Term
|
Definition
anything that brings about a change in a biological system
Any agent approved by the Food and Drug Administration for the treatment or prevention of disease. |
|
|
Term
what is phase 1 in drug testing on humans? |
|
Definition
study of 20-100 seeing if a drug is even safe in humans, pharmacokinetics |
|
|
Term
what is phase two of human testing trial? |
|
Definition
is it safe in PATIENTS with the disorder
100-200 |
|
|
Term
|
Definition
does it WORK in patients, double blind studies
1000-6000 patients |
|
|
Term
what is phase 4 of testing? |
|
Definition
post marketing surviellance
selling to the population at large |
|
|
Term
what is studied when a drug is researched on animal testing? |
|
Definition
acute toxicity, possibly LD testing
chonic toxicity testing if drug is for long term
effect on reproduction
carcinogenisis/mutagenesis
mechanisms of toxicity |
|
|
Term
what is pharmacodynamics? |
|
Definition
effecs of a drug on the body
(mechanism of action magnitude of effect)
receptor binding
signaling mechanism
physiological effect |
|
|
Term
what is pharmacokinetics? |
|
Definition
effects of the body when on a drug
-absorption
-distribution
-metabolism
-excretion |
|
|
Term
the majority of drugs are between 100-1000 MW size. What dictates this? |
|
Definition
lower limit set by requirement for sufficient selectivity
upper limit dictated by poor absorption nd distribution in the body |
|
|
Term
what are 3 requirements of a drug when binding to its designated receptor? |
|
Definition
must be selective in their binding properties
receptor becomes altered or modified when bound
binding of the drug initiates a series of events that leads to the observed functional change |
|
|
Term
what is the most common target receptors? |
|
Definition
proteins
ion channels
enzymes
transport proteins
protiens that bind neurotransmitters or hormones |
|
|
Term
what are teh top 4 protein receptors targeted? |
|
Definition
GPCR
nucear
ligand gated ion
voltaged gated ion |
|
|
Term
how does non-nuclear estrogen signaling work? |
|
Definition
estrogen binds which causes the phosphorylation of endothelial nitric oxide synthase (eNOS) leading to:
increase eNOS activity
increase NO production
Increase dilation |
|
|
Term
how do receptor tyrosine kinases work? |
|
Definition
in the ex. of epidermal growth factor receptor:
ligand binds extracellular domain which can then bind with another bound momomeric receptor to become active dimers
intracellular cytoplasmic enzyme domains then become activated and phosphorylate other substrate proteins |
|
|
Term
how does a JAK kinase work? |
|
Definition
just the same as a tyrosine kinase but kinase activity is not intrinsic to the receptor
•Cytokine binding promotes receptor dimerization, phosphorylation and activation of associated Janus kinase (JAK)
•JAK phosphorylates signal transducers and activation of transcription (STAT) molecules, which dimerize, travel to the nucleus and regulate gene expression |
|
|
Term
|
Definition
acts as a decoy receptor for TNF alpha which promotes inflammation, thus reducing inflammation for rheumatory arthritis |
|
|
Term
what is the magnitude of a drugs effect? |
|
Definition
the amount of drug:receptor complexes formed.
Maximum effect is achieved when all receptors are occupied. |
|
|
Term
|
Definition
drug dose that produces 50% of the max effect |
|
|
Term
how does the EC50 relate to potency? |
|
Definition
the lower the EC50 the higher the potency |
|
|
Term
what is affinity?
How is potency affected by affinity? |
|
Definition
affinity is the ability of the drug to bind its receptor site
The potency of a drug is dependent on the affinity of the drug for its receptor.
Drugs with a higher affinity are usually more potent. |
|
|
Term
|
Definition
Kd = drug concentration required to bind 50% of the receptor sites in the system
the smaller the Kd the greater the drugs affinity --> potency = less drug needed to get a desired affect |
|
|
Term
how does a competative antagonist shift the dose-response curve? |
|
Definition
It shifts it to the R, increasing the EC50.
it is surmountable if enough agonist is added |
|
|
Term
can partial agonists have antagonistic activity? |
|
Definition
YES
if a partial agonist is added with a full agonist, it will act as an antagonist to the full agonist causing a max partial agonist response rather than a full agonist response |
|
|
Term
what are the effects of a noncompetitive antagonist on a dose-response curve? |
|
Definition
Irreversible antagonist shifts the response curve downwards, reducing the maximal response.
The effects of an irreversible antagonist are insurmountable and cannot be overcome by adding more agonist |
|
|
Term
what is the effect and binding properties of allosteric modulators? |
|
Definition
•Bind to a site on the receptor that is distinct from the agonist binding site
•Have no direct activity on their own
•Modulate the affinity of the agonist for the receptor or the level of receptor activation after agonist binding
- Increase response : Positive modulator
- Decrease response : Negative modulator |
|
|
Term
how does a GABA receptor work? |
|
Definition
chloride channel expressed on neurons that
inhibits neuron function when activated |
|
|
Term
what's an example of physiological antagonism? |
|
Definition
Catabolic actions of glucocorticoid hormones can lead to increased blood sugar
Insulin acts at the insulin receptor to decrease blood sugar levels
Clinicians sometimes administer insulin to oppose the hyperglycemic effects of elevated glucocorticoid hormones levels. |
|
|
Term
how do chemical antagonists work? |
|
Definition
•Interact directly with the drug/agonist
- Remove the drug from the system
-Prevent the drug from binding to its receptor
-
•Examples:
-Protamine sulfate: reverses anticoagulant effects of heparin by stably binding to it, dissociates heparin from antithrombin III
-Dimercaprol: binds to and chelates lead, arsenic and other toxic metals, chelated metal is excreted in the urine |
|
|
Term
what are chemical and physiological antagonists classified as? |
|
Definition
|
|
Term
what is the effect of spare receptors and irreversable antagonists? |
|
Definition
When 5 receptors need to be filled to reach a max response and if the irreversable antagonists have bound 3 out of 10 receptors you can increase the drug to still reach that response, this is true even if 5 of ten are bound.
When there are less than 5 receptors available to be bound due to irreversable antagonists, you can no longer reach a max response and your curve is further pushed to the right. |
|
|
Term
why are there spare receptors? |
|
Definition
•Allows the maximal response to occur without total receptor occupancy which increases the sensitivity of the system
•
•Spare receptors can bind up any extra agonist preventing an excessive response if too much agonist is present |
|
|
Term
in a system with spare receptors what is the ED50 to Kd ratio?
What does this mean? |
|
Definition
ED50< Kd
there is more sensitivity to the agonistand maximum effect is achieved before all receptors are occupied
when there are no spare receptors
ED50 = Kd |
|
|
Term
|
Definition
the maximum effect a drug/agonist can produce |
|
|
Term
what is drug threshold?
