Drugs - First Aid 2014 Flashcards

1
Q

What do these drugs do and what are their side effects? Lispro, Aspart, Glulisine

A

Insulin, rapid acting. Binds insulin receptor (Tyrosine kinase activity).
Liver: increased glucose stored as glycogen
Muscle: increased glycogen, protein synthesis; increased K+ uptake.
Fat: increased TG storage.
For DM1, DM2, GDM (post prandial control).
Toxicity: Hypoglycemia, rare hypersensitivity reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Which are the long acting insulin drugs?

A

Glargine, Detemir. Used for DM1, DM2, GDM (basal level control).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What does metformin do and what is its side effects?

A

Buguanide class. Decrease gluconeogenesis, increased glycolysis, and increased peripheral glucose uptake (insulin sensitivity).
First line therapy in DM2. Can be used in patients without islet function.
Causes GI upset; most serious adverse effect is lactic acidosis (cant use in renal failure)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What do tolubatmide and chlorpropamide do?

A

First generation Sulfonylureas. Close K+ channel in Beta cell membrane so cell depolarizes and causes insulin release. Stimulates release of endogenous insulin in type 2 DM. Requires some islet function so cant use in DM1. Risk of hypoglycemia when there is renal failure. Has disulfram like side effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What do glyburide, glimepiride, and glipizide do?

A

Second generation Sulfonylureas. Close K+ channel in Beta cell membrane so cell depolarizes and causes insulin release. Stimulates release of endogenous insulin in type 2 DM. Requires some islet function so cant use in DM1. Risk of hypoglycemia when there is renal failure. Hypoglycemia side effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What do Pioglitazone and rosiglitazone do?

A

Glitazones/Thiazolidinediones. Increased insulin sensitivity in peripheral tissue. Binds to PPAR-gamma nuclear transcription regulator (this activates genes for regulating fatty acid storage and glucose metabolism. Increases insulin sensitivty. Also increased adiponectin). Used as monotherapy in type 2 DM or combined with above agents. Side effects: weight gain, edema, hepatoxicity, heart failure. From fluid retention.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What do acarbose and miglitol do?

A

Alpha-glucosidase inhibitors. Inhibit intestinal brush-border alpha-glucosidases. Delayed sugar hydrolysis and glucose absorption resulting in decreased postprandial hyperglycemia. Used as monotherapy in type 2 DM or in combination with other agents. Side effects include GI disturbances.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What does pramlintide do?

A

Amylin analog. Decreases gastric emptying and decreases glucagon. Type 1 and type 2 DM. Side effects: hypoglycemia, nausea, diarrhea.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What does exenatide and liraglutide do?

A

GLP-1 analogs. Increased insulin and decreased glucagon release. Type 2 DM. Nausea, vomiting, pancreatitis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What do linagliptin, saxagliptin, and sitagliptin do?

A

DPP-4 inhibitors. (DPP-4 breaks down GLP-1) Increased insulin and decreased glucagon release. Type 2 DM. Mild urinary or respiratory infections.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What do propylthiouracil and methimazole do?

A

They block thyroid peroxidase, inhibiting the oxidation of iodide and the organification (coupling) to iodine. Inhibition of thyroid hormone synthesis. Propylthiouracil also blocks 5’-deiodinase, which decreases peripheral conversion of T4 to T3. Used for hypoerthroidism. PTU blocks peripheral conversion and is used in pregnancy. Side effects: skin rash, agranulocytosis, aplastic anemia, hepatotoxicity (propylthiouracil). Methimazole is a possible teratogen (aplastia cutis).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What do levothyroxine and triiodothyronine do?

A

Thyroxine replacements. Used for hypothyroidism and myxedema. Side effects: Tachycardia, heat intolerance, tremors, arrhythmias.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is somatostatin (octreotide) used for?

A

Acromegaly, carcinoid, gastrinoma, glucagonoma, esphageal varices

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is oxytocin used for?

A

Stimulates labor, uterine contractions, milk let-down; controls uterine hemorrhage.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is ADH used for?

A

central DI, but not nephrogenic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is demeclocycline used for?

A

Is an ADH antagonist and is used for SIADH. Side effects: nephrogenic DI, photosensitivity, abnormalities of bone and teeth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are glucocorticoids do?

A

Examples: Hydrocortisone, prednisone, triamcinolone, dexamethasone, beclomethasone, fludrocortisone (mineralocorticoid and glucocorticoid activity).
metabolic, catabolic, anti-inflammatory, and immunosuppressive effects mediated by interaction with glucocorticoid response elements and inhibition of transcription factors such as NFkB. Used for Addison disease, inflammation, immune suppression, and asthma. Side effects: iatrogenic Cushing syndrome- buffalo hump, moon facies, truncal obesity, muscle wasting, thin skin, easy bruisability, osteoporosis (treat with bisphosphonates), adrenocortical atrophy, peptic ulcers, diabetes (if chronic). Adrenal insufficiency when drug stopped abruptly after chronic use (from inhibition of ACTH).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are cimetidine, ranitidine, famotidine, nizatidine?

A

H2 blockers. Reversible block of histamine H2-receptors and decreases H+ secretion by parietal cells.
Clinical use: peptic ulcer, gastritis, mild esophageal reflux
Toxicity: cimetidine is a potent inhibitor of cytochrome P-450. Antiandrogenic effects (prolactin release, gynecomastia, impotence, decreased libido in males). Crosses blood brain barrier (confusion, dizziness, headaches). Cimetadine and ranitidine decrease renal excretion of creatinine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are omeprazole, lansoprazole, esomeprazole, pantoprazole, dexlansoprazole?

A

Irreversibly inhibit H+/K+ ATPase in stomach parietal cells.
Clinical use: peptic ulcer, gastritis, esophageal reflux, Zollinger-Ellison syndrome.
Toxicity: increased risk of C. difficile infection, pneumonia. Hip fractures, decreased serum Mg+2 with long term use.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are Bismuth, sucralfate?

A

Bind to ulcer base, providing physical protection and allowing HCO3- secretion to reestablish pH gradient in the mucous layer.
Clinical use: increases ulcer healing, fixes traveler’s diarrhea, and for constipation.
Increases motility.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is misoprostol?

A

A PGE1 analog. Increased production and secretion of gastric mucous barrier. Decreased acid production.
Clinical use: prevention of NSAID induced peptic ulcers (NSAIDs block PGE1 production); maintenance of a PDA. Also used to induce labor (ripens cervix)
Toxicity: Diarrhea. Contraindicated in women of childbearing potential (abortifacient).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is octreotide?

A

Long-acting somatostatin analog.
Clinical use: acute variceal bleeds, acromegaly, VIPoma, and carcinoid tumors.
Toxicity: nausea, cramps, steatorrhea.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What do antacids do?

A

Can affect absorption, bioavailability, or urinary excretion of other drugs by altering gastric and urinary pH or by delaying gastric emptying.
All can cause hypokalemia.
Overuse can also cause the following problems.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is Aluminum hydroxide?

A

It’s an antacid that causes constipation and hypophosphatemia; proximal muscle weakness, osteodystrophy, seizures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is Calcium carbonate?

A

Antacid. Causes hypercalcemia, rebound acid increase, constipation. Can chelate and decrease effectiveness of other drugs (e.g. tetracycline)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is Magnesium hydroxide?

A

Antacid. Causes diarrhea, hyporeflexia, hypotension, cardiac arrest.

Think magnesium and diarrhea. Magnesium salts will do this in general.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are magnesium hydroxide, magnesium citrate, polyethylene glycol, lactulose?

A

Provides osmotic load to draw water out. Lactulose also treats hepatic encephalopathy since gut flora degrade it into metabolites (lactic acid and acetic acid) that promote nitrogen excretion as NH4+.
Clinical use: constipation
Toxicity: diarrhea, dehydration; may be abused by bulimacs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is infliximab?

A

monoclonal antibody to TNFalpha.
Clinical use: Crohn disease, ulcerative colitis, rheumatoid arthritis, ankylosing spondylitis, psoriasis.
Toxicity: infection (including reactivation of latent TB), fever, hypotension

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is sulfasalazine?

A

A combination of sulfapyridine (anti-bacterial) and 5-aminosalicylic acid (anti-inflammatory). Activated by colonic bacteria.
Clinical use: Ulcerative colitis, Crohn disease
Toxicity: Malaise, nausea, sulfonamide toxicity, reversible oligospermia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is ondansetron?

A

It is a 5-HT3 antagonist; decreased vagal stimulation. Powerful central-acting antiemetic.
Clinical use: Control vomiting postoperatively and in patients undergoing cancer chemotherapy.
Toxicity: Headache, constipation.

Note: all -etron drugs are serotonin antagonists useful for anti-emetic purposes. e.g. granisetron and dolasetron.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is metoclopramide?

A

It is a D2 receptor antagonist. Increased resting tone, contractility, LES tone, motility. Does not influence colon transport time.
Clinical use: diabetic and post-surgery gastrparesis, antiemetic.
Toxicity: increased parkinsonian effects. Restlessness, drowsiness, fatigue, depression, nausea, diarrhea. Drug interaction with digoxin and diabetic agents. Contraindicated in patients with small bowel obstruction or Parkinson disease (D1-receptor blockade)

Note: another prokinetic drug is erythromycin (that’s why it has GI mobility side effects!)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are the alpha-agonist glaucoma drugs?

A

Epinephrine (alpha-1): Decreased aqueous humor synthesis via vasoconstriction.
Toxicity: Mydriasis and do not use in closed angle glaucoma.

Brimonidine (alpha-2): Decreased aqueous humor synthesis (inhibit cAMP production and thus less secretion).
Toxicity: Blurry vision, ocular hyperemia, foreign body sensation, ocular allergic reactions, ocular pruritis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the general principle for Glaucoma drugs?

A

decrease IOP by decreasing amount of of aqueous humor. (by inhibiting synthesis/secretion or increasing drainage).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are the beta-blocker glaucoma drugs?

A

Timolol, betaxolol, carteolol.
Decreased aqueous humor synthesis.
Toxicity: no pupillary or vision changes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What are the diuretics used for glaucoma drugs?

A

Acetazolamide. Decreased aqueous humor synthesis via inhibition of carbonic anhydrase.
Toxicity: no pupillary or vision changes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are the cholinomimetics glaucoma drugs?

A

Direct: pilocarpine, carbachol. Increased outflow of aqueous humor via contraction of ciliary muscle and opening of trabecular meshwork. Toxicity: miosis, and cyclospasm (contraction of ciliary muscle).
Use pilocarpine in emergencies- very effective at opening meshwork into canal of Schlemm.

Indirect: physostigmine (reversible), echothiophate (irreversible).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are the prostaglandin glaucoma drugs?

A

Latanoprost (PGF2alpha).

Toxicity: darkens color of iris (browning).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are morphine, fentanyl, codeine, loperamide, methadone, meperidine, dextromethorphan, diphenoxylate?

A

Opioid analgesics.
Act as agonists at opioid receptors (mu = morphine, delta = enkephaline, kappa = dynorphin) to modulate synaptic transmission - open K+ channels, close Ca+2 channels. Decreased synaptic transmission.
Inhibit release of ACh, norepinephrine, 5-HT, glutamate, substance P.
Clinical use: pain, cough suppression (dextromethorphan), diarrhea (loperamide and diphenoxylate), acute pulmonary edema, maintenance programs for heroin addicts (methadone).
Toxicity: addiction, respiratory depression, constipation, miosis (pinpoint pupils), additive CNS depression with other drugs.
Tolerance does not develop to miosis and constipation.
Toxicity treated with naloxone or naltrexone (opioid receptor antagonist).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is butorphanol?

A

Mu-opioid receptor partial agonist and kappa-opioid receptor agonist; produces analgesia.
Clinical use: severe pain (migraine, labor, etc). Causes less respiratory depression than full opioid agonists.
Toxicity: can cause opioid withdrawal symptoms if patient is also taking full opioid agonist (competition for opioid receptors). Overdose not easily reversed with naloxone.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is tramadol?

A

Very weak opioid agonist; also inhibits serotonin and norepinephrine reuptake (works on multiple neurotransmitters).
Clinical use: chronic pain
Toxicity: similar to opioids. Decreases seizure threshold. Serotonin syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is ethosuximide used for?

A

Treating generalized absence seizures.
Mech: blacks thalamic T-type Ca+2 channels.
Side effects: EFGHIJ
Ethosouximide causes Fatigue, GI distress, Headache, Itching, Stevens-Johnson syndrome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are benzodiazepines used for in epilepsy? And which ones?

A

Diazepam, Lorazepam. Used for first-line acute generalized status epilepticus. Increases GABA-A action.
Side effects: Sedation, tolerance, dependence, respiratory depression.
(Also for eclampsia seizures, but 1st line is MgSO4).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What is phenytoin used for?

A

First line in generalized tonic-clonic and prophylaxis in status epilepticus. Also used for partial simple and complex seizures.
Increased Na+ channel inactivation zero-order kinetics.
Side effects: nystagmus, diplopia, ataxia, sedation, gingival hyperplasia, hirsutism, peripheral neuropathy (from B6 deficiency), megaloblastic anemia, teratogenesis (fetal hydantoin syndrome), SLE-like syndrome, induction of cytochrome P450, lymphadenopathy, Stevens-Johnson syndrome, osteopenia.
Fosphenytoin for parenteral use.
FLOG MASSH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are carbamazepines used for in seizures?

A

First line for partial simple and complex seizures and generalized tonic-clonic seizures.
Increased sodium channel inactivation.
Side effects: diplopia, ataxia, blood dyscrasias (agranulocytosis, aplastic anemia), liver toxicity, teratogenesis, induction of cytochrome P450, SIADH, Stevens-Johnson syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What does valproic acid do for epilepsy?

A

First line for generalized tonic-clonic. Otherwise used for partial simple and complex seizures and generalized absence. Increased sodium channel inactivation and increased GABA concentration by inhibiting GABA transaminase.
Side effects: GI distress, rare but fatal hepatotoxicity (measure LFTs), neural tube defects in fetus (spina bifida), tremor, weight gain, contraindicated in pregnancy.
Also for myoclonic seizures and bipolar disorder.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What does gabapentin for epilepsy?

A

Used for partial simple and complex seizures and generalized tonic-clonic seizures. Primarily inhibits high-voltage-activated Ca+2 channels; designed as GABA analog.
Side effects: sedation, ataxia.
Also used for peripheral neuropathy, postherpetic neuralgia, migraine prophylaxis, bipolar disorder.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What does phenobarbital do for epilepsy?

A

Used for partial simple and complex seizures and generalized tonic-clonic seizures.
Increased GABA-A action.
Side effects: sedation, tolerance, dependence, induction of cytochrome P450, cardiorespiratory depression.
1st line in neonates.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What does topiramate do in epilepsy?

A

Used for partial simple and complex seizures and generalized tonic-clonic seizures.
Blocks sodium channels, increased GABA action.
Side effects: sedation, mental dulling, kidney stones, weight loss.
Also used for migraine prevention.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What does lamotrigine in epilepsy?

A

Used for partial simple and complex seizures and generalized tonic-clonic and absence seizures.
Blocks voltage-gated sodium channels.
Side effects: Stevens-Johnson syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What does levetiracetam do in epilepsy?

A

Used for partial simple and complex seizures and generalized tonic-clonic seizures.
Unknown mech; may modulate GABA and glutamate and release.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What does tiagabine do in epilepsy?

A

Used in partial simple and complex seizures.

Increased GABA by inhibiting re-uptake.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What does vigabatrin do in epilepsy?

A

Used in partial simple and complex seizures.

Increased GABA by irreversibly inhibiting GABA transaminase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is Stevens-Johnson syndrome?

A

Prodrome of malaise and fever followed by rapid onset of erythematous/purpuric macules (oral, ocular, genital). Skin lesions progress to epidermal necrosis and sloughing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What are the inducers of cytochrome P-450?

A
Inducers:
Chronic alcohol use
Modafinil
St. John's wort
Phenytoin
Phenobarbital
Nevirapine
Rifampin
Griseofulvin
Carbamazepine

Mnemonic: chronic alcohol Mona steals phen-phen and never refuses greasy carbs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What are the inhibitors of the cytochrome P-450 system?

A
Inhibitors: 
Acute alcohol abuse
Gemfibrozil
Ciprofloxacin
Isoniazid
Grapefruit juice
Quinidine 
Amiodarone
Ketoconazole (all azoles: fluconazole, itraconazole, voriconazole)
Macrolides
Sulfonamides 
Cimetidine
Ritonavir 

A cute gentleman Cipped iced Grapefruit juice quickly and kept munching on soft cinammon rolls

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What are the sulfa drugs?

A

Probenecid, Furosemide, Acetazolamide, Celecoxib, Thiazides, Sulfonamide antibiotics, Sulfasalazine, Sulfonylureas.
Patients with sulfa allergies may develop:
fever, urinary tract infection, Stevens-Johnson syndrome, hemolytic anemia, thrombocytopenia, agranulocytosis, and urticaria (hives).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What are the common barbiturates and what do you they?

