• 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

1/733

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;

733 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)
Which antibiotics block cell wall synthesis by inhibition of peptidoglycan cross-linking?
Penicillin, ampicillin, ticarcillin, piperacillin, imipenem, aztreonam,
cephalosporins
Which antibiotics block peptidoglycan synthesis?
Bacitracin, vancomycin
Which antibiotic disrupts bacterial cell membranes?
Polymyxins
Which antibiotics block nucleotide synthesis?
Sulfonamides, trimethoprim
Which antibiotic blocks D N A topoisomerases?
Fluoroquinolones
Which antibiotic blocks mRNA synthesis?
Rifampin
Which antibiotics block protein synthesis at 50S ribosomal subunit?
Chloramphenicol, macrolides, clindamycin, streptogramins (quinupristin, dalfopristin), linezolid
Which antibiotics block protein synthesis at 30S ribosomal subunit
Aminoglycosides, tetracyclines
Which antibiotics are bacteriostatic?
"We're ECSTaTiC about bacteriostatics."

Erythromycin, Clindamycin, Sulfamethoxazole, Trimethoprim, Tetracyclines, Chloramphenicol.
Which antibiotics are bactericidal?
"Very Finely Proficient At Cell Murder."

Vancomycin, Fluoroquinolones, Penicillin, Aminoglycosides, Cephalosporins, Metronidazole.
Mechanism of Penicillin? (G- IV, V-oral)
1. Bind penicillin-binding proteins
2. Block transpeptidase cross-linking of cell wall
3. Activate autolytic enzymes
Clinical use of penicillin?
Mostly used for gram-positive organisms (S. pneumoniae, S.pyogenes, Actinomyces) and syphilis.
Toxicity of penicillin?
Hypersensitivity reactions, hemolytic anemia
Mechanism of methicillin/nafcillin?
Same as penicillin. Narrow spectrum; penicillinase resistant because of bulkier R group.
Clinical use of nafcillin?
S. aureus (except MRSA; resistant because of altered penicillin-binding protein target site).
Toxicity of Methicillin/naficillin?
Methicillin- interstitial nephritis
hypersensitivity
Mechanism of ampicillin/amoxicillin?
Wider spectrum; penicillinase sensitive. Also combine with clavulanic acid to enhance spectrum. AmOxicillin has greater Oral bioavailability than ampicillin.
Clinical use of ampicillin/amoxicillin?
Ampicillin HELPS kill enterococci

Extended-spectrum penicillin- certain gram-positive bacteria and gram-negative rods (Haemophilus influenzae, E. coli, Listeria monocytogenes, Proteus mirabilis, Salmonella, enterococci).
Toxicity of ampicillin/amoxicillin?
Hypersensitivity reactions; ampicillin rash; pseudomembranous colitis.
Mechanism of ticarcillin, carbenicillin, piperacillin?
Same as penicillin. Extended spectrum.
Clinical use of ticarcillin, carbenicillin, piperacillin?
TCP- Takes Care of Pseudomonas

Pseudomonas spp. and gram-negative rods;
susceptible to penicillinase; use with clavulanic
acid.
Toxicity of ticarcillin, carbenicillin, piperacillin
Hypersensitivity reaction
What are the B-lacatamase inhibitors?
CAST

clavulanic acid, sulbactam, tazobactam.
Mechanism of cephalosporins?
j3-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases. Bactericidal.
Clinical use of 1st generation cephalosporins? (cefazolin, cephalexin)
PEcK

gram-positive cocci, Proteus mirabilis, E. coli, Klebsiella pneumoniae.
Clinical use of 2nd generation cephalosporins?(cefoxitin, cefaclor, cefuroxime)
HENS PEcK

Gram-positive cocci, Haemophilus influenzae, Enterobacter aerogenes Neisseria spp., Proteus mirabilis, E. coli, Klebsiella pneumoniae, Serratia marcescens.
Clinical use of 3rd generation cephalosporins? (ceftriaxone, cefotaxime, ceftazidime)
Serious gram-negative infections resistant to other B lactams.
(Gonorrhea and Meningitis)
Clinical use of 4th generation cephalosporins? Cefepime
p for pseudomonas

Increased activity against
Pseudomonas and gram-positive organisms.
Toxicity of cephalosporins?
Vitamin K deficiency
Cross-hypersensitivity with penicillins occurs in 5-10% of patients.
Increased nephrotoxicity of aminoglycosides
Disulfiram-like reaction with ethanol
Mechanism of ticarcillin, carbenicillin, piperacillin?
Same as penicillin. Extended spectrum.
Clinical use of ticarcillin, carbenicillin, piperacillin?
TCP- Takes Care of Pseudomonas

Pseudomonas spp. and gram-negative rods;
susceptible to penicillinase; use with clavulanic
acid.
Toxicity of ticarcillin, carbenicillin, piperacillin
Hypersensitivity reaction
What are the B-lacatamase inhibitors?
CAST

clavulanic acid, sulbactam, tazobactam.
Mechanism of cephalosporins?
j3-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases. Bactericidal.
Clinical use of 1st generation cephalosporins? (cefazolin, cephalexin)
PEcK

gram-positive cocci, Proteus mirabilis, E. coli, Klebsiella pneumoniae.
Clinical use of 2nd generation cephalosporins?(cefoxitin, cefaclor, cefuroxime)
HENS PEcK

Gram-positive cocci, Haemophilus influenzae, Enterobacter aerogenes Neisseria spp., Proteus mirabilis, E. coli, Klebsiella pneumoniae, Serratia marcescens.
Clinical use of 3rd generation cephalosporins? (ceftriaxone, cefotaxime, ceftazidime)
Serious gram-negative infections resistant to other B lactams.
(Gonorrhea and Meningitis)
Clinical use of 4th generation cephalosporins? Cefepime
p for pseudomonas

Increased activity against
Pseudomonas and gram-positive organisms.
Toxicity of cephalosporins?
Vitamin K deficiency
Cross-hypersensitivity with penicillins occurs in 5-10% of patients.
Increased nephrotoxicity of aminoglycosides
Disulfiram-like reaction with ethanol
Mechanism of Aztreonam?
A monobactam resistant to B lactamases. Inhibits cell wall synthesis (binds to PBP3 ). Synergistic with aminoglycosides.
Clinical use of Aztreonam?
Gram-negative rods only-No activity against gram-positives or anaerobes. For penicillin-allergic patients and those with renal insufficiency who cannot tolerate aminoglycosides.
Toxicity of Aztreonam?
Usually nontoxic; occasional GI upset. No cross-sensitivity with penicillins or cephalosporins.
Mechanism of lmipenem/cilastatin, meropenem?
lmipenem is a broad-spectrum, j3-lactamase-resistant carbapenem. Always administered with cilastatin (inhibitor of renal dihydropeptidase I) to decrease inactivation of drug in renal tubules.
Clinical use of lmipenem/cilastatin, meropenem?
Gram-positive cocci, gram-negative rods, and anaerobes. Wide spectrum, but the significant side effects limit use to life-threatening infections, or after other drugs have failed. Meropenem, however, has a reduced risk ofseizures and is stable to dihydropeptidase I.
Toxicity of lmipenem/cilastatin, meropenem?
GI distress, skin rash, and CNS toxicity (seizures) at high plasma levels.
Mechanism of Vancomycin?
Inhibits cell wall mucopeptide formation by binding D-ala D-ala portion of cell wall precursors. Bactericidal.
Clinical use of Vancomycin?
Gram positive only-serious, multidrug-resistant organisms, including S. aureus, enterococci and Clostridium difficile (pseudomembranous colitis).
Toxicity of Vancomycin?
NOT

Nephrotoxicity, Ototoxicity, Thrombophlebitis, diffuse flushing- "red man syndrome"
(prevent with antihistamines and slow infusion rate).
Well tolerated
What are the aminoglycosides?
Gentamicin, Neomycin, Amikacin, Tobramycin, Streptomycin
Mechanism of Aminoglycosides?
Bactericidal; inhibit formation of initiation complex and cause misreading of mRNA. Require oxygen for uptake; therefore ineffective against anaerobes.
Clinical use of Aminoglycosides?
Severe gram-negative rod infections. Synergistic with B lactam antibiotics. Neomycin for bowel surgery.
Toxicity of Aminoglycosides?
Nephrotoxic (esp with cephalosporins)
Ototoxicity (esp with loop diuretics)
Tertagoenic
Resistance to Aminoglycosides?
Transferase enzymes that inactivate the drug by acetylation, phosphorylation, or adenylation.
Mechanism of tetracycline?
Bacteriostatic; bind to 30S and prevent attachment of aminoacyl-tRNA; limited CNS penetration. Doxycycline is fecally eliminated and can be used in patients with renal failure. Must NOT take with milk, antacids, or iron-containing preparations because divalent cations inhibit its absorption in the gut.
Clinical use of tetracycline?
Borrelia burgdorferi, H. pylori, M. pneumoniae. Drug's ability to accumulate intracellularly makes it very effective against Rickettsia and Chlamydia.
Toxicity of tetracycline?
GI distress, discoloration of teeth and inhibition of bone growth in children, photosensitivity.
Resistance to tetracycline?
decrease uptake into cells or increased effiux out of cell by plasmid-encoded transport pumps.
Mechanism of macrolides?
Inhibit protein synthesis by blocking translocation; bind to the 23S rRNA of the 50S
ribosomal subunit. Bacteriostatic.
Clinical use of macrolides?
Atypical pneumonias (Mycoplasma, Chlamydia, Legionella), URis, STDs,
gram-positive cocci (streptococcal infections in patients allergic to penicillin), and Neisseria.
Toxicity of macrolides?
Prolonged QT interval (especially erythromycin), GI discomfort (most common cause of noncompliance), acute cholestatic hepatitis, eosinophilia, skin rashes. Increases serum concentration of theophyllines, oral anticoagulants.
Resistance to macrolides?
Methylation of 23S rRNA binding site.
Mechanism of Chloramphenicol?
Inhibits 50S peptidyltransferase activity. Bacteriostatic.
Clinical use of Chloramphenicol?
Meningitis (Haemophilus influenzae, Neisseria meningitidis, Streptococcus pneumoniae).
Conservative use owing to toxicities but often still used in developing countries due to
low cost.
Toxicity of Chloramphenicol?
Anemia (dose dependent), aplastic anemia (dose independent), gray baby syndrome (in
premature infants because they lack liver UDP-glucuronyl transferase).
Resistance to Chloramphenicol?
Plasmid-encoded acetyltransferase that inactivates drug.
Mechanism of Clindamycin?
Blocks peptide bond formation at 50S ribosomal subunit. Static
Clinical use of Clindamycin?
Anaerobic infections (above diaphragm)
Toxicity of Clindamycin?
Pseudomembranous colitis (C. difficile overgrowth), fever, diarrhea.
Mechanism of sulfonamides?
PABA antimetabolites inhibit dihydropteroate synthetase. Bacteriostatic.
Clinical use of sulfonamides?
Gram-positive, gram-negative, Nocardia, Chlamydia. Triple sulfas or SMX for simple UTI. :z:
Toxicity of sulfonamides?
Hypersensitivity reactions, hemolysis if G6PD deficient, nephrotoxicity (tubulointerstitial nephritis), photosensitivity, kernicterus in infants, displace other drugs from albumin "'
(e.g., warfarin).
Mechanism of trimethoprim?
Inhibits bacterial dihydrofolate reductase. Bacteriostatic.
Clinical use of trimethoprim?
Combination used for recurrent UTis, Shigella, Salmonella , Pneumocystis iiroveci pneumonia.
Toxicity of trimethoprim?
Megaloblastic anemia, leukopenia, granulocytopenia.
What are the sulfa drugs?
sulfasalazine, sulfonylureas, thiazide diuretics, acetazolamide, furosemide, celecoxib, or probenecid.
Mechanism of Fluoroquinolones?
Inhibit DNA gyrase (topoisomerase II). Bactericidal. Must not be taken with antacids.
Clinical use of Fluoroquinolones?
Gram-negative rods of urinary and GI tracts (including Pseudomonas), Neisseria, some gram- positive organisms.
Toxicity of Fluoroquinolones?
GI upset, superinfections, skin rashes, headache, dizziness. Contraindicated in pregnant women and in children because animal studies show damage to cartilage. Tendonitis and tendon rupture in adults; leg cramps and myalgias in kids.
Mechanism of Metronidazole?
Forms free radical toxic metabolites in the bacterial cell that damage DNA. Bactericidal, antiprotozoal.
Clinical use of Metronidazole?
GET GAP on the Metro!