How is it the inverse of spare receptors? |
|
Definition
•Allows the maximal response to occur without total receptor occupancy which increases the sensitivity of the system
•Spare receptors can bind up any extra agonist preventing an excessive response if too much agonist is present
. As you bind more receptors you don’t produce an effect at all until a certain percentage of receptors are bound.
drug threshold decreases the sensitivity of a system to agonist
|
|
|
Term
how does a drug threshold affect ED50 to Kd? |
|
Definition
With a drug threshold, EC50 > Kd
drug threshold decreases the sensitivity of a system to agonist |
|
|
Term
|
Definition
•Frequent or continuous exposure to agonists over a short period of time often can result in a rapid reduction in the receptor response = DESENSITIZATION |
|
|
Term
what is pharmacokinetic tolerance? |
|
Definition
change of responsiveness at the drug level
Decreased absorption
Decreased penetration to site of action
Increased metabolism
Increased clearance
The amount of drug that reaches the site of action is decreased |
|
|
Term
what is pharmacoynamic tolerance? |
|
Definition
responsiveness change at the receptor level
Change in receptor
Receptor uncoupled from signaling pathway
Total number of receptors in the system is decreased |
|
|
Term
what is the effect of Beta arrestin binding to a GPCR? |
|
Definition
prevents G protein from from interacting with the receptor. Uncoupling the receptor . The receptors can also be internalized and be degraded by lysosomes. This leads to an overall decrease in the total number of receptors.
|
|
|
Term
how are quantal drug doses different from graded drug doses? |
|
Definition
Quantal doses give an all or none effect
graded increases doses give a incremental increase in effect |
|
|
Term
how is a quantal dose discovered? |
|
Definition
testing the population and finding the mean minimal dose required to produce a specified effect for all members of a population |
|
|
Term
how is an ED50 determined for a quantal-dose reponse plot? |
|
Definition
ED50 = effective dose at which 50% of subjects respond a drug with a high potency has a low ED50
|
|
|
Term
how is a therapeutic index calculated? |
|
Definition
dose toxic effect
= _________________
dose therapeutic effect
The higher the TI, the safer the drug |
|
|
Term
If TI ≤ 2.0, the drug is considered to have a narrow therapeutic index (NTI) What are 4 drugs in use with a NTI? |
|
Definition
Lithium - bipolar disorder
Warfarin - prevents blood clots
Digoxin – congestive heart failure
Theophylline- respiratory diseases such as asthma |
|
|
Term
what is a theraputic window? |
|
Definition
Index used for estimation of drug dosage
Defined range of drug concentration at which the desired effect occurs within the safety range.
Below the therapeutic window there is little effect and above the toxicity is greater than the desired effect
Typically the range is between ED50 and TD50 |
|
|
Term
what is the calculation for Kd?
What is it for EC50? |
|
Definition
Kd= 1/affinity
EC50 = 1/potency |
|
|
Term
in Pharamacokinetics what are the 4 main ways the body acts on a drug? |
|
Definition
absorption
distribution
metabolism
excretion |
|
|
Term
bioavailability is a measure of the drug available to the systemic circulation over time after administration. How is it measured?
|
|
Definition
|
|
Term
what things determine a drugs ability to diffuse? |
|
Definition
1.Size
2.Area
3.Lipid solubility
4.Thickness
5.Concentration gradient |
|
|
Term
|
Definition
pKa is the pH value at which [protonated]=[nonprotonated] |
|
|
Term
if you want to give a drug as a weak acid, where is the best environment for it? |
|
Definition
For weak acids, if pH is low (< pKa), lots of H+ are around, which drives A− toward HA and absorption is favored. |
|
|
Term
what if weak bases are added to an acidic environment? |
|
Definition
For weak bases, if pH is low (< pKa), lots of H+ are around, which will drive B toward BH+ and
absorption is disfavored |
|
|
Term
what controls a drugs distribution through the body? |
|
Definition
1. Different tissues have different blood perfusion rates;
2. For the same tissue, different drugs have different partition ratio;
3. For the same drug, different tissues have different partition ratio.
4. Blood flow can dictate how fast a drug saturates a tissue.
5. Partition ratio Kp determines the maximum amount of distribution at a tissue
|
|
|
Term
what is the tissue/blood partition ration Kp? |
|
Definition
Kp = Ctissue/Cvenous-blood (at equilibrium) |
|
|
Term
what dictates drug distribution to the brain? |
|
Definition
|
|
Term
what is the PK concept of volume of distribution (Vd)?
how is it calculated? |
|
Definition
Vd: The volume of fluid in which a drug would need to be dissolved (homogenously) in order to have the same concentration in that volume as it does in plasma.
Vd= amount of drug in body/plasma [drug] |
|
|
Term
what is the major site of 1st pass metabolism? |
|
Definition
|
|
Term
what is the reason for Phase I metabolism? |
|
Definition
functionalization:
oxidation
reduction
hydrolysis |
|
|
Term
what happens in phase II metabolism of drugs? |
|
Definition
The products of phase I functionalization are conjugated, commonly with endogenous molecules like glucuronic acid. The reason for this is to make a hydrophilic compound that can be excreted.
|
|
|
Term
do all metabolic processes result in a diminution of pharmacological activity for a given drug? |
|
Definition
No
some such as 5FU become active. This is known as bioactivation |
|
|
Term
what is metabolic toxicity? |
|
Definition
when a drug becomes a toxin due to metabolism
This is the case for acetominophen being metabolized to NAPQI. Sulfhydryl groups can be attacked by NAPQI. Large amounts can lead to liver damage when it nucleophilicaly attacks proteins
|
|
|
Term
what type of enzymes are responsible for phase II metabolism? |
|
Definition
|
|
Term
what are the three major liver CYP40's? |
|
Definition
CYP2C9 - metabolizes weakly acidic drugs
CYP2D6 - metabolizes weakly basic drugs
CYP3A4 - metabolizes practically everything else
|
|
|
Term
what is the main function of cytochrom P450 enzymes?
Where are they found? |
|
Definition
Catalyzing oxidative metabolism reactions (Phase I)
found in the liver sitting on the ER membrane of hepatocytes |
|
|
Term
in metabolic rxns for phase I, where do oxidation rxns occur mostly? |
|
Definition
at carbon, nitrogen, and sulfur centers |
|
|
Term
for metabolic rxns in Phase I, where do hydrolysis rxns occur? |
|
Definition
mainly on esters which serve as delivery systems for prodrugs |
|
|
Term
for metabolic rxns in Phase I, where do reduction rxns occur mostly? |
|
Definition
the same as oxidation rxns, at carbon nitrogen and sulfur centers |
|
|
Term
what are the 6 major conjugation rxns in phase II metabolism? |
|
Definition
1.Glucuronidation
2.Sulfation
3.Acetylation
4.Methylation
5.Glutathione conjugation
6.Amino acid conjugation |
|
|
Term
what is enzyme induction? |
|
Definition
An increase in the steady-state concentration of enzyme following exposure to an appropriate stimulus
ex. for someone who drinks a lot of alcohol, they will have raised CYP levels for dealing with OH reduction |
|
|
Term
what are causes of enzyme induction? |
|
Definition
Chemicals | drugs | xenobiotics
· Diet | Nutritional factors
· Biological rhythms (stress/pregnancy) |
|
|
Term
what are some mechanisms of P450 induction (which presumably could be the same for other enzymes)? |
|
Definition
(1) Increased rate of transcription/gene expression of P450s
(2) Decreased rate of degradation of P450 protein products
(3) Increased rate of translation of P450 protein products |
|
|
Term
what is enzyme inhibition? |
|
Definition
Loss or decrease of the drug-metabolizing enzymatic activity following exposure to stimuli
ex: GFJ induces CYP3A4 protein degradation
|
|
|
Term
what are major excretory routes |
|
Definition
urine
bile
feces
expired air |
|
|
Term
what are minor excretory routes? |
|
Definition
|
|
Term
what does the bile system typically excrete?
what are 2 examples? |
|
Definition
highly polar organic anions and cations (and some neutral molecules) with molecular weights greater than 300-500
ex: Glucuronate and sulfate derivatives of steroids |
|
|
Term
how are drugs that are excreted by the bile reabsorbed into circulation and returned to the liver? |
|
Definition
Intestinal glucuronidases and sulfatases can convert drug metabolites back to the free drug.