A

Phenobarbital, pentobarbital, thiopental, secobarbital.
Facilitate GABA-A action by increased duration of Cl- channel opening thus decreasing neuron firing (barbidurates increase duration). Contraindicated in porphyria.
Clinical USE: Sedative for anxiety, seizures, insomnia, induction of anesthesia (thiopental).
Toxicity: respiratory and CV depression (can be fatal); CNS depression (can be exacerbated by EtOH use); dependence; drug interactions (induces cytochrome P-450). Overdose treatment is supportive (assist respiration and maintain BP).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are the common benzodiazepines and what do they do?

A

Diazepam, lorazepam, triazolam, temazepam, oxazepam, midazolam, chloridazepoxide, alprazolam.
Facilitate GABA-a action by increased frequency of Cl- channel opening. Decreases REM sleep. Most have long half-lives and active metabolites (exceptions: triazolam, oxazepam, and midazolam are short acting -> higher addictive potential).
Clinical USE: Anxiety, spasticity, status epilepticus (lorazepam and diazepam), detoxification (especially alcohol withdrawal-DTs), night terrors, sleepwalking, general anesthetic (amnesia, muscle relaxation), hypnotic (insomnia).
Toxicity: 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 benzo receptor).

Frenzodiazepines increased frequency. Benzos, barbs, and EtOH all bind GABA-a receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are the Nonbenzodiazepine hypnotics and what do they do?

A

Zolpidem (Ambien), Zaleplon, esZopiclone. All ZZZs put you to sleep.
Act via the BZ1 subtype of GABA receptor. Effects reversed by flumazenil (competitive antagonist at GABA benzo receptor).
Clinical USE: Insomnia.
Toxicity: 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. Decreased dependence risk than benzodiazepines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What are the inhaled anesthetics and what do they do?

A

Halothane, enflurane, isoflurane, sevoflurane, methoxyflurane, nitrous oxide.
Mechanism unknown
Effects: myocardial depression, respiratory depression, nausea/emesis, increased cerebral blood flow (decreased cerebral metabolic demand).
Toxicity: Hepatotoxicity (halothane), nephrotoxicity (methoxyflurane), proconvulsant (enflurane), expansion of trapped gas in a body cavity (nitrous oxide).
Malignant hyperthermia: rare, life-threatening hereditary condition in which inhaled anesthetics (except nitrous oxide) and succinylcholine induce fever and severe muscle contractions.
Tx: dantrolene.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

How do barbiturates act as IV anesthetics?

A

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 (i.e. skeletal muscle) and fat. Decreased cerebral blood flow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

How do benzodiazepines act as IV anesthetics?

A

Midazolam most common drug used for endoscopy; used adjunctively with gaseous anesthetics and narcotics. May cause severe postoperative respiratory depression, decreased BP (treat overdose with flumazenil), and anterograde amnesia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

How does Arylcyclohexylamines/Ketamine work as an IV anesthetic?

A

PCP analog that act as dissociative anesthetics. Block NDMA receptors (non-competitive antagonist of glutamate binding). CV stimulants. Cause disorientation, hallucination, and bad dreams. Increased cerebral blood flow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

How do Opioids work as IV anesthetic?

A

Morphine, fentanyl used with other CNS depressants during general anesthesia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

How do Propofol work as IV anesthetic?

A

Used for sedation in ICU, rapid anesthesia induction, and short procedures. Less postoperative nausea than thiopental. Potentiates GABA-A.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What are the local anesthetics esters?

A

Esters - procaine, cocaine, tetracaine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What are the local anesthetics amides?

A

lidocaine, mepivacine, bupivacaine, (amides have two I’s in them)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What are the mechanism of local anesthetics and what do they do?

A

Mechanism: 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. Tertiary amine local anesthetics penetrate membrane in uncharged form, then bind to ion chanels as charged form.
Principle: can be given with vasoconstrictors (usually epinephrine) to enhance local action - decreased bleeding, increased anesthesia by decreased systemic concentration.
In infected (acidic) tissue, alkaline anesthetics are charged and cannot penetrate membrane effectively -> need more anesthetic.
Order of nerve blockade: small-diameter fibers > large diameter. Myelinated fibers > unmyelinated fibers. Overall, size factor predominates over myelination such that small myelinated fibers > small unmyelinated fibers > large myelinated fibers > large unmyelinated fibers.
Order of loss: 1. pain 2. temperature 3. touch 4. pressure.
Clinical use: minor surgical procedures, spinal anesthesia. If allergic to esters, give amides.
Toxicity: CNS excitation, severe CV toxicity (bupivacaine), hypertension, hypotension, and arrhythmias (cocaine).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What are Neuromuscular blocking drugs used for?

A

Used for muscle paralysis in surgery or mechanical ventilation. Selective for motor (vs. autonomic) nicotinic receptor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What are the depolarizing neuromuscular blocking drugs and what do they do?

A

Succinylcholine - strong Ach receptor agonist; produces sustained depolarization and prevents muscle contraction.
Reversal blockade:
Phase 1 (prolonged depolarization): no antidote. Block potentiated by cholinesterase inhibitors.
Phase 2 (repolarized but blocked; ACh receptors are available, but desensitized): antidote consists of cholinesterase inhibitors.
Complications include hypercalcemia, hyperkalemia, and malignant hyperthermia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are the nondepolarizing neuromuscular blocking drugs and what do they do?

A

Tubocurarine, atracurium, mivacurium, pancuronium, vecuronium, rocuronium - competitive antagonists - compete with ACh for receptors.
Reversal of blockade: neostigmine (must be given with atropine to prevent muscarinic effects such as bradycardia), edrophonium, and other cholinesterase inhibitors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is dantrolene and what does it do?

A

prevents the release of Ca+2 from the sarcoplasmic reticulum of skeletal muscle.
Clinical use: used to treat malignant hyperthermia and neuroleptic malignant syndrome (a toxicity of antipsychotic drugs).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What are the dopamine agonists?

A

Bromocriptine (ergot), pramipexole, ropinirole (non-ergot); non-ergots are preferred.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What are the drugs that increase dopamine release?

A

Amantadine (may increase dopamine release); also used as an antiviral against influenza A and rubella; toxicity = ataxia.
L-dopa/carbidopa (converted to dopamine in CNS). Carbidopa is a peripheral decarboxylase inhibitor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What drugs prevent dopamine breakdown?

A

Selegiline (selective MAO type B inhibitor); entacopone, tolcapone (COMT inhibitors- prevent L-dopa degradation -> increased dopamine availability)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What drugs curb excess cholinerigc activity? In the context of Parkinson’s.

A

Benztropine (antimuscarinic; improves tremor and rigidity but has little effect on bradykinesia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

How does L-dopa/carbidopa?

A

increased 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. Carbidopa, a peripheral decarboxylase inhibitor, is given with L-dopa to increase the bioavailabity of L-dopa in the brain and to limit peripheral side effects.
Clinical use: Parkinson disease.
Toxicity: arrhythmias from increased peripheral formation of catecholamines. Long-term use can lead to dyskinesia following administration (on-off phenomonen), akinesia between doses.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What does selegiline do?

A

Selectively inhibits MAO-B, which preferentially metabolizes dopamine over norepinephrine and 5-HT, thereby increased availability of dopamine.
Clinical use: adjunctive agent to L-dopa in treatment of Parkinson disease.
Toxicity: may enhance adverse effects of L-dopa.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

How does memantine work in the context of Alzheimer?

A

NDMA receptor antagonist; help prevent excitotoxicity (mediated by Ca+2 )
Toxicity: dizziness, confusion, hallucinations.

From wiki: 
The drug belongs to a class of drugs called NMDA receptor antagonists, which reduce certain types of brain activity by binding to NMDA receptors on brain cells and blocking the activity of the neurotransmitter glutamate. At normal levels, glutamate aids in memory and learning, but if levels are too high, glutamate appears to overstimulate nerve cells, killing them through excitotoxicity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What do donepezil, galantamine, and rivastigmine do?

A

They are AChE inhibitors and are used for Alzheimer.

Toxicity: nausea, dizziness, insomnia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are the huntington drugs?

A

Neurotransmitter changes in Huntington disease: decreased GABA, decreased ACh, increased dopamine.
Tx:
Tetrabenazine and reserpine: inhibit vesicular monoamine transporter (VMAT); limit dopamine vesicle packaging and release.
Halperidol-dopamine receptor antagonist.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What does sumatriptan do?

A

It’s a 5-HT-1B/1D agonist. Inhibits trigeminal nerve activation; prevents vasoactive peptide release (can result in vasodilation and headaches); induces vasoconstriction. Half-life <2 hours.
Clinical USE: acute migraine, cluster headache attacks.
Toxicity: coronary vasospasm (contraindicated in patients with CAD or Prinzmetal angina), mild tingling.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

How do you treat metal toxicity? Think about Arsenic, Mercury, Lead, Copper, and Iron

A

Dimercaprol - Arsenic, Mercury, Lead

Pencillamine - copper toxicity, (also gold, arsenic, lead)

CaNA2 - EDTA - lead poisoning

Dimercaptosuccinic acid - lead poisoning

Deferoxamine/Deferasirox - acute iron poisoning and overload

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

How do calcium channel blockers work? Toxicity?

A

BLock voltage-dependent L-type Calcium channels of cardiac/smooth muscle, thereby reducing contractility
Toxicity: cardiac depression, AV block, peripheral edema, flushing, dizziness, hyperprolactinemia, constipation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What are the Ca channel blockers for vessels?

A

Vascular smooth muscle: amlodipine = nifedipine > diltiazem > verapamil
Except for Nimodipine the Dihydropyridines are used for hypertension, angina (including Prinzmetal), and Raynaud’s
Nimodipine used for Subarachnoid hemorrhage (prevents cerebral vasospasm).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are the Ca channel blockers for heart muscle?

A

Heart- verapamil > diltiazem > amlodipine = nifedipine

Non-dihydropyridine used for hypertension, angina, atrial fib/flutter.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

How does hydralazine used for and toxicity?

A

increases cGMP -> smooth muscle relaxation -> Vasodilates arterioles more than veins. Reduces afterload.
Clinical use: severe hypertension, CHF. First-line therapy for hypertension in pregnancy, with methyldopa. Frequently co-administered with a Beta-blocker to prevent reflex tachycardia.
Toxicity: compensatory tachycardia (contraindicated in angina/CAD), fluid retention, nausea, headache, angina. Lupus-like syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What drugs for hypertensive emergency?

A

nitroprusside, nicardipine, clevidipine, labetalol, and fenoldopam,

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

How does nitroprusside work?

A

short acting; increased cGMP via direct release of NO. can cause cyanide toxicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

How does fenoldopam work?

A

Dopamine D1 receptor agonist - coronary, peripheral, renal, and splanchnic vasodilation. Decreased BP and Increased natriuresis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What does nitroglycerin, isosorbide dinitrate do and toxicity?

A

Vasodilate by increased NO in vascular smooth muscle by increased cGMP and smooth muscle relaxation. Dilates veins more than arteries so decreased preload.
Clinical use: Angina, acute coronary syndrome, pulm edema.
Toxicity: Reflex tachycardia (treat with Beta blockers), hypotension, flushing, headache. Monday disease is due to loss of tolerance over the weekend such that these toxic symptoms occur on Monday with tachycardia, dizziness, and headache on re-exposure.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What does HMG-CoA reductase do and what are the drugs/uses/toxicity?

A

Lovastatin, pravastatin, simvastatin, atorvastatin, rosuvastatin.
Inhibit conversion of HMG-CoA to mevalonate, a cholesterol precursor.
Decreases LDL the most with slight decrease in TGs and slight increase in HDL.
Toxicity: Hepatotoxicity (increased LFTs), rhabdomyolysis (esp when used with fibrates and niacin).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What does Niacin do/uses/toxicity?

A

Vitamin B3. It inhibits lipolysis in adipose tissues and reduces hepatic VLDL synthesis.
Decreases LDL and increases HDL and slightly decreases TG.
Toxicity: Red, flushed face, which is decreased by aspirin or long-term use. Hyperglycemia (aconthosis nigricans). Hyperuricemia (exacerbates gout).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What do bile acid resins do/uses/toxicity?

A

Cholestyramine, colestipol, colesevelam
Decreased LDL and slightly increased HDL/TGs.

Prevent intestinal reabsorption of bile acids; liver must use cholesterol to make more (thus increasing LDL receptors and taking LDL into the liver from the blood).
Toxicity: Patients hate it - tastes bad, causes GI discomfort, decreased absorption of fat soluble vitamins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What do cholesterol absorption blockers do/uses/toxicity?

A

Ezetimibe
Decreased LDL and no effect on HDL/TGs.

Prevent cholesterol absorption at small intestine brush border.

Rare increased LFTs, diarrhea.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What do fibrates do/uses/toxicity?

A

Gemifibrozil, clofibrate, bezafibrate, fenofibrate
Bigg decrease in TGs and decrease in TGs and increase in HDL.

Upregulate LPL -> increased TG clearance.
Activates PPAR-alpha to induce HDL synthesis.

Toxicity: Myositis (increased risk with concurrent statins), hepatotoxicity (increased LFTs), cholesterol gall stones (suppresses cholesterol 7alpha-hydroxylase, especially with concurrent bile acid resins).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What do cardiac glycosides do, uses, toxicity?

A

Digoxin (75% bioavailabiity, 20-40% protein bound). Long half life at 40 hrs by urine.

Direct inhibition of Na/K ATPase leads to indirect inhibition of Na/Ca exchanger/antiport.
Increased Ca+2 leads to positive inotropy. Stimulates vagus nerve leading to decreased HR.
Uses: CHF (increased contractility); atrial fib (decreased conduction at AV node and depression of SA node).

Toxicity: Cholinergic - nausea, vomiting, diarrhea, blurry yellow vision (think Van Gogh).
ECG - increased PR interval, decreased QT, ST scooping, T-wave inversion, arrhythmia, AV block.
Can lead to hyperkalemia, which indicates poor prognosis.

Factors predisposing to toxicity - renal failure (decreased excretion), hypokalemia (permissive for digoxin binding at K+-binding site on Na/K ATPase), verapamil, amiodarone, quinidine (decreased digoxin clearance; displaces digoxin from tissue binding site).

Antidote: slowly normalize K+, cardiac pacer, anti-digoxin Fab fragments, Mg+2.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

How do Class 1 anti-arrythmics do in general?

A

Slow conduction in depolarized cells, decrease slope of phase 0 depolarization and increase threshold for firing in abnormal pacemaker cells. State dependent: affects most frequently depolarizing cells
Hyperkalemia increases toxicity because it leaves cells partially depolarized and so more sodium channels will be inactivated. Putting a Class 1 on top of this will make the situation even worse.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

How do Class 1A work, uses, toxicity?

A

Disopyramide, Quinidine, Procainamide,
Double Quarter Pounder.

Increases AP duration, Increases effective refractory period, increases QT interval.
Uses: Atrial/ventricular arrhythmias, especially re-entrant and ectopic SVT and VT.

Toxicity: heart failure (disopyramide), Cinchonism (headache, tinnitus with quinidine), reversible SLE-like syndrome (procainamide), thrombocytopenia, torsades de pointes due to increased QT interval.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

How do Class 1B work, uses, toxicity?

A

Lidocaine, Phenytoin, Mexiletine
Lettuce, Pickles, Mayo

Decreased AP duration. Preferentially affect ischemic or depolarized Purkinje and ventricular tissue.

Uses: Acute ventricular arrhythmias (especially post MI), digitalis-induced arrhythmias.
1B is Best for post-MI.

Toxicity: CNS stimulation/depression, CV depression.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

How do Class 1C work, uses, toxicity?

A

Flecainide, Propafenone
Fries with that Please.

Significantly prolongs refractory period in AV node. Minimal effect on AP duration.

Uses: SVTs, including A-fib, Last resort in refractory ventricular tachycardias.

Toxicity: proarrhythmic, especially post MI (contraindicated). IC is Contraindicated post MI in structural and ischemic heart tissue.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

How do class II blockers work, uses, toxicity?

A

Metoprolo, propranolol, esmolol, atenolol, timolol, carvedilol.
Decrease SA/AV nodal activity by decreased cAMP, decreased Ca+2 activity. Suppresses abnormal pacemakers by decreased slope 4.
AV node particularly sensitive. Increased PR interval. Esmolol very short acting.

Uses: SVT, slowing ventricular rate during atrial fibrillation and atrial flutter.

Toxicity: Impotence, exacerbation of COPD and asthma, CV effects (bradycardia, AV block, CHF), CNS effects (sedation, sleep alterations). May mask signs of hypoglycemia. Metoprolol can cause dyslipidemia. Propranolol can exacerbate vasospasm in Prinzmetal angina.
Contraindicated in cocaine users (risk of unopposed alpha-adrenergic receptor agonist activity). Treat overdose with glucagon.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

How do Class III work, uses, toxicity?