Treats Giardia, Entamoeba, Trichomonas, Gardnerella vaginalis, Anaerobes (Bacteroides, Clostridium). Used with bismuth and amoxicillin (or tetracycline) for "triple therapy" against H. Pylori.
Toxicity of Metronidazole?
Disulfiram-like reaction with alcohol; headache, metallic taste.
Mechanism of polymyxin?
Bind to cell membranes of bacteria and disrupt
their osmotic properties. Polymyxins are cationic, basic proteins that act like detergents.
Clinical use of polymyxin?
Resistant gram-negative infections.
Toxicity of polymyxin?
Neurotoxicity, acute renal tubular necrosis.
Treatment of M. leprae?
Dapsone, rifampin, clofazimine
Treatment of M. avium- intracellulare
Azithromycin, rifampin,
ethambutol, streptomycin
What is the mechanism of ethambutol?
Decreased carbohydrate - "a
polymerization of mycobacterium cell wall by blocking arabinosyltransferase.

Red green color blindness
Mechanism of isoniazid?
Decreases synthesis of mycolic acids. Bacteria catalase- peroxidase needed to convert INH to active metabolite
Toxicity of isoniazide?
Neurotoxicity, hepatotoxicity, lupus. Pyridoxine (vitamin B6) can prevent neurotoxicity, lupus.
Clinical use of rifampin?
Mycobacterium tuberculosis; delays resistance to
dapsone when used for leprosy. Used for meningococcal prophylaxis and chemoprophylaxis in contacts of children with Haemophilus influenzae type B.
Toxicity of Rifampin?
Minor hepatotoxicity and drug interactions (P-450); orange body fluids (nonhazardous side effect).
Treatment of VRE?
Linezolid and quinupristin/dalfopristin
Mechanism of amphotericin B?
Binds ergosterol (unique to fungi); forms membrane pores that allow leakage of electrolytes.
Clinical use of amphotericin B?
Serious, systemic mycoses.
lntrathecally for fungal
meningitis; does not cross blood-brain barrier.
Toxicity of amphotericin B?
Fever/chills ("shake and bake"), hypotension,
nephrotoxicity, arrhythmias, anemia, IV phlebitis ("amphoterrible"). Hydration reduces nephrotoxicity. Liposomal amphotericin reduces toxicity.
Mechanism of nystatin?
Same as amphotericin B. Topical form because too toxic for systemic use.
Clinical use of nystatin?
"Swish and swallow" for oral candidiasis (thrush); topical for diaper rash or vaginal
candidiasis.
Mechanism of Azoles?
Inhibit fungal sterol (ergosterol) synthesis, by inhibiting the P-450 enzyme that converts
lanosterol to ergosterol.
Clinical use of azoles?
Systemic mycoses. Fluconazole for cryptococcal meningitis in AIDS patients (because it can cross blood-brain barrier) and candida! infections of all types.
Ketoconazole for Blastomyces, Coccidioides, Histoplasma, Candida albicans;
Toxicity of azoles?
Hormone synthesis inhibition (gynecomastia), liver dysfunction (inhibits cytochrome
P-450), fever, chills.
Mechanism of flucocytosine?
Inhibits DNA synthesis by conversion to 5-fluorouracil.
Clinical use of flucocytosine?
Used in systemic fungal infections (e.g., Candida, Cryptococcus) in combination with amphotericin B.
Toxicity of flucocytosine?
Nausea, vomiting, diarrhea, bone marrow suppression.
Mechanism of caspfungin?
Inhibits cell wall synthesis by inhibiting synthesis of B-glucan.
Clinical use of caspfungin?
Invasive aspergillosis.
Toxicity of caspfungin?
G l upset, flushing.
Mechanism of Terbinafine?
Inhibits the fungal enzyme squalene epoxidase.
Clinical use of Terbinafine?
Used to treat dermatophytoses (especially onychomycosis-fungal infection of finger or toe nails)
Mechanism of Griseofulvin?
Interferes with microtubule function; disrupts mitosis. Deposits in keratin-containing tissues (e.g., nails).
Clinical use of Griseofulvin?
Oral treatment of superficial infections; inhibits growth of dermatophytes (tinea, ringworm).
Toxicity of Griseofulvin?
Teratogenic, carcinogenic, confusion, headaches, increased P-450 and warfarin metabolism.
Mechanism of amantadine?
Blocks viral penetration/uncoating (M2 protein). Also causes the release of dopamine from intact nerve terminals.
Clinical use of amantadine?
Prophylaxis and treatment for influenza A only; Parkinson's disease.
Toxicity of amantadine?
Ataxia, dizziness, slurred speech.
Resistance to amantadine?
Mutated M2 protein. 90% of all influenza A strains are resistant to amantadine, so not used.

Rimantidine used instead- less CNS side effects
Mechanism of oseltamivir?
Inhibit influenza neuraminidase, decreasing the release of progeny virus.

Both influenza A and B
Mechanism of ribavirin?
Inhibits synthesis of guanine nucleotides by competitively inhibiting IMP dehydrogenase.
Clinical use of ribavirin?
RSV, chronic hepatitis C.
Toxicity of ribavirin?
Hemolytic anemia. Severe teratogen.
Mechanism of acyclovir?
Monophosphorylated by HSVNZV thymidine kinase. Guanosine analog. Triphosphate formed by cellular enzymes. Preferentially inhibits viral DNA polymerase by chain termination.
Clinical use of acyclovir?
HSV, VZV, EBV. Used for HSV-induced mucocutaneous and genital lesions as well as for encephalitis. Prophylaxis in immunocompromised patients. For herpes zoster, use a related agent, famciclovir.
Resistance to acyclovir?
Lack of viral thymidine kinase.
Mechanism of ganciclovir?
5'-monophosphate formed by a CMV viral kinase or HSVNZV thymidine kinase. Guanosine analog. Triphosphate formed by cellular kinases. Preferentially inhibits viral DNA polymerase.
Clinical use of ganciclovir?
CMV , especially in immunocompromised patients.
Toxicity of ganciclovir?
Leukopenia, neutropenia, thrombocytopenia, renal toxicity. More toxic to host enzymes
than acyclovir.
Resistance to ganciclovir?
Mutated CMV DNA polymerase or lack of viral kinase.
Mechanism of foscarnet?
Viral DNA polymerase inhibitor- does not require viral kinase
Clinical use of foscarnet?
CMV retinitis in immunocompromised patients when ganciclovir fails; acyclovir-resistant HSV.
Mechanism of cyclosporine?
Binds to cyclophilins. Complex blocks the differentiation and activation of T cells by inhibiting calcineurin, thus preventing the production of IL-2 and its receptor.
Clinical use of cyclosporine?
Suppresses organ rejection after transplantation; selected autoimmune disorders.
Toxicity of cyclosporine?
Predisposes patients to viral infections and lymphoma; nephrotoxic (preventable with
mannitol diuresis).
Mechanism of tacrolimus?
Similar to cyclosporine; binds to FK-binding protein, inhibiting secretion of IL-2 and other cytokines.
Clinical use of tacrolimus?
Potent immunosuppressive used in organ transplant recipients.
Toxicity of tacrolimus?
Significant-nephrotoxicity, peripheral neuropathy, hypertension, pleural effusion,
hyperglycemia.
Mechanism of Sirolimus (rapamycin)?
Binds to mTOR. Inhibits T-cell proliferation in response to IL-2.
Toxicity of sirolimus?
Hyperlipidemia, thrombocytopenia, leukopenia.
Mechanism of daclizumab?
Monoclonal antibody with high affinity for the IL-2 receptor on activated T cells.
Mechanism of Azathioprine?
Antimetabolite precursor of 6-mercaptopurine that interferes with the metabolism and synthesis of nucleic acids. Toxic to proliferating lymphocytes.
Toxicity of azathioprine?
Bone marrow suppression. Toxic effect with allopurinol.
Mechanism of Muromonab-CD3?
Monoclonal antibody that binds to CD3 (epsilon chain) on the surface of T cells. Blocks cellular interaction with CD3 protein responsible forT-cell signal transduction.
Toxicity of Muromonab-CD3?
Immunosuppression after kidney transplantation.
Clinical use of Muromonab-CD3?
Cytokine release syndrome, hypersensitivity reaction.
Clinical use of aldesleukin?
IL-2: Renal cell carcinoma, metastatic melanoma
Clinical use of erythropoietin?
Anemia
Clinical use of filgrastim?
Recovery of bone marrow- GCSF
Clinical use of a-interferon?
Hepatitis Band C, Kaposi's sarcoma, leukemias, malignant melanoma
Clinical use of B-interferon?
Multiple sclerosis
Clinical use of gamma-interferon?
Chronic granulomatous disease
Clinical use of Oprelvekin?
Il-11: Thrombocytopenia
Clinical use of infliximab?
TNF-alpha-Crohn's disease, rheumatoid arthritis, psoriatic arthritis
Clinical use of abciximab?
Glycoprotein IIb/IIIa- Prevent cardiac ischemia in unstable angina and in
patients treated with percutaneous coronary intervention
Clinical use of trastuzumab (herceptin)?
erb-B2- HER-2-Dverexpressing breast cancer
Clinical use of Rituximab?
CD20- B-cell non-Hodgkin's lymphoma
What are the 1st generation H1 blockers?
Diphenhydramine, dimenhydrinate, chlorpheniramine.
Clinical uses of 1st generation H1 blockers?
Allergy, motion sickness, sleep aid.
Toxicity of 1st generation H1 blockers?
Sedation, antimuscarinic, anti-a-adrenergic.
Clinical uses of 2nd generation H1 blockers?
Allergies
Toxicity of 2nd generation H1 blockers?
Far less sedating than lst generation because of decreased entry into CNS.
What are the 2nd generation H1 blockers?
Loratadine, fexofenadine, desloratadine, cetirizine.
Mechanism (and type) of isoproterenol?
Relaxes bronchial smooth muscle (B2). Adverse effect is tachycardia (B1).
Mechanism (and type) of albuterol?
Relaxes bronchial smooth muscle (B2). Use during acute exacerbation.
Mechanism (and type) of albuterol?
Long-acting agent for prophylaxis. Adverse effects are tremor and arrhythmia.
Mechanism of theophylline?
Likely causes bronchodilation by inhibiting phosphodiesterase, thereby .decreases cAMP hydrolysis.
Toxicity of Theophylline?
Narrow therapeutic index- cardiotoxic, neurotoxic, blocks actions of adenosine
Mechanism of ipratropium?
Muscarinic antagonist- competitive block of muscarinic receptors, preventing bronchoconstriction. Also used for COPD.
Mechanism of Cromolyn?
Prevents release of mediators from mast cells. Effective only for the prophylaxis of asthma. Not effective during an acute asthmatic attack. Toxicity is rare.
Mechanism of zileuton?
5-lipoxygenase pathway inhibitor. Blocks conversion or arachidonic acid to leukotrienes.
Mechanism of zafirlukast/montelukast?
Block leukotriene receptors. Especially good for aspirin-induced.
Mechanism of Bosentan?
Used to treat pulmonary hypertension. Competitively antagonizes endothelin-1 receptors, decreasing pulmonary vascular resistance.
Use of Guaifenesin?
Expectorant- removes excess sputum; does not suppress cough reflex.
Use of N-acetylcysteine?
Acetaminophen overdose
Mucolytic- loosen mucous plugs in CF patients
Mechanism of sildenafil?
Inhibit cGMP phosphodiesterase, causing increases cGMP, smooth muscle relaxation in the corpus cavernosum, I blood flow, and penile erection.
Toxicity of sildenafil?
Headache, flushing, dyspepsia, impaired blue-green color vision. Risk of life-threatening hypotension in patients taking nitrates.
Mechanism of tamsulosin?
A1-antagonist used to treat BPH by inhibiting smooth muscle contraction. Selective for a 1A,D receptors (found on prostate)
Mechanism of Ritodrine/terbutaline?
B2 agonists that relax the uterus; reduce premature uterine contractions.
Mechanism and action of Dinoprostone?
PGE2 analog causing cervical dilation and uterine contraction, inducing labor.
Advantages of oral contraception?
Reliable
Decreased risk of endometrial and ovarian cancer
lower ectopic pregnancy
Decreased PID
Regulation of menses
Contraindicaiton of oral contraception?
mokers > 35 years of age (increased risk of cardiovascular events), patients with history of thromboembolism and stroke or history of estrogen-dependent tumor.
Mechanism of Mifepristone (RU-486)
Competitive inhibitor of progestins at progesterone receptors.
Clinical use of Mifepristone (RU-486)?
Termination of pregnancy. Administered with misoprostol (PGE 1) .
Mechanism of Progestins?
Bind progesterone receptors, reduce growth, and increase vascularization of endometrium.
Clinical use of progestins?
Used in oral contraceptives and in the treatment of endometrial cancer and abnormal
uterine bleeding.
Clinical use of hormone replacement therapy?
Used for relief or prevention of menopausal symptoms (e.g., hot flashes, vaginal atrophy) and osteoporosis (increase estrogen, decreased osteoclast activity).
Mechanism/action of raloxifene?
Partial estrogen agonist

