• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/492

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

492 Cards in this Set

  • Front
  • Back
What is the difference between the words affinity and efficacy with relation to receptors/ligands? What is a partial agonist?
Affinity is the tendency of a drug to bind to receptors

Efficacy is the tendency for the ligand, once bound, to activate the receptor

Partial agonist is one with intermediate levels of efficacy (therefore submaximal tissue response)
Which receptors show an appreciable level of activation even when no ligand is present?
Cannabinoids
Benzodiazepines
Serotonin

*Others exist as well*
What is the efficacy of each of the following?
1) Inverse agonist
2) Agonist
3) Competitive antagonist
1) Negative
2) Positive
3) Zero
What is an inverse agonist?
One that reduces the level of constitutive activation. ( If drug has a higher affinity for the nonactive state)
How does a competitive antagonist work in terms of efficacy?
When there is no preference between the active and nonactive form of the receptor
What state do most receptors exist in?
Inactive. Very few remain in active state.
Spare receptors are commonly involved with which types of drugs?
Ones that elicits smooth muscle contraction.
What is chemical antagonism?
It occurs when two substances combine in solution and as a result the effect of the active drug is lost. ( Ex. the use of chelating agents that bind to heavy metals)
What is pharmacokinetic antagonism?
Occurs when the antagonist effectively reduces the concentration of the active drug at its site of action through affecting its absorption, metabolism, excretion etc.
What are the classifications of antagonism by receptor block ?
- Reversible (equilibrium) or irreversible (non-equilibrium)
- Reversible is the most commonest and most important type of antagonism
What are the two main characteristics of competitive antagonism?
1- in the presence of the antagonist, agonist log [ ] - effect curve is shifted to the right with no change in slope or max. (Hallmark!)

2- Dose ratio increases linearly with antagonist concentration (slope of line is affinity)
What is important about competitive antagonism that differs from other forms of antagonism?
This type of antagonism is said to be surmountable. This means that because the two are in competition, raising the agonist concentration can restore the agonist occupancy
What is a dose ratio?
It expresses the shift in a log concentration curve and is defined by the ratio of agonist concentration which has to be increased in presence to an antagonist, to restore response
What is characteristic of non-competitive antagonism?
Reduction in slope and max of agonist log [ ] curve. Generally , it is the situation where the antagonist blocks at some point the chain of events leading to response
What is physiological antagonism?
A term used loosely to describe the interaction of two drugs whose opposing actions in the body tend to cancel each other
What is desensitisation or tachyphylaxis? Tolerance? Drug resistance
The effect of a drug gradually diminishes when it is given continuously. (develops in a few minutes)

Tolerance is used to describe a more gradual decrease in responsiveness to a drug (days to weeks to develop)

Drug resistance is used to describe the loss of effectiveness of antimicrobial or antitumor drugs.
What occurs during prolonged exposure to agonists?
Gradual decrease in number of receptors expressed ( internalisation of receptors)
What does the pronounced tolerance to nitrovasodilators result from?
Decreased metabolism

(as opposed to ethanol and barbiturates to which tolerance develops to due to increased metabolic degradation)
What is physiological adaptation?
When a drug's effects are reduced because it is nullified by a homeostatic response
Which law does binding of drugs obey?
Law of Mass Action
Which drugs bind to accessory sites on ion channel proteins, thus affecting channel gating?
Dihydropyridine (vasodilator - inhibits opening of L-type Ca Ch.)

Benzodiazepines (bind to region of GABA receptor-chloride channel complex)

Sulfonylureas ( act on ATP-sensitivty potassium channels of pancreatic B-cells - used in treatment of diabetes (^ insulin production)
List the types of receptors in order of fastest reaction time to slowest
Type 1:Ligand-gated[ionotropic] (ms)
Type 2: G-protein-coupled [metabotropic] (s)
Type 3: Kinase-linked (hrs)
Type 4: Nuclear (hrs)
Which receptors constitute the largest family?
G-protein
What are the families of G-protein receptors?
Family A (rhodopsin fam): Largest. Receptors for most things. Short N terminal tail (extracell.)

Family B (secretin/glucagon fam): Receptors for peptide hormones (including secretin, glucagon, calcitonin). Intermediate extracell. tail

Family C. Small. Includes: GABAb receptors, Ca-sensing receptors , metabotropic glu receptors. Long extracellular tail.
What are some examples alternative mechanims of receptor activation?
1) Rhodopsin being activated by light inducing cis-trans isomerisation of retinal

2) PARs, when activated, bind to receptor domains and act as tethered agonists.

3) Receptor mutations
What are the G-protein subtypes and their main effects
G(alpha)s - Stimulates adenylyl cyclase (^cAMP)

G(alpha)i - Inhibits adenylyl cyclase (Dec. cAMP)

G(alpha)o - Limited effects of a subunit

G(alpha)q - Activates PLC

G(beta)(gamma) - Same effects as alpha subunits but also other effects.
What are characteristics of G protein receptors?
- 7 membrane-spanning a-helices
- One intracellular loop is larger than the others which interacts with G proteins
- G-proteins are made up of a,b,y subunits and a subunit has the GTPase activity
How are the effects of G proteins turned on and off?
Turned on:
- GDP-GTP exchange causing