zoster virus

- an acyclic guanosine derivative that causes DNA chain termination, inhibits the viral DNA polymerases by preventing chain elongation

-CI DOC for herpes zoster (shingles) and recurrent genital herpes (HSV-2). HSV-encephalitis and varicella zoster
what's the difference between acyclovir and valacyclovir?
valacyclovir is a prodrug activated by stomach acid, acyclovir is just eh drug

acyclovir can be used for herpes encephalities and varicella zoster, they both work for herpes zoster and genital herpes
what si the difference between ganciclovir and valganciclovir
1. valganciclovir - can be used in transplant pts in addiiton to immunocompromised pts
-valganciclovir to produg of ganciclovir, oral administration results in higher plasma levels

2. different side effects (ganciclovir causes bone marrow toxicity, myelosuppresion, and CNS toxcitiy)

Valganciclovir hematologic dysfunction + renal function impariment
zidovudine
-AZT
-Antiretroviral, NRTI
-a deoxythymidine analog
-HIV infections (give with HAART)
-avoid concurrent myelosuppresion
drug of choice for enterobacter?
type of bug
imipenem

enteric gram negative bugs
Drug: Chloramphenicol
Drug Class: Antibiotic (wide spectrum & bacteriostatic)

Mechanism of Action: Binds to 50S ribosomal subunit and inhibits peptidyl transferase and blocks protein synthesis

Indications: Used outside of the United States.

Side Effects: Bone marrow suppression and idiosyncratic aplastic anemia. Gray baby syndrome. Superinfection.
sulfonamides MOA
Mechanism of Action: Competitively inhibit incorporation of para-aminobenzoic acid (PABA) into dihydropteroic acid, a precursor of folic acid. Bacteriostatic. Resistance can occur due to decreased drug uptake, altered target enzyme and escape mechanism by alteration in cell permeability.
Which drugs bind ot eh 50s ribosomal subunit
chloramphenicol
macrolides
azithromycin
clindamycin
which drugs bind to the 30 s subunit
aminoglycosides
tetracyclines
ciprofloxacin
Mechanism of Action: Interference with the activity of DNA gyrase Bactericidal. Chromosomally mediated acquired resistance.

Indications: Methicillin susceptible and methicillin resistant strains of Staph. aureaus, Streptococcus.

Side Effects: GI (nausea & vomiting), CNS (mainly at high concentrations), skin reactions, cartilage toxicity and joint swelling in children, tendonitis (not recommended in pregnant woman and infants < 8 yr old).
amantadine
Drug Class: Antiviral, anti-influenza-A virus

Mechanism of Action: Interferes with uncoating of the viral RNA of influenza A within infected host cells, thus preventing viral replication.

Clinical Indications: A Prophylactic DOC for influenza-A virus. Is not as effective if taken after viral infection.

Contraindications: Contraindicated in pregnancy due to teratogenic effects in animals.
name two non-specific anti-influenza A and B
zanamivir and oseltamivir
sulfonamides are contraindicated, when?
newborns
during the last two months of pregnancy
foscarnet
Clinical Indications:A DOC for acyclovir resistant HSV or VZV infections. Alternative drug against CMV infections.

Major drug Interactions: Nephrotoxic drugs (e.g. aminoglycoside, amphotericin B, pentamidine).
What is the HAART and why do we use it?
Highly Active Antiretroviral Therapy. A cocktail of 3 or 4 drugs used to produce a more effective reduction in plasma HIV as well as an increase in CD4 cell counts. Typical drug combinations include 2 "acceptable" NRTI's plus either an NNRTI or a PI.
Note: Not all NRTI's can be combined to produce adequate antiviral efficacy, e.g. don't combine stavudine with zidovudine (see below).
DOC for neisseia meningitis (meningococcus) type of bug?
penicillin G

gram negative cocci
DOC for neisseria gonorrhoeae (gonococcus)

what type of bug?
ceftriaxone
ciproflozacin

gram negative cocci
DOC for clostridium difficle

type of bug?
metronidazole

gram positive bacilli
DOC for klebsiella pneumoniae?