A

Amiodarone, Ibutilide, Dofetilide, Sotalol. AIDS
K+ channel blockers
Increased AP duration, increased effective refractory period. Used when other anti-arrhythmics fail. Increased QT interval.

Uses: Atrial Fib, atrial flutter, ventricular tachycardia (amiodarone, sotalol).

Toxicity: Sotalol - torsades de pointes, excessive Beta blockade.
Ibutilide - torsades de pointes.
Amiodarone - pulmonary fibrosis, hepatoxicity, hypothyroidism/hyperthyroidism (amiodarone is 40% iodine by weight), corneal deposits, skin deposits (blue/grey) resulting in photodermatitis, neurologic defects, constipation, CV effects (bradycardia, heart block, CHF).

Must check PFTs, LFTs, TFTs, when using amiodarone. 
Amiodarone has class I, II, III, IV effects and alters lipid membrane.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

How do Class IV work, uses, toxicity?

A

Verapamil, diltiazem.
Calcium channel blockers.

Decreased conduction velocity, increased effective refractory period, increased PR interval.

Uses: Prevention of nodal arrhythmias (e.g. SVT), rate control in atrial fib.

Toxicity: Constipation, flushing, edema, CV effects (CHF, AV block, sinus node depression). Verapamil has gingival hyperplasia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What does adenosine do?

A

Increased K+ out of cells -> hyperpolarizing cells and decreasing Calcium current. Can be used for slowing HR in nodal cells.
Drug of choice in diagnosing/abolishing SVT. Very short acting (15 seconds).

Adverse effects include flushing, hypotension, chest pain. Effects blocked by theophylline and caffeine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What does Mg+2 doo

A

Effective in torsades de pointes and digoxin toxicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What are the H1 first-gen/reversible blockers and what do they do?

A

Reversible inhibitors of H1 histamine receptors
Diphenhydramine, dimenhydrinate, chlorpheniramine.
Names contain en/ine or en/ate
Uses: allergy, motion sickness, sleep aid.
Toxicity: Sedation, antimuscarinic, anti-alpha-adrenergic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What are the 2nd generation H1 blockers and what do they do?

A

Loratadine, fexofenadine, desloratadine, cetirizine.
Names usually end in “adine”.
Uses: allergy.
Toxicity: far less sedating than 1st generation because of decreased entry into CNS.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

What are the expectorants and how do they work?

A

Guaifenesin - Expectorant: thins respiratory secretions; does not suppress cough reflex.
N-acetylcysteine - Mucolytic: can loosen mucous plugs in CF patients. Also used as an antidote for acetaminophen overdose.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What does Dextromethorphan do?

A

Antitussive (suppresses cough by antagonizing NMDA glutamate receptors). Synthetic codeine analog. Has mild opioid effect when used in excess. Naloxone can be given for overdose. Mild abuse potential.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What do psudoephedrine, phenylephrine do and what are its uses?

A

Sympathomimetic alpha-agonistic nonprescription nasal decongestants.
Use: Reduce hyperemia, edema, and nasal congestion. open obstructed eustachian tubes. Pseudoephedrine also illicitly used to make methamphetamine.
Toxicity: hypertension. can also cause CNS stimulation/anxiety (psuedophedrine).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What are the B2 agonists and how do they work? list 3

A

Albuterol - relaxes bronchial smooth muscle (Beta-2). Use during acute exacerbation.
Salmeterol, formoterol - long-acting agents for prophylaxis. Adverse effects are tremor and arrhythmia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are the methylxanthines and what do they do?

A

Theophylline - likely causes bronchodilation by inhibiting phosphodiesterase -> increases cAMP levels due to decreased cAMP hydrolysis.
Usage limited because of narrow therapeutic index (cardiotoxicity, neurotoxicity); metabolised by cytochrome P-450.
Blocks actions of adenosine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What are the muscarinic antagonists and what do they do? list 2.

A

Ipratropium - competitive block of muscarinic receptors, preventing bronchoconstriction. Also used for COPD, as is tiotropium, a long-acting muscarinic antagonist.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are the corticosteroids and what do they do?

A

Beclomethasone, fluticasone - inhibit the synthesis of virtually all cytokines. Inactivate NF-kB, the TF that induces the production of TNF-alpha and other inflammatory agents. 1st line therapy for chronic asthma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What are the anti-leukotrienes and what do they do?

A

Montelukast, zafirlukast - block leukotriene receptors. Especially good for aspiring induced asthma.
Zileuton - a 5-lipoxygenase pathway inhibitor. Blocks conversion of arachidonic acid to leukotrienes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is omalizumab and what does it do?

A

Monoclonal anti-IgE antibody. Binds mostly unbound serum IgE and blocks binding to FceRI. Used in allergic asthma resistant to inhaled steroids and long-acting B2 agonists.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What does methacoline do?

A

Muscarinic receptor agonist. Used in bronchial provocation challange to help diagnose asthma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What does Bosentan do?

A

Used to treat pulmonary arterial hypertension. Competitively antagonizes endothelin-1 receptors, decreased pulmonary vascular resistance.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What inhibits lipoxygenase?

A

Zileuton

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What inhibits LTC4, LTD4, LTE4?

A

Zafirlukast, montelukast. Receptor antagonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What is the function of LTB4 vs LTC4/LTD4/LTE4?

A

LTB4: Neutrophil chemotaxis (neutrophils arive B4 others)
LTC4/LTD4/LTE4: Increased bronchial tone, vasoconstriction, contraction of smooth muscle, and increased vascular permeability.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What does Prostacyclin do (PGI2)?

A

Decreases platelet aggregation, vascular tone, bronchial tone, uterine tone.
Platelet Gathering Inhibitor (mnemonic)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What do PGE2/PGF2alpha do?

A

Increased uterine tone and decreased bronchial tone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What does thromboxane do?

A

Increased platelet aggregation, vascular tone, and bronchial tone.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What does aspirin do and side effects?

A

Irreversibly inhibits COX1/COX2 by covalent acetylation. Decreased TXA2 and prostaglandins and increased bleeding time until new platelets are produced (about 7 days). No effect on PT/PTT. Type of NSAID.

With increasing doses you get, decreased platelet aggregation, antipyretic/analgesic, and antiinflammatory actions (in order of increasing dosage)

Toxicity: gastric ulceration, tinnitus (CNVIII). Chronic use can lead to acute renal failure, interstitial nephritis, and upper GI bleeding. Risk of Reye syndrome in children treated with aspirin for viral infection. Also stimulates respiratory centers, causing hyperventilation and respiratory alkalosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What are common NSAIDs and what do they do/toxicity?

A

Ibuprofen, naproxen, indomethacin, ketorolac, diclofenac.

Reversibly inhibit COX1/COX2.
Antipyretic, analgesic, anti-inflammatory. Indomethacin used to close a PDA.

Toxicity: interstitial nephritis, gastric ulcer (PGs protect gastric mucosa), renal ischemia (PGs vasodilate afferent arteriole).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What do COX2 inhibitors do and what are they used for/toxicity?

A

Reversible inhibition of COX2. Blocks pain/inflammation but spares platelet function (TXA2 dependent on COX1).
Use: Rheumatoid arthritis, osteoarthritis; patients with gastric ulcers.

Toxicity: increased risk of thrombosis. Sulfa allergy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What is Acetominophen used for/toxicity?

A

Reversibly inhibits COX, mostly in CNS. Inactivated peripherally.

Use: antipyretic, analgesic, but not anti-inflammatory. Used instead of aspirin to avoid Reye syndrome in children with viral infection.

Toxicity: overdose produces hepatic necrosis; acetominophen metabolite (NAPQI) depletes glutathione and forms toxic tissue adducts in liver. N-acetylcysteine is antidote - regenerates glutathione.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What are bisphosphonates used for?

A

Alendronate, other -dronates.
Mechanism: Pyrophosphate analogs; bind hydroxyapatite in bone, inhibiting osteoclast activity (and induces apoptosis).

Use: Osteoporosis, hypercalcemia, Paget disease of bone.

Toxicity: Corrosive esophagitis (patients are advised to take with water and remain upright for 30 minutes), osteonecrosis of the jaw.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What are the chronic gout drugs?

A

Allopurinol: inhibits xanthin oxidase, decreases xanthine to uric acid. Also used for tumor lysis syndrome. You see increase in azothioprine and 6-MP (before xanthine oxidase). Don’t give salicylates because they depress uric acid clearance.

Febuxostat: inhibits xanthine oxidase.

Probenecid: Inhibits reabsorption of uric acid in PCT (also inhibits secretion of penicillin).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What are the acute gout drugs?

A

NSAID: naproxen, indomethacin.
Glucocorticoids: oral/intra-articular
Colchicine: binds and stabilizes tubulin to inhibit microtubule polymerization, impairing leukocyte chemotaxis and degranulation. Acute and prophylactic value. GI side effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What do the TNF-alpha inhibitors do/toxicity?

A

Etanercept: fusion protein of TNF-alpha receptor + IgG1 FC. Decoy receptor.
Use: RA, psoriasis, ankylosing spondylitis.

Inflixamab/adalimumab - anti-TNF-alpha monoclonal antibody.
Use: IBD, RA, psoriasis, ankylosing spondylitis.

All TNF-alpha inhibitors predispose to an infection, including re-activation of TB, since TNF blockade prevents activation of macrophages and destruction of phagocytosed microbes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What does Leuprolide do/toxicity?

A

Mechanism: GnRH analog with agonist properties when used in pulsatile function; antagonist properties when used in continuous function (downregulates GnRH receptor in pituitary leading to decreased FSH/LH).
Use: infertility (pulsatile), prostate cancer (continuous - use with flutamide), uterine fibroids (continuous), precocious puberty (continuous)
Toxicity: antiandrogen, nausea, vomiting

Leuprolide in Lieu of GnRH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

what do the estrogens do/use/toxicty?

A

Ethinyl estradiol, DES, mestranol
Binds estrogen receptors
Use: hypogonadism or ovarian failure, menstrual abnormalities, HRT postmenopausal women; use in men with androgen-dependent prostate cancer
Toxicity: 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. Contraindications- ER positive breast cancer or history of DVTs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is clomiphene used for?

A

SERM.
Antagonist at estrogen receptors in hypothalamus. Prevents normal feedback inhibition and increased release of LH/FSH from pituitary, which stimulates ovulation.
Used to treat infertility due to anovulation (PCOS).
Toxicity: may cause hot flashes, ovarian enlargement, multiple simultaneous pregnancies, and visual disturbances.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is Tamoxifen used for?

A

Antagonist on breast tissue; agonist at uterus, bone;
Associated with endometrial cancer and thromboembolic events.
Primarily used to treat and prevent recurrence of ER positive breast cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is raloxifene used for?

A

Agonist on bone; antagonist at uterus.
Causes decreased resorption of bone and so is used to treat osteoporosis.
Toxicity: increased risk of thromboembolic events.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What is hormone replacement therapy?

A

Used for relief or prevention of menopausal symptoms(e.g. hot flashes, vaginal atrophy) and osteoporosis (it increases estrogen and decreases osteoclast activity)
Unopposed estrogen replacement therapy (ERT) increases the risk of endometrial cancer, so progesterone is added.
Possible increased CV risk.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What are anastrozole/exemestane?

A

Aromatase inhibitors used in postmenopausal women with breast cancer. Decreases estrogen production.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are progestins used for?

A

Bind progesterone receptors, decreases growth and increased vascularization of endometrium.
Use: used in oral contraceptives and in treatment of endometrial cancer and abnormal uterine bleeding.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What is MIfepristone used for? it’s other name?

A

RU-486.
Competitive inhibitor of progestins at progesterone receptors.
Use: termination of pregnancy. Administered with misoprostol (PGE1).
Toxicity: heavy bleeding, GI effects (nausea, vomiting, anorexia), abdominal pain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

what is oral contraception?

A

Estrogen and progestins inhibit LH/FSH and thus prevent estrogen surge. No estrogen surge means no LH surge and no ovulation.
Progestins cause thickening of the cervical mucus, thereby limiting access of sperm to uterus. Progestins also inhibit endometrial proliferation, thus making endometrium less suitable for the implantation of embryo.
Contraindications: smokers > 35 yrs old (increased risk of CV events), patients with history of thromboembolism and stroke or history of estrogen-dependent tumor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What is terbutaline used for?

A

B2-agonist that relaxes the uterus; used to decrease contraction frequency in women during labor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What is danazol used for?

A

Synthetic androgen that acts as partial agonist at androgen receptors.
Use: Endometriosis and hereditary angioedema.
Toxicity: weight gain, edema, acne, hirsuitism, masculinization, decreased HDL levels, hepatotoxicity.

From wiki:
Danazol exhibits hypoestrogenic, hyperandrogenic effects that cause atrophy of the endometrium, which can alleviate the symptoms of endometriosis.

Danazol prevents ovulation by suppressing the increase of luteinizing hormone during the middle of the menstrual cycle.[4][5] Danazol inhibits ovarian steroidogenesis resulting in decreased secretion of estradiol and may increase androgens. Danazol displaces testosterone from sex hormone-binding globulin (SHBG), displacing it and increasing serum testosterone levels.[4] Danazol also directly stimulates androgen and progesterone receptors.[4]

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What is testosterone/mehtyltestosterone used for?

A

Agonist at androgen receptors
Use: treats hypogonadism and promotes development of secondary sex characteristics; stimulation of anabolism to promote recovery after burn or injury.
Toxicity: causes masculinization in females; decreased intratesticular testosterone in males by inhibiting release of LH (via neg feedback) and resultant gonadal atrophy. Premature closure of epiphyseal plates. Increased LDL and decreased HDL.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What is finasteride used for?

A

5-alpha reductase inhibitor (decreased conversion of testosterone to DHT).
Used in BPH. Also promotes hair growth-used to treat male pattern baldness.

To prevent male pattern hair loss, give a drug that will encourage female breast growth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What is flutamide used for?

A

A nonsteroidal competitive inhibitor of androgens at the testosterone receptor.
Used in prostate carcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What is ketoconazole used for?

A

Inhibits steroid synthesis (inhibits 17,20 desmolase)

Uses: Ketaconazole and spironolactone are used in the treatment of PCOS to prevent hirsutism. Both have side effects of gynecomastia and amenorrhea.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

what is spironolactone used for?

A

Inhibits steroid binding, 17-alpha hydroxylase, and 17,20 desmolase

Uses: Ketaconazole and spironolactone are used in the treatment of PCOS to prevent hirsutism. Both have side effects of gynecomastia and amenorrhea.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What is tamsulosin used for?

A

alpha-1-antagonist used to treat BPH by inhibiting smooth muscle contraction. Selective for alpha1A,D receptors (found on prostate) vs. vascular alpha1B receptors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What are sildenafil, vardenafil used for?

A

Inhibit phosphodiesterase 5, causing increased cGMP, smooth muscle relaxation in the corpus cavernosum, increased blood flow, and penile erection.
SildaneFIL and vardenaFIL fill the penis.

Use: treatment of erectile dysfunction.

Toxicity: headache, flushing, dyspepsia, impaired blue green color vision.
Risk of life threatening hypotension in patients taking nitrates.
“Hot and sweaty” but then Headache, Heartburn, and Hypotension. All the Hs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is Penicillin G,V, mechanism, use and toxicity?

A

Penicillin G (IV/IM forms), penicillin V (oral). Prototype B-lactam antibiotics.
Mechanism: bind penicillin-binding proteins (transpeptidases). Block transpeptidase cross-linking of peptidoglycan. Activate autolytic enzymes.
Use: mostly used for gram-pos organisms (S. pneumoniae, S. pyogenes, Actinomyces). Also used for N. meningitidis and T pallidum. Bactericidal for gram-pos cocci, gram-positive rods, gram-negative cocci, and sphirochetes. Penicillinase sensitive.
Toxicity: hypersensitivity reactions, hemolytic anemia.
Resistance: pencillinase in bacteria (a type of B-lactamase) cleaves B-lactam ring.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What is ampicillin, amoxicillin, mechanism, use, toxicity, and resistance?

A

Amino penicillins, penicillinase-sensitive penicillins.
Mechanism: Same as penicillin. Wider spectrum; penicillinase sensitive. Also combine with clavulanic acid to protect against B-lactamase.
Use: Extended spectrum penicillin - Haemophilus influenzae, E. coli, Listeria Monocytogenes, Proteus, Salmonella, Shigella, enterococci. HELPSS mnemonic.
Toxicity: Hypersensitivty reactions; rash; pseudomembranous colitis.
Mechanism of Resistance: Penicillinase in bacteria (a type of B-lactamase) cleaves B-lactam ring.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What is oxacillin, nafcillin, dicloxacillin? mech, use, toxicity

A

Pencillinase-resistance penicillins
Mech: same as penicillin. Narrow spectrum; pencillinase resistant because bulky R group blocks access of B-lactamase to B lactam ring.
Use: Staph aureus (except MRSA; resistant because of altered penicillin-binding protein target site). Use NAFcillin for staph.
Toxicity: Hypersensitivity reactions, interstitial nephritis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What are ticarcillin, piperacillin? mechanism, use, toxicity?