Agonist on bone; reduces resorption of bone; used to treat osteoporosis.
Mechanism/action of tamoxifen?
Antagonist on breast tissue; used to treat and prevent recurrence of ER-positive breast cancer.
Mechanism/action of clomiphene?
Partial agonist at estrogen receptors in hypothalamus. Prevents normal feedback inhibition and i release of LH and FSH from pituitary, which stimulates ovulation. Used to treat infertility and PCOS. May cause hot flashes, ovarian enlargement, multiple simultaneous pregnancies, and visual disturbances.
Clinical uses of estrogens?
(EE, DES, mestranol)
Hypogonadism or ovarian failure, menstrual abnormalities, HRT in postmenopausal
women; use in men with androgen-dependent prostate cancer.
Toxicity of estrogen?
Increased risk of endometrial cancer, bleeding in postmenopausal women, clear cell adenocarcinoma of vagina in females exposed to DES in utero, increased risk of thrombi.
Mechanism of leuprolide?
GnRH analog with agonist properties when used in pulsatile fashion; antagonist properties when used in continuous fashion.
Clinical use of leuprolide?
Infertility (pulsatile), prostate cancer (continuous- use with flutamide), uterine fibroids.
Clinical use of Testosterone?
Treat hypogonadism and promote development of zosex characteristics; stimulation of
anabolism to promote recovery after burn or injury; treat ER-positive breast cancer
Toxicity of Testosterone?
Causes masculinization in females; reduces intratesticular testosterone in males by inhibiting release of LH (via negative feedback), leading to gonadal atrophy. Premature closure of epiphyseal plates. increased LDL, decreased HDL.
Mechanism of finasteride?
A 5a-reductase inhibitor (.!-conversion of testosterone
to dihydrotestosterone) Useful in BPH. Also promotes hair growth- used to treat male-pattern baldness.
Mechanism of flutamide?
A nonsteroidal competitive inhibitor of androgens at the testosterone receptor. Used in prostate carcinoma.
Reproductive effect of ketoconazole?
Inhibits desmolase- decreased testosterone
Reproductive effect of spironolactone?
Inhibits steroid binding
Mechanism of mannitol?
Osmotic diuretic, increased tubular fluid osmolarity, producing increased urine flow.
Clinical use of mannitol?
Shock, drug overdose, increased intracranial/intraocular pressure.
Toxicity of mannitol?
Pulmonary edema, dehydration. Contraindicated in anuria, CHF
Mechanism of acetazolamide?
Carbonic anhydrase inhibitor. Causes self-limited NaHC03 diuresis and reduction in total-body HC03- stores.
Clinical use of acetazolamide?
Glaucoma, urinary alkalinization, metabolic alkalosis, altitude sickness.
Toxicity of acetazolamide?
Hyperchloremic metabolic acidosis, neuropathy, NH3 toxicity, sulfa allergy.
Mechanism of furosemide?
Sulfonamide loop diuretic. Inhibits cotransport system (Na+, K+, 2 Cl-) of thick ascending limb of loop of Henle. Abolishes hypertonicity of medulla, preventing concentration of urine. Increased Ca2+ excretion. Loops Lose calcium.
Clinical use of furosemide?
Edematous states (CHF, cirrhosis, nephrotic syndrome, pulmonary edema), hypertension, hypercalcemia.
Toxicity of furosemide?
OH DANG

Ototoxicity, Hypokalemia, Dehydration, Allergy (sulfa), Nephritis (interstitial), Gout.
Mechanism of Ethacrynic acid?
Same action as furosemide.
Clinical use of Ethacrynic acid?
Diuresis in patients allergic to sulfa drugs.
Toxicity of Ethacrynic acid?
Ototoxicity, Hypokalemia, Dehydration, Nephritis (interstitial)..
Mechanism of Hydrochlorothiazide?
Thiazide diuretic. Inhibits NaCl reabsorption in early distal tubule, reducing diluting capacity of the nephron. .decreased Ca2+ excretion.
Clinical use of Hydrochlorothiazide?
Hypertension, CHF, idiopathic hypercalciuria, nephrogenic diabetes insipidus.
Toxicity of Hydrochlorothiazide?
Hypokalemic metabolic alkalosis, hyponatremia, hyperglycemia, hyperlipidemia, hyperuricemia, and hypercalcemia. Sulfa allergy.
What are the Potassium sparing diuretics?
Spironolactone, Triamterene, Amiloride, eplerenone.
Mechanism of Potassium sparing diuretics?
Spironolactone is a competitive aldosterone receptor
antagonist in the cortical collecting tubule. Triamterene and amiloride act at the same part of the tubule by blocking Na+channels in the CCT.
Clinical use of K sparing diuretics?
Hyperaldosteronism, K+ depletion, CHF
Toxicity of K sparing diuretics?
Hyperkalemia (can lead to arrhythmias), endocrine
effects with aldosterone antagonists (e. g., spironolactone causes gynecomastia)
Mechanism of ACE inhibitors?
(captopril, enalapril, lisinopril)
Inhibit angiotensin-converting enzyme, reducing
levels of angiotensin II and preventing inactivation of bradykinin, a potent vasodilator. Renin release is increased due to loss of feedback inhibition.
Mechanism of losartan?
Angiotensin II receptor antagonist.
(Does not cause cough)
Toxicity of ACE inhibitors?
Cough, Angioedema, Proteinuria, Taste changes, Hypotension, Fetal renal damage

Avoid with renal artery stenosis- ACE inhibitors significantly. decreased GFR by preventing constriction of efferent arterioles.
What are the H2 blockers?
Cimetidine, ranitidine, famotidine, nizatidine.
Mechanism of H2 blockers?
Reversible block of histamine Hz receptors~ decreased H+
secretion by parietal cells.
Clinical use of H2 blocker?
Peptic ulcer, gastritis, mild esophageal reflux. (esp. nocturnal)
Toxicity of H2 blocker?
Cimitidine (others aren't as bad)
P-450 inhibitor
Anti-androgenic effect
Confusion, headache, dizziness
Mechanism of Proton pump inhibitors?
Irreversibly inhibit H+fK+-ATPase in stomach parietal cells
Clinical use of Proton pump inhibitors?
Peptic ulcer, gastritis, esophageal reflux, Zollinger-Ellison syndrome.
Mechanism of bismuth/ sucraflate?
Bind to ulcer base, providing physical protection, and allow HC03secretion to reestablish pH gradient in the mucous layer.
Clinical use of of bismuth/ sucraflate?
Increases ulcer healing, traveler's diarrhea.
Mechanism of misoprostol?
A PGE 1 analog. increased production and secretion of gastric mucous barrier,decrease acid production.
Clinical use of misoprostol?
Prevention of NSAID-induced peptic ulcers; maintenance of a patent ductus arteriosus. Also used to induce labor.
Toxicity of Misoprostol?
Diarrhea. Contraindicated in women of childbearing potential (abortifacient).
Muscarinic antagonist used in GI problems?
Pirenzepine, propantheline.
Mechanism of GI muscarinic antagonist?
Block Ml receptors on ECL cells (decreased histamine secretion) and M3 receptors on parietal cells (decreased H+secretion).
Toxicity of GI muscarinic antagonist?
Tachycardia, dry mouth, difficulty focusing eyes.
Mechanism of octreotide?
Somatostatin analog
Clinical use of octreotide?
Acute variceal bleeds, acromegaly, VIPoma, and carcinoid tumors.
Toxicity of octreotide?
Nausea, cramps, steatorrhea.
Toxicity of aluminum hydoxide antacid?
Aluminium-minimum feces

Constipation and hypophosphatemia; proximal muscle weakness, osteodystrophy, seizures
Toxicity of magnesium hydoxide antacid?
Mg- must got to the bathroom

Diarrhea, hyporeflexia,
hypotension, cardiac arrest
Toxicity of calcium carbonate antacid?
Hypercalcemia, rebound acid

Chelates drugs like tetracycline
Mechanism of lnfliximab?
A monoclonal antibody to TNF

INFLIX pain on TNF
Clinical use of lnfliximab?
Crohn's disease, rheumatoid arthritis.
Toxicity of lnfliximab?
Respiratory infection (including reactivation of latent TB), fever, hypotension.
Mechanism of sulfasalazine?
A combination of sulfapyridine (antibacterial) and 5-aminosalicylic acid (anti-inflammatory). Activated by colonic bacteria.
Clinical use of sulfasalazine?
Ulcerative colitis, Crohn's disease.
Toxicity of sulfasalazine?
Malaise, nausea, sulfonamide toxicity, reversible oligospermia.
Mechanism of ondansetron?
5-HT3 antagonist. Powerful central-acting antiemetic.
Clinical use of ondansetron?
Control vomiting postoperatively and in patients undergoing cancer chemotherapy.
Toxicity of ondansetron?
Headache, constipation.
Mechanism of metoclopramide?
D2 receptor antagonist. I resting tone, contractility, LES tone, motility. Does not influence colon transport time.
Clinical uses of metoclopramide?
Diabetic and post-surgery gastroparesis.
Toxicity of metoclopramide?
Increases parkinsonian effects. Restlessness, drowsiness, fatigue, depression, nausea,
diarrhea. Drug interaction with digoxin and diabetic agents. Contraindicated in patients with small bowel obstruction.
Mechanism of hydralazine?
Increased cGMP ~smooth muscle relaxation. Vasodilates arterioles> veins; afterload reduction.
Clinical use of hydralazine?
Severe hypertension, CHF. First-line therapy for hypertension in pregnancy, with methyldopa. Frequently coadministered with a B-blocker to prevent reflex tachycardia.
Toxicity of hydralazine?
Compensatory tachycardia (contraindicated in angina/CAD), fluid retention, nausea, headache, angina. Lupus-like syndrome.
What are the calcium channel blockers?
Nifedipine, verapamil, diltiazem.
Vascular smooth muscle-nifedipine > diltiazem > verapamil (Verapamil =Ventricle).
Heart-verapamil > diltiazem > nifedipine.
Mechanism of calcium channel blockers?
Block voltage-dependent L-type calcium channels of cardiac and smooth muscle and thereby reduce muscle contractility.
Clinical use of calcium channel blockers?
Hypertension, angina, arrhythmias (not nifedipine), Prinzmetal's angina, Raynaud's.
Toxicity of calcium channel blockers?
Hypertension, angina, arrhythmias (not nifedipine), Prinzmetal's angina, Raynaud's.
Mechanism of nitroglycerin/isosorbide dinitrate?
Vasodilate by releasing nitric oxide in smooth muscle, causing I in cGMP and smooth muscle relaxation. Dilate veins>> arteries. Decreases preload.
Clinical use of nitroglycerin/isosorbide dinitrate?
Angina, pulmonary edema. Also used as an aphrodisiac and erection enhancer.
Toxicity of nitroglycerin/isosorbide dinitrate?
Reflex tachycardia, hypotension, flushing, headache, "Monday disease" in industrial
exposure; development of tolerance for the vasodilating action during the work week and loss of tolerance over the weekend, resulting in tachycardia, dizziness, and headache on reexposure.
Mechanism/use of nitroprusside?
Malignant hypertension