Type of bug?
cefotaximine, ceftizoxime, ceftriaxone, ceftazidime, or cefapime

enteric gram negative bascila
zanamivir
Drug Class: Antiviral, anti-influenza virus (A and B)

Mechanism of Action: Inhibits influenza neuraminidase which blocks the cellular release of the virus particle

Clinical Indications: Treatment of acute uncomplicated influenza A and B (oral inhalation).
which enzyme activates ganciclovir, valganciclovir, valacyclovir, and acyclovir (guanosine analogs)
viral thymidine kinase (TK)
what does the ending -dine indiccate? Name two drugs with this ending and indicate their MOA
-dine indicates a thymidine analog

-antiviral thymidine analgos are idoxuridine and trifluridine

Mechanism of Action: Tri-fluoro thymidine analog, activated intracellularly to tri-phosphates which then inhibit DNA polymerases.
Name two thymidine analogs (2)
idoxuridine and trifluridine
name a purine analog
vidarabine
name guanosine analogs
acyclovir, valacylovir, ganciclovir, valganciclovir
what is the difference between oseltamivir and zanamivir?

and what aer they classified as?
zanamivir is orally inhaled with no systemic absorption

Oseltamivir is administrered orally and is readily absorbed

2. they are both non-specific anti-influenza anti-virals that block neuroamidaze
rimantadine
Mechanism of Action: Interferes with uncoating of virus

Indications: Prophylactic for influenza-A virus. Preferred over amantadine due to lower CNS toxicity & increased metabolism.

Contraindications: Contraindicated in pregnancy due to teratogenic effects in animals.

-prefered over amantatdine because of less CNS toxicity
why is rimantadine prefered over amantadine as a prophylactic

and what is it a prophylactic for?
less CNS toxicity than amantadine

prophylactic for influenzae-A
which drugs can treat CMV? cytolomegalovirus


which drugs cannot be used against CMV? MOA for drugs (2)?
1. CAN - ganciclovir and valganciclovir -alternative is foscarnet

2. CANNOT - acyclovir cannot be used

3. MOA - DNA chain termination also inhibtis viral DNA polymerase
name two anti-influenza A antivirals (specific)
amantadine and rimantadine
ribavirin
Drug Class: Antiviral, anti Respiratory Syncytial Virus (RSV).

Mechanism of Action: A triazine riboside analog. Converted intracellularly to a 5'-triphosphate derivative that inhibits viral RNA polymerase and capping of viral mRNA at the 5' position.

Clinical Indications: Used as an aerosol DOC for treating infections by the respiratory syncytial virus (RSV) and as a DOC in combination with Interferon-alpha-2b for hepatitis-C virus infection.
what is the difference between idoxuridine and trifluridine?
Idoxuridine can eb incorporated into viral DNA, resulting in faulty DNA and the inability of the virus to infect tissue and reproduce

Both are topically applied but trifluridine is the DOC for the topical treatment of HSV keratitis and is used to treat cutaneous infections by acyclovir resistant HSV strains
trifluridine
-antiviral, antiherpes, HSV 1 and 2

-phosphyorylated to trifluoro thymidine analog which then inhibit DNA polymerase

-DOC for topical treatment of HSV Keratitis and cutaneous infections by acyclovir resistant strains of HSV

- topically applied in eye drops ophthalmic preps
MOA for non selective (A+B) antiinfluenza virus
zanamivir and oseltamivir are anti-influenza virus antivirals

MOA - blocks virus particle release and aggregation

-inhibits influenza neuraminidase
vidarabine
-purine analog, antiviral, antiherpes simplex virus (HSV 1 and 2

-purine analog is phosphorylated and activated intracellularly which inhibits DNA polymerase activity