Intestinal bacteria can facilitate this process |
|
|
Term
what is the kinetic order of drug elimination equation? |
|
Definition
|
|
Term
what is a zero order elimination? |
|
Definition
when n = 0
this means that A (amount of drug) has a decreasing LINEAR relationship with t at any given time.
The rate of drug elimination is constant
Zero order kinetics suggests that
the elimination process is saturated
|
|
|
Term
what is a 1st order elimination? |
|
Definition
when n = 1
The amount of drug decreases exponentially as time goes
The rate of drug elimination (dA/dt) is proportional to the (remaining) amount of drug
|
|
|
Term
If drug elimination has first-order kinetics, how long does it take to have 95% of the drug to be removed after a single dose? |
|
Definition
0 = 100
1 = 50
2= 25
3 = 12.5
4 = 6.25
5 = 3.125 |
|
|
Term
what are the differences between zero order and 1st order kinetics? |
|
Definition
rate of elimination:
zero = constant, 1st = dependent on A
limination rate constant:
zero = mg/hr, 1st = hr
half life (t1/2):
zero = n/a, 1st = constant
relationship between A and T
zero = linear, 1st = exponential |
|
|
Term
what's the practical way to monitor drug in the body? |
|
Definition
Measure plasma drug concentration Cplasma (Cp).
What is Cp equal to?
Amount plasma/volume plasma |
|
|
Term
is the amount of the drug in the plasma equal to the amount of the drug in the body? |
|
Definition
|
|
Term
because the amount of drug in plasma isn't equal to the amount of drug in the body, how do we measure the amount of drug in the body? |
|
Definition
|
|
Term
if we want to know the amount of drug in the volume of the body, what equation do we use? |
|
Definition
volume of distribution =
Vd = amount of drug in the body/ plasma drug concentration |
|
|
Term
what is the concept of Clearance? |
|
Definition
a measure of the efficiency of irreversible removal of a drug by an eliminating organ
CL = rate of elimination/plasma [drug]
|
|
|
Term
what are the rules of clearance? |
|
Definition
CL is a constant for first-order kinetics
CL is dependent on the drug and the condition of the eliminating organ |
|
|
Term
what are the two ways of determining renal clearance of a drug in relationship to the GFR? |
|
Definition
if excretion > creatinine clearance = active secretion
if excretion < creatinine clearance = reabsorption or plasma protein bound |
|
|
Term
|
Definition
CL = (dA/dt)/Cp = −KA / (A/Vd) = − KVd ß ml min−1
so CL can be considered as a fraction of Vd that
gets “cleared” in unit of time. |
|
|
Term
how does clearance relate to half-life? |
|
Definition
Since t½ = 0.693 / K & CL= K Vd
CL = 0.693Vd / t½ or t½ = 0.693Vd / CL |
|
|
Term
what does t½ = 0.693Vd / CL
mean? |
|
Definition
the half life is proportional to the volume of distribution and inversely proportional to the clearance |
|
|
Term
what does CL = 0.693Vd / t½
mean? |
|
Definition
For two drugs with the same half-life, if one has a larger volume of distribution, it will require higher clearance for elimination |
|
|
Term
how many half lives are needed to reach a steady state? |
|
Definition
4−5 half-lives are required to achieve a steady state level.
3−4 half-lives are adequate to achieve an effect indistinguishable to the steady state effect |
|
|
Term
If Css is the target Cp what should the dosing rate be? |
|
Definition
Dosing ratess = CL X Css
meaning that in a steady state the dosing rate must be equal to the rate of elimination |
|
|
Term
what is the goal of the loading dose? |
|
Definition
to rapidly achieve therapeutic concentration
Loading dose = Vd X Ctarget
(Cp=A / Vd) |
|
|
Term
what are the differences in dosing rate between:
IV
intermintent
Oral |
|
Definition
Iv: dosing rate = infusion rate
intermittent: dosing rate =
maintenance dose/dose interval
Oral dose: dosing ratess= dosing rateoralx bioavailability |
|
|
Term
what is the enzyme used to make acetylcholine? |
|
Definition
choline acetyl transferase
takes choline and Acetyl CoA and makes Ach |
|
|
Term
what is the function of Hemicholinium-3? |
|
Definition
blocks choline uptake, depletes ACh |
|
|
Term
what is the difference between botulinum toxin A and toxin B? |
|
Definition
A cleaves a protein on the nerve terminal
B cleaves a protein on the synaptic vessicle |
|
|
Term
what are some uses for botulinum? |
|
Definition
Persistent muscle spasms
Poststroke spasticity
Treatment of hyperhidrosis
Healing of anal fissure
Migraine headaches
Neurogenic detrusor overactivity
Elimination of facial wrinkles |
|
|
Term
what happens if you denervate a muscle fiber? |
|
Definition
the AchR on the fibers increase x20 |
|
|
Term
what are therapeutic uses of neuromusclar blockers? |
|
Definition
•Bind to nAChR and block neuromuscular transmission
•Muscle relaxants during surgery
•Facilitate manipulations during realignment of fractures
•Decrease physical trauma during electroshock therapy
•Usually given IV; not active after oral administration
•Doesn’t act on CNS |
|
|
Term
what are the toxic effects of neuromusclar blockers? |
|
Definition
•Neuromuscular blockade- respiratory paralysis
•
While relatively specific for neuromuscular nAChR, can block nAChR in autonomic ganglia- arrhythmia (tachycardia) and hypotension
•
d-tubocurarine can cause histamine release-hypotension and bronchial spasm
•
Aminoglycosides and tetracyclines interfere with Ca influx and ACh release and act synergistically with N-M blockers
RESPIRATORY FAILURE AND CV COLLAPSE!!!!!!
|
|
|
Term
What are Tx options for NM blocks? |
|
Definition
•Competitive antagonists: Treat with AChE Inhibitors: Increase ACh levels, overcome competitive blockade |
|
|
Term
what have non-competitive AchE inhibitors been used for? What are two examples? |
|
Definition
the Tx of Alzheimers (counter act the effect of the loss of cholinergic neurons)
Tacrine- first drug approved for treatment of Alzheimer’s; many actions in addition to AChE inhibition; high incidence of liver toxicity
Donepezil- more specific for inhibition of AChE; delays progression of symptoms; side effects- diarrhea, etc.- expected based on cholinergic stimulation |
|
|
Term
myasthenia gravis is an autoimmune disease that forms Abs against the nAchR decreasing # and function at the NMJ. What are tx options?