Short acting; increases cGMP via direct release of NO. Can cause cyanide toxicity (releases CN).
Mechanism/use of fenoldopam?
Malignant hypertension

Dopamine D 1 receptor agonist-relaxes renal vascular smooth muscle.
Mechanism/use of diazoxide?
Malignant Hypertension

K+ channel opener-hyperpolarizes and relaxes vascular smooth muscle. Can cause hyperglycemia (reduces insulin release).
Mechanism of lovastatin?
HMG CoA reductase inhibitor

Inhibit cholesterol precursor, mevalonate
Side effects of statins?
Hepatotoxic
Rhabdomyolisis
Mechanism of niacin?
Inhibits lipolysis in adipose tissue; reduces hepatic VLDL secretion into circulation

Increases HDL!!!!
Side effects of niacin?
Redness and flushing (give aspirin)
Hyperglycemia
Hyperuricemia (gout worsened)
Mechanism of cholestyramine/ colestipol/ colesevelam?
Prevent intestinal
reabsorption of bile acids; liver must use cholesterol
to make more (can increase TGs)
Toxicity of cholestyramine/ colestipol/ colesevelam?
Tastes bad and causes GI discomfort, decreases absorption of fat-soluble vitamins
Cholesterol gallstones
Mechanism of ezetimibe?
Prevent cholesterol reabsorption at small intestine brush border
Mechanism of fibrates?
Upregulates LPL leading to increased TG clearance

Major effect is on TGs
Mechanism of digoxin?
Direct inhibition ofNa+fK+ATPase leads to indirect inhibition ofNa+JCa2+ exchanger/
antiport. Increases [Ca2+L positive inotropy. Stimulates vagus nerve
Clinical use of digoxin?
CHF (increases contractility); atrial fibrillation (decreases conduction at AV node and depression of SA node).
Cholinergic toxicity of digoxin?
Cholinergic-nausea, vomiting, diarrhea, blurry yellow vision (think Van Gogh).
ECG toxicity of digoxin
Increased PR, Decreased QT, T wave inversion, arrhythmia, hyperkalemia

Worsened by renal failure (decreased excretion), hypokalemia, quinidine
Antidote for digoxin?
Slowly normalize K+, lidocaine, cardiac pacer, anti-dig Fab fragments, Mg2+.
Mechanism of fibrates?
Upregulates LPL leading to increased TG clearance

Major effect is on TGs
Mechanism of digoxin?
Direct inhibition ofNa+fK+ATPase leads to indirect inhibition ofNa+JCa2+ exchanger/
antiport. Increases [Ca2+L positive inotropy. Stimulates vagus nerve
Clinical use of digoxin?
CHF (increases contractility); atrial fibrillation (decreases conduction at AV node and depression of
SA node).
Cholinergic toxicity of digoxin?
Cholinergic-nausea, vomiting, diarrhea, blurry yellow vision (think Van Gogh).
ECG toxicity of digoxin
Increased PR, Decreased QT, T wave inversion, arrhythmia, hyperkalemia

Worsened by renal failure (decreased excretion), hypokalemia, quinidine
Antidote for digoxin?
Slowly normalize K+, lidocaine, cardiac pacer, anti-dig Fab fragments, Mg2+.
Mechanism of fibrates?
Upregulates LPL leading to increased TG clearance

Major effect is on TGs
Mechanism of digoxin?
Direct inhibition ofNa+fK+ATPase leads to indirect inhibition ofNa+JCa2+ exchanger/
antiport. Increases [Ca2+L positive inotropy. Stimulates vagus nerve
Clinical use of digoxin?
CHF (increases contractility); atrial fibrillation (decreases conduction at AV node and depression of
SA node).
Cholinergic toxicity of digoxin?
Cholinergic-nausea, vomiting, diarrhea, blurry yellow vision (think Van Gogh).
ECG toxicity of digoxin
Increased PR, Decreased QT, T wave inversion, arrhythmia, hyperkalemia