A

Anti-pseudomonals.
Mechanism: same as penicillin, extended spectrum.
Use: Pseudomonas spp. and gram-neg rods; susceptible to penicillinase; use with B-lactamase inhibitors.
Toxicity: hypersensitivity reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What are the B-lactamase inhibitors?

A

CAST.
Include clavulanic acid, sulbactam, tazobactam. Often added to penicillin antibiotics to protect the antibiotic from destruction by B-lactamase (penicillinase).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is the mechanism of cephalosporins? Toxicity?

A

B-lactam drugs that inhibit cell wall synthesis but are less susceptible to penicillinases. Bactericidal.
Organisms typically not covered by cephalosporins are LAME: Listeria, Atypicals (Chlamydia, Mycoplasma), MRSA, and Enterococci. Exception: ceftaroline covers MRSA.
Toxicity: Hypersensitivity reactions, vitamin K deficiency.
Low cross-reactivity with penicillins. Increased nephrotoxicity of aminoglycosides.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What are cefazolin, cephalexin?

A

1st generation cephalosporins
Gram positive cocci: Proteus mirabilis, E. Coli, Klebsiella pneumoniae. Cefazolin used prior to surgery to prevent S. aureus wound infections. PEcK.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What are cefoxitin, cefaclor, cefuroxime?

A

2nd generation cephalosporins.
Gram-positive cocci: Haemophilus influenzae, Enterobacter aerogenes, Neisseria spp., Proteus mirabilis, E. coli, Klebsiella pneumoniae, Serratia marcescens.
HEN PEcKS.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What are ceftriaxone, cefotaxime, ceftazidime?

A

3rd generation cephalosporins.
Serious gram-negative infections resistant to other B-lactams.
Ceftriaxone: meningitis and gonorrhea.
Ceftazidime: Pseudomonas.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What is cefepime?

A

4th generation cephalosporin

Increased activity against Pseudomonas and gram-positive organisms.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What is ceftaroline?

A

5th Generation cephalosporin.

Broad gram-positive and gram-negative organism coverage, including MRSA; does not cover Pseudomonas.

164
Q

What does aztreonam do? mechanism, use, toxicity

A

Mechanism: a monobactam; resistant to B-lactamases. Prevents peptidoglycan cross-linking by binding to penicillin-binding protein 3. Synergistic with aminoglycosides. No cross-allergenicity with penicillins.
Clinical use: 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: Usually non toxic; occasional GI upset.

165
Q

What are Imipenem, meropenem, ertapenem, doripenem? use, mechanism, toxicity

A

Carbapenems.
Imipenem is a broad-spectrum, B-lactamase-resistant carbapenem. Always administered with cilastatin (inhibitor of renal dehydropeptidase I) to decrease inactivation of drug in renal tubules.
With imipenem, “the kill is lastin’ with cilastatin”.
Use: Gram-positive cocci, gram negative rods, and anaerobes. Wide spectrum, but significant side effects limit use to life-threatening infections or after other drugs have failed. Meropenem has a decreased risk of seizures and is stable to dehydropeptidase I.
Toxicity: GI Distress, skin rash, and CNS toxicity (seizures) at high plasma levels.

166
Q

What is vancomycin? mechanism, use, toxicity.

A

Mechanism: Inhibits cell wall peptidoglycan formation by binding D-ala D-ala portion of cell wall precursors. Bactericidal.
Use: gram positive only- serious, multidrug-resistant organisms, including MRSA, enterococci, and Clostridium difficile (oral dose for pseudomembranous colitis).
Toxicity: well-tolerated in general, but NOT trouble free. Nephrotoxicity, Ototoxicity, Thrombophlebitis, diffuse flushing - red man syndrome (can largely prevent by pre-treatment with antihistamines and slow infusion rate).
Resistance mechanism: Occurs in bacteria via amino acid modification of D-ala D-ala to D-ala D-lac. Pay back 2 D-alas (dollars) for vandalizing (vancomycin).

167
Q

Which are the 30S and 50S inhibitors and label them as bacteriostatic or bateriocidal. And what part of the cycle they act at.

A

30S Inhibitors:
Aminoglycosides (bactericidal) -block Initiation complex formation (causes misreading of mRNA)
Tetracyclines (bacteriostatic) - blocks A-site tRNA binding.

50S inhibitors:
Chloramphenicol (bacteriostatic) - blocks peptidyl transferase
Clindamycin (bacteriostatic) - blocks translocation
Erythromycin (macrolides) (bacteriostatic) - blocks translocation
Linezolid (variable) - blocks binding of everything to mRNA and initiation complex.

buy AT 30, CCEL at 50

168
Q

What are gentamicin, neomycin, amikacin, tobramycin, and streptomycin? mech, use, toxicity, and resistance

A

Aminoglycosides. Mean (aMINoglycoside) GNATS caNNOT kill anaerobes.
Mechanism: Bactericidal; inhibit formation of initiation complex and cause misreading of mRNA. Also block translocation. Require O2 for uptake; therefore ineffective against anaerobes.
Use: severe gram-negative rod infections. Synergistic with Beta-lactam antibiotics. Neomycin for bowel surgery.
Toxicity: Nephrotoxicity (especially with cephalosporins), Neuromuscular blockade, Ototoxicity (especially when used with loop diuretics). Teratogen.
Resistance: bacterial transferase enzymes inactivate the drug by acetylation, phosphorylation, or adenylation.

169
Q

What are tetracycline, doxycycline, and minocycline? mech, use, toxicity, and resistance

A

Mechanism: 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. Do not take with milk (Ca+2), antacids (Ca+2 or Mg+2) or iron containing preparations because divalent cations inhibit its absorption in the gut.
Use: Borrelia Burgodorferi, M. pneumoniae. Drug’s ability to accumulate intracellularly makes it very effective against Rickettsia and Chlamydia. Also used to treat acne.
Toxicity: GI distress, discoloration of teeth and inhibition of bone growth in children, photosensitivity. Contraindicated in pregnancy.
Resistance: decreased uptake or increased efflux out of bacterial cells by plasmid-encoded transport pumps.

170
Q

What are azithromycin, clarithromycin, erythromycin? mechanism, clinical use, toxicity, mech of resistance

A

Macrolides. Inhibit protein synthesis by blocking translocation (macroslides); bind to the 23S rRNA of the 50S ribosomal subunit. Bacteriostatic.
Use: atypical pneumonias (mycoplasma, chlamydia, legionella), STDs (for Chlamydia), and gram-positive cocci (streptococcal infections in patients allergic to penicillin).
Toxicity: MACRO: GI Motility issues (it’s prokinetic), Arrhythmia caused by prolonged QT, acute Cholestatic hepatitis, Rash, eOsinophilia. Increases serum concentration of theophyllines, oral anti-coagulants.
Resistance: methylation of 23S rRNA-binding site prevents binding of drug.

171
Q

What is chloramphenicol? mech, use, toxicity, resistance

A

Blocks peptidyltransferase at 50S ribosomal subunit. Bacteriostatic.
Use: Meningitis (H. influenzae, Neisseria meningitidis, Strep pneumoniae) and Rocky mountain spotted fever (rickettsia rickettsii).
Limited use owing to toxicities but often still used in developing countries because of low cost.
Toxicity: Anemia (dose dependent), aplastic anemia (dose independent), gray baby syndrome (in premature infants because they lack liver UDP-glucoronyl transferase).
Resistance: plasmid-encoded acetyltransferase inactivates the drug.

172
Q

What is clindamycin? mechanism, use, toxicity

A

Blocks peptide transfer (translocation) at 50S ribosomal subunit. Bacteriostatic.
Use: anaerobic infections (e.g. bacteroides spp. Clostridium perfringes) in aspiration pneumonia, lung abscesses, and oral infections. Also effective against invasive group A streptococcal (GAS) infection. Also can be used for acne.
Treats anaerobes above the diaphragm vs. metronidazole (anaerobic infections below diaphragm).
Toxicity: Pseudomembranous colitis (C. difficile overgrowth which should be treated with metronidazole or vancomycin), fever, diarrhea.

173
Q

What is sulfamethoxazole (SMX), sulfisoxazole, sulfadiazine? mech, use, toxicity, resistance

A

Inhibit folate synthesis. Para-aminobenzoic acid (PABA) antimetabolites inhibit dihydropteroate synthase. Bacteriostatic.
Use: gram-pos, gram-neg, Nocardia, Chlamydia. Triple sulfas or SMX for simple UTI.
Toxicity: Hypersensitivity reactions, hemolysis if G6PD deficient, nephrotoxicity (tubulointerstitial nephritis), photosensitivity, kernicterus in infants, displace other drugs from albumin (e.g. warfarin).
Resistance: altered enzyme (bacterial dihydropteroate synthase), decreased uptake, or increased PABA synthesis.

174
Q

What is trimethoprim?

A

Inhibits bacterial dihydrofolate reductase. Bacteriostatic.
Use: used in combo with sulfonamides (TMP-SMX), causing sequential block of folate synthesis. Combination used for UTIs, Shigella, Salmonella, Pneumocystis jirovecii pneumonia treatment and prophylaxis, toxoplasmosis prophylaxis.
Toxicity: megaloblastic anemia, leukopenia, granulocytopenia, (may alleviate with folic acid supplementation). TMP: treats marrow poorly.

175
Q

What are ciprofloxacin, norfloxacin, levofloxacin, ofloxacin, sparfloxacin, moxifloxacin, gemifloxacin, enoxacin, and nalidixic acid?

A

All are fluoroquinolones, except for nalidixic acid which is a quinalone.
Mech: inhibit DNA gyrase (topoisomerase II) and topoisomerase IV. Bactericidal. Must not be taken with antacids.
Use: gram neg rods of urinary and GI tracts (including pseudomonas), Neisseria, some gram pos organisms.
Toxicity: GI upset, superinfections, skin rashes, headache, dizziness. Less commonly, can cause tendonitis, tendon rupture, leg cramps, and myalgias. Contraindicated in pregnant women, nursing mothers, and children under 18 years old due to possible damage to cartilage. Some may cause prolonged QT interval. May cause tendon rupture in people > 60 years old and in patients taking prednisone.
FluoroquinoLONES hurt attachments to your BONES.

Resistance: chromosome-encoded mutation in DNA gyrase, plasmid-mediated resistance, efflux pumps.

176
Q

What is metronidazole?

A

Mechanism: Forms free radical toxic metabolites in the bacterial cell that damages DNA. bactericidal, antiprotozoal.
Use: Treats Giardia, Entamoeba, Trichomonas, Gardnerella vaginalis, Anaerobes (Bacteroides, C. difficile). Used with a proton pump inhibitor and clarithromycin for “triple therapy” against H. pylori.
GET GAP on the Metro with METROnidazole! Treat anaerobic infection below the diaphragm vs clindamycin (which is anaerobic infections above the diaphragm).
Toxicity: Disulfiram-like reaction (severe flushing, tachycardia, hypotension) with alcohol; headache, metallic taste.

177
Q

What is the drug for M. tuberculosis? prophylaxis and treatment

A

Prophlaxis: Isoniazid
Treatment: Rifampin, Isoniazid, Pyrazinamide, Ethambutol
RIPE for treatment.

178
Q

What is the drug for M. avium-intracellulare? prophylaxis and treatment

A

Prophylaxis: Azithromycin, Rifabutin
Treatment: More drug resistant than M. tuberculosis. Azithromycin or clarithromycin + ethambutol. Can add rifabutin or ciproflaxacin.

179
Q

What is the drug for M. leprae?

A

Long term treatment with dapsone and rifampin for tuberculoid form. Add clofazimine for lepromatous form.

180
Q

What is isoniazid? Mech, use, toxicity.

A

Mech: decreased synthesis of mycolic acids. Bacterial catalase-peroxidase (encoded by KatG) needed to convert INH to active metabolite.
Use: Mycobacterium tuberculosis. The only agent as solo prophylaxis against TB. Different INH half-lives in fast vs. slow acetylators.
Toxicity: Neurotoxicity, hepatotoxicity. Pyridoxine (B6) can prevent neurotoxicity, lupus.
INH injures Neurons and Hepatocytes.

181
Q

What are rifampin and rifabutin?

A

Rifamycins
Mech: inhibits DNA-dependent RNA polymerase
Use: M. tuberculosis; delays resistance to dapsone when used fro leprosy. Used for menigococcal prophylaxis and chemoprophylaxis in contacts of children with H. influenzae type B.
Rifampin’s four Rs: RNA polymerase inhibitor, Ramps up microsomal cytochrome P450, Red/orange body fluids, Rapid resistance if used alone, Rifampin ramps up cytochrome P-450, but rifabutin does not.

182
Q

What is pyrazinamide? mech, use and toxicity

A

Mechanism uncertain. Thought to acidify intracellular environment via conversion to pyrazinoic acid. Effective in acidic pH of phagolysosomes, where TB engulfed by macrophages is found.
Use: Mycobacterium tuberculosis.
Toxicity: Hyperuricemia, hepatotoxicity.

183
Q

What is ethambutol? mech, use, toxicity

A

Decreased carbohydrate polymerization of mycobacterium cell wall by blocking arbinosyltransferase.
Use: mycobacterium tuberculosis.
Toxicity: optic neuropathy (red-green color blindness).

184
Q
What is used for prophylaxis in HIV patients? 
For the below list drug and infection
CD4 < 200
CD4 < 100
CD4 < 50
A

200: Pneumocystis pneumoniae with TMP-SMX
100: Pneumocystis pneumonia and toxoplasmosis with TMP-SMX
50: Mycobacterium avium complex with Azithromycin

185
Q

Antimicrobial prophylaxis:

1) Endocarditis with surgical/dental procedures
2) Gonorrhea
3) History of recurrent UTIs
4) Meninogoccal infection
5) Pregnant woman carrying group B strep
6) Prevention of gonococcal or chlamydial conjunctivitis in newborn
7) Prevention of postsurgical infection due to S. aureus
8) Prophylaxis of strep pharyngitis in child with prior rheumatic fever
9) Syphilis

A

1) Penicillins
2) Ceftriaxone
3) TMP-SMX
4) Ciprofloxacin (drug of choice), rifampin for children
5) Ampicillin
6) Erythromycin ointment
7) Cefazolin
8) Oral penicillin
9) Benzathine penicillin G

186
Q

What is the treatment of highly resistant bacteria?
MRSA - three
VRE - two

A

MRSA - vancomycin, daptomycin, linezolid (can cause serotonin syndrome, optic neuritis, thrombocytopenia), tigecycline, ceftaroline
VRSA - linezolid and streptogramins (quinupristin/dalfopristin)

Note: according to wikipedia regarding action of streptogramins -> Quinupristin and dalfopristin are protein synthesis inhibitors in a synergistic manner. While each of the two is only a bacteriostatic agent, the combination shows bactericidal activity.
Dalfopristin binds to the 23S portion of the 50S ribosomal subunit, and changes the conformation of it, enhancing the binding of quinupristin by a factor of about 100. In addition, it inhibits peptidyl transfer.
Quinupristin binds to a nearby site on the 50S ribosomal subunit and prevents elongation of the polypeptide, as well as causing incomplete chains to be released.

187
Q

What is Amphotericin B? mech, use, toxicity

A

Mechanism: binds ergosterol (unique to fungi); forms membrane pores that allow leakage of electrolytes. AmphoTERicin “tears” holes in the fungal membrane by forming pores.

Use: serious, systemic mycoses. Cryptococcus (amphotericin B with/without flucytosine for cryptococcal meningitis), Blastomyces, Coccidioides, Histoplasma, Candida, Mucor. Intrathecally for fungal meningitis. Supplement K+ and Mg+2 because of altered renal tubule permeability.

Toxicity: Fever/chills (“shake and bake”), hypotension, nephrotoxicity, arrhythmias, anemia, IV phlebitis (“amphoterrible”). Hydration decreases nephrotoxicity. Liposomal amphotericin decreases toxicity.

Phlebitis means inflammation of vessel walls.

188
Q

What is nystatin? mechanism and clinical use

A

Mech: same as amphotericin B. Topical form because too toxic for systemic use.
Use: “swish and swallow” for oral candidiasis (thrush); topical for diaper rash or vaginal candiasis.

189
Q

What are fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole? mech, use, toxicity

A

Mech: Inhibit fungal sterol (ergosterol) synthesis, by inhibiting the cytochrome P-450 enzyme (14-alpha-demehtylase) that converts lanosterol to ergosterol.

Use: local and less serious systemic mycoses. Fluconazole for chronic suppression of cryptococcal meningitis in AIDS patients and candidal infections of all types. Itraconazole for Blastomyces, Coccidioides, Histoplasma. Clotrimazole and miconazole for topical fungal infections.