What should be given concurrently? |
|
Definition
AChE Inhibitors- increase [ACh] at the NMJ- improves efficiency of NM transmission (neostigmine, pyridostigmine)
Also: use a muscarinic receptor antagonist (e.g., atropine) to counter effects of increased ACh at muscarinic synapses |
|
|
Term
what is a good diagnosisng drug for Myas. gravis? |
|
Definition
endrophonium is a short acting AchE inhibitor causing transient improvement |
|
|
Term
what are the two sympathetic systems that don't follow the normal rules? |
|
Definition
sweat glands
adrenal medulla |
|
|
Term
what is the affect of Ach by the parasympathetic on the SA node? |
|
Definition
mAchR increases K+ permeamiability slowing rate of diastolic depolarization
Ach decreases the rate of the atrial action potential
(decreases Ca entry & force of contraction) |
|
|
Term
how does the sympathetic cause dilation of the eye? |
|
Definition
relaxation of the ciliary muscles decreasing curvature of the lens
opposite effect for PSNS |
|
|
Term
What are indicators of abnormal autonomic function? |
|
Definition
•Resting Heart Rate > 90 beats/min
•Inability to achieve 85% of predicted maximal HR (220-age) on treadmill testing
•Abnormal HR recovery (failure to decrease HR≥ 12 bpm during first minute after peak exercise)
•Abnormal HR variability (failure to change HR by ≥10 bpm during 1 minute of slow deep breaths) |
|
|
Term
is ChAT made only by neurons? |
|
Definition
No
-Airway epithelial cells
-Vascular endothelial cells
-Keratinocytes
-T-cells
-Urothelial cells
-Placenta
-Various human cancer cell lines and tumors |
|
|
Term
what are 4 choline esters? |
|
Definition
acetylcholine
carbachol
bethanecol
methacholine |
|
|
Term
what are the actions of choline esters? |
|
Definition
•Vasodilation- blood vessels have mAChR
•Heart: low doses cause reflex tachycardia; very high doses can cause bradycardia
•Increase tone, motility, secretions from GI tract
•Bronchoconstriction
•Cause miosis
•Increase secretions from exocrine glands |
|
|
Term
what 3 ways does mAChR cause vasodilation? |
|
Definition
causes release of NO which diffuses to SM where it activates cGMP which lowers cell [Ca]
also causes release of endothelial derived hyperpolarizing factor. EDHF is a c type naturietic peptide that activates K+ channels causing hyperpolarization of SM causing relaxation of small diameter vessels
release of H2S causes smooth muscle relaxation |
|
|
Term
what are therapeutic uses of choline esters? |
|
Definition
•GI stimulant (bethanechol)
•Miosis- extraction of cataracts and treatment of glaucoma
•Diagnosis (MeCh):
–Airways hyperreactivity |
|
|
Term
pilocaprine, a muscarinic agonist, can be used for what therapeutic uses? |
|
Definition
•Miosis- treatment of glaucoma
•Increase salivation for treatment of dry mouth |
|
|
Term
what are the benefits of using AchE inhibitors? |
|
Definition
•Ophthalmology- produce miosis to increase drainage of aqueous humor in glaucoma
•GI & Urinary Tracts- increase tone and motility (e.g., in paralytic ileum and acute colonic pseudo-obstruction)
•Treat toxic effects of muscarinic antagonists
|
|
|
Term
Why does Atropine have a biphasic dose-response curve? |
|
Definition
1. CNS- Low doses of atropine may act preferentially in the CNS to increase parasympathetic outflow
2. Presynaptic effect- Low doses of atropine may act preferentially on presynapticmAChR on parasympathetic terminals, resulting in increased ACh release onto the heart |
|
|
Term
how do low doses of Atropine affect the mAchR? |
|
Definition
they block it causing more Ach to be available for the mAchR leading to bradycardia |
|
|
Term
what do high doses of atropine cause? |
|
Definition
Block mAChR on heart-
Block effects of ACh, increases HR |
|
|
Term
what are toxic effects of tertiary mAchR antagonists? |
|
Definition
•Visual problems
•Constipation and urinary retention
•Glaucoma in predisposed individuals
•Hallucinations and delirium
•Decreased sweating and salivation
•(Erectile problems/impaired vaginal lubrication) |
|
|
Term
what are differences between quaternary antagonists vs tertiary |
|
Definition
1. 4° antagonists not lipid soluble (tertiary more soluble):
-don’t act on CNS after systemic administration
-more poorly absorbed from the GI tract
2. 4° antagonists may block nAChR at:
-NMJ, interfere with respiration
-autonomic ganglia, can cause hypotension |
|
|
Term
what is the history between mAchR antagonists and asthma? |
|
Definition
•mAChR antagonists were a main therapy in asthma
–Bronchodilation
–But: decrease secretions and mucous clearance- more prone to infections
•Ipatropium:
–4° ammonium, inhaled
–Bronchodilation without inhibiting ciliary clearance of mucous |
|
|
Term
what are the effects of Ipratropium? |
|
Definition
•Quaternary ammonium mAChR antagonist
•Causes bronchodilation
•Poorly absorbed into general circulation after inhalation (so- little effect on bronchial secretions, heart, CNS, etc.)
•Can be used for asthma and COPD (chronic obstructive pulmonary disease)
•Sometimes combined with ß2-adrenergic agonist |
|
|
Term
what are the main subtypes of mAchR and what are their functions? |
|
Definition
•M1- in sympathetic ganglia (and adrenal medulla), activated by McN-A-343, blocked by pirenzepine
•M2- cardiac mAChR; can contribute to contraction of some smooth muscles; a presynaptic receptor on some nerve terminals
•M3- mediates contraction of smooth muscle, relaxation of vasculature, and secretion from many glands |
|
|
Term
what is the main use of cevimeline? |
|
Definition
•Selective M3 agonist
•Used for treatment of xerostomia and Sjorgren’s syndrome
•Long-lasting sialogogic agent
•May have fewer side effects than pilocarpine |
|
|
Term
what is the main use of Tiotropium? |
|
Definition
•Selective M3 antagonist
–Very slow dissociation from M3mAChR
–4° antagonist
–like ipratropium, is an inhaled bronchodilator
•Used for treatment of COPD |
|
|
Term
what cells store histamine? |
|
Definition
basophils, mast cells, enterochromafin cells |
|
|
Term
what turns histadine into histamine? |
|
Definition
histidinedecarboxylase (HDC) |
|
|
Term
what are the 3 types of GPCRs and what are their functions? |
|
Definition
•Gaq -> phospholipase C -> PIP2 hydrolysis -> IP3/DAG --> PKC/Ca2+
•Gas --> increase adenylylcyclase --> cAMP --> PKA
•Gai -> decrease adenylylcyclase -> decrease cAMP --> decrease in PKA |
|
|
Term
where are H1 receptor subtypes found?
What type of receptor is H1? |
|
Definition
smooth muscle
endothelium
brain
Gq |
|
|
Term
what type of histamine subtype receptor is H2?