Worsened by renal failure (decreased excretion), hypokalemia, quinidine
Antidote for digoxin?
Slowly normalize K+, lidocaine, cardiac pacer, anti-dig Fab fragments, Mg2+.
What are the rapid acting insulins?
Lispro, Aspart
Toxicity of insulin?
Hypoglycemia, hypersensitivity reaction (very rare).
What are the long acting insulin?
Glargine, detemir
Mechanism of sulfonylureas?
C lose K+ channel in
B cell membrane, so
cell depolarizes, triggering of insulin release via increased Ca influx
Clinical use of sulfonylureas?
Stimulate release of endogenous insulin in type 2 DM. Require some islet function, so useless in type l DM
Toxicity of sulfonylureas?
First generation: disulfiram-like effects.
Second generation: hypoglycemia.
What is the mechanism of metformin?
Decreased gluconeogenesis, increased glycolysis, increased peripheral glucose uptake (insulin sensitivity)
Toxicity of metformin?
Most grave adverse effect is lactic acidosis (contraindicated in renal failure).
Mechanism of rosiglitazone?
Increased insulin sensitivity in peripheral tissue. Binds to PPAR -y nuclear transcription regulator.
Toxicity of rosiglitazone?
Weight gain, edema. Hepatotoxicity,
CV toxicity.
Mechanism of acarbose/ miglitol?
Inhibit intestinal brush- border a-glucosidases. Delayed sugar hydrolysis and glucose absorption lead to decreased postprandial hyperglycemia.
Mechanism of Pramlintide?
Decreased glucagon
Toxicity of Pramlintide?
Hypoglycemia, nausea, diarrhea.
Mechanism of exenatide?
GLP-1 analog
Increases insulin, decrease glucagon post meal
Mechanism of propylthiouracil and methimazole?
Inhibit organification of iodide and coupling of thyroid hormone synthesis. Propylthiouracil also decreases peripheral conversion ofT4 to T 3.
Clinical use of propylthiouracil and methimazole?
Hyperthyroidism
Toxicity of propylthiouracil and methimazole?
Skin rash, agranulocytosis (rare), aplastic anemia. Methimazole is a possible teratogen.
Clinical use of GH?
GH deficiency, Turner syndrome
Clinical use of somatostatin (octreolide)?
Acromegaly, carcinoid, gastrinoma, glucagonoma
Clinical use of oxytocin?
Stimulates labor, uterine contractions, milk let-down; controls uterine hemorrhage
AOH (desmopressin)
Pituitary (central, not nephrogenic) 0I
Mechanism of demeclocycline?
AOH antagonist (member of the tetracycline family).
Clinical use of demeclocycline?
SIADH
Toxicity of demeclocycline?
Nephrogenic DI, photosensitivity, abnormalities of bone and teeth.
Mechanism of glucocorticoids?
Decrease the production ofleukotrienes and prostaglandins by inhibiting phospholipase A2
and expression of COX-2.
Toxicity of glucocorticoids?
Iatrogenic Cushing's syndrome-buffalo hump, moon facies, truncal obesity, muscle
wasting, thin skin, easy bruisability, osteoporosis, adrenocortical atrophy, peptic
ulcers, diabetes (if chronic). Adrenal insufficiency when drug stopped after chronic use.
Mechanism of aspirin?
Irreversibly inhibits cyclooxygenase by covalent binding, which decreases synthesis of both thromboxane and prostaglandins. A type of NSAID.
Clinical use of aspirin?
Low dose (< 300 mg/day): .!-platelet aggregation. Intermediate dose (300-2400 mg/day): antipyretic and analgesic. High dose (2400-4000 mg/day): anti-inflammatory.
Toxicity of aspirin?
Gastric upset. Chronic use can lead to acute renal failure, interstitial nephritis, and upper GI bleeding. Reye's syndrome in children with viral infection.
What are the important NSAIDs?
Ibuprofen, naproxen, indomethacin, ketorolac.
Mechanism of NSAIDs?
Reversibly inhibit cyclooxygenase (both COX-1 and COX-2). Block prostaglandin
synthesis.
Clinical use of NSAIDs?
Antipyretic, analgesic, anti-inflammatory. Indomethacin is used to close a PDA.
Toxicity of NSAIDs?
Renal damage, fluid retention, aplastic anemia, GI distress, ulcers.
Mechanism of celecoxib?
Reversibly inhibit specifically the cyclooxygenase (COX) isoform 2, which is found in inflammatory cells and vascular endothelium and mediates inflammation and pain; spares COX-1, which helps maintain the gastric mucosa. Thus, should not have the corrosive effects of other NSAIDs on the GI lining.
Toxicity of celecoxib?
Increased risk of thrombosis. Sulfa allergy. Less toxicity to GI mucosa (lower incidence of ulcers,
bleeding than NSAIDs).
Mechanism of acetaminophen?
Reversibly inhibits cyclooxygenase, mostly in CNS. Inactivated peripherally.
Clinical use of celecoxib?
Rheumatoid and osteoarthritis; patients with gastritis or ulcers.
Clinical use of acetaminophen?
Antipyretic, analgesic, but lacking anti-inflammatory properties. Use in children.
Toxicity of acetaminophen?
Overdose produces hepatic necrosis; acetaminophen metabolite depletes glutathione
and forms toxic tissue adducts in liver. N-acetylcysteine is antidote-regenerates glutathione.
Mechanism of bisphosphonates? (dronate)
Inhibit osteoclastic activity; reduce both formation and resorption of hydroxyapatite.
Clinical use of bisphosphonates?
Malignancy-associated hypercalcemia, Paget's disease of bone, postmenopausal
osteoporosis.
Toxicity of bisphosphonates?
Corrosive esophagitis (except zoledronate), nausea, diarrhea, osteonecrosis of the jaw.
Mechanism of colchicine?
Acute gout (with NSAIDs). Binds and stabilizes tubulin to inhibit polymerization, impairing leukocyte chemotaxis and degranulation. GI side effects, especially if given orally.
Mechanism of probenecid?
Chronic gout. Inhibits reabsorption of uric acid in PCT (also inhibits secretion of penicillin).
Mechanism of allopurinol?
Chronic gout. Inhibits xanthine oxidase, decreased conversion of xanthine to uric acid. Also used in lymphoma and leukemia to prevent tumor lysis-associated urate nephropathy. increased concentrations of azathioprine and 6-MP
Mechanism of Etanercept?
Recombinant form of human TNF receptor that binds TNF
Mechanism of adalimumab?
Anti-TNF antibody
Mechanism of heparin?
Cofactorfortheactivationofantithrombin,decrease thrombin,and Xa.Short half-life.
Clinical use of heparin?
Immediate anticoagulation for pulmonary embolism, stroke, acute coronary syndrome, MI, DVT. Used during pregnancy (does not cross placenta). Follow PTT.
Toxicity of heparin?
Bleeding, thrombocytopenia (HIT), osteoporosis, drug-drug interactions. For rapid
reversal (antidote), use protamine sulfate (positively charged molecule that
binds negatively charged heparin).
Mechanism of Heparin-induced thrombocytopenia?
Act more on Xa, have better
bioavailability and 2-4 times longer half-life. Can be administered subcutaneously
and without laboratory monitoring. Not easily reversible.
Mechanism of low-molecular weight heparin?
Heparin binds to platelet factor IV, causing
antibody production that binds to and activates platelets leading to their clearance and resulting in a thrombocytopenic, hypercoagulable state.
Mechanism of lepirudin/ bivalirudin?
Hirudin derivatives; directly inhibit thrombin. Used as an alternative to heparin for anticoagulating patients with HIT.
Mechanism of warfarin?
Interferes with normal synthesis and y-carboxylation of vitamin K-dependent clotting factors II, VII, IX, and X and protein C and S. Metabolized by the cytochrome P-450 pathway. In laboratory assay, has effect on EXtrinsic pathway and increase PT. Long half-life.
Clinical use of warfarin?
Chronic anticoagulation. Not used in pregnant women (because warfarin, unlike heparin, can cross the placenta). Follow PT/INR values.
Toxicity of warfarin?
Bleeding, teratogenic, skin/tissue necrosis, drug-drug interactions.
Mechanism of alteplase?
Directly or indirectly aid conversion of plasminogen to plasmin, which cleaves thrombin and fibrin clots. increases PT, increases PTT, no change in platelet count.
Toxicity of alteplase?
Bleeding. Contraindicated in patients with active bleeding, history of intracranial
bleeding, recent surgery, known bleeding diatheses, or severe hypertension. Treat toxicity with aminocaproic acid, an inhibitor of fibrinolysis.
Mechanism of clopidogrel/ ticlopidine?
Inhibit platelet aggregation by irreversibly blocking ADP receptors. Inhibit fibrinogen binding by preventing glycoprotein lib/Ilia expression.
Clinical use of clopidogrel?
Acute coronary syndrome; coronary stenting, decreases incidence or recurrence of thrombotic stroke.
Mechanism of abciximab?
Monoclonal antibody that binds to the glycoprotein receptor IIb/IIIa on activated platelets, preventing aggregation.
Clinical use of abciximab?
Acute coronary syndromes, percutaneous transluminal coronary angioplasty.
Toxicity of abciximab?
Bleeding, thrombocytopenia.
What phase of the cell cycle does bleomycin effect?
G2 (synthesis of components needed for mitosis)
What phase of the cell cycle does Vinca alkaloids and taxols effect?
Mitosis
What phase of the cell cycle does antimetabolites effect?
Synthesis
What phase of the cell cycle does etoposide effect?
Synthesis and G2
Mechanism of Methotrexate (MTX)?
Folic acid analog that inhibits dihydrofolate reductase-> decreases dTMP ~> decreases DNA and
J- protein synthesis.
Clinical use of Methotrexate (MTX)?
Cancers: Leukemias, lymphomas, choriocarcinoma, sarcomas.
Non-neoplastic: Abortion, ectopic pregnancy, rheumatoid arthritis, psonas1s.
Toxicity of Methotrexate (MTX)?
Myelosuppression, which is reversible with leucovorin (folinic acid) "rescue." Macrovesicular fatty change in liver. Mucositis. Teratogenic.
Mechanism of 5-fluorouracil (5-FU)?
Pyrimidine analog bioactivated to 5F-dUMP, which covalently complexes folic acid.
This complex inhibits thymidylate synthase ~> decrease dTMP -> decreased DNA and decreased protein synthesis.
Clinical use of 5-fluorouracil (5-FU)?
Colon cancer and other solid tumors, basal cell carcinoma (topical).
Synergy with MTX.
Toxicity of 5-fluorouracil (5-FU)?
Myelosuppression, which is not reversible with leucovorin. Overdose: "rescue" with thymidine. Photosensitivity.
Mechanism of 6-mercaptopurine (6- MP)?
Purine (thiol) analog ~> decreased de novo purine synthesis.
Activated by HGPRTase.
Clinical use of 6-mercaptopurine (6- MP)?
Leukemias, lymphomas (not CLL or Hodgkin's).
Toxicity of 6-mercaptopurine (6- MP)?
Bone marrow, GI,liver. Metabolized by xanthine oxidase; thus increased toxicity with allopurinol.
Mechanism of 6-thioguanine (6-TG)?
Purine (thiol) analog ~> decreased de novo purine synthesis.
Activated by HGPRTase.
Clinical use of 6-thioguanine (6-TG)?
Acute lymphoid leukemia.
Toxicity of 6-thioguanine (6-TG)?
Bone marrow depression, liver.
Can be given with allopurinol.
Mechanism of Cytarabine (ara-C)
Pyrimidine antagonist ~> inhibition of DNA polymerase.
Clinical use of Cytarabine (ara-C)?
AML, ALL, high- grade non-Hodgkin's lymphoma.
Toxicity of Cytarabine (ara-C)?
Leukopenia,
thrombocytopenia, megaloblastic anemia.
Mechanism of Dactinomycin?
Intercalates in DNA.
Clinical use of Dactinomycin?
Wilms' tumor, Ewing's sarcoma, rhabdomyosarcoma. Used for childhood tumors (children ACT out).
Toxicity of Dactinomycin?
Myelosuppression.
Mechanism of Doxorubicin?
Generate free radicals. Noncovalently intercalate
in DNA ---> breaks in DNA ---> decreased replication.
Clinical use of Doxorubicin?
Hodgkin's lymphomas; also for myelomas, sarcomas, and solid tumors (breast, ovary, lung).
Toxicity of Doxorubicin?
Cardiotoxicity, myelosuppression, and alopecia. Toxic to tissues with extravasation.
Mechanism of Bleomycin?
Induces free radical formation, which causes breaks in DNA strands.
Clinical use of Bleomycin?
Testicular cancer,
Hodgkin's lymphoma.
Toxicity of Bleomycin?
Pulmonary fibrosis, skin changes. Minimal myelosuppression.
Mechanism of Etoposide/teniposide?
Inhibits topoisomerase II ---> increased DNA degradation.
Clinical use of Etoposide/teniposide?
Small cell carcinoma of the lung and prostate, testicular carcinoma.
Toxicity of Etoposide/teniposide?
Myelosuppression, Gl irritation, alopecia.
Mechanism of Cyclophosphamide?
Covalently X-link (interstrand) DNA at guanine N-7. Require bioactivation by liver.
Clinical use of Cyclophosphamide?
Non-hodgkin's lymphoma
Breast cancer
Ovarian cancer
Immunosuppressant
Toxicity of Cyclophosphamide,?
Myelosuppression; hemorrhagic cystitis, partially prevented with mesna (thiol group of mesna binds toxic metabolite).
Mechanism of Nitrosoureas (carmustine, lomustine)?
Require bioactivation. Cross blood-brain barrier
---7 CNS.
Clinical use o Nitrosoureas
(carmustine, lomustine)f?
Brain tumors (including glioblastoma multiforme).
Toxicity of Nitrosoureas
(carmustine, lomustine)?
CNS toxicity (dizziness, ataxia).
Mechanism of busulfan?
Alkylates DNA
Clinical use of bulsufan?
CML. Also used to ablate patient's bone marrow before bone marrow transplantation.
Toxicity of busulfan?
Pulmonary fibrosis, hyperpigmentation.
Mechanism of Vincristine/ vinblastin?
Alkaloids that bind to tubulin in M-phase and block polymerization of microtubules so that mitotic spindle cannot form. Microtubules are the vines of your cells.
Clinical use of Vincristine/ vinblastin?
Hodgkin's lymphoma, Wilms' tumor, choriocarcinoma.
Toxicity of Vincristine/ vinblastin?
V incristine- neurotoxicity (areflexia, peripheral neuritis), paralytic ileus.
VinBLASTine BLASTs Bone marrow (suppression).
Mechanism of Paclitaxel?
Hyperstabilize polymerized microtubules in M-phase so that mitotic spindle cannot break down (anaphase cannot occur).
It is TAXing to stay polymerized.
Clinical use of Paclitaxel?
Ovarian and breast cancer
Toxicity of Paclitaxel?
Myelosuppression and hypersensitivity.
Mechanism of Cisplatin, carboplatin?
Cross-link DNA.
Clinical use of Cisplatin, carboplatin?
Testicular, bladder, ovary, and lung carcinomas.
Toxicity of Cisplatin, carboplatin?
Nephrotoxicity and acoustic nerve damage.
Mechanism of Hydroxyurea?
Inhibits Ribonucleotide Reductase ~ decreases DNA Synthesis (S-phase specific).
Clinical use of Hydroxyurea?
Melanoma, CML, sickle cell disease (increases HbF).
Toxicity of Hydroxyurea?
Bone marrow suppression, GI upset.
Mechanism of Prednisone (in cancer)?
May trigger apoptosis. May even work on nondividing cells.
Clinical use of Prednisone (in cancer)?
Most commonly used glucocorticoid in cancer chemotherapy. Used in CLL, Hodgkin's lymphomas (part ofthe MOPP regimen).
Mechanism of Tamoxifen, raloxifene?
SERMs-receptor antagonists in breastand agonists in bone. Block the binding ofestrogen to estrogen receptor-positive cells.
Toxicity of Tamoxifen, raloxifene?
Tamoxifen-may incr. the risk of endometrial carcinoma via partial agonist effects; "hot
flashes." Raloxifene-no incr. in endometrial carcinoma because it is an endometrial antagonist.
Clinical use of Tamoxifen, raloxifene?
Breast cancer. Also useful to prevent osteoporosis.
Mechanism of Trastuzumab (Herceptin)?
Monoclonal antibody against HER-2 (erb-B2 ). Helps kill breast cancer cells that overexpress HER-2, possibly through antibody-dependent cytotoxicity.
Clinical use of Trastuzumab (Herceptin)?
Metastatic breast cancer.
Toxicity of Trastuzumab (Herceptin)?
Cardiotoxicity.
Mechanism of lmatinib (Gieevec)?
Philadelphia chromosome bcr-abl tyrosine kinase inhibitor.
Clinical use of lmatinib (Gieevec)?
CML, GI stromal tumors.
Toxicity of lmatinib (Gieevec)?
Fluid retention.
Mechanism of Rituximab?
Monoclonal antibody against CD20, which is found on most B-cell neoplasms.
Clinical use of Rituximab?
Non-Hodgkin's lymphoma, rheumatoid arthritis (with methotrexate).
Mechanism of Trastuzumab (Herceptin)?
Monoclonal antibody against HER-2 (erb-B2 ). Helps kill breast cancer cells that overexpress HER-2, possibly through antibody-dependent cytotoxicity.
Clinical use of Trastuzumab (Herceptin)?
Metastatic breast cancer.
Toxicity of Trastuzumab (Herceptin)?
Cardiotoxicity.
Mechanism of lmatinib (Gieevec)?
Philadelphia chromosome bcr-abl tyrosine kinase inhibitor.
Clinical use of lmatinib (Gieevec)?
CML, GI stromal tumors.
Toxicity of lmatinib (Gieevec)?
Fluid retention.
Mechanism of Rituximab?
Monoclonal antibody against CD20, which is found on most B-cell neoplasms.
Clinical use of Rituximab?
Non-Hodgkin's lymphoma, rheumatoid arthritis (with methotrexate).
Antiarrhythmic effect of adenosine?
Increases K+ out of cells ~> hyperpolarizingthe cell+ decreases Ica. Drug of choice in diagnosing/ abolishing supraventricular tachycardia. Very short acting (~ 15 sec). Toxicity includes flushing, hypotension, chest pain. Effects blocked by theophylline.
Antiarrhythmic effect of K+?
Depresses ectopic pacemakers in hypokalemia (e.g., digoxin toxicity).
Antiarrhythmic effect of Mg2+?
Effective in torsades de pointes and digoxin toxicity.
Mechanism of Ca blocker antiarrhythmic effect?
Decreased conduction velocity, Increased ERP, Increased PR interval.
Used in prevention of nodal arrhythmias (e.g. SVT)
What are the important class III antiarrhythmics?
Sotalol, ibutilide, bretylium, dofetilide, amiodarone.
Mechanism of class III antiarrhythmics?
Increased AP duration, Increased ERP. Used when other antiarrhythmics fail. Increased QT interval.
Toxicity of amiodarone?
Pulmonary fibrosis
Hepatoxicity
Hyporthyroidism/hyperthyroidism
Skin deposits (blue/gray)- photodermitis
Corneal deposits
Bradycardia, heartblock, CHF
What are the Class II antiarrhythmics?
Beta blockers
Propranolol, esmolol, metoprolol, atenolol, timolol.
Mechanism of Class II antiarrhythmics?
Decreased cAMP, decr. Ca2+ currents. Suppress abnormal pacemakers by decr. slope of phase 4. AV node particularly sensitive- incr. PR interval. Esmolol very short acting.
Clinical use of Class II antiarrhythmics?
V-tach, SVT, slowing ventricular rate during atrial fibrillation and atrial flutter.
Toxicity of Beta blockers?
Impotence, exacerbation of asthma, cardiovascular effects (bradycardia, AV block, CHF), CNS effects (sedation, sleep alterations).

Metoprolol can cause dyslipidemia. Treat overdose with glucagon.
Mechanism of Class I antiarrhythmics?
Slow or block conduction (especially in depolarized cells). decr. slope of phase 0 depolarization and incr. threshold for firing in abnormal pacemaker cells. Are state dependent (selectively depress tissue that is frequently depolarized, e.g., fast tachycardia).
Action of of Class IA antiarrhythmics?
Increases AP duration, Decr. ERP, Incr. QT interval.