Toxicity: Testosterone synthesis inhibition (gynecomastia, esp. with ketoconazole), liver dysfunction (inhibits cytochrome P-450).

190
Q

What is flucytosine? mech, use, toxicity

A

Mech: inhibits DNA and RNA biosynthesis by conversion to 5-flurouracil by cytosine deaminase.

Use: Systemic fungal infections (esp. meningitis caused by Cryptococcus) in combination with amphotericin B.

Toxicity: Bone marrow suppression.

191
Q

What are capsofungin, micafungin, anidulafungin? mech, use, toxicity

A

mech: inhibits cell wall synthesis by inhibiting synthesis of B-glucan.
Use: invasive aspergillosis, Candida.
Toxicity: GI upset, flushing (by histamine release).

192
Q

What is terbinafine? mech, use, toxicity

A

Mech: inhibits the fungal enzyme squalene epoxidase.
Use: dermatophytoses (especially onychomycosis - fungal infection of finger or toe nails).
Toxicity: GI upset, headaches, hepatotoxicity, taste disturbance.

193
Q

What is griseofulvin? mech, use, toxicity

A

Mech: interferes with microtubule function: disrupts mitosis. Deposits in keratin-containing tissues (e.g. nails).
Use: Oral treatment of superficial infections; inhibits growth of dermatophytes (tinea, ringworm).
Toxicity: teratogenic, carcinogenic, confusion, headaches, increased P-450, and warfarin metabolism.

194
Q
Antiprotozoan therapy:
uses for -
Pyrimethamine
Suramin and Melarsoprol
Nifurtimox
Sodium stibogluconate
A

Pyrimethamine - toxoplasmosis
Suramin and melarsoprol - Trypanosoma brucei
Nifurtimox - T. cruzi
Sodium stibogluconate - leishmaniasis

195
Q

What is chloroquine used for? mech, use, toxicity

A

Mech: blocks detoxification of heme into hemozoin. Heme accumulates and is toxic to plasmodia.

Use: Treatment of plasmodial species other than P. falciparum (frequency of resistance in P. falciparum is too high). Resistance due to membrane pump that decreases intracellular concentration of drug. Treat P. falciparum with artemether/lumefantrine or atovaquone/proguanil. For life-threatening malaria, use quinidine in U.S. (quinine elsewhere) or artesunate.
Primaquine is used for dormant hypnozoites.

Toxicity: retinopathy; pruritis (especially in dark-skinned individuals).

196
Q

What are common Anti-helminthic therapies?

A

mebendazole, pyrantel pamoate, ivermectin, diethylcarbamazine, praziquantel…immobilize helminths.

Use praziquantel against flukes (trematodes) such as Schistosoma.

197
Q

What are bendazoles or pyrantel pamoate used for

A

Nematodes (roundworms): Enterobius vermicularis (pinworm), ascaris lumbricoides (giant roundworms), Ancylostoma duodenale/Necator Americanus (hookworms).
Also albendazole/mebendazole used for Toxocara canis.
Albendazole used in Echniococcus granulosus.

198
Q

What is ivermectin used for?

A

Strongyloides stercoralis
Onchocerca volvulus

Both nematodes (roundworms)

199
Q

What is diethylcarbamazine used for?

A
Loa loa and Wuchereria bancrofti
Both Nematodes (roundworms)
200
Q

What is praziquantel used for?

A

Cestodes (tapeworms)
Taenia solium (although albendazole for neurocysticercosis)
Diphyllobothrium latum

Tramatodes (flukes)
Schistosoma
Clonorchis sinensis

201
Q

What are zanamivir, oseltamivir?

A

Inhibit influenza neuraminidase -> decreases the release of progeny virus.
Use: treatment and prevention of both influenza A and B.

202
Q

What is ribavirin? mech, use, and toxicity

A

Mech: inhibit synthesis of guanine nucleotides by competitively inhibiting inosine monophosphate.
Use: RSV, chronic hepatitis C.
Toxicity: Hemolytic anemia. Severe teratogen.

203
Q

What are acyclovir, famciclovir, valacyclovir used for? mech, use, toxicity, and resistance

A

Mechanism: monophosphorylated by HSV/VZV thymidine kinase and not phosphorylated in uninfected cells -> few adverse effects. Guanosine analog. Triphosphate formed by cellular enzymes. Preferentially inhibits viral DNA polymerase by chain termination.

Use: HSV and ZVZ. weak activity against EBV and no activity against CMV. Used for HSV-induced mucocutaneous and genital lesions as well as for encephalitis. Prophylaxis in immunocompromised patients. No effect on latent forms of HSV and VZV. Valacyvlovir, a prodrug of acyclovir, has better oral bioavailability.
For herpes zoster, use a related agent, famciclovir.

Toxicity: obstructive crystalline nephropathy and acute renal failure if not adequately hydrated.

Resistance: mutated viral thymidine kinase

204
Q

What is gangciclovir used for? mech, clinical use, toxicity, mech of resistance

A

Mechanism: 5’-monophosphate formed by a CMV viral kinase. Guanosine analog. Triphosphate formed by cellular kinases. Preferentially inhibit viral DNA polymerase.

Use: CMV, especially in immunocompromised patients. Valganciclovir, a prodrug of ganciclovir, has better oral bioavailability.

Toxicity: leukopenia, neutropenia, thrombocytopenia, renal toxicity. More toxic to host enzymes than acyclovir.

Mech of resistance: mutated CMV DNA polymerase or lack of viral kinase.

205
Q

What is foscarnet used for? mech, use, toxicity, and mech of resistance

A

Mech: Viral DNA polymerase inhibitor that binds to the pyrophosphate-binding site of the enzyme. Does not require activation by viral kinase.
FOScarnet = pyroFOSphate analog.

Use: CMV retinitis in immunocompromised patients when ganciclovir fails; acyclovir-resistant HSV.

Toxicity: nephrotoxicity

Mech of resistance: mutated DNA polymerase.

206
Q

What is cidofovir used for? mech, use, toxicity

A

Mech: preferentially inhibits viral DNA polymerase. Does not require phosphorylation by viral kinase.

Use: CMV retinitis in immunocompromised patients; acyclovir-resistant HSV. long-half life.

Toxicity: nephrotoxicity (co-administer with probenecid -decreases excretion- and IV saline to decrease toxicity)

207
Q

What do you do for paraoxysmal supraventricular tachycardia?

A

carotid massage

208
Q

What is the typical regiment for HIV therapy?

A

Highly active antiretroviral therapy (HAART): initiated when patients present with AIDS-defining illness, low CD4 cell counts (<500cells/mm3), or high viral load. Regimen consists of 3 drugs to prevent resistance:
[2 NRTIs] + [1 NNRTI or 1 protease inhibitor or 1 integrase inhibitor]

209
Q

What are atazanavir, darunavir, fosamprenavir, indinavir, lopinavir, ritonavir, saquinavir? mech and toxicity

A

Protease inhibitors. Assembly of virions depends on HIV-1 protease (pol gene), which cleaves the polypeptide products of HIV mRNA into their functional parts. Thus, protease inhibitors prevent maturation of new viruses.
Ritonavir can “boost” other drug concentrations by inhibiting cytochrome P-450.
All protease inhibitors end in -navir.
Navir (never) tease a protease.

Toxicity: Hyperglycemia, GI intolerance (nausea, diarrhea), lipodystrophy.
Nephropathy, hematuria (indinavir).

210
Q

What are Abacavir (ABC), Didanosine (ddI), Emtricitabine (FTC), Lamivudine (3TC), Stavudine (d4T), Tenofavir (TDF), Zidovudine (ZDV, formerly AZT)? mech and toxicity

A

NRTIs.
Competitively inhibit nucleotide binding to reverse transcriptase and terminate the DNA chain (lack a 3’ OH group). Tenofovir is a nucleoTide; the others are nucleosides and need to be phosphorylated to be active. ZDV is used for general prophylaxis and during pregnancy to decrease risk of fetal transmission.
Have you dined (vudine) with my nuclear (nucleosides) family?

Toxicity: bone marrow suppression ( can be reversed with granulocyte colony-stimulating factor [G-CSF] and EPO), peripheral neuropathy, lactic acidosis (nucleosides), rash (non-nucleosides), anemia (ZDV), pancreatitis (didanosine).

211
Q

What are efavirenz, nevirapine, delavirdine? mech and toxicity

A

NNRTIs.
Bind to reverse transcriptase at site different from NRTIs. Do not require phosphorylation to be active or compete with nucleotides.

Toxicity: Rash and hepatotoxicity are common to all NNRTIs. Vivid dreams and CNS symptoms are common with efavirenz. Delavirdine and efavirenz are contraindicated in pregnancy. SJS and toxic epidermal necrolysis with efavirenz and nevirapine.

212
Q

What does raltegravir do? mech and toxicity.

A

Integrase inhibitor.
Inhibits HIV genome integration into host cell chromosome by reversibly inhibiting HIV integrase.

Toxicity: Hypercholesteremia

213
Q

What is enfurvirtide? mech and toxicity

A

Fusion inhibitor.
Binds gp41 and inhibits viral entry.

Toxicity: Skin reaction at injection sites

214
Q

What is Maraviroc? mech

A

Fusion inhibitor

Binds CCR-5 on surface of T cells/monocytes inhibiting interaction with gp120.

215
Q

What do the interferons do? List uses for IFN-alpha, INF-beta, IFN-gamma. mech, use, and toxicity

A

Mech: Glycoproteins normally synthesized by virus-infected cells, exhibiting a wide range of antiviral and antitumoral properties.

Clinical use:
IFN-alpha - chronic hepatitis B and C, Kaposi sarcoma, hairy cell leukemia, condyloma acuminatum, renal cell carcinoma, malignant melanoma.
IFN-beta - multiple sclerosis
IFN-gamma - chronic granulomatous disease.

Toxicity: neutropenia, myopathy.

216
Q

what are the antibiotics to avoid in pregnancy?

A

Sulfonamides (kernicterus), Aminioglycosides (ototoxicity), fluoroquinolones (cartilage damage), clarithromycin (embryotoxic), tetracyclines (discolored teeth/inhibition of bone growth), Ribavirin (anti-viral and teratogenic), Griseofulvin (anti-fungal and teratogenic), Chloramphenicol (“gray baby”)
SAFe Children Take Really Good Care.

217
Q

What does mannitol do? mech, use, toxicity

A

Mech: osmotic diuretic, increased tubular fluid osmolarity, producing increased urine flow, decreased intracranial/intraocular pressure.

Use: drug overdose, increased intracranial/intraocular pressure

Toxicity: pulmonary edema, dehydration. Contraindicated in anuria, CHF.

218
Q

What does acetazolamide? mech, use, toxicity

A

Mech: Carbonic anhydrase inhibitor. Causes self-limited NaHCO3 diuresis and decreased total-body HCO3- stores.

Use: glaucoma, urinary alkalinization, metabolic alkalosis, altitude sickness, pseudotumor cerebri.

Toxicity: hyperchloremic metabolic acidosis, paresthesias, NH3 toxicity, sulfa allergy.

219
Q

What does furosemide do? mech, use, toxicity

A

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. Stimulates PGE release (vasodilatory effect on afferent arteriole); inhibited by NSAIDs. Increased Ca+2 excretion. Loops Lose calcium.

Use: Edematous states (CHF, cirrhosis, nephrotic syndrome, pulmonary edema), hypertension, hypercalcemia.

Toxicity: Ototoxicity, Hypokalemia, Dehydration, Allergy (sulfa), Nephritis (interstitial), Gout. OH DANG.

220
Q

What is ethacrynic acid and what does it do? mech, use, toxicity

A

Loop diuretics
Mech: Phenoxyacetic acid derivative (not a sulfonamide). Essentially same action as furosemide.

Use: Diuresis in patients allergic to sulfa drugs.

Toxicity: Similar to furosemide; can cause hyperuricemia; never use to treat gout.

221
Q

What does hydrochlorothiazide do? mech, use, toxicity

A

Mech: thiazide diuretic. Inhibits NaCl reabsorption in early distal tubule. Decreased diluting capacity of the nephron. Decreased Ca+2 excretion.

Use: Hypertension, CHF, idiopathic hypercalcuria, nephrogenic diabetes insipidus, osteoporosis.

Toxicity: Hypokalemic metabolic alkalosis, hyponatremia, hyperGlycemia, hyperLipidemia, hyperUricemia, and hyperCalcemia. Sulfa allergy.
HyperGLUC

222
Q

What do spironolactone and eplerenone, triamterene, and amiloride do? mech, use, toxicity

A

K+ sparing diuretics. The K+ STAys.

Mech: spironolactone and eplerenone are competitive aldosterone receptor antagonists 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: hyperaldosteronism, K+ depletion, CHF.

Toxicity: Hyperkalemia (can lead to arrhythmias), endocrine effects with spironolactone (e.g. gynecomastia, antiandrogen effects).

223
Q

Which diuretics change urine NaCl levels?

A

Increased by all diuretics except acetazolamide. Serum NaCl may decrease too.

224
Q

Which diuretics change urine K+?

A

increased with loop and thiazide diuretics. Serum K+ may decrease as a result.

225
Q

Which diuretics cause changes in Blood pH?

A

decreased (acidemia) by carbonic anyhydrase inhibitors- > decreased HCO3- reabsorption. K+ sparing-aldosterone blockade prevents K+ secretion and H+ secretion. Additionally, hyperkalemia leads to K+ entering all cells (via H/K exchange) in exchange for H+ exiting cells.

Increased (alkaemia) by loop diuretics and thiazides cause alkalemia through several mechanisms:

1) volume contraction -> increased ATII -> increased Na/H+ exchange in proximal tubules -> increased HCO3- reabsorption (contraction alkalosis)
2) K+ loss leads to K+ exiting all cells (via H/K exchanger) in exchange for H+ entering cells.
3) in low K+ state, H+ (rather than K+) is exchanged for Na+ in cortical collecting tubule -> alkalosis and “paradoxical aciduria”

226
Q

Which diuretics affect urine Ca+2?

A

Increased with loop diuretics: decreased paracellular Ca+2 reabsorption leading to hypocalcemia.
Decreased with thiazides: enchanced paracellular Ca+2 reabsorption in distal tubule.

227
Q

What do captopril, enalapril, and lisinopril do? mech, use, toxicity

A

ACE inhibitors
Mech: inhibit ACE leads to decreased angiotensin II and then decreased GFR by preventing constriction of efferent arterioles. Levels of renin increased as a result of loss of feedback inhibition. Inhibition of ACE also prevents inactivation of bradykinin, a potent vasodilator.

Use: hypertension, CHF, proteinuria, diabetic nephropathy. Prevent unfavorable heart remodeling as a result of chronic hypertension.

Toxicity: Cough, Angioedema (contraindicated in C1 esterase inhibitor deficiency), Teratogen (fetal renal malformations), Increased Creatinine (decreased GFR), Hyperkalemia, and Hypotension. Avoid in bilateral renal artery stenosis, because ACE inhibitors will further decrease GFR leading to renal failure.

Captopril’s CATCHH.

228
Q

What is the treatment for ADHD?

A

methylphenidate

229
Q

What is the treatment for alcohol withdrawal?

A

benzodiazepines

230
Q

What is the treatment for anxiety?

A

SSRIs, SNRIs, buspirone

231
Q

What is the treatment for Bipolar disorder?

A

“Mood stabilizers” (e.g. lithium, valproic acid, carbamazepine), atypical antipsychotics.

232
Q

What is the treatment for Bulimia?

A

SSRIs

233
Q

What is the treatment for Depression?

A

SSRIs, SNRIs, TCA, bupropion, mirtazapine (especially with insomnia)

234
Q

What is the treatment for OCD?

A

SSRIs, clomipramine

235
Q

What is the treatment for panic disorder?

A

SSRIs, venlafaxine, benzodiazepines.

236
Q

What is the treatment for PTSD?

A

SSRIs

note: prazosin also (according to pharm section in FA)

237
Q

What is the treatment for schizoprenia?

A

Antipsychotics

238
Q

What is the treatment for social phobias?

A

SSRIs, Beta-blockers

239
Q

What is the treatment for Tourette syndrome?

A

Antipsychotics

240
Q

What are methylphenidate, dextroamphetamine, methamphetamine, phentermine?

A

CNS stimulants
Increased catecholamines at the synaptic cleft, especially norepinephrine and dopamine (by promoting vesicular release, also some reuptake inhibition).

USE: ADHD, narcolepsy, appetite control.

241
Q

What are haloperidol, trifluoperazine, fluphenazine, thioridazine, chlorpromazine? Mech, use, toxicity

A

Typical Antipsychotics.
All typical antipsychotics block Dopamine D2 receptors (increased cAMP).

Use: Schizophrenia (primarily positive symptoms), psychosis, acute mania, Tourette syndrome.