SE toxicity = bone marrow toxicity
name two guanosine prodrug analogs

name two guanosine analogs
prodrugs : valacyclovir + valganciclovir

regular drug: acyclovir and ganciclovir
name two non-specific influenza anti-virals
zanamivir and oseltmavir
which antifungals can be used to treat candida fungi
nystatin & flucytosine

difference: flucytosine has diff mechanism and also inhibits cryptococcus
amphotericin B
-selective fungal effect - binds to ergosterol in the cell membrane with a resultant change in permeability allowing leakage of intracellular components

infusion related toxicity and slower onset of toxicity

IV admin
What is a pharmacokinetic advantage of administratering fluconazole compared to itraconazole or ketoconazole
it has fewer hepatic enzyme interactions and the widest therapeutic index, therefore you can aggressively dose
SE of amphotericin B
Infusion - related toxicity: fever chills, muscle spasm, vomiting, headache & hypotension

slower onset of toxicity: some degree fo renal damage If above > 4 g cucmulative
which antifungal agent has a large impact on mammalian cells? What is the drug interactions
ketoconazole

(some mammalian cell toxicity if high concentration of amphotericin B

-inhibit mammalian cyt p450
what is special about fluconazole that is not true of itraconazole or ketoconazole
-fluconazole crosses the BBB, and is therefore the azole of choice int eh treatment and secondary prophylaxis of cryptococcal meningitis

itraconazole + ketoconazole have poor CSF penetration
what are the three mechanisms of action for antifungal agents?
name examples from each type
1. binds ergosterols or sterols in the cell membrane results in a change in membrane permeability and leakage of intracellular components e.g. amphotericin B and nystaitin

2. prodrug gets converted to FUTP& FdUMP which inhibits RNA and DNA synthesis e.g. flucytosine

3. inhibit cyt-p450 dependant synthesis of ergosterol, a vital coponent of cell membranes e.g. ketoconazole, itraconazole and fluconazole
what is the difference between nystatin and amphotericin B?
roughly the same mechanism but

amphotericin B has selective fungal effects, amphotercin B binds to ergosterol, resulting in a change in membrane permeability
(leaky intracellular contents). Nystatin binds to sterols in also not effective against bacteria, protozoa, viruses, wheras amphotericin B has a broad spectrum
which drugs inhibit cyt p450?

which drugs inhibits induces cyt p450
inhibit - erythromycin, clarithromycin & ketoconazole

-induce rifampin
name 2 synergistic drug combinations and explain why they are synergistic
-CWSI + aminoglycosides --> CWSI weakens cell wall allowing easier penetratition penetration by aminoglycosides

-trimethoprim + sulfa drugs (block two steps in the synthesis of folic acid, which bacterial cells need
alterative drugs in patients with a history of severe (anaphylatic) penicillin allergry
-aztreonam
-macrolides
-vancomycin
-clindamycin
drugs contraindicated in pregnancy + children
-tetracycline (bone growth retardation, teeth stain)
-fluoroquinoles (cartilage toxicity)
-thalidomide (teratogenic birth defects)
-metronidazole (mutagenic & carcinogenic potential)
-chloramphenicol (gray baby syndrome)
-sulfonamides (contraindication in new borns + late)
- amantadine
-rimantadine
what drug is associated with gray baby syndrome
chlorampheniol (infant doesn't make enough glucoronyl transferase )
mupirocin
topical antibacterial

MOA: inhibit isolucyl tRNA synthetase

Indication : topical treatment of minor skins infections, such as impetigo, also indicated for intranasal application for elimination of methicillin-resistant s. aureus
1. whcih produgs and drug guanosine analog are used to treat HSV (herpes simplex virus) and VZV (varicella zoster virus)

2. prodrugs and drug used to treat HSV and CMV cytomegalo virus
1. acyclovir and valacyclovir
2. ganciclovir and valganciclovir
1. route of administration for ZORA
2. which has a different administration
1. ORAL MAN!
2. zanamivir has oral inhalation
nitrofurantoin
urinary antiseptic

lower urinary tract infection --> exerts antibacterial activity in urine but little systemic antibacterial action
what is the one drug that ends in -avir but is not an antiretroviral protease inhibitor?