Affect both atrial and ventricular arrhythmias, especially reentrant and ectopic supraventricular and ventricular tachycardia.
What are the Class IA antiarrhythmics?
"The Queen Proclaims Diso's pyramid."

Quinidine, Procainamide, Disopyramide.
Toxicity of Class IA antiarrhythmics?
quinidine (cinchonism-headache, tinnitus); thrombocytopenia; torsades de pointes due to incr. QT interval; procainamide (reversible SLE-like syndrome).
Action of Class IB antiarrhythmics?
Decreases AP duration
Preferentially affect ischemic or depolarized Purkinje and ventricular tissue. Useful in acute ventricular arrhythmias (especially post-MI) and in digitalis-induced arrhythmias.
What are the Class IB antiarrhythmics?
''I'd Buy Lidy's Mexican Tacos."

Lidocaine, Mexiletine, Tocainide.
Toxicity of Class IB antiarrhythmics?
Local anesthetic. CNS stimulation/depression, cardiovascular depression.
Action of of Class IC antiarrhythmics?
No effect on AP duration. Useful in V-tachs that progress to VF and in intractable SVT. Usually used only as last resort in refractory tachyarrhythmias. For patients without structural abnormalities.
What are the Class IC antiarrhythmics?
"Chipotle's Food has Excellent Produce."

Flecainide, Encainide, Propafenone.
Toxicity of Class IC antiarrhythmics?
proarrhythmic, especially post-MI (contraindicated). Significantly prolongs refractory period in AV node.
What is the volume of distribution? Calculation?
Vd=amount of drug/plasma drug concentration

Relates the amount ofdrug in the body to the plasma concentration. Vd ofplasma protein-bound drugs can be altered by liver and kidney disease.
How would drugs with low/ medium/ high Vd distribute?
Low Vd (4-8 L) distribute in blood.
Medium Vd distribute in extracellular space or body water.
High Vd (>body weight) distribute into all tissues.
What is the clearance? Calculation?
CL= rate of elimination/ plasma concentration
CL=Vd* Ke (elimination constant)
Relates the rate of elimination to the plasma concentration.
Calculation for Half-life?
t(1/2)= (0.7*Vd)/CL

Drugs infused at a constate rate take 4-5 half lives to reach steady state
Calculation for Loading dose?
LD= Cp*Vd/F (F=bioavaliability)
Calculation for maintenance dose?
MD= Cp*CL/F (F=bioavaliability)
What is a physiologic antagonist?
Substance that produces the opposite physiologic effect of an agonist but does not act at the same receptor.
Major function of A1 receptor?
Incr. vascular smooth muscle contraction, Incr. pupillary dilator muscle contraction (mydriasis), Incr. intestinal and bladder sphincter muscle contraction
Major function of A2 receptor?
Decreases sympathetic outflow
Decreases insulin release
Major function of B1 receptor?
Increases heart rate
Increases contractility
Increases renin release
Increases lipolysis
Major function of B2 receptor?
Vasodilation, bronchodilation, incr. heart rate, incr. contractility, incr. lipolysis, incr. insulin release, incr. uterine tone
Major function of M1 receptor?
CNS, enteric nervous system
Major function of M2 receptor?
Decreases heart rate and contractility of atria
Major function of M3 receptor?
Incr. excrine gland secretion (e.g. sweat, gastric acid) Incr. gut peristalsis, Incr. bladder contraction Bronchoconstriction, pupillary constriction, ciliary contraction (accommodation)
Major function of D1 receptor?
Relaxes renal vascular smooth muscle
Major function of Ds receptor?
Modulates transmitter release, especially in brain
Major function of H1 receptor?
Increases nasal and bronchial mucus production, contraction of bronchioles, pruritus, and pain
Major effect of H2 receptor?
Increases gastric acid secretion
Major effect of V1 receptor?
Increases vascular smooth muscle contraction
Major effect of V2 receptor?
Increases H20 permeability and reabsorption in the collecting tubules of the kidney
Which receptors are Gq (Phospolipase C, PKC)?
H1, A1, V1, M1, M3

HAVe 1 M&M
Which receptors are Gs (Adenylyl cyclase, cAMP)?
B1, B2, D1, H2, V2
Which receptors are Gi (Adenylyl cyclase, cAMP)?
M2, A2, D2

MAD 2's
Mechanism of hemicholinium?
Blocks reuptake of choline
Major effect of H2 receptor?
Increases gastric acid secretion
Major effect of V1 receptor?
Increases vascular smooth muscle contraction
Major effect of V2 receptor?
Increases H20 permeability and reabsorption in the collecting tubules of the kidney
Which receptors are Gq (Phospolipase C, PKC)?
H1, A1, V1, M1, M3

HAVe 1 M&M
Which receptors are Gs (Adenylyl cyclase, cAMP)?
B1, B2, D1, H2, V2
Which receptors are Gi (Adenylyl cyclase, cAMP)?
M2, A2, D2

MAD 2's
Mechanism of hemicholinium?
Blocks reuptake of choline
What are important sulfa drugs?
Celecoxib, furosemide, probenecid, thiazides, TMP-SMX, sulfasalazine, sulfonylureas, acetazolamide, sulfonamide antibiotics.
Toxicity of sulfa drugs?
Patients with sulfa allergies may develop fever, pruritic rash, Stevens-Johnson syndrome, hemolytic anemia, thrombocytopenia, agranulocytosis, and urticaria
Mechanism of alcohol toxicity
lcohol metabolism depletes NAD+, which is needed for fatty acid oxidation in the liver and conversion of pyruvate to lactate-> fatty liver and lactic acidosis
Important Inducers of P-450?
Queen Barb Steals Phen-phen and Refuses Greasy Carbs Chronically.

Quinidine, Barbiturates, St. John's wort, Phenytoin, Rifampin, Griseofulvin, Carbamazepine, Chronic alcohol use
Important inhibitors of P-450?
Inhibit yourself from drinking
beer from a KEG because it makes you Acutely SICk.

HIV protease inhibitors Ketoconazole Erythromycin Grapefruit juice Acute alcohol use Sulfonamides Isoniazid Cimetidine
What drugs cause disulfram-like reaction?
Metronidazole, certain cephalosporins, procarbazine, 1st-generation sulfonylureas
What drug causes nephrotoxicity and neurotoxicity?
Polymyxins
What drug causes nephrotoxicity and ototoxicity?
Aminoglycosides, vancomycin, loop diuretics, cisplatin
What drugs causes cinchonism?
Quinidine, quinine
What drugs cause diabetes insipidus?
Lithium, demeclocycline
What drugs cause parkinson-like syndrome?
Haloperidol, chlorpromazine, reserpine, metoclopramide
What drugs cause seizures?
Bupropion, imipenem/cilastatin, isoniazid
What drug cause tardive dyskinesia?
Antipsychotics
What drug causes fanconi's syndrome?
Expired tetracycline
What drugs causes interstitial nephritis?
Methicillin, NSAIDs, furosemide
What drugs causes hemorrhagic cystitis?
Cyclophosphamide, ifosfamide (prevent by coadministering with mesna)
What drugs causes tendonitis?
Fluoroquinolones
What drugs causes
Hydralazine, INH, Procainamide, Phenytoin (it's not HIPP to have lupus)
What drugs causes Stevens Johnson syndrome?
Ethosuximide, lamotrigine, carbamazepine, phenobarbital, phenytoin, sulfa drugs,
penicillin, allopurinol
What drugs causes photosensitivity?
Sulfonamides, Amiodarone, Tetracycline (SAT for a photo)
What drugs cause osteoporosis?
Corticosteroids, heparin
What drugs cause gout?
Furosemide, thiazides
What drugs cause gingival hyperplasia?
Phenytoin
What drugs cause hypothyroidism?
Lithium, amiodarone
What drugs cause hot flashes?
Tamoxifen, clomiphene
What drugs cause gynecomastia?
Spironolactone, Digitalis, Cimetidine, chronic Alcohol use, estrogens, Ketoconazole (Some Drugs Create Awesome Knockers)
What drugs cause pseudomembranous colitis?
Clindamycin, ampicillin
What drugs cause hepatitis?
INH
What drugs cause hepatic necrosis?
Halothane, valproic acid, acetaminophen, Amanita phalloides
What drugs cause acute cholestatic?
Macrolides
What drugs cause cough?
ACE inhibitors (note: ARBs like losartan-no cough)
What drugs cause pulmonary fibrosis?
BLeomycin, Amiodarone, Busulfan (it's hard to BLAB when you have pulmonary
fibrosis )
What drugs cause thrombotic complications?
OCPs (e.g., estrogens and progestins)
What drugs cause megaloblastic anemia?
Phenytoin, Methotrexate, Sulfa drugs (having a blast with PMS)
What drugs cause hemolysis in G6PD deficient patients?
Isoniazid (INH), Sulfonamides, Primaquine, Aspirin, Ibuprofen, Nitrofurantoin
(hemolysis IS PAIN)
What drugs cause Gray baby syndrome?
Chloramphenicol
What drugs cause direct coombs positive hemolytic anemia?
Methyldopa
What drugs cause Aplastic anemia?
Chloramphenicol, benzene, NSAIDs, propylthiouracil, methimazole
What drugs cause agrunolcytosis?
Clozapine, carbamazepine, colchicine, propylthiouracil, methimazole, dapsone
What drugs cause torsades de pointes?
Class III (sotalol), class lA (quinidine) antiarrhythmics
What drugs cause Dilated cardiomyopathy?
Doxorubicin (Adriamycin), daunorubicin
What drugs cause cutaneous flushing?
VANC: Vancomycin, Adenosine, Niacin, Ca2+ channel blockers
What drugs cause coronary vasospasm?
Cocaine, sumatriptan
What drugs cause atropine-like side effects?
TCAs
What is the antidote for acetaminophen?
N-acetylcysteine
What is the antidote for Salicylates?
NaHC03 (alkalinize urine), dialysis
What is the antidote for Amphetamines (basic)?
NH4Cl (acidify urine)
What is the antidote for Acetylcholinesterase inhibitors,
organophosphates?
Atropine, pralidoxime
What is the antidote for antimuscarinic, anticholinergic agents?
Physostigmine salicylate
What is the antidote for Beta blockers?
Glucagon
What is the antidote for iron?
Deferoxamine
What is the antidote for lead?
CaEDTA, dimercaprol,
succimer, penicillamine
What is the antidote for mercury, arsenic, gold?
Dimercaprol (BAL),
Succimer
What is the antidote for copper, arsenic, gold?
Penicillamine
What is the antidote for cyanide?
Nitrite, hydroxocobalamin, thiosulfate
What is the antidote for methemoglobin?
Methylene blue, vitamin C
What is the antidote for methanonl, ethylene glycol?
Ethanol, dialysis, fomepizole
What is the antidote for opioid?
Naloxone/naltrexone
What is the antidote for benzodiazepine?
Flumazenil
What is the antidote for TCAs?
NaHC03 (plasma
alkalinization)
What is the antidote for heparin?
Protamine
What is the antidote for warfarin?
Vitamin K, fresh frozen
plasma
What is the antidote for tPA, streptokinase?
Aminocaproic acid
What is the antidote for theophylline?
Beta blocker
Action of Bethanechol?
Postoperative and neurogenic ileus and urinary retention
Clinical application of Bethanechol?
Activates Bowel and Bladder
smooth muscle; resistant to AChE.
Beth Anne, call (bethanechol) me if you want to activate your Bowels and Bladder.
Toxicity of cholinesterase inhibitors or direct agonist?
DUMBBELSS.
Causes Diarrhea, Urination, Miosis, Bronchospasm, Bradycardia, Excitation ofskeletal muscle and CNS, Lacrimation, Sweating, and Salivation.
Action of Carbachol?
CARBon copy of acetylcholine.
Clinical application of Carbachol?
Glaucoma, pupillary contraction, and relief of intraocular pressure
Action of Pilocarpine?
Contracts ciliary muscle of eye (open angle), pupillary sphincter (narrow angle); resistant to AChE. PILE on the sweat and tears.
Clinical application of Pilocarpine?
Potent stimulator of sweat, tears, saliva
Action of Methacholine
Stimulates muscarinic receptors in airway when inhaled.
Clinical application of Methacholine?
Challenge test for diagnosis of asthma
Action of Neostigmine?
Increased endogenous ACh; no CNS penetration.
NEO CNS =NO CNS penetration.
Clinical use of neostigmine?
Postoperative and neurogenic ileus and urinary retention, myasthenia gravis, reversal of neuromuscular junction blockade (postoperative)
Clinical use of Pyridostigmine?
Myasthenia gravis (long acting); does not penetrate CNS
Action of Pyridostigmine?
Anticholinesterases
Clinical use of Edrophonium?
Diagnosis of myasthenia gravis (extremely short acting)
Action of Edrophonium?
Anticholinesterases
Clinical use of Physostigmine?
Glaucoma (crosses blood-brain barrier ~> CNS) and atropine overdose
Action of Physostigmine?
Anticholinesterases
Clinical use of Echothiophate
Glaucoma
Action of Echothiophate?
Irreversible Anticholinesterases
Action of atropine/ tropicamide?
Muscuranic Antagonist