Toxicity: Highly lipid soluble and stored in body fat; thus, very slow to be removed from body. Extrapyramidal symptoms side effects (e.g. dyskinesias). Treatment: benztropine or diphenhydramine.
Endocrine side effects (e.g. dopamine receptor antagonism -> hyperprolactinemia -> galactorrhea).
Side effects arising from blocking muscarinic (dry mouth, constipation), alpha-1 (hypotension), and histamine (sedation) receptors.

242
Q

Which are the high potency antipsychotics? side effects

A

Trifluoperazine, Fluphenazine, Haloperidol (Try to Fly High) - neurologic side effects (EPS symptoms).

Haloperidal - NMS, tardive dyskinesia.

243
Q

Which are the low potency antipsychotics? and side effects

A

Chlorpromazine, Thioridazine (Cheating Thieves are low) - non-neurologic side effects (anticholinergic, antihistamine, and alpha-1-blockade effects) - non-neurologic side effects (anticholinergic, antihistamine, and alpha-1-blockade effects)

Chlorpromazine- Corneal deposits
Thioridazine - reTinal deposits;

244
Q

What are the EPS side effects?

A

4 hr acute dystonia (muscle spasm, stiffness, oculogyric crisis)
4 day akathisia (restlessness)
4 week bradykinesia (parkinsonism)
4 month tardive dyskinesia

245
Q

What is neuroleptic malignant syndrome (NMS)

A

caused by typical antipsychotics
Rigidity, myoglobinuria, autonomic instability, hyperpyrexia. Treatment: dantrolene, D2 agonists (e.g. bromocriptine)

For NMS, think FEVER
Fever, Encephalopathy, Vitals unstable, Enzymes increased, Rigidity of muscles.

246
Q

What is tardive dyskinesia?

A

stereotypic oral-facial movements as a result of long term anti-psychotic use. Potentially irreversible.

247
Q

What are olanzapine, clozapine, quetiapine, risperidone, aripiprazole, ziprasidone?

A

Atypical antipsychotics
Mech: not completely understood. Varied effects on 5-HT2, dopamine, and alpha and H1-receptors.

Use: Schizophrenia - both positive and negative symptoms. Also used for bipolar disorder, OCD, anxiety disorder, depression, mania, Tourette syndrome.

Toxicity: Fewer EPS and anticholinergic side effects than traditional antipsychotics. Olanzapine/clozapine may cause significant weight gain. Clozapine may cause agranulocytosis (requires weekly WBC monitoring) and seizure. Risperidone may increase prolactin (causing lactation and gynecomastia) decrease GnRH, LH, and FSH (causing irregular menstruation and fertility issues). Ziprasidone may prolong the QT interval.

248
Q

What is lithium used for? mech, use, toxicity

A

Mech: not established; possibly related to inhibition of phosphoinositol cascade.

Use: mood stabilizer for bipolar disorder; blocks relapse and acute manic events. Also SIADH.

Toxicity: Tremor, sedation, edema, heart block, hypothyroidism, polyuria (ADH antagonist causing nephrogenic diabetes insipidus), teratogenesis. Fetal cardiac defects include Ebstein anomaly and malformation of the great vessels. Narrow therapeutic window requires close monitoring of serum levels. Almost exclusively excreted by the kidneys; most is reabsorbed at the proximal convoluted tubules following Na+ reabsorption.

LMNOP - Lithium side effects - Movement (tremor), Nephrogenic diabetes insipidus, hypOthyroidism, Pregnancy problems.

249
Q

What does buspirone do?

A

Mech: Stimulates 5-HT1A receptors.
Use: Generalized anxiety disorders. Does not cause sedation, addiction, or tolerance. Takes 1-2 weeks to take effect. Does not interact with alcohol (vs. barbiturates, benzodiazepines).

I’m alway anxious if the BUS will be ON time, so I take BUSpirONe.

250
Q

What do fluoxetine, paroxetine, sertraline, citalopram do? Mech, use, toxicity

A

Flashbacks paralyze senior citizens.
Mech: 5-HT specific reuptake inhibitors
Use: Depression, generalized anxiety disorder, panic disorder, OCD, bulimia, social phobias, PTSD.

Toxicity: Fewer than TCAs. GI distress, sexual dysfunction (anorgasmia and decreased libido).
Serotonin syndrome with any drug that increases 5-HT (e.g. MAO inhibitors, SNRIs, TCAs) - hyperthermia, confusion, myoclonus, cardiovascular collapse, flushing, diarrhea, seizures.
Treatment is cyproheptadine (5-HT2 receptor antagonist).

251
Q

What are venlafaxine, duloxetine? mech, use, toxicity

A

SNRIs.
Mech: inhibit 5-HT and norepinephrine reuptake.
Use: Depression. Venlafaxine is also used in generalized anxiety disorder and panic disorders; duloxetine is also indicated for diabetic peripheral neuropathy.
Toxicity: increased BP most common; also stimulant effects, sedation, nausea.

252
Q

What are amitriptyline, nortriptyline, imipramine, desipramine, clomipramine, doxepin, amoxapine (all TCAs end in -iptyline or -ipramine except doxepin and amoxapine)?

A

TCAs.
Mech: Block re-uptake of norepinephrine and 5-HT.
Use: Major depression, OCD (clomipramine), fibromyalgia.
Toxicity: Sedation, alpha-1-blocking effects, including postural hypotension, and atropine-like (anticholinergic) side effects (tachycardia, urinary retention, dry mouth). Tertiary TCAs (amitriptyline) have more anti-cholinergic effects than secondary TCAs (nortriptyline) have. Desipramine is less sedating, but has a higher seizure incidence.
Tri-C’s: Convulsions, Coma, Cardiotoxicity (arrhythmias); also respiratory depression, hyperpyrexia. Confusion and hallucinations in elderly due to anticholinergic side effects (use nortriptyline). Treatment: NaHCO3 for cardiovascular toxicity.

253
Q

What Tranylcypromine, Phenelzine, Isocarboxazid, Selegiline?

A

MAO Takes Pride In Shanghai.
(Selegiline is a selective MAO-B inhibitor)
Mech: Nonselective MAO inhibition increases levels of amine neurotransmitters (norepinephrine, 5-HT, dopamine).
Use: atypical depression, anxiety, hypochondriasis.

Toxicity: Hypertensive crisis (most notably with ingestion of tyramine, which is found in many foods such as wine and cheese); CNS stimulation. Contraindicated with SSRIs, TCAs, St. John’s wort, meperidine, and dextromethorphan (to prevent serotonin syndrome).

254
Q

What is Bupropion?

A

Atypical antidepressant.
Also used for smoking cessation. Increased norepinephrine and dopamine via unknown mechanism. Toxicity: stimulant effects (tachycardia, insomnia), headache, seizure in bulimic patients. No sexual side effects.

255
Q

What is mirtazapine?

A

Atypical antidepressants.
alpha-2-antagonist (increased release of norepinephrine and 5-HT) and potent 5-HT2 and 5-HT3 receptor antagonist. Toxicity: sedation (which may be desirable in depressed patients with insomnia), increased appetite, weight gain (which may be desirable in elderly or anorexic patients), dry mouth.

256
Q

What is trazodone?

A

Primarily blocks 5-HT2 and alpha-1-adrenergic receptors. Used primarily for insomnia as high doses are needed for antidepressant effects.

Toxicity: sedation, nausea, priapism, postural hypotension.
called trazoBONE due to male-specific side effects.

257
Q

What is dobutamine?

A

Sympathomimetic drug with direct stimulation of B1 receptors of the sympathetic nervous system. Positive inotropic action. Weak B2 and alpha 1 activity.

Uses: heart failure and cardiogenic shock.

Side effects: hypertension, angina, arrhythmia, and tachycardia. Less likely to induce hypertension than dopamine is (dopamine acts on dopamine receptors to also release norepinephrine which is alpha-1-agonist)

258
Q

Where does Tamoxifen act and what stage does it stop cells in?

A

Stops them in G1. activates nuclear receptor that goes and degrades DNA.

Antagonist in breast and bone and activator in endometrium.

259
Q

What is cyclosporine? mech, use, toxicity

A

Mech: Calcineurin inhibitor; binds cyclophilin. Blocks T cell activation by preventing IL-2 transcription.
Use: transplant rejection prophylaxis, psoriasis, rheumatoid arthritis.
Toxicity: nephrotoxicity, hypertension, hyperlipidemia, hyperglycemia, tremor, hirsutism, gingival hyperplasia.
Notes: both calcineurin inhibitors are very nephrotoxic.

260
Q

What is Tacrolimus? mech, use, toxicity

A

Mech: Calcineurin inhibitor; binds FK506 binding protein (FKBP). Blocks T cell activation by preventing IL-2 transcription.
Use: transplant rejection prophylaxis.
Toxicity: similar to cyclosporine, increased risk of diabetes and neurotoxicity; no gingival hyperplasia or hirsuitism.
-limus drugs binds FKBP.

261
Q

What is Sirolimus do? Rapamycin. mech, use, toxicity

A

mTOR inhibitor; binds FKBP. Blocks T cell activation and B cell differentiation by preventing IL-2 signal transduction.
Use: kidney transplant rejection prophylaxis.

Toxicity: anemia, thrombocytopenia, leukopenia, insulin resistance, hyperlipidemia, non-nephrotoxic.

Notes: Kidney “sir-vives”. Synergistic with cyclosporine. Also used in drug-eluting stents.

262
Q

What is Basiliximab? mech ,use, toxicity

A

Mech: monoclonal antibody; blocks IL-2R.
Use: Kidney transplant rejection prophylaxis.
Toxicity: edema, hypertension, tremor.

263
Q

What is azathioprine? mech, use, toxicity

A

Antimetabolite precursor of 6-mercaptopurine. Inhibits lymphocyte proliferation by blocking nucleotide synthesis.
Use: Transplant rejection prophylaxis, rheumatoid arthritis, Crohn disease, glomerulonephritis, other autoimmune conditions.
Toxicity: leukopenia, anemia, thrombocytopenia.
Note: 6-MP degrade by xanthine oxidase; toxicity increased by allopurinol. Pronounce azathio-PURINE.

264
Q

What are the glucocorticocoids? mech, use, toxicity

A

Mech: Inhibit NF-kB and suppress both B and T cell function by decreased transcription of many cytokines.

Use: Transplant rejection prophylaxis (immune suppression), many autoimmune disorders, inflammation.

Toxicity: hyperglycemia, osteoporosis, central obesity, muscle breakdown, psychosis, acne, hypertension, cataracts, peptic ulcers.

265
Q

What are thrombopoietin and Oprelvekin (IL-11) used for?

A

thrombocytopenia. Thrombopoitetin stimulates platelet production and is mainly produced in liver and kidney. Stimulates production and differentiation of megakaryocytes.

266
Q

What are Filgrastim (GMCSF) and Sargramostim (GMCSF) used for?

A

recovery of bone marrow

267
Q

What are Aldesleukin (IL-2) used for?

A

Renal cell carcinoma, metastatic melanoma

268
Q

What is IFN-alpha used for?

A

Chronic Hep B and C; kaposi sarcoma, hairy cell leukemia, condyloma acuminatum, renal cell carcinoma, malignant melanoma.

269
Q

What is IFN-beta used for?

A

Multiple sclerosis

270
Q

What is IFN-gamma used for?

A

chronic granulomatous disease

271
Q

What is Alemtuzumab used for?

A

CD52 is target. Use is CLL

aLYMtuzumab - chronic LYMphocytic leukemia

272
Q

What is Bevacizumab used for?

A

VEGF is target.

Colorectal cancer, renal cell carcinoma

273
Q

What is Cetuximab used for?

A

EGFR is target

Stage IV colorectal cancer, head and neck cancer

274
Q

what is Rituximab used for?

A

CD20 is target.

B-cell non-Hodgkin lymphoma, rheumatoid arthritis (with MTX), ITP

275
Q

What is trastuzumab used for?

A

HER2/neu is target.

Breast cancer, gastric cancer.

276
Q

What are infliximab and adalimumab used for?

A

TNF-alpha is target
IBD, rheumatoid arthritis, ankylosing spondylitis, psoriasis.
RA “inflix” pain in “DA LIMbs”

277
Q

What is natalizumab used for?

A

alpha-4-integrin is target.
MS, crohn disease.
Alpha-4-integrin is involved in leukocyte adhesion. Risk of PML in patients with JC virus.

278
Q

What is abciximab used for?

A

Glycoprotein IIb/IIIa is the target.
Anti-platelet agent for prevention of ischemic complications in patients undergoing percutaneous coronary intervention.
IIb times IIIa equals “abSIXimab”

279
Q

What is Denosumab used for?

A

RANKL is the target.
Osteoporosis; inhibits osteoclast maturation (mimics osteoprotegrin).
DenOSumab affects OSteoclasts.

280
Q

What does Digoxin immune Fab do?

A

Digoxin is the target

Antidote for digoxin toxicity

281
Q

What is Omalizumab used for?

A

IgE is the target.

Allergic asthma; prevents IgE binding to FceRI

282
Q

What is Palivizumab used for?

A

RSV F protein is target
RSV prophylaxis for high-risk infants.
PaliVIzumab - VIrus.

283
Q

What is the treatment for essential tremor?

A

propranolol - nonspecific Beta blocker

284
Q

What is Daptomycin used for? mech, use, toxicity

A

Mech: Depolarization of membrane. Can only penetrate gram positive membranes and causes the creation of transmembrane channels that depolarize it and inhibit cellular function causing death.

Use: invasive MRSA infections

Adverse actions: Myopathy and CPK elevation. Inactivated by pulmonary surfactant.

285
Q

What is cromolyn sodium used for?

A

Used for preventing mast cell degranulation in asthma prophylaxis.

286
Q

How do you treat lead toxicity?

A

LEAD - Lead lines on gingivae and on metaphyses of long bones on xray
Encephalopathy and Erythrocyte basophilic stipling (rRNA degradation blocked)
Abdominal colic and sideroblastic anemia
Drops - wrist and foot drop. Dimercaprol and eDta are first line of treatment.
Succimer used for chelation in kids.

lead inhibits ferrochelatase and ALA dehydratase.

287
Q

Which drugs cause macrocytic, non-megaloblastic anemia?

A

5-FU, zidovudine, hydroxyurea

Also liver disease, alcoholism (vacuolization), and reticulocytosis

288
Q

Which are the low-molecular weight heparins?

A

Enoxaparin, dalteparin
Act more on factor Xa.
They have better bioavailability and 2-4 times longer half-life.
Can be administered subcu and without lab monitoring. Not easily reversible.

289
Q

What is heparin induced thrombocytopenia?

A

development of IgG antibodies against heparin bound to platelet factor (PF4). Antibody-heparin-PF4 complex activates platelets -> thrombosis and thrombocytopenia.

290
Q

What are argatroban, bivalirudin?

A

Derivatives of hirudin, the anti-coagulant used by leeches; inhibit thrombin directly. Used instead of Heparin for anti-coagulating patients with HIT.

291
Q

What is warfarin? mech, use, toxicity

A

Mech: blocks normal synthesis and gamma carboxylation of vitamin K-dependent clotting factors II, VII, IX, and X and proteins C and S. Metabolized by the cytochrome P-450 pathway. Increases PT…long half-life.

Use: Chronic anticoagulation (after STEMI, venous thromboembolism prophylaxis, and prevention of stroke in atrial fibrillation). Not used in pregnant women (because warfarin, unlike heparin, can cross the placenta). Follow PT/INR values.

Toxicity: Bleeding, teratogenic, skin/tissue necrosis, drug-drug interactions.

292
Q

What are apixaban, rivaroxaban?

A

Direct factor Xa inhibitors
Bind and directly inhibit the activity of factor Xa.
Use: treatment and prophylaxis of DVT and PE (rivaroxaban), stroke prophylaxis in patients with atrial fibrillation.
Oral agents do not usually require coagulation monitoring.

Toxicity: bleeding (no specific reversal agent available).

293
Q

What is the toxicity for heparin? and how do you stop it

A

Bleeding, thrombocytopenia (HIT), osteoporosis, drug-drug interactions. For rapid reversal (antidote), use protamine sulfate (positively charged molecule that binds negatively charged heparin).

294
Q

What are alteplase (tPA), reteplase (rPA), tenecteplase (TNK-tPA)? mech, use, toxicity

A

Directly or indirectly aid conversion of plasminogen to plasmin, which cleaves thrombin and fibrin clots. Increased PT, PTT, no change in platelet count.

Use: Early MI, early ischemic stroke, direct thrombolysis of severe PE.

Toxicity: 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. Fresh frozen plasma and cryoprecipitate can also be used to correct factor deficiencies.

295
Q

What is Aspirin (ASA)?

A

Mech: irreversibly inhibits cyclooxygenase (both COX1 and COX2) enzyme by covalent acetylation. Platelets cannot synthesize new enzyme, so effect lasts until new platelets are produced. Increased bleeding time, decreased thromboxane and prostaglandins. No effect on PT or PTT.