Where is it found? |
|
Definition
gastric mucosa, heart, mast cells, brain
Gs |
|
|
Term
where are H3 receptors found, what type are they? |
|
Definition
brain, myenteric plexus
Gi |
|
|
Term
where are H4 receptors found, what type are they?
|
|
Definition
|
|
Term
what is histamines affect on the CV system? |
|
Definition
anaphylaxis
lower blood pressure
H1, H2 both lead to vasodilation
H2 in SA node increases HR and increases spontaneous action potential |
|
|
Term
what is Histamines affect in the nervous system? |
|
Definition
it has all subtypes but H4
•Stimulates sensory nerve endings
•Pain/itching
•Sleep regulation: posterior hypothalamus releases histamine ècortex/medial forebrain bundle èarousal/wakefulness
•Memory consolidation/loss
Drinking, temperature regulation, appetite |
|
|
Term
what is H1's function in the lungs and GI? |
|
Definition
increase bronchoconstriction
increase GI contraction |
|
|
Term
what is H2's effect on gastric secretions? |
|
Definition
increase gastric acid and pepsin |
|
|
Term
what is H3's effect on metabolism? |
|
Definition
as an inhibitor it lowers food intake |
|
|
Term
what is the functional use of Betahistadine? |
|
Definition
is an H1 agonist
H1 stimulation in inner ear -> vasodilation -> vascular permeability -> increases endolymphatic drainage |
|
|
Term
what is betazole used for? |
|
Definition
H2 receptor agonist which is diagnostic for H2-stimulated gastric secretion |
|
|
Term
how are H1 receptor antagonists separated? |
|
Definition
between 1st generation and 2nd gen.
1st generation antagonists cause sedative effects |
|
|
Term
what are the main important properties of H1 1st generation antagonists? |
|
Definition
most are OTC and safe
rapidly absorbed (1-2h) with a wide range of duration (2-24h)
readily penetrate CNS
have mild to marked sedative effects |
|
|
Term
what are 1st generation antihistamine uses? |
|
Definition
• H1 antagonism - anti-allergenic
• anti-cholinergic: muscarinic receptor antagonism
• sedation/drowsiness ®sleep aid
• inhibit Parkinson-like side effects of D2 antagonists
• anti-emetic/nausea: prevent motion sickness |
|
|
Term
what are 1st generation anti histamine side effects? |
|
Definition
hallucinations, convulsions, flushing, urinary retention, tachycardia, dry mouth, coma |
|
|
Term
ethanolamines are a 1st generation antihistamine.
What are 3 commonly used ones? |
|
Definition
•carbinoxamine (Clistin)
•dimenhydrinate (Dramamine)
•diphenhydramine (Benadryl)
high amount of sedation involved |
|
|
Term
what type of drug is piperazine? |
|
Definition
1st gen antihistamine
hydroxyine
used for motion sickness and anxiety
non OTC |
|
|
Term
what type of drug is ethylenediamine? |
|
Definition
a 1st gen anti histamine
active drug is pyrilamine
moderate sedation
MIDOL
|
|
|
Term
what type of drug are alkylamines? |
|
Definition
1st generation antihistamine
•OTC for allergy, common cold, rhinitis
• slight sedation: best for daytime use
•potentiate analgesia |
|
|
Term
under what category is brompheniramine? |
|
Definition
|
|
Term
what type of drugs are phenothiazine? |
|
Definition
PROMETHAZINE
1st gen anti histamine
•phenothiazine derivative
• strong sedation
• non-OTC
• pre-operative anesthetic: Na+ channel blocker
•α-adrenergic blockade: orthostatic hypotension
• used for severe motion sickness |
|
|
Term
|
Definition
• 1st generation piperidine
• anti-serotonin effects
• used for adolescent migraine
• used for Cushing’s syndrome
• lead compound for 2nd generation H1-antagonists |
|
|
Term
what are the functional uses for 2nd generation anti histamines? |
|
Definition
•drugs of choice for allergy
• increased selectivity for H1-receptor
• non-sedating
•zwitterionic at pH 7.4
• poorly cross blood brain barrier |
|
|
Term
what are the uses of H3 antagonists? |
|
Definition
• stimulant, nootropic effects
• on trial to treat neurodegeneration (AD), ADHD, schizophrenia, sleep disorders (i.e. narcolepsy), obesity
clobenpropit, ABT-239, ciproxifan, A-349,821 |
|
|
Term
what are drug interactions that can occur with H1 antagonists from 2nd gen drugs? |
|
Definition
• 2nd generation anti-histamines: lethal ventricular arrhythmias
•terfenadine (Seldane®), astemizole (Hismanal®)
• DDI’s with ketoconazole, itraconazole, macrolide antibiotics
•bergamottin®grapefruit juice
• CYP3A4 |
|
|
Term
what is direct regulation of gastric acid secretion? |
|
Definition
1. vagus nerve increases Ach to M3 receptor activating Gq increasing calcium which increases H+ pump
2. In the antrum, g cells are stimulated to release gastrin which binds to CCK2 receptors and CCKB receptors which stimulate Gs causing an increasein Ca and a release of H+
3. ECL cells release histamine onto H2 receptors of parietal cells causing them to stimulate Gs receptors which increase cAMP and increase H/K pump activity |
|
|
Term
what are indirect ways (non parietal cells) of gastric acid secretion? |
|
Definition
vagus nerves increase antrum Ach which increase GRP
This leads to increase in gastrin and a decrease in somatostatin which indirectly causes an increase in gastrin
increased levels of gastrin and Ach lead ECL cells to increase histamine release |
|
|
Term
what is negative feedback for GA secretion? |
|
Definition
increased acid in the antrum leads to activation of D cells to release somatostatin which binds to the ST2 receptor (Gi) on g cells lowering cAMP levels & gastrin release
The ST2 receptor is also on ECL cells causing a decreased release of histamine
these actions lower the acid in the fundus |
|
|
Term
what are examples of acid peptic disorders? |
|
Definition
1.GastroesophagealReflux Disease (GERD)
2.Peptic/Gastric Ulcer
3.Stress-related mucosal injury |
|
|
Term
where are the targets of gastrin? |
|
Definition
CCK-B receptors on ECL cells and parietal cells |
|
|
Term
what are the symptoms of GERD? |
|
Definition
heart burn
acid regurgitation
sore throat
causes a lower anti-reflux barrier and esophageal sphincter function |
|
|
Term
what is the physiology of a peptic ulcer? |
|
Definition
•damage to gastroduodenal mucosa, erosion of lining (gastritis)
• aggressive factors > defensive factors |