Produce mydriasis and
cycloplegia
Action of benztropine?
Muscuranic Antagonist

PARKinson's disease-PARK my BENZ
Action of scopolamine?
Muscuranic Antagonist

Motion sickness
Action of ipratropium?
Muscuranic Antagonist

Asthma, COPD (I pray I can
breathe soon!)
Action of Oxybutynin/ glycopyrolate?
Muscuranic Antagonist

Reduce urgency in mild
cystitis and reduce bladder
spasms
Action of methscopolamine, pirenzepine, propantheline?
Muscuranic Antagonist

Peptic Ulcer treatment
Systemic action of atropine?
Incr. pupil dilation, cycloplegia.
Decr. secretions.
Decr. acid secretion.
Decr. motility
Decr. urgency in cystitis.
Toxicity of atropine?
Incr. body temperature (due to decr. sweating); rapid pulse; dry mouth; dry, flushed skin; cycloplegia; constipation; disorientation.
Acute-closure glucoma, BPH
Action of hexamethonium?
Ganglionic blocker. Used in experimental models
to prevent vagal reflex responses to changes in blood pressure-e.g., prevents reflex bradycardia caused by NE.
Toxicity of hexamethonium?
Severe orthostatic hypotension, blurred vision, constipation, sexual dysfunction.
Mechanism/selectivity of epinephrine?
Sympathomimetics
a 1, a2, B1, B2, low doses selective for B1
Application of epinephrine?
Anaphylaxis, glaucoma (open angle), asthma, hypotension
Mechanism/selectivity of norepinephrine?
Sympathomimetics
a1, a2> B1
Application of norepinephrine?
Hypotension (but decr. renal perfusion )
Mechanism/selectivity of Isoproterenol?
Sympathomimetics
B1=B2
Application of Dopamine?
Shock (increases renal perfusion), heart failure
Mechanism/selectivity of Dobutamine?
Sympathomimetics
D1=D2>B>a, inotropic but not chronotropic
Application of Dobutamine?
B1>B2, inotropic but not chronotropic
Mechanism/selectivity of Phenylephrine?
Sympathomimetics
A1>A2
Application of Phenylephrine?
Pupillary dilation,
vasoconstriction, nasal
decongestion
Mechanism/selectivity of Metaproterenol, albuterol, salmeterol, terbutaline?
Selective B2 agonist
Clinical use of Metaproterenol, albuterol, salmeterol, terbutaline?
MAST: Metaproterenol and
Albuterol for acute asthma; Salmeterol for long-term treatment; Terbutaline to reduce premature uterine contractions
Action of ritodrine?
B2 agonist
Reduces premature uterine contractions
Mechanism of amphetamine?
Indirect general agonist, releases stored catecholamines
Clinical use of amphetamine?
Narcolepsy, obesity, attention deficit disorder
Mechanism of ephedrine?
Indirect general agonist, releases stored catecholamines
Clinical use of ephedrine?
Nasal decongestion, urinary incontinence, hypotension
Mechanism of cocaine?
Indirect general agonist, uptake inhibitor
Clinical use of cocaine?
Causes vasoconstriction and local anesthesia
Mechanism of clonidine/ methyldopa?
Centrally acting A2-agonist, decreases central adrenergic outflow
Clinical use of clonidine/ methyldopa?
Hypertension, especially with renal disease (no decreased blood flow to kidneys)
Application/selectivity of Phenoxybenzamine (irreversible) and phentolamine (reversible)?
Nonselective A blocker

Pheochromocytoma (use phenoxybenzamine
before removing tumor, since high levels of released catecholamines will not be able to overcome blockage)
Toxicity of alpha blocker?
Orthostatic hypotension, reflex tachycardia
Application/selectivity of prazosin?
A1 blocker

Hypertension, urinary retention in BPH
Toxicity of prazosin?
1st-dose orthostatic hypotension, dizziness, headache
Application/selectivity of mirtazapine?
A2 blocker
Depression
Toxicity of mirtazapine?
Sedation, increased serum cholesterol, increased appetite
Clinical use of Beta blockers?
Hypertension
Angina pectoris
MI
SVT
CHF
Glaucoma
Migranes
Effects of Beta blockers?
Decr. cardiac output ( decr. heart rate, contractility) decr. renin secretion, decr. AV conduction velocity, slows chronic failure, decrease secretion of aqueous humor
Toxicity of Beta blockers?
Impotence, exacerbation of asthma, cardiovascular
adverse effects (bradycardia, AV block, CHF), CNS adverse effects (sedation, sleep alterations); use with caution in diabetics
What are the nonselective Beta antagonist?
ropranolol, timolol, nadolol, and pindolol
What are the Beta 1 antagonist?
Acebutolol (partial agonist), Betaxolol, Esmolol (short acting), Atenolol, Metoprolol

Cardioselective
A BEAM of B1 blockers. Advantageous in patients with comorbid pulmonary disease and diabetes
What are the nonselective alpha-1 and beta antagonist?
Carvedilol, labetalol

Hypertension
What are the partial B- agonist?
Pindolol, Acebutolol
Mechanism of epinephrine in glaucoma?
A agonist

Decrease aqueous humor synthesis due to vasoconstriction
Mechanism of brimonidine in glaucoma?
Decreased aqueous humor synthesis
Mechanism of timolol in glaucoma?
Decreased aqueous humor secretion
Mechanism of acetazolamide in glaucoma?
Decreased aqueous humor secretion due to decrease HCO3 via inhibition of carbonic anhydrase
Mechanism of cholinomimentics in glaucoma?
Decreased outflow of aqueous humor; contract ciliary muscle and open trabecular meshwork; use pilocarpine in emergencies; very effective at opening meshwork into canal of Schlemm
Side effect of epinephrine in glaucoma?
Mydriasis, stinging; do not use in closed-angle glaucoma
Side effect of brimonidine in glaucoma?
No pupillary or vision changes
Side effect of timolol in glaucoma?
No pupillary or vision changes
Side effect of acetazolamide in glaucoma?
No pupillary or vision changes
Side effect of cholinomimetics in glaucoma?
Miosis, cyclospasm
Mechanism of latanoprost? Side effect?
PGF(2A) Increased outflow of aqueous humor

Darkens color of iris (browning)
What are the opioid analgesics?
Morphine, fentanyl, codeine, heroin, methadone, meperidine, dextromethorphan.
Mechanism of opioid analgesics?
Act as agonists at opioid receptors to modulate synaptic transmission-open K+ channels, close Ca2+ channels-> Decreased synaptic transmission. Inhibit release of ACh, NE, 5-HT, glutamate, substance P.
Clinical use of opioid analgesics?
Pain, cough suppression (dextromethorphan), diarrhea (loperamide and diphenoxylate), acute pulmonary edema, maintenance programs for addicts (methadone).
Toxicity of opioid analgesics?
Addiction, respiratory depression, constipation, miosis (pinpoint pupils), additive CNS depression with other drugs. Tolerance does not develop to miosis and constipation.
Mechanism of butorphanol?
Partial agonist at opioid mu receptors, agonist at kappa receptors.
Clinical use of butorphanol?
Pain; causes less respiratory depression than full agonists.
Toxicity of butorphanol?
Causes withdrawal if on full opioid agonist.
Mechanism of Tramadol?
Very weak opioid agonist; also inhibits serotonin and NE reuptake (works on multiple neurotransmitters- "tram it all" in).
Clinical use of Tramadol?
Chronic pain
(Can decrease seizure threshold)
Mechanism of Phenytoin?
Increase Na channel inactivation
Clinical use of Phenytoin?
1st line treatment of tonic clonic seizures
Also for simple and partial
Fosphenytoin for parenteral use
Mechanism of carbamazepine?
Increase Na channel inactivation
Clinical use of carbamazepine?
1st line treatment of tonic clonic seizures

Trigeminal neuralgia
Mechanism of Lamotrigine?
Blocks voltage gated Na channels
Clinical use of Lamotrigine?
Tonic clonic, simple, and partial
Mechanism of Gabapentin?
Designed as GABA analog, but primarily inhibits HVA Ca2+ channels
Clinical use of Gabapentin?
Adjuvant seizure treatment

Also used for peripheral
neuropathy, bipolar disorder
Mechanism of Topiramate?
Blocks Na channels, Increase GABA action
Clinical use of Topiramate?
Tonic clonic, simple, and partial
Mechanism of phenobarbital?
Increases GABA(A) action
1st line in pregnant, children
Clinical use of phenobarbital?
Tonic clonic,simple and partial seizures
Mechanism of Valproic acid?
Increases Na+ channel inactivation, Increases GABA concentration
Clinical use of Valproic acid?
Tonic clonic, absence, simple, and partial seizures

Myoclonic seizures
Mechanism of Ethosuximide?
Blocks thalamic T-type Ca2+ channels
Clinical use of Ethosuximide?
Absence seizures (1st line)
Mechanism of tiagabine?
Inhibits GABA re-uptake
Clinical use of tiagabine?
Simple and partial seizures
Mechanism of Vigabatrin?
Irreversibly inhibits GABA transaminase --> Increase GABA
Clinical use of Vigabatrin?
Simple and partial seizures
Mechanism of phenytoin?
Use-dependent blockade of Na+ channels; Increases refractory period; inhibition of glutamate release from excitatory presynaptic neuron.
Toxicity of carbamazepine?
Diplopia, ataxia, blood dyscrasias (agranulocytosis,
aplastic anemia), liver toxicity, teratogenesis, induction of cytochrome P-450, SIADH, Stevens-Johnson syndrome.
Toxicity of ethosuximide?
GI distress, fatigue, headache, urticaria, Stevens- Johnson syndrome.
Toxicity of phenobarbital?
Sedation, tolerance, dependence, induction of cytochrome P-450.
Toxicity of phenytoin?
Nystagmus, diplopia, ataxia, sedation, gingival hyperplasia, hirsutism, megaloblastic anemia, teratogenesis (fetal hydantoin syndrome), SLE-like syndrome, induction of cytochrome P-450 .
Toxicity of valproic acid?
GI distress, rare but fatal hepatotoxicity (measure LFTs), neural tube defects in fetus (spina bifida), tremor, weight gain. Contraindicated in pregnancy.
Toxicity of lamotrigine?
Stevens-Johnson syndrome.
Toxicity of gabapentin?
Sedation, ataxia.
Toxicity of topiramate?
Sedation, mental dulling, kidney stones, weight loss.
Mechanism of barbiturates?
Facilitate GABAA action by Increase duration o f Cl- channel opening, thus decreases neuron firing.
Clinical use of barbiturates?
Sedative for anxiety, seizures, insomnia, induction of anesthesia (thiopental).
Mechanism of benzodiazepines?
Facilitate GABAA action by Increasing frequency of Cl- channel opening decreases REM sleep. Most have long half-lives and active metabolites.
Clinical use of benzodiazepines?
Anxiety, spasticity, status epilepticus (lorazepam and diazepam), detoxification (especially alcohol withdrawal-DTs), night terrors, sleepwalking, general anesthetic (amnesia, muscle relaxation), hypnotic (insomnia).
What are the most addictive benzodiazepine?
Short acting= TOM- Triazolam, Oxazepam, Midazolam.
Toxicity of benzodiazepine?
Dependence, additive CNS depression effects with alcohol. Less risk of respiratory depression and coma than with barbiturates.
Treat overdose with flumazenil (competitive antagonist at GABA benzodiazepine receptor).
What are the nonbenzodiazepine hypnotics?
Zolpidem (Ambien), zaleplon, eszopiclone.
Mechanism of nonbenzodiazepine hypnotics?
Act via the BZl receptor subtype and is reversed by flumazenil.
Toxicity of nonbenzodiazepine hypnotics?
Ataxia, headaches, confusion. Short duration because of rapid metabolism by liver ..
enzymes. Unlike older sedative-hypnotics, cause only modest day-after psychomotor depression and few amnestic effects. Lower dependence risk than benzodiazepines.
Which anesthetics act most rapidly?
Those with decreased solubility in the blood
What is the toxicity of inhaled anesthetics?
Hepatotoxicity (halothane), nephrotoxicity (methoxyflurane), proconvulsant
(enflurane), malignant hyperthermia (rare), expansion of trapped gas (nitrous oxide)
Mechanism of sumatriptan?
5-HT(lB/ID) agonist. Causes vasoconstriction, inhibition of trigeminal activation and vasoactive peptide release. Half-life < 2 hours.
Clinical use of sumatriptan?
Acute migraine, cluster headache attacks.
Toxicity of sumatriptan?
Coronary vasospasm (contraindicated in patients
with CAD or Prinzmetal's angina), mild tingling.
Mechanism/ use of memantine?
NMDA receptor antagonist; helps prevent excitotoxicity (mediated by Ca2+).