Clinical use: antipyretic, analgesic, anti-inflammatory, antiplatelet (decreased aggregation)

Toxicity: Gastric ulceration, tinnitus (CN VIII). Chronic use can lead to acute renal failure, interstitial nephritis, and upper GI bleeding. Reye syndrome in children with viral infection Overdose causes respiratory alkalosis initially, which is then superimposed by metabolic acidosis.

296
Q

What are clopidogrel, ticlopidine, prasugrel, ticagrelor? mech, use, toxicity

A

Mech: Inhibit platelet aggregation by irreversibly blocking ADP receptors. Inhibit fibrinogen binding by preventing glycoprotein IIb/IIIa from binding to fibrinogen (through the mechanism of reduced glycoprotein receptors)

Use: acute coronary syndrome; coronary stenting. Decreased incidence or recurrence of thrombotic stroke.

Toxicity: neutropenia (ticlopidine). TTP/HUS may be seen.

297
Q

What is cilostazol, dipyridamole?

A

Mech: Phosphodiesterase III inhibitor; cAMP increased in platelets, thus inhibiting platelet aggregation.

Use: intermittent claudication, coronary vasodilation, prevention of stroke or TIAs (combined with aspirin), angina prophylaxis.

Toxicity: Nausea, headache, facial flushing, hypotension, abdominal pain. CORONARY STEAL PHENOMENON a

298
Q

What are abciximab, eptifibatide, tirofiban? mech, use, toxicity

A

Mech: bind to the glycoprotein receptor IIb/IIIa on activated platelets, preventing aggregation (blocks binding of fibrinogen). Abciximab is made from monoclonal antibody Fab fragments.

Use: Unstable angina, percutaneous transluminal coronary angioplasty.

Toxicity: bleeding, thrombocytopenia.

299
Q

What is methotrexate? mech, use, toxicity

A

Mech: folic acid analog that inhibits dihydrofolate reductase -> decreased dTMP -> decreased DNA and decreased protein synthesis.

Use: cancers: leukemias, lymphomas, choriocarcinoma, sarcomas. Non-neoplastic: abortion, ectopic pregnancy, rheumatoid arthritis, psoriasis, IBD.

Toxicity: myelosuppression, which is reversible with leucovorin (folinic acid) “rescue”
Macrovesicular fatty change in liver.
Mucositis.
Teratogenic.

300
Q

What is 5-fluorouracil (5-FU)? mech, use, toxicity

A

Mech: Pyrimidine analog bioactivated to 5F-dUMP, which covalently complexes folic acid. This complex inhibits thymidylate synthase -> decreased dTMP -> decreased DNA and decreased protein synthesis.

Use: colon cancer, pancreatic cancer, basal cell carcinoma (topical)

Toxicity: myelosuppression, which is not reversible with leucovorin. Overdose: “rescue” with uridine.
Photosensitivity.

301
Q

What is cytarabine (arabinofuranosyl cytidine)? mech, use, toxicity

A

Pyrimidine analog -> inhibition of DNA polymerase

Use: leukemias, lymphomas

Toxicity: leukopenia, thrombocytopenia, megaloblastic anemia, CYTarabine causes panCYTopenia.

302
Q

What is azathioprine, 6-mercaptopurine, 6-thioguanine? mech, use, toxicity

A

Purine (thiol) analogs -> decreased de novo purine synthesis. Activated by HGPRT.

Use: preventing organ rejection, RA, SLE (azathioprine). Leukemia, IBD (6-MP, 6-TG).

Toxicity: bone marrow, GI, liver. Azathioprine and 6-MP are metabolized by xanthine oxidase; thus both have increased toxicity with allopurinol, which inhibits their metabolism.

303
Q

What is dactinomycin (actinomycin D)? mech, use, toxicity

A

Intercalates in DNA
Use: Wilms tumor, Ewing sarcoma, rhabdomyosarcoma. Used for childhood tumors (children ACT out).

Toxicity:
myelosuppression

304
Q

What is doxorubicin (adriamycin), daunorubicin? mech, use, toxicity

A

Generate free radicals. Intercalate in DNA -> breaks in DNA -> decreased replication.

Use: solid tumors, leukemias, lymphomas.

Toxicity: Cardiotoxicity (dilated cardiomyopathy), myelosuppression, alopecia. Toxic to tissues following extravasation.
Dexrazoxane (iron chelating agent), used to prevent cardiotoxicity.

305
Q

What is bleomycin? mech, use, toxicity

A

Induces free radical formation, which causes breaks in DNA strands.

Use; testicular cancer, Hodgkin lymphoma

Toxicity: pulmonary fibrosis, skin changes, mucositis. Minimal myelosuppression.

306
Q

What is cyclophosphamide, ifosfamide?

A

covalently X-link (interstrand) DNA at guanine N-7. Require bioactivation by liver.

Use: solid tumors, leukemia, lymphomas, and some brain cancers.

Toxicity: Myelosuppression; hemorrhagic cystitis, partially prevented with mesna (thiol group of mesna binds toxic metabolites).

307
Q

What are nitrosoureas (carmustine, lomustine, semustine, streptozocin)?

A

Require bioactivation. Cross blood brain barrier into CNS and cross link DNA.

Use: brain tumors (including glioblastoma multiforme)

Toxicity: CNS toxicity (convulsions, dizziness, ataxia)

308
Q

What is busulfan?

A

Cross links DNA.

USed for: CML. Also used to ablate patient’s bone marrow before bone marrow transplantation.

Toxicity: severe myelosuppression (in almost all cases), pulmonary fibrosis, hyperpigmentation.

309
Q

What is Vincristine, vinblastine?

A

Vinca alkaloids that bind Beta-tubulin, inhibit its polymerization into microtubules, thereby preventing mitotic spindle formation (M-phase arrest)

Use: solid tumors, leukemias, and lymphomas.

Toxicity: vincristine - neurotoxicity (areflexia, peripheral neuritis), paralytic ileus.
Vinblastine blasts bone marrow (suppression).

310
Q

What is paclitaxel, other taxols?

A

Hyperstabilize polymerized microtubules in M phase so that mitotic spindle cannot break down (anaphase cannot occur). It is TAXing to stay polymerized.

Use: ovarian and breast carcinomas.

Toxicity: myelosuppression, alopecia, hypersensitivity.

311
Q

What is cisplatin, carboplatin?

A

mech: cross-link DNA

Use: testicular, bladder, ovary, and lung carcinomas.

Toxicity: nephrotoxicity, and acoustic nerve damage. Prevent nephrotoxicity with amifostine (free radical scavenger) and chloride diuresis.

312
Q

What is etoposide, teniposide?

A

Mech: eTOPOside inhibits TOPOisomerase II -> DNA degradation

Use: solid tumors (particularly testicular and small cell lung cancer), leukemias, lymphomas

Toxicity: myelosuppression, GI irritation, alopecia

313
Q

What is irontecan, topotecan?

A

mech: inhibit topoisomerase I and prevent DNA unwinding and replication

Use: colon cancer (irinotecan); ovarian and small cell lung cancers (troptecan)

Toxicity: severe myelosuppression, diarrhea.

314
Q

What is hydroxyurea?

A

Inhibits ribnoucleotide reductase -> DNA synthesis decreased (S-phase specific)

Use: Melanoma, CML, sickle cell disease (by increasing HbF)

Toxicity: Bone marrow suppression, GI upset.

315
Q

What is prednisone, prednisolone?

A

May trigger apoptosis. May even work on nondividing cells

Most commonly used glucocorticoids in cancer chemotherapy. Used in CLL, non-hodgkin lymphomas (part of combo therapy), also used as immunosuppressants in autoimmune diseases and others.

Toxicity: Cushing like symptoms; weight gain, central obesity, muscle breakdown, cataracts, acne, osteoporosis, hypertension, peptic ulcers, hyperglycemia, psychosis, water retention, hypernatremia, hypokalemia, hypocalcemia.

316
Q

What is tamoxifen, raloxifene?

A

Mech: SERMs - receptor antagonists in breast and agonists in bones. Block the binding of estrogen to ER + cells.

Use: breast cancer treatment (tamoxifen only) and prevention. Raloxifene is also used to prevent osteoporosis.

Toxicity: tamoxifen - partial agonist in endometrium, which has the increased risk of endometrial cancer. “hot flashes”. Raloxifene - no increase in endometrial carcinoma because it is an endometrial antagonist.

317
Q

What is trastuzumab?

A

Herceptin. Monoclonal antibody against HER-2 (e-erbB2), a tyrosine kinase receptor. Helps kill breast cancer cells that overexpress HER-2, through inhibition of HER2-initiated cellular signaling and antibody-dependent cytotoxicity.

Use: HER-2 positive breast cancer and gastric cancer (tras2zumab).

Toxicity: cardiotoxicity. “HEARTceptin” damages the HEART.

318
Q

What is imatinib?

A

Gleevec. Tyrosine kinase inhibitor of bcr-abl (Philadelphia chromosome fusion gene in CML) and c-Kit (common in GI stromal tumors).

Use: CML, GI stromal tumors.

Toxicity: fluid retention

319
Q

What is Rituximab?

A

Monoclonal antibody against CD20, which is found on most B-cell neoplasms.

Use: non-hodgkin lymphoma, rheumatoid arthritis (with MTX), ITP.

Toxicity: increased risk of progressive multifocal leukoencephalopathy.

320
Q

What is vemurafenib?

A

Small molecular inhibitor of forms of the B-raf kinase with the V600E mutation

Use; Metastatic melanoma

321
Q

What is Bevacizumab?

A

Mech: monoclonal antibody against VEGF. Inhibits angiogenesis.

Use: solid tumors (colorectal cancer, renal cell carcinoma)

Toxicity: hemorrhage and impaired wound healing.

322
Q

What is Baclofen?

A

Baclofen is a muscle relaxant that affects GABAb receptors at the level of the spinal cord.

323
Q

What is atomoxetine?

A

It’s a selective norepinephrine reuptake inhibitor for ADHD.
Good because there is little abuse potential.

324
Q

What is the intoxication and withdrawal symptoms for Alcohol?

A

Intoxication: emotional lability, slurred speech, ataxia, coma, blackouts. Serum gamma-glutamyltransferase (GGT) - sensitive indicator of alcohol use. Lab AST value is twice ALT level.

Withdrawal: symptoms similar to other depressants. Severe alcohol withdrawal can cause autonomic hyperactivity and DTs (5-15% mortality rate). Treatment for DTs: benzodiazepines.

325
Q

What is the intoxication and withdrawal symptoms for Opioids (eg. morphine, heroin, methadone)?

A

Intoxication: euphoria, respiratory and CNS depression, decreased gag reflex, pupillary constriction (pinpoint pupils), seizures (overdose). Constipation too. Treatment: naloxone, naltrexone.

Withdrawal: Sweating, dilated pupils, piloerection (“cold turkey” from pilae erector muscle), fever, rinorrhea, yawning, nausea, stomach cramps, diarrhea (flu like symptoms). Treatment: long-term support, methadone, buprenorphine.

326
Q

What is the intoxication and withdrawal symptoms for Barbiturates?

A

Low safety margin, marked respiratory depression. Treatment: symptom management (assist respiration, increased BP).

Withdrawal: delirium, life-threatening cardiovascular collapse.

327
Q

What is the intoxication and withdrawal symptoms for Benzodiazepines?

A

Intox: greater safety margin. Ataxia, minor respiratory depression. Tx: supportive care; consider flumazenil (competitive benzodiazepine antagonist).

Withdrawal: sleep disturbance, depression, rebound anxiety, seizure (can be triggered by reversal with flumazenil).

328
Q

What is the intoxication and withdrawal symptoms for Amphetamines?

A

Intoxication: Euphoria, grandiosity, pupillary dilation, prolonged wakefulness and attention, hypertension, tachycardia, anorexia, paranoia, fever. Severe: cardiac arrest, seizure.

Withdrawal: anhedonia, increased appetite, hypersomnolence, existential crisis.

329
Q

What is the intoxication and withdrawal symptoms for Cocaine?

A

Intox: impaired judgement, pupillary dilation, hallucinations (including tactile), paranoid ideations, angina, sudden cardiac death. Tx: benzodiazepines

Withdrawal: hypersomnolence, malaise, severe psychological craving, depression/suicidality.

330
Q

What is the intoxication and withdrawal symptoms for caffeine?

A

Intoxication: Restlessness, increased diuresis, muscle twitching

Withdrawal: lack of concentration, headache.

331
Q

What is the intoxication and withdrawal symptoms for Nicotine?

A

Intox: restlessness.

Withdrawal: irritability, anxiety, craving.
Tx: nicotine patch, gum, or lozenges. bupropion (norepi/dopamine reuptake inhibitor and nicotinic antagonist)/varenicline (partial agonist of nicotinic receptors)

332
Q

What is the intoxication and withdrawal symptoms for PCP?

A

Intox: belligerence, impulsiveness, fever, psychomotor agitation, analgesia, vertical and horizontal nystagmus, tachycardia, homicidality, psychosis, delirium, seizures.
Tx: benzodiazepines, rapid-acting antipsychotic.

Withdrawal: Depression, anxiety, irritability, restlessness, anergia, disturbances of thought and sleep.

333
Q

What is the intoxication and withdrawal symptoms for LSD?

A

Intox: perceptual distortion (visual, auditory), depersonalization, anxiety, paranoia, psychosis, possible flashbacks.

334
Q

What is the intoxication and withdrawal symptoms for Marijuana (cannabinoid)?

A

Intox: euphoria, anxiety, paranoid delusions, perception of slowed time, impaired judgement, social withdrawal, increased appetite, dry mouth, conjunctival injection, hallucinations.

Prescription form is dronabinol (tetrahydrocannabinol isomer): used as anti-emetic (chemotherapy) and appetite stimulant (in AIDS). used when conventional therapies have failed.

Withdrawal: irritability, depression, insomnia, nausea, anorexia. Most symptoms peak in 48 hours and last for 5-7 days. Generally detectable in urine for 4-10 days.

335
Q

What are you at risk for in heroin addiction and what are the treatments?

A

Users at increased risk for hepatitis, abscesses, overdose, hemorrhoids, AIDS, and right-sided endocarditis. Look for track marks (needle sticks in veins).
Treatments:
Methadone: long acting oral opiate; used for heroin detox or long term maintenance
Naloxone and buprenorphine: Partial agonist; long acting with fewer withdrawal symptoms than methadone. Naloxone is not active when taken orally, so withdrawal symptoms occur only if injected (lower abuse potential).
Naltrexone: long-acting opioid antagonist used for relapse prevention once detoxified.

336
Q

What are the complications and treatment for alcoholism?

A

Complications: alcoholic cirrhosis, hepatitis, pancreatitis, peripheral neuropathy, testicular atrophy.
Treatment: disulfiram (inhibitor of acetylaldehyde…to condition the patient to abstain from alcohol use), naltrexone, supportive care. AA and peer support groups are helpful too.

Delirium tremens: life-threatening and peaks 2-5 days after last drink. Symptoms in order of apperance: autonomic system hyperactivity (tachycardia, tremors, anxiety, seizures), psychotic symptoms (hallucinations, delusions), confusion. Treatment is benzodiazepines.

337
Q

What is the treatment for gestational hypertension?

A

hypertension that is high after 20th week of gestation (no existing hypertension and no proteinuria or organ damage)

Treat with anti-hypertensives (alpha-methyldopa, labetalol, hydralazine, nifedipine), deliver at 39 weeks.

338
Q

What is phenoxybenzamine?

A

it’s an irreversible competitive antagonist on alpha receptors.

339
Q

What does hemicholinium do?

A

Inhibits uptake of choline into pre-synaptic cholinergic terminal.

Remember: choline is taken up and then converted with Acetyl-CoA to ACh by ChAT.

340
Q

What does Vesamicol do?

A

It inhibits the uptake of ACh into pre-synpatic vesicles.

341
Q

What does Botulinum do?

A

It inhibits the release of pre-synaptic ACh vesicles.

342
Q

What does AChE convert ACh to?

A

Choline and acetate.

343
Q

What does Metyrosine do?

A

it inhibits the conversion of tyrosine to DOPA in the pre-synaptic nor-adrenergic receptor.

This step is carried out by tyrosine hydroxylase with co-factor BH4

344
Q

What does Reserpine do?

A

It blocks the uptake of dopamine into pre-synaptic storage vesicles.

345
Q

What does Bretylium and guanethidine do ?

A

They block pre-synaptic release of nor-epinephrine.

346
Q

What does amphetamine do?

A

It promotes the pre-synpatic release of nor-epinephrine.

347
Q

What do cocaine, TCAs, and amphetamines do?

A

they prevent reuptake of the nor-epinephrine into the pre-synaptic terminal.

348
Q

What are the regulators of nor-epinephrine at the pre-synaptic terminal?