|
|
Term
what are causes of peptic ulcers? |
|
Definition
h. pylori, alcohol abuse, NSAIDs, radiation, smoking, Zollinger-Ellison syndrome, stress (?) |
|
|
Term
what are treatment goals for a peptic ulcer? |
|
Definition
• neutralize stomach pH
• lower GA secretion
• lower H. pylori infection
• provide mucosal protection
• promote mucosal healing
• lifestyle changes |
|
|
Term
what are the two types of antacid formulations? |
|
Definition
Carbonates: NaHCO3, CaCO3
• Alka-Seltzer®, Maalox®, Pepto-Bismol®, Rolaids®, Tums®
•CO2 in stomach ®belching/gas
• Toxicity: metabolic alkalosis, kidney stones, milk-alkali syndrome
Aluminum/Magnesium Hydroxides:
Al(OH)3, Mg(OH)2
• Al(OH)3 = Gaviscon®, Equate®, Mylanta®
• Mg(OH)2 = Milk of Magnesia®, Rolaids®, Maalox®
• increased gas/belching, alkalosis
• AlCl3 = increased constipation
• MgCl2 = increased diarrhea |
|
|
Term
H2 receptor antagonist work as modifaction structures for histamine. What are two well known ones? |
|
Definition
•Ranitidine (Zantac®)
•Famotidine (Pepcid®) |
|
|
Term
what is the clinical use for H2 antagonists? |
|
Definition
• highly selective: no effects at H1, H3, H4
• inhibit 60-70% of GA secretion in 24 hr period
• heartburn, GERD, peptic/gastric ulcer, stress-related bleeding |
|
|
Term
what are the pharmacokinetics for H2 antagonists? |
|
Definition
• serum t 1/2 = 1-4 hours
• 1st pass metabolism in liver; 50% bioavailability
• minimal side effects (<3% patients)
• diarrhea, headache, fatigue, myalgias,
constipation |
|
|
Term
what are the current drugs of choice for GA disorders above H2 receptor antagonists? |
|
Definition
proton pump inhibitors
irreversible inhibition of action at H/K pump |
|
|
Term
what are two main proton pump inhibitors? |
|
Definition
omeprazole (prilosec)
pantoprazole (protium) |
|
|
Term
what is the clinical use for PPIs? |
|
Definition
•inhibit 90-98% of 24 hour acid secretion
• heartburn, GERD, peptic/gastric ulcer (NSAID, H. Pylori induced), stress-related bleeding, gastrinoma
• rapid symptom relief, ulcer healing
• GERD, peptic ulcers: effective in 80-90% of patients |
|
|
Term
what are the pharmacokinetics of PPIs? |
|
Definition
serum t 1/2 = 0.5 - 2 hours•minimal side effects (<5% patients)
• diarrhea, headache, abdominal pain
•¯vitamin B12, Ca2+, Iron, Zinc absorption
|
|
|
Term
what are mucosal protective agents used for? |
|
Definition
•potentiate endogenous mucosal repair/defence mechanisms |
|
|
Term
what are the 3 main mucosal rotective agents? |
|
Definition
bismuth
sucralfate
prostaglandin analogs
|
|
|
Term
what's the deal with sucralfate? |
|
Definition
•sucrose sulfate-aluminum salt complex
• reacts with HCl in stomach
• forms viscous paste acid buffer
• forms complexes with albumin/fibrinogen ® protective barrier
• Dose: 1 g/qd
• Uses: gastric/duodenal ulcers, stress-related GI bleeding
• Side effects: constipation, flatulence |
|
|
Term
misoprotol is a prostaglandin analog. How does it work? |
|
Definition
• increase mucus/bicarbonate secretion
•increase mucosal blood flow
• MOA: activate PG-E3/E4 receptors on parietal cells= Gai --> decrease levels of cAMP --> counter histamine effects
• Use: NSAID-induced ulcers; gastric/peptic ulcer patient using NSAIDs
Adverse Effects: diarrhea, abdominal pain |
|
|
Term
bismuth compounds such as peptobismol are used when? |
|
Definition
• Oral, liquid/gel caps; antacid
•¯inflammation/irritation of GI lining; protective coat
• Use: H.pylori eradication cocktail (Bismuth, PPI, tetracycline, metronidazole); dyspepsia, acute diarrhea
• Adverse Effects: black tongue/stools; Reye’s syndrome |
|
|
Term
what are prokinetic agents and what are they used for? |
|
Definition
•Uses: GERD, irritable bowel syndrome, gastritis, dyspepsia
• Metoclopramide (Maxolon®)
• 5-HT4 receptor agonist/D2 receptor antagonist
• increases intestinal motility, releases pyloric sphincter
• Toxicity: extrapyramidal effects |
|
|
Term
what is the function of lipoprotien lipase? |
|
Definition
hydrolysis of lipids, chylomicrons - VLDL, into capillaries
upon hydrolysis, TGs are removed |
|
|
Term
what is the fate of VLDLs upon hydrolysis? |
|
Definition
TGs are removed and become IDLs. IDLs can either go to the liver or give up more TGs and become LDLs.
LDLs are high in cholesterol, low in TGs |
|
|
Term
what are good levels for total cholesterol, LDL, HDL and TG's? |
|
Definition
cholesterol = <200
LDL <130
TG < 120
HDL = > 40 for men > 50 for woemen |
|
|
Term
|
Definition
abnormal amounts of lipids/cholesterol in the blood
these could be hyper or hypo |
|
|
Term
what are the primary reasons for a hypolipidemia? |
|
Definition
Primary causes :
•abetalipoproteinemia
•hypobetalipoproteinemia
•Marfan syndrome
• Smith-Lemli-Opitz syndrome
• Tangier disease
low growth and development |
|
|
Term
what's a diagnosis and symptoms of hyperlipidemia? |
|
Definition
•diagnosis: triglycerides (LDL + VLDL) >150-200 mg/dL
symptoms: risk CAD/CHF, acute pancreatitis (TG > 900 mg/dL), eruptive xanthomas, obesity |
|
|
Term
what leads to hypercholesterolemia? |
|
Definition
high LDL
• familial hypercholesteromia
•ligand-defective Apolipoprotein-B
deficient HDL |
|
|
Term
what causes a hyperTRIglyerademia? |
|
Definition
high chylomicrons, VLDL
•chylomicronemia
•hypertriglyceridemia
• severe
• moderate |
|
|
Term
what are treatment options for hyperlipidemia? |
|
Definition
•Diet/Lifestyle changes
•HMG-coAreductase inhibitors (Statins)
•Niacin (nicotinic acid)
•Fibric Acid Derivatives (Fibrates)
•Bile-Acid Binding Resins
•Intestinal Sterol absorption inibitors |
|
|
Term
what are the "statin" drugs? |
|
Definition
HMG CoA reductase inhibitors
Structural analogs of HMG-CoA
|
|
|
Term
|
Definition
• lower LDL plasma levels >60%; less effective at lower TG, raise HDL
most effective for patients with cardiovascular disease (CAD, CHF)
used prophylatically for patients with lipidemias/risk factors to prevent atherosclerosis
may lower risk of heart attack in patients with elevated C-reactive protein levels
starting dose range: 10-40 mg SID; monitor lipid levels every 1-3 months
most effective at night; use in late evening |