what type of drug is it?
abacavir

it is an NRTI antiretroviral nucleoside reverse transcriptase inhibitor
antiviral that cause bone marrow toxicity?

that cause serious nephrotoxicity
-idoxuridine + trifluridine
high doses of interferon alpha
-vidarabine
-ganciclovir

foscarnet: serious nephrotoxicity
antiviral with severe teratogenic effects?
-Amantadine + rimantadine
-ganciclovir
-ribavirin
the side effects of this antiviral are flu-lke symptoms
interferon alpha used for hepatitis B and C infections
side effects if fatal dysfunction such as pancreatitis, sepsis, and multiple organ failure
valganciclovir
difference between (doxcyclin + minocyclin) and tetracycline
-doxycyclin and minoccline are more lipophilic therefore they have alonger serum 1/2 life

-they are give 1X day
-eliminated biliary route --> treat biliary infection

contrast : tetracyclin = 2X day + renal elimination
what are doxycyclin and minocyclin NOT useful for?
UTIs
what interferes with tetracyclin absorption and why?
calcium or magnesiu, salts in antacids chelate tetracylins forming an insoluble complex metal
can tetracyclins be used to treat meningitis
no, tetracyclins do not cross the BBB
can chloramphenicol be used to treat meningitis caused by H. influenzae
yes, bacteriocidal effects against H. influenzae

b/c highly sensitive at low concentration
how is chloramphenicol metabolized?
metabolized by hepatic metabolism via the enzyme glucuronyl transferase

--> puts glucuronic acid olecule @ hydroxyl group of chloramphenicol --> more polar --> more elimination
which organisms are sensitive to chloramphenicol
-clostridium perferingens

-bacteroides
-h influenzae
are fluoroquinolenes bacteriostatic or bacteriocidal
bacteriocidal inhibits DNA gyrase
what should not be taken concurrently with fluoroquinoles? why?
don't take antacids
they reduce bioavailability of fluoroquinoles
can fluoroquinoolones be used for prostatitis?

for meningitis
yes - concentrates in prostate

no-does not cross the BBB
enterobacter
-imipenem
-2nd line, 3rd gen cephs and carbenicillin
DOC for staph
1. pen G
2. penicillinase producing if MRSA
3. vancomyin or cephalosporins (1st gneration) if allergic rxn --> vancomycin or fluoroquinolones (oral, better)
can fluoroquinolones be used on anaerobes ? which ones?
no ineffective against clostridium and bacteroides
indications for fluoroquinolones?
UTI with gram-organisms
STDs gonorrhea - gonococcal organisms are b-lactamase prodrugs
fluoroquinolone side effects
-GI discomfort
-CNS toxicity
-cartilage toxicity

-long QT elongation (like erythromycin)
c. difficle
metronidazole
therapeutic indications of metronidazole
anaerobes
closterium difficle

closterium perfringes
bacteoides
h. pylori
do sulfonamides cross the BBB?
yes crosses the BBB
how are antifungial drugs delivered
lysomes are used to carry antifungal drugs directly to the infections site which reduces cellular toxicity
what fungal species are typically seen in HIV patients?
-candidiasis, cyptococcosis + histoplamosis
-opportunisitc fuckers
toxicity of protease inhibitors
1. dyspepsia, nausea, vomitting, diarrhea
2. insulin resistance, glucose intolerance, diabetes mellitus
3. hyperlipidemia: hypertriglyceridemia, hyperchlolesterolemica
4. liver toxicity
antiretroviral drug interactions
1. PI+ NNRTI - liver metabolized
ritonavir = PI-booster
2. NNRTI induces + nevirapine induce + inhibits efavirenz