A

Nor-epinpehrine modulates itself by binding to alpha-2 receptors which prevent further vesicle release.

Angiotensin II acts to promote release of nor-epinephrine presynaptic vesicles.

349
Q

What is Bethanechol? clinical uses and action

A

Cholinomimetic agent - direct agonist
Gq mechanism so increases phosphoinositide turnover.

Uses: Postoperative ileus, neurogenic ileus, urinary retention

Action: Activates Bowel and Bladder smooth muscle; resistant to AChE. “Bethany, call (bethanechol) me, maybe if you want to activate your Bowels and Bladder”
Think all the B’s for this drug.

350
Q

What does Carbachol do? uses and action

A

Cholinomimetic agent - direct agonist

Uses: glaucoma, pupillary constriction, and relief of intraocular pressure.

CARBon copy of of AcetylCHOLine.

351
Q

What does pilocarpine do? uses and action

A

Cholinomimetic agent - direct agonist

Uses: Potent stimulator of sweat, tears, and saliva. Open angle and closed angle glaucoma.

Action: Contracts ciliary muscle of eye (open angle glaucoma), pupillary sphincter (closed angle glaucoma); resistant to AChE. “You cry, drool, and sweat on your “pilow”.

352
Q

What does methacholine do? uses and action

A

Cholinomimetic agent - direct agonist

Use: Challenge test for diagnosis of asthma.

Action: stimulates muscarinic receptors in airway when inhaled.

353
Q

What does Neostigmine do? uses and action

A

Cholinomimetic agent - indirect agonist (anticholinesterases)

Uses: postoperative and neurogenic ileus and urinary retention, myasthenia gravis, reversal of neuromuscular junction blockade (postoperative).

Action: Increases endogenous ACh. Neo CNS = NO CNS Penetration.

354
Q

What does pyridostigmine do?

A

Cholinomimetic agent - indirect agonist (anticholinesterases)

Uses: Myasthenia gravis (long acting); does not penetrate CNS.

Action: Increased endogenous ACh, increased strength. PyRIDostiGMine gets RID of Myasthenia Gravis.

355
Q

What does physostigmine do? uses and action

A

Cholinomimetic agent - indirect agonist (anticholinesterases)

Uses: Anti-cholinergic toxicity (crosses blood brain barrier -> CNS).

Action: increased endogenous ACh. PHYsostigmine “PHYxes” atropine overdose.

356
Q

What does Donepezil, rivastigmine, and galantamine do? uses and action

A

Cholinomimetic agent - indirect agonist (anticholinesterases)

Uses: Alzheimer disease

Action: Increased endogenous ACh.

357
Q

What does Edrophonium do? uses and action

A

Cholinomimetic agent - indirect agonist (anticholinesterases)

Uses: Historically diagnosis of myasthenia gravis (extremely short acting); Myasthenia now diagnosed by anti-AChR Ab (anti-acetylcholine receptor antibody) test.

358
Q

What do you need to worry about for all cholinomimetic agents?

A

Watch for exacerbation of COPD, asthma, and peptic ulcers when giving to susceptible patients.

M3 causes smooth muscle contraction (so bronchoconstriction, stimulation of acid secretion).

359
Q

Describe cholinesterase inhibitor poisoning?

A

Often due to organophosphates, such as parathion, that irreversibly inhibit AChE. Causes Diarrhea, Urination, Miosis, Bronchospasm, Bradycardia, Excitation of skeletal muscle and CNS, Lacrimation, Sweating, and Salivation.

DUMBBELSS.
Organophosphates are a component of insecticides; poisoning usually seen in farmers.
Antidote is atropine (competitive inhibitor) + pralidoxime (regenerates AChE if given early enough).

360
Q

What are atropine, homatropine, tropicamide? use and applications

A

Muscarinic antagonist

Acts at eyes. Produces mydriasis and cycloplegia. Atropine used as antidote for AChE inhibitor poisoning.

361
Q

What does atropine do in more detail? Actions on which systems. Toxicity?

A

Muscarinic antagonist.
Used to treat bradycardia and for ophthalmic applications.

Action: Increases pupil dilation, cycloplegia (Eye), decreased secretions (Airway), decreased acid secretion (stomach), decreased motility (gut), decreased urgency in cystitis (bladder).

Blocks DUMBBeLSS. Note it does not affect the E, excitation of Skeletal muscles and CNS. Those are mediated by nicotinic receptors NOT muscarinic. This is relevant because you can treat myasthenia gravis with AChE inhibitor and give atropine to block unwanted muscarnic side effects, without taking away the therapeutic effect on muscles.

Toxicity: Increased body temperature (due to decreased sweating); rapid pulse; dry mouth; dry flushed skin; cycloplegia; constipation; disorientation.
Can cause acute angle-closure glaucoma in elderly (due to mydriasis), urinary retention in men with prostatic hyperplasia, and hyperthermia in infants.
Hot as a Hare, Dry as a Bone, Red as a Beet, Blind as a Bat, Mad as a Hatter.

Also Jimson weed (Datura)…causes gardeners pupil (mydriasis due to plant alkaloids). It’s made of atropine and scopolamine.

362
Q

What does Benztropine do? usess

A

Muscarinic antagonist

Acts in the CNS system and used for Parkinson disease - “PARK my BENZ”

363
Q

What is scopolamine used for? uses

A

Muscarinic antagonist

Motion sickness

364
Q

What is Ipratropium, tiotropium used for? uses

A

Muscarinic antagonist
In respiratory system for COPD, asthma (I PRAy I can breathe soon).

Ipratropium (short), tiotropium (long)

365
Q

What is Oxybutyin, darifenacin, and solifenacin used for?

A

Muscarinic antagonist
In genitourinary system for reducing urgency in mild cystitis and reducing bladder spasms.

Other agents: tolterodine, fesoterodine, trospium.

366
Q

What does glycopyrrolate do? uses

A

Muscarinic antagonist

Used in Gastrointestinal and respiratory systems for preoperative use to reduce airway secretions (parenteral) and for drooling and peptic ulcers (orally).

367
Q

What is the receptor effect of epinephrine? and uses

A

Direct sympathomimetic
Beta greater than alpha
Uses: anaphylaxis, open angle glaucoma, asthma, hypotension; alpha effects predominate at high doses

368
Q

What is the receptor effect of norepinephrine? and uses

A

Direct sympathomimetic
alpha1 > alpha2 > Beta1
Uses: hypotension (but decreased renal perfusion).

369
Q

What is the receptor effect of isoproterenol? and uses

A

Direct sympathomimetic
Beta1 = Beta2
uses: electrophysiologic evaluation of tachyarrhythmias. Can worsen ischemia.

370
Q

What is the receptor effect of Dopamine? and uses

A

Direct sympathomimetic
D1 = D2 > Beta > alpha

Uses: unstable bradycardia, heart failure, shock;
inotropic and chronotropic alpha effects predominate at high doses.

371
Q

What is the receptor effect of Dobutamine? and uses

A

Direct sympathomimetic
B1 > B2, alpha.

Uses: Heart failure (inotropic > chronotropic), cardiac stress testing.

372
Q

What is the receptor effect of Phenylephrine? and uses

A

Direct sympathomimetic
alpha1 > alpha 2.

Uses: Hypotension (vasoconstrictor), ocular procedures (mydriatic), rhinitis (decongestant) .

373
Q

What is the receptor effect of Albuterol, salmeterol, terbutaline? and uses

A

Direct sympathomimetic
B2 > B1.

Uses: Albuterol for acute asthma; salmeterol for long-term asthma or COPD control; terbutaline to reduce premature uterine contractions.

374
Q

What is the effect of Amphetamine? and uses

A

Indirect sympathomimetic.

Action: indirect general agonist, reuptake inhibitor, also released stored catecholamines.

Uses: narcolepsy, obesity, attention deficit disorder.

375
Q

What is the effect of Ephedrine? and uses

A

Indirect sympathomimetic

Action: Indirect general agonist, released stored catecholamines.

Uses: nasal decongestion, urinary incontinence, hypotension.

376
Q

What is the effect of Cocaine? and uses

A

indirect sympathomimetic
Action: Indirect general agonist, reuptake inhibitor.

Uses: causes vasoconstriction and local anesthesia; never give B-blockers if cocaine intoxication is suspected ( can lead to unopposed alpha1 activation and extreme hypertension).

377
Q

What does clonidine do? uses and toxicity

A

Sympatholytic (alpha2- agonist)

Uses: Hypertensive urgency (limited siutations); does not decrease renal blood flow. ADHD, severe pain, and a variety of off-label indications (e.g. ethanol and opioid withdrawal).

Toxicity: CNS depression, bradycardia, hypotension, respiratory depression, and small pupil size.

378
Q

What does alpha-methyldopa do? uses and toxicity

A

Sympatholytic (alpha2- agonist)

Uses: Hypertension in pregnancy. Safe in pregnancy.

Toxicity: Direct Coombs + hemolytic anemia, SLE-like syndrome.

379
Q

What does Phenoxybenzamine do? uses and toxicity

A

nonselective alpha blocker. Irreversible.

Uses: pheochromocytoma (used preoperatively) to prevent catecholamine (hypertensive) crisis.

Toxicity: orthostatic hypotension, reflex tachycardia.

380
Q

What does phentolamine do? uses and toxicity.

A

nonselective alpha blocker. Reversible.

Uses: Give to patients on MAO inhibitors who eat tyramine containing foods.

381
Q

What do prazosin, terazosin, doxazosin, and tamsulosin do? uses and toxicity.

A

alpha-1 selective blocker. Think all the -osin endings.

Uses: urinary symptoms of BPH; PTSD (prazosin); hypertension (except tamsulosin).

Toxicity: 1st dose orthostatic hypotension, dizziness, headache.

382
Q

What does mirtazapine do? uses and toxicity

A

Alpha-2 selective inhibitor.

Uses: depression.

Toxicity: sedation, increased serum cholesterol, increased appetite.

383
Q

What’s the difference between high dose epinephrine and phenylephrine in terms of blood pressure response before and after application of alpha blockade (like phentolamine)?

A

Epinephrine: Before it has an increase in blood pressure because of the high dose effects on alpha receptors. After alpha blockade, the Beta effects win out and you get a drop in blood pressure (primarily B2).

Phenylephrine: Before it has an increase in blood pressure because it is alpha 1 agonist (more than alpha 2). After there is a suppressed change (i.e. the pressure does not change), because it is only an alpha 1 agonist and nothing else.

384
Q

what do metoprolol, acebutolol, betaxolol, carvedilol, esmolol, atenolol, nadolol, timolol, pindolol, labetalol do? uses, effects, toxicity.

A

Beta blockers.

Angina pectoris: decreased heart rate and contractility, result in decreased O2 consumption
MI: B-blockers (metoprolol, carvedilol, bisoprolol) decreased mortality
SVT: Metoprolol, esmolol. Decreased AV conduction velocitiy (class 2 antiarrhythmic).
Hypertension: decreased cardiac output, decreased renin secretion (Beta 1 blockade on JGA cells).
CHF: slows progression of chronic failure
Glaucoma: Timolol. decreased secretion of aqueous humor.

Toxicity: Impotence, cardiovascular effects (bradycardia, AV block, CHF), CNS adverse effects (seizures, sedation, sleep alterations), dyslipidemia (metoprolol), and asthmatics/COPDers (may cause exacerbation).
Avoid in cocaine users due to risk of unopposed alpha adrenergic receptor agonist activity. Despite theoretical concern of masking hypoglycemia in diabetics, benefits likely outweigh risks; not contraindicated.

385
Q

How do Beta blockers mask the “effects of hypoglycemia”?

A

They mask the effects in diabetics taking insulin.
When a person becomes hypoglycemia they begin experience sympathetic symptoms (tremors, sweating, palpitations). Beta blockers (specifcally through B1) will block these effects and a person will not be aware their blood glucose is dropping. Also, through effects on blocking B2 on muscle and liver (which is normally stimulated by epinephrine), there will be decreased glycogenolysis and gluconeogenesis and it will be harder to correct for hypoglycemia.

386
Q

Which beta blockers are B1-selective?

A

B1 selective antagonists (B1 > B2): acebutolol (partial agonist), atenolol, betaxolol, esmolol, metoprolol.
Selective antagonists mostly go from A to M (B1 with first half of alphabet).

387
Q

Which beta blockers are non-selective?

A
nonselective antagonists (B1 = B2): nadolol, pindolol (partial agonist), propranolol, timolol. 
Non-selective antagonists mostly go from N to Z (Beta 2 with second half of the alphabet).
388
Q

Which beta blockers are non-selective alpha and beta antagonists?

A

nonselective alpha and beta antagonists: carvedilol, labetalol.
Nonselective alpha- and beta-antagonists have modified suffixes (instead of -“olol”).

389
Q

What does nebivolol do?

A

Combines cardiac-selective B1-adrenergic blockade with stimulation of B3 receptors, which activate NO synthase in vasculature.

390
Q

What is pegvisomat?

A

Growth hormone receptor antagonist. Used for treating acromegaly.

391
Q

How do you treat nephrogenic DI?

A

HCTZ, indomethacin, amiloride (the diuretics work by causing diuresis and promoting reabsorption at the proximal tubule)
Hydration

392
Q

What is the treatment for SIADH?

A

Treatment: fluid restriction, IV hypertonic saline, conivaptan, tolvaptan, demeclocycline (these last three drugs are ADH antagonists).

393
Q

What is the treatment for post-partum hemorrhage?

A

oxytocin has been shown to help by inducing uterine contraction.

If not available, misoprostol can be used to (PGE analog)

394
Q

Which drugs prolong the QT and what is the treatment for torsades de pointes?

A
Sotalol
Risperidone (anti-psychotics) 
Macrolides
Chloroquine 
Protease inhibitors (-navir)
Quinidine (class Ia..also class III)
Thiazides

Some Risky Meds Can Prolong QT

Treatment is with Magnesium Sulfate.

Remember: torasdes de pointes can progress to VT. Decrease K+ or Mg+ can also prolong QT.

395
Q

What is nesiritide?

A

a recombinant form of B-type natriuretic peptide for heart failure.

Remember: this is normally released from ventricular myocytes (vs ANP which is released from atrial myocytes) in response to increased tension. Similar physiological action as ANP but with longer half life: it causes vasodilation, decreased Na+ absorption in the collecting tubule, and constriction of efferent arterioles and dilation of afferent arterioles via cGMP. thus promotes diuresis and contributes to aldosterone escape. BNP is also used for diagnosing heart failure (has very good negative predictive value).

396
Q

Which hypertensive drugs are ok for pregnancy?

A

methyldopa
hyralazine
nifedipine
Labetol

My Hypertension No Likey

397
Q

What drug can you use for premature ejaculation?

A

SSRIs like paroxetine.

398
Q

how do you treat bedwetting?

A

oral desmopressin acetate (DDAVP).

Can also be used for central nephrogenic insipidus and von willebrands disease or hemophilia A (because of its effect on vasculature V2 to increase von willebrand factor release and consequently factor VIII).

399
Q

What can you use to treat hyperammonia?

A

lactulose to acidify GI tract and trap NH4+ for excretion.
Benzoate or phenylbutrate bind amino acids and lead to excretion, decreasing ammonia levels.

Note: this happens usually because of liver disease or hereditary urea cycle enzyme deficiencies. Results in excess NH4+ which depletes alpha ketoglutarate and blocks TCA cycle.

400
Q

What is used for prophylaxis and treatment of pneumocystis jirovecii?

A

TMP-SMX and pentamidine are used for both.

Dapsone and atovaquone are used for prophylaxis only.

401
Q

What is the treatment for serontonin syndrome?

A

Cyproheptadine (5-HT2 receptor antagonist)

402
Q

What is trimethaphan?

A

Trimetaphan camsilate (INN) or trimethaphan camsylate (USAN), trade name Arfonad, is a drug that counteracts cholinergic transmission at the ganglion type of nicotinic receptors of the autonomic ganglia and therefore blocks both the sympathetic nervous system and the parasympathetic nervous system. It acts as a non-depolarizing competitive antagonist at the nicotinic acetylcholine receptor, is short-acting, and is given intravenously.

Uses: It is occasionally used to treat a hypertensive crisis and dissecting aortic aneurysm, to treat pulmonary edema, and to reduce bleeding during neurosurgery

403
Q

What’s an appropriate initial therapy for ulcerative collitis?

A

mesalamine (5-ASA compound)

404
Q

How can type 2 Crigler Najar syndrome be treated ?

A

Phenobarbital which increases enzyme synthesis.

Note: Type 2 is already a milder form.

405
Q

What is prochlorperazine?

A

Phenothiazine antiemetic that acts by blocking dopamine receptors in the chemoreceptor trigger zone (floor of the fourth ventricle in the medulla). Helps with severe nausea and vomiting as well as nonpsychotic anxiety.

Side effects: tardive dyskinesia, neuroleptic malignant syndrome, blood dsycrasias, lupus.

Basically one of the high potency antipsychotics.