|
|
Term
what are two main statins used? |
|
Definition
atorvastatin, simvastatin |
|
|
Term
statins work by inhibiting HMG CoA reductase. What is the result? |
|
Definition
liver produces less cholesterol
hepatocytes increase LDL receptor synthesis
more receptors means more LDL/VLDL bound and turned into bile salts |
|
|
Term
what are adverse effects of the statins? |
|
Definition
• serum liver enzyme concentration (aminotransferase); exacerbated with history of alcohol abuse
• cramps, myalgias, GI
• malaise
• anorexia |
|
|
Term
What is the mechanism of action for Niacin? |
|
Definition
• directly and non-competitively inhibits diacylglycerolacyl-transferase 2 in hepatocytes
|
|
|
Term
what results does Niacin produce? |
|
Definition
lower TG synthesis and increase ApoB degredation
decrease VLDL secretion
lower HDL breakdown |
|
|
Term
what are clofibrate and Fenofibrate? |
|
Definition
Fibrates (fibric acid derivatives) |
|
|
Term
|
Definition
• nuclear receptor/transcription factor
•heterodimerize with retinoic acid X receptor (RXR)
stimulates increased production of proteins involved in Free fatty acid breakdown
less LDL and VLDL and more HDL |
|
|
Term
what are the uses of fibrates? |
|
Definition
effective for hypertriglyceridemia and dysbetalipoproteinemia
use in combination with statin for maximal efficacy |
|
|
Term
what are side effects of fibrates? |
|
Definition
•myopathy
• arrhythmias
• rashes
•hypokalemia
• gastric irritation/nausea
• cholesterol gall stones |
|
|
Term
|
Definition
|
|
Term
how do bile acid binding resins work? |
|
Definition
•disrupt enterohepatic bile acid circulation
• bind bile in small intestine
prevent bile reabsorbtion and lower plasma cholesterol |
|
|
Term
what are nasty side effects of bile acid binding resins? |
|
Definition
•GI effects
• constipation
• diarrhea
• flatulence
• bloating
• bad taste |
|
|
Term
what is the use for a bile acid binding resin? |
|
Definition
effective for primary hypercholesterolemia, pruritus
use in combination with statin for maximal efficacy
prevents absorbtion of a lot of drugs |
|
|
Term
why is ezetimibe a last resort drug option with statins? |
|
Definition
it can increase carotid arteriol thickness and CV events |
|
|
Term
what is the MoA with intestinal sterol absorption inhibitors? |
|
Definition
• inhibits cholesterol absorption in brush border of small intestine
• inhibits cholesterol transporter NPC1L1 in GI tract and hepatocytes
•LDL receptors on hepatocytes
•LDL uptake into cells
• used with statins to efficacy |
|
|
Term
what are the side effects of intestinal sterol absorption inhibitors? |
|
Definition
•GI effects/diarrhea
• headache
•myalgia |
|
|
Term
what is a quick way to find NNT? |
|
Definition
inverse of Absolute Risk reduction |
|
|
Term
what is the experimental event rate? |
|
Definition
the proportion of patients in the experimental group demonstrating the defined event |
|
|
Term
what is the control event rate? |
|
Definition
proportion of patients in the control group experiencing the defined event |
|
|
Term
how do you calculate the RRR? |
|
Definition
|
|
Term
how do you calculate the ARR? |
|
Definition
|
|
Term
What are the 4 types of catecholamines? |
|
Definition
isoproterenol
NE
epi
dopamine |
|
|
Term
what is released more by the adrenals, epi or NE? |
|
Definition
|
|
Term
what is the rate limiting step for the conversion of Epi to NE?
What is the enzyme that does this? |
|
Definition
the conversion of tyrosine to DOPA
tyrosine hydroxylase |
|
|
Term
what happens to most NE upon release? |
|
Definition
80-90% it is taken back into the cell |
|
|
Term
how does alpha-methyl tyrosine disrupt the production of NE? |
|
Definition
it inhibits tyrosine hydroxylase |
|
|
Term
what does cocain or tricyclic anti depressants do? |
|
Definition
inhibit the reuptake of NE into the cell |
|
|
Term
what is the function of reserptine? |
|
Definition
inhibit the uptake of DA and NE into the synaptic vessicle |
|
|
Term
what is Bretylium's function? |
|
Definition
Not only does it block the release of NE into the synapse but it also acts like cocaine |
|
|
Term
how does guanethidine work? |
|
Definition
displaces NE from the vessicle so it cannot be released (like bretylium) also acts like cocaine (like bertylium!) |
|
|
Term
how do amphetamines and tyramines work? |
|
Definition
induce the release of NE into the synapse |
|
|
Term
what are alpha 1 receptors responsible for doing? |
|
Definition
contraction of smooth muscle |
|
|
Term
what are beta 1 receptors for? |
|
Definition
heart and juxtaglomerular cells |
|
|
Term
what are alpha 2 receptors for? |
|
Definition
presynaptic receptors that decrease NE release |
|
|
Term
what are Beta 2 receptors for? |
|
Definition
relaxation of smooth muscles
(some B2 are in heart) |
|
|
Term
what are the orders of potency for alpha receptors between Epi, NE, and Iso?
for beta receptors? |
|
Definition
A: EPI > NE > ISO
B: ISO > EPI > NE |
|
|
Term
what is the function of the as family? |
|
Definition
mediates stimulation of adenylylcyclase
(ß-AdR) |
|
|
Term
How does the ai receptor work? |
|
Definition
mediates inhibition of adenylylcyclase; activates
potassium channels in heart
(M2, M4mAChR; a2-AdR) |
|
|
Term
how does the aq family work? |
|
Definition
activate certain forms of phospholipase C
(M1, M3, M5mAChR; a1-AdR) |
|
|
Term
what is the function of a g protein coupled receptor kinase? |
|
Definition
regulate the activity of GPCRs by phosphorylating their intracellular domains. They can up or down regulate the activity.
Upon phosphorylation of serine or threonine residues binding sites are made for arrestins which prevent the reassociation of the G proteins with their receptors, thereby preventing reactivation of the signaling pathway |
|
|
Term
if you treat a heart with chronic ISO, what happens? |
|
Definition
the beta AdR # goes down and you cannot reach the same responsiveness |
|
|
Term
how can you increase Cardiac AdR #? |
|
Definition
|
|
Term
what type of g protein do Beta 1 receptors couple to? |
|
Definition
|
|
Term
what type of g protein do Beta 2 receptors couple to?