(e.g. ritonavir + saquinavir)
feever + confusion : pts has dental disease with history of heart murmur no penicillin allergy, echocardiogram shows vegitatins on the calcified aortic arch
ampicillin and gentamicin
symptoms: urinary frequency, dysurea, low grade fever, pyuria and bacilli
sulfamethoxazole + trimethoprim
fever , pumlent wound drainage with gram + cocci 5 days post-colostomy for diverticular abscess
ampicillin and gentamicin
symptoms and severe headache rigid neck, sensitivity to bright lights
symptoms of meningitis

use 3rd gen ceph ceftrioxone
most suitable for a pts with renal insufficiency
1. chlortetracycline
b. demedocycline
c. doxycycline
d. methacycline
e. oxytetracycline
doxycyline
dentist is going to remove toooth in patient with abnormal heart valve
ampicillin
prostatic infection
trimethoprim
which two bacteria cannot be treated by aminoglycosides and why?
B. fragilis and clostridium their anaerobic
didanosine
-deoxyadenosine analog
antiretroviral, NRTI

-fatal & non-fatal pancreatitis
-myelosuppression, neutropenia, severe anemia, lactic acidosis + severe hepatomegaly
which antivirals when activated inhibit DNA polymerases?
inhibit RNA polymerase?
inhibit both
DNA polymerase:
idoxuridine (thymidine analog)
vidarabine (purine analog)

RNA polymerase
ribavirin - riboside analog
foscarnet (HIV reverse transcriptast)
ganciclovir and valganciclovir

MOA: indications, major drug interactions
MOA: activated by tK, inhibits viral DNA polymerases. Produces DNA chain termination

CI: DOC for CMV retinitis in immuno-compromised patients

SE: can results in organfailure, sepsis, etc. fatal dysfunction
which antiviral inhibits RNA polymerase instead of DNA polymerase?
ribavirin acts on RNA polymerase insterads of DNA polymerase
which antivriral acts on DNA polymerases, RNA polymerase and HIV reverse transcriptase
foscarnet
metronidazole
-ANTIPROTOZOAL and antibacterial against anaerobes

-selective for bacteria and protozoa
-forms nitro anion radical and hydroxylazine which binds with DNA causing single strand breaks. Bacteriocidal

-contraindicated in pregnacy
-SE: GI discomfort, metallic taste, disulfiram like reactions
pharmacokinetics of sulfonamides
metabolism of hepatic acetylation
side effects of sulfonamides ]

name sulfonamides and the drug they are administered with
SE: stevens johnson syndrome, hemolytic anemia, kernicterus (a toxic encephalopathy in infants)), contraindicated in new borns and last two months

2. sulisoxazole, sulfamethoxazole, and pregnancy, trimethoprim, administered with
sulfonamides MOA
use of sulfamethoxazole clinically is in combination with trimethoprim in a 5:1 ratio, known as cotrimoxazole, trimethoprim is an inhibitor of dihydrofolate reductase and provides a sequneital blockade of synthesis of tetrahydrofolate
aminoglycosides-how excreted?
renal
salmonella typh in bile duct
ans. ampicillin, why? ampicillin achieves high [] in bile ducts
enterobacter UTI
carbenicillin (not covered by ampicillin + amoxicilllin)
when would you use carbenicillin instead of amoxicillin & ampicillin
pseudomona & enterbacter UTI
therapeutic indications for aminoglycosides
-UTI pseudomonas
-prophylaxis for endocarditis
-primarily for gram negative bugs
-when combined with CWI - uses --> staph, strep, viridans, MRSA, enterococcal
i'm going in for surgery - need surgery prophylaxysis but allergic to penicillin
vancomycin
gram + bascilla (rods)
-anthrax
-castilium perfringens
-castilium difficle
pharmacokinetics of aminoglycosides (3 things)
1. administered only in a hospital with therapeutic monitoring must monitor co
2. minimal metabolism
3. extracellular distribution
vancomycin MOA
vancomycin inhibits peptidoglycan synthase thereby inhibiting cell wall cross linking

peptidoglycan synthase - brings peptidoglycans outside membrane cross-linking
pharmacokinetics of vancomycin
-parental
-can be given orally to treat for local GI tract infection because not absorbed
when is oral vancomycin used?
i have a growth of c difficule in my GI tract
not useful against gram negative
what is vancomycin not useful for
MRSA
-use penicilllin resistant penicillins

cephalosporins don't work
macrolide toxicity
GI upset esolate salt of erythromycin causes cholestatic jaundice
-cyp450 inhibit (not azithromycin)
advantage of clarithromycin over erythromycin
-respiratory infections more effective against gram negative