Alzheimer's disease
Mechanism/ use of donepezil?
Acetylcholinesterase inhibitor

Alzheimer's disease
Mechanism of selegiline?
Parkinson's disease

Selectively inhibits MAO-B, which preferentially metabolizes dopamine over NE and 5-HT, thereby increasing the availability of dopamine.
Mechanism of L-dopa/carbidopa?
Increases level of dopamine in brain. Unlike dopamine, L-dopa can cross blood-brain barrier and is converted by dopa decarboxylase in the CNS to dopamine.
Toxicity of L-dopa/carbidopa?
Arrhythmias from peripheral conversion to dopamine. Long-term use can -> dyskinesia following administration, akinesia between doses. Carbidopa, a peripheral decarboxylase inhibitor, is given with L-dopa in order to 1' the bioavailability of L-dopa in the brain and to limit peripheral side effects.
Mechanism of tolcapone?
COMT inhibitors-prevent L-dopa degradation, thereby increasing dopamine availability
Mechanism of bromocriptine?
Dopamine receptor Agonist
Action of dantrolene?
Used in the treatment of malignant hyperthermia, which is caused by inhalation anesthetics (except N20) and succinylcholine. Also used to treat neuroleptic malignant syndrome.

Prevents the release of Ca2+ from the sarcoplasmic reticulum of skeletal muscle.
Mechanism of succinylcholine?
Depolarizing
Phase I (prolonged depolarization)-no antidote. Block potentiated by cholinesterase inhibitors.
Phase II (repolarized but blocked)-antidote consists of cholinesterase inhibitors (e.g., neostigmine ).
Mechanism of tubocurarine (and other curiums)?
Antagonist of acetylcholine receptor
What are the local anesthetics?
Esters-procaine, cocaine, tetracaine;
amides-lldocalne, meplvacalne, buplvacalne (amldes have 2 I's in name).
Mechanism of local anesthetics?
Block Na+ channels by binding to specific receptors on inner portion of channel. Preferentially bind to activated Na+ channels, so most effective in rapidly firing neurons. 3° amine local anesthetics penetrate membrane in uncharged form, then bind to ion channels as charged form.
Does infected tissue require more or less local anesthetics?
More.

Alkaline anesthetics are charged and cannot penetrate membrane effectively. More anesthetic is needed in these cases.
What is the order of nerve blockade for local anesthetics?
Small-diameter fibers> large diameter. Myelinated fibers > unmyelinated fibers.
pain (lose first)> temperature> touch> pressure (lose last).
Toxicity of local anesthetics?
CNS excitation, severe cardiovascular toxicity (bupivacaine), hypertension,
hypotension, and arrhythmias (cocaine).
Clinical use of propofol?
Used for rapid anesthesia induction and short procedures. Less postoperative nausea than thiopental. Potentiates GABA(A).
Clinical use of Arylcyclohexylamines (Ketamine)?
PCP analogs that act as dissociative anesthetics. Block NMDA receptors. Cardiovascular stimulants. Cause disorientation, hallucination, and bad dreams. increases cerebral blood flow.
Clinical use of thiopental?
Thiopental-high potency, high lipid solubility, rapid entry into brain. Used for induction of anesthesia and short surgical procedures. Effect terminated by rapid redistribution into tissue and fat.
Drug used for alcohol withdrawal?
Benzodiazepines
Drug used for anorexia/bulemia
SSRIs
Drug used for anxiety?
Benzodiazepines Buspirone
SSRis
Drug used for ADHD?
Methylphenidate (Ritalin) Amphetamines (Dexedrine)
Drug used for atypical depression?
MAO inhibitors SSRis
Drug used for bipolar disorder?
"Mood stabilizers":
Lithium Valproic acid Carbamazepine
Atypical antipsychotics
Drug used for depression?
SSRis, SNRis TCAs
Drug used for depression with insomnia?
Mirtazapine
Drug used for obessive-compulsive disorder?
SSRis Clomipramine
Drug used for panic disorder?
SSRis TCAs Benzodiazepines
Drug used to treat PTSD?
SSRIs
Drug used to treat schizophrenia?
Antipsychotics
Drug used to treat tourette's syndrome?
Antipsychotics (haloperidol)
Drug used to treat social phobias?
SSRIs
Mechanism/action of Trazodone?
Primarily inhibits serotonin reuptake. Used for atypical depression, insomnia, as high doses are needed for antidepressant effects. Toxicity: sedation, nausea, priapism, postural hypotension.
Mechanism/action of maprotiline?
Blocks NE reuptake.
Atypical depression
Toxicity: sedation, orthostatic hypotension.
Mechanism/action of mirtazapine?
A2 antagonist (incr. release of NE and 5HT) and potent 5-HT2 and 5-HT3 receptor antagonist
Atypical depression
Toxicity: sedation, orthostatic hypotension.
Mechanism/action of buproprion?
Also used for smoking cessation. Incr. NE and dopamine via unknown mechanism. Toxicity: stimulant effects (tachycardia, insomnia), headache, seizure in bulimic patients. No sexual side effects.
What are the monoamine oxidase inhibitors?
Phenelzine, tranylcypromine, isocarboxazid, selegiline (selective MAO-B inhibitor).
Clinical use of MAOIs?
Atypical depression, anxiety, hypochondriasis.
Toxicity of MAOIs?
Hypertensive crisis with tyramine ingestion (in many foods, such as wine and cheese) and B-agonists; CNS stimulation. Contraindicated with SSRis or meperidine (to prevent serotonin syndrome).
What are the SNRIs?
Venlafaxine, duloxetine.
Mechanism of SNRIs?
Inhibit serotonin and NE reuptake.
Clinical use of SNRIs?
Depression. Venlafaxine is also used in generalized anxiety disorder; duloxetine is also indicated for diabetic peripheral neuropathy. Duloxetine has greater effect on NE.
Toxicity of SSRIs?
Fewer than TCAs. Gl distress, sexual dysfunction
(anorgasmia). "Serotonin syndrome" with any drug that Incr. serotonin (e.g., MAO inhibitors) hyperthermia, muscle rigidity, cardiovascular collapse, flushing, diarrhea, seizures. Treatment: cyproheptadine (5-HT2 receptor antagonist).
Clinical use of SSRIs?
Depression, OCD, bulimia, social phobias.
What are the SSRIs?
Fluoxetine, paroxetine, sertraline, citalopram.
Mechanism of buspirone?
Stimulates 5-HT lA receptors (partial agonist)
What are the TCAs?
Imipramine, amitriptyline, desipramine, nortriptyline, clomipramine, doxepin, amoxapme.
Mechanism of TCAs?
Block reuptake of NE and serotonin.
Side effects of TCAs?
Sedation, a-blocking effects, atropine-like (anticholinergic) side effects (tachycardia,
urinary retention). 3° TCAs (amitriptyline) have more anticholinergic effects than do zoTCAs (nortriptyline). Desipramine is the least sedating and has lower seizure threshold.
Toxicity of TCAs?
Convulsions, Coma, Cardiotoxicity (arrhythmias); also respiratory depression, hyperpyrexia. Confusion and hallucinations in elderly due to anticholinergic side effects (use nortriptyline).
Mechanism of Lithium?
Not established; possibly related to inhibition of phosphoinositol cascade.
Clinical use of Lithium?
Mood stabilizer for bipolar disorder; blocks relapse and acute manic events. Also SIADH.
Toxicity of lithium?
Movement (tremor) Nephrogenic diabetes insipidus, Hypothyroidism Pregnancy problems (Ebstein anomaly and malformation of the great vessels.)
What are the atypical antipsychotics?
It's atypical for old closets to quietly risper from A to Z.

Olanzapine, clozapine, quetiapine, risperidone, aripiprazole, ziprasidone.
Mechanism of atypical antipsychotics?
Block 5-HT2, dopamine, A, and H1 receptors.
Clinical use of atypical antipsychotics?
Schizophrenia- both positive and negative
symptoms. Olanzapine is also used for OCD, anxiety disorder, depression, mania, Tourette's syndrome.
Toxicity of atypical antipsychotics?
Fewer extrapyramidal and anticholinergic side effects than traditional antipsychotics. Olanzapine/clozapine may cause significant weight gain. Clozapine may cause agranulocytosis (requires weekly WBC monitoring).
What are the typical (neuroleptic) antipsychotics?
Haloperidol, trifluoperazine, fluphenazine, thioridazine, chlorpromazine (haloperidol + "-azine"s).
What are the high and low potency antipsychotics?
High potency: haloperidol, trifluoperazine, fluphenazine -neurologic side effects (extrapyramidal symptoms).

Low potency: thioridazine, chlorpromazine- non-neurologic side effects (anticholinergic, antihistamine, and A blockade effects)
Clinical use of antipsychotics?
Schizophrenia (primarily positive symptoms), psychosis, acute mania, Tourette's syndrome.
Mechanism of antipsychotics?
All typical antipsychotics block dopamine D2 receptors (Incr. [cAMP]).
Toxicity of neuroleptics
Highly lipid soluble and stored in body fat; thus, very slow to be removed from body 2. Extrapyramidal system (EPS) side effects
3. Endocrine side effects (e.g., dopamine receptor antagonism -> hyperprolactinemia -> galactorrhea)
4. Side effects arising from blocking muscarinic (dry mouth, constipation), a (hypotension), and histamine (sedation) receptors
Neuroleptic malignant syndrome (NMS)-rigidity, myoglobinuria, autonomic instability, hyperpyrexia. Treatment: dantrolene, agonists (e.g., bromocriptine).

Tardive dyskinesia- stereotypic oral-facial movements due to long-term antipsychotic use. Often irreversible.
What are the typical (neuroleptic) antipsychotics?
Haloperidol, trifluoperazine, fluphenazine, thioridazine, chlorpromazine (haloperidol + "-azine"s).
What are the high and low potency antipsychotics?
High potency: haloperidol, trifluoperazine, fluphenazine -neurologic side effects (extrapyramidal symptoms).

Low potency: thioridazine, chlorpromazine- non-neurologic side effects (anticholinergic, antihistamine, and A blockade effects)
Clinical use of antipsychotics?
Schizophrenia (primarily positive symptoms), psychosis, acute mania, Tourette's syndrome.
Mechanism of antipsychotics?
All typical antipsychotics block dopamine D2 receptors (Incr. [cAMP]).
Toxicity of neuroleptics
1. Highly lipid soluble and stored in body fat; thus, very slow to be removed from body
2. Extrapyramidal system (EPS) side effects
3. Endocrine side effects (e.g., dopamine receptor antagonism -> hyperprolactinemia -> galactorrhea)
4. Side effects arising from blocking muscarinic (dry mouth, constipation), a (hypotension), and histamine (sedation) receptors
Neuroleptic malignant syndrome (NMS)-rigidity, myoglobinuria, autonomic instability, hyperpyrexia. Treatment: dantrolene, agonists (e.g., bromocriptine).

Tardive dyskinesia- stereotypic oral-facial movements due to long-term antipsychotic use. Often irreversible.