|
|
Definition
|
|
Term
what happens to different receptor Beta receptor levels in the heart during heart failure? |
|
Definition
Beta 1 levels go down and Gi go up
•Failing heart has increased expression and activity of GRK, which increases ß1 desensitization and degradation and also increases coupling of ß2 to Gi
•Therefore, ß2-AdR has less stimulatory and more inhibitory effects in a failing heart than in a non-failing heart
•The decreased level of ß1-AdR and increased ß2-AdR coupling to Gi both contribute to decreased ß-adrenergic stimulation of contractility in failing heart |
|
|
Term
what are the receptors where epi is more potent? |
|
Definition
|
|
Term
because epi has two effects on different receptors, what is its effect on the circulation? |
|
Definition
B2 receptors at low volume cause vasodilation and lower TPR
A1 cause vasoconstriction and increase TPR |
|
|
Term
what are the 4 main effects of Epi? |
|
Definition
Causes relaxation of intestinal and bronchial smooth muscle
Cardiac stimulation
Treatment of anaphylatic shock
Slows absorption of local anesthetics |
|
|
Term
what is the effect of NE on the heart and vascular system? |
|
Definition
in the heart it causes are "stronger" heart beat by increasing contractility, but lowers heart rate
in the vasculature it stimulates alpha1 receptors and causes vasodilation which increases TPR |
|
|
Term
what are the effects of Iso on the heart and vascular system? |
|
Definition
it increases heart rate and cardiac output but also has a strong interaction with Beta 2 receptors causing vasodilation. This leads to a decrease in TPR = diastolic pressure |
|
|
Term
what is the order of affinity of different receptors for Dopamine? |
|
Definition
|
|
Term
what does dopamine activate? |
|
Definition
(1) vasodilatory dopamine (D1) receptors in renal, mesenteric, and coronary vascular beds
(2) beta receptors in heart (greater effect on
contractile force than rate)
(3) stimulates NE release from nerve
terminals (contributes to cardiac effects)
(4) high doses can activate vascular a1 receptors |
|
|
Term
what are the effects of phenylepherine on the CV? |
|
Definition
|
|
Term
what is the function of albuterol, salmeterol? |
|
Definition
it is a B2 agonist = vasodilation |
|
|
Term
what has a better strength of bronchodilation between salmeterol and albuterol? |
|
Definition
salmeterol, it has a more longlasting effect |
|
|
Term
what is Dobutamine? what are it's effects/uses? |
|
Definition
One isomer is ß1 agonist and alpha1 agonist
Other isomer is ß1 agonist (and apparently weak a1 antagonist) Increases contractile force, little effect on heart rate or TPR
Used to increase cardiac output (e.g., CHF)
Why does dobutamine have little effect on HR and TPR?
1. Human atria: 40- 50% ß1; human ventricle: 70-85%ß1
2. Little ß2- mediated vasodilation, so no reflex tachycardia
3. alpha1 agonist activity may also contribute to direct stimulation of ventricles and lack of vasodilation |
|
|
Term
|
Definition
D1 agonist
IV used in rapid vasodilation for emergency hypertension |
|
|
Term
|
Definition
it is a sympathomimetic amine whose use |
|
|
Term
what type of drug is phenoxybenzamine? |
|
Definition
a COVALENT non-selective alpha adrenergic antagonist |
|
|
Term
as an alpha adrenergic antagonist what are the uses of phenoxybenzamine? |
|
Definition
–Postural hypotension
–Reflex tachycardia
–Miosis
–Impaired ejaculation
–Can act on the CNS (nausea and sedation) |
|
|
Term
what type of drug is phentolamine? |
|
Definition
a NON-COVALENT alpha adrenergic antagonist |
|
|
Term
what type of drug is prazosin? |
|
Definition
this drug is selective for A1 adrenergic antagonism |
|
|
Term
what are the uses of prazosin? |
|
Definition
-Blocks a1-AdR in vasculature- cause vasodilation
-Doesn’t block a2-AdR on sympathetic nerve terminals-
therefore- less NE released on heart than if a2-AdR were blocked
therefore- less tachycardia than if a2-AdR were blocked
-Used in the treatment of hypertension
-Also used in the treatment of BPH (benign prostatic hyperplasia) |
|
|
Term
what is yohimbime and it's uses? |
|
Definition
it is selective for A2 adrenergic antagonism thus it increases the release of NE |
|
|
Term
what type of drug is propranalol? |
|
Definition
a NON-SELECTIVE beta adrenergic antagonist
|
|
|
Term
what would a beta blocker do to benefit cardiovascular problems? |
|
Definition
–Angina Pectoris
–Arrhythmias
–Hypertension
–Recurrence of heart attack |
|
|
Term
what are major adverse effects of beta blockers? |
|
Definition
heart failure
bronchospasm
bradycardia
heart block
hypotension
claudication
fatigue
constipation
diarrhea |
|
|
Term
why would you not want to quit propranalol right away? |
|
Definition
propranolol leads to the increase of beta receptors on the heart thus stoping would leave the heart susceptible to massive stimulation |
|
|
Term
|
Definition
it seems that it's sole purpose is to see if it blocks beta 2 adrenergic receptors |
|
|
Term
what are atenolol, metoprolol and esmerol used for? |
|
Definition
specific for Beta 1 adrenergic antagonists
REDUCE HEART RATE |
|
|
Term
what are labetelol & Carvedilol special for? |
|
Definition
both are non selective beta blockers as well as alpha 1 antagonists |
|
|
Term
other than blocking dopamine and NE transfer into vessicles, what else does Reserpine do? |
|
Definition
Blocks reuptake into vesicle of previously released NE
Blocks reuptake of NE that leaks out of vesicle |
|
|
Term
what is tyramines function? |
|
Definition
it acts as a catecholamine releaser |
|
|
Term
what effects does Reserpine have on the body? |
|
Definition
•Decreases blood pressure and heart rate
•Increases GI tone and motility
•Causes:
–Postural hypotension
–Diarrhea
–Sexual dysfunction
–CNS effects: sedation, depression
•Can be used for treatment of hypertension |
|
|
Term
what are the effects of Guanethidine? |
|
Definition
Causes increases Beta adrenergic receptors on the heart
Decrease blood pressure (used in the past for hypertension; guanadrel still used occasionally)
-Postural hypotension
-Diarrhea
-Sexual dysfunction |
|
|
Term
what are two drugs that decrease sympathetic activity by acting on the CNS? |
|
Definition
CLONIDINE
alpha-methyldopa |
|
|
Term
on what receptors does clondine target? |
|
Definition
•Selective a2 agonist
Acts in the CNS to decrease sympathetic outflow |
|
|
Term
how does alpha-methyldop decrease sympathetic activity? |
|
Definition
a-methyldopa converted in nerve terminal to a-methylnorepinephrine
a-methylnorepinephrine is stored in vesicles and released with nerve stimulation
a-methylnorepinephrine is an a2-adrenergic agonist:
acts in CNS to decrease sympathetic outflow
|
|
|
Term
what is the use of alpha-methylTYROSINE? |
|
Definition
Inhibits tyrosine hydroxylase activity- decreases
catecholamine synthesis
Used occasionally for treatment of
pheochromocytoma |
|
|
Term
What is the function of MAO? |
|
Definition
degrades catacholamines in the cell |
|
|
Term
what are the effects of a MAOI? |
|
Definition
•Cause increased levels of catecholamines in both CNS and periphery
•Introduced for the treatment of depression
•Can cause hypotension |
|
|
Term
What is MAOI affect on tyramine? |
|
Definition
turns it to octopamine
this causes less NE to be released!
if one eats foods high in tyramine, it exacerbates the effect
|
|
|
Term
how can a MAOI cause HYPOtension? |
|
Definition
•Cause hypotension- dietary tyramine converted to octopamine in nerve terminal
Octopamine acts as false transmitter |
|
|
Term
what happens in a baroreflux failure?
what drug can alleviate this problem? |
|
Definition
volatile HR and BP
CLONIDINE |
|
|
Term
what is the function of 6-hydroxydopamine? |
|
Definition
•Taken up by adrenergic nerves
•Oxidized to toxic compounds
•Causes “chemical sympathectomy”:
–Causes destruction of nerve terminals
–In newborns, also destroys cell bodies |
|
|