-h. influenza
-MAC mycobacterium avium complex in HIV patients
erythromycin macrolide indications
1. upper respiratory with staph and strep
2. legionnaires
3. alternative to penicillin hypersensitive pts
pen V
first oral penicillin
which penicillin is often combined with clavulanate acid and why?
ticarcillin - we combine for inhibition of b-lactamases
plasma mediated alteration of binding site
macrolide mech of resistance
erythromycin
drug used in colonrectal surgery b/c no absorption
clindamycin crossess BBB?
no, no, meningitis
alternative prophylaxis for endocarditis
clindamycin
bacterial endocarditisi use
1st generation ceph like cephalexin or cefazolin
or penicillin G benzatnin
i have siphilus, give me an antibiotic
ceftriaxone
antibiotic for abdominal surgical operations
use cefoxitan
gram positive rod
anthrax is a ...
gram + rod
clostilum difficile is a ...
gram negative cocci
gonococcus & meningococcus
castilium perfringens is a ...
gram + rod
what type of bacteria is meningococcus?
gam negative cocci?
gram negative cocci
what type of bacteria gonococci
list extended and broad spectrum penicillins
-ampicillin
-amoxicillin
-carbenicillin
-ticarcillin
-piperacillin
PEN G is adminstered
IV or IM (parental)
what are three isoxazole penicillins?
oxacillin, cloxacillin, dicloxacillin
penicillinase resistant penicillins name them
methicillin (not used clinically)
isoxazole penicillins
naficillin
h. influenzae respiratory infections
ampicillin or amoxicillin
2 carbapenems
imipenem
cilastatin
name a monobactam and what is it used for?
aztreonam active against gram negative rods
first generation cephalosporins
cephalexin
cefazolin
second generation cephalosporins
cefoxitin
cefaclor
cefprozil
cefuroxime axetil
infection with castilium perfringens
pen G
UTI with pseudomonas
carbenicillin or cephedrine (3rd genration), can't use amoxicillin or ampicillin
infections with pseudomonas no b-lactamases
carbenicillin
pneumonococcal pneumonia use
cephalexin
difference between penicilins and cephalosporins
penicilins are effective against enterococci, cephalosporins don't work on enterococci
h. influenza with b-lactamaze resistnat what is no longer useful and what do you use?
cefaclor
proteus mirabilis UTI
ampicillin and amoxicillin
e coli UTi
ampicillin or amoxicillin (extended spectrum)
reflex tachycardia
non selective alpha 1 and alpha 2 blockers

b2 agonists
phenylephrine
a1 agonist
hypertension treatment
ace inhibitors
b blockers
diuretics
clonidine
a2 agonist (antihypertensive)
albuterol
B2 agonist
Terbutyline
B2 agonist
dobutamine
b1 agonist
doxazosin
long activng a1 selective blocker
ends in sin
alpha 1 selective blocker
treatment of pheochromocytoma
phentolamine or pehnoxybenzamine (nonselective irreversible alpha 1 and alpha2 blocker)
methoxamine
alpha one agonist
Reserpine
antihypertensive
SE+ suicidal tendencies
inhibitor of catecholamines vesicular uptake transporter
resulting in depletion of neuronal catecholamine stores, decreased CO, decreased PVR
imipramine
antidepressant
-tricyclic amine
blocks repuptake of norepinephrine serotonin
edrophonium
-anticholinesterase
-short and rapid-acting
diagnosis of myasthemia gravis & whenever need curare antagonist for cholinergic reactions
methyldopa
-antihypertensive
-alpha 2 agonist prodrug
-lowers arterial pressure
decrease false neurotransmission
phenoxybenzamine
alpha non-selective antagonist
irreversible binder, binder, binds convalently vasodilator
for pheochromocytoma for hypertension
propranolol
nonselective beta blocker
block b1 & B2 competitive
for hypertension arrhytmias angina prevents sudden death + reinfarction & migraine
guanethidine
antihypertensive, adrenergic neuron-blocking agents
4 - mech -> tyramine-like, reserpine-like cocaine-like, betrylium-like
alpha blockers
terazosin
tamsulosin
doxazosin
prazosin
yohimibine
alpha 2 blockers
increased sympathetic outflow
increased bp, no clinical role
antihypertensive
reserpine, bretylium, guanethidine, methyldopa, prazosin, clonidine
prazosin
alpha 1 bocker antihypertensive
not accompanied by reflextachy cardia
phenylephrine
synthetic alpha, agonist, vasal constriction, nasal decongestion, pressor agent, maintain bp
bretylium
-antihypertensive & antiarrhythmics
-interferes with release of catecholamines for attempted resentation from vfib
Terbutaline
b2 adrenergic agonist prevent bronchospasm in pts with asthma
SC administration
doxazosin
alpha selective blocker
once a day dosing
succinylcholine
-nicotinic cholinomimetic and NMJ blocker (depolarizing type)
uses: adjunct to general anesthesia
contraind: genetic disorders of pseudocholinesterase from history of malignant hyperthermia
dopamine
low dose d, intermed b, increases HR, high d1 = vasoconstriction including renal

for: shock due to ML, septic, surgery, with MAOI lasts longer
phentolamine
non-selective alpha antagonist blocks alpha 1 &2 - leads to tachycardia IM or IV
physostgmine
-reversible cholisterase inhibitor
-to reverse effects upon CNS caused by toxic doses of drugs
-crosses BBB
tubocurarine
-nondepolarizing neuromuscular blocking agent
- muscle relaxant during surgery for setting fractures & dislocations
-most likely to cause histamine release
ipratropium
-antimuscarinic (like atropine)
-treatment of bronchospasm associated with asthma, COPD
-inhalation, aerosol,
- a local site specific effect
origin of preganglionic parasympathetic
craniosacral
sympathetic
thoracolumbar
trimethaphan
-nondepolarizing ganglionic blockers, + doesn't cross BBB
-blocks nicotinic receptor for hypertnsive emergencies & discesting aortic aneurysim
SE= cyclopegia, decrease BP and increase HR decrease GI
zerostygmia
dry mouth - symptoms of atropine salivary glands are blocked
side effects of nicotinic antagonists
orthostatic hypertension postural hypertension, urinary retension, sexual dysfunction
metoprolol
b1 selective blocking agent
for hypertension + angina ectoris following abrupt cessation of therapy
cocaine
-CNS stimulant blocks monamines reuptake channels
-blocks Na channels + cardiac K
tyramine
indirect acting sympathomimetic
-MAO I prolongs affects
-cocaine blocks reuptake
dopamine in fermented food cheese and beer
ephedrine
direct + indrectly act sympath
-dierect a + B indirect tyramine like
-diet supression, bronchodilator
what's in a dense core vesicle
NE, DOP, dop B-hydroxylase,
chromaranin
ATP
loop b-hydronon converts DOP --> NE
isoproterenol
nonselective Beta agonist B1, B2
for heart blocker or cardiac arrests
SE: tachycardia
CNS effects
sedation, confusion, ataxia, (decreased pathway), loss of reflex, slurred speech, convulsion
angiotensin II
vasoactive peptide a potent pressor agent
atropine
-preanesthetic medication
- to restore cardiac rate
and arterial pressure during
anesthesia
-muscarinic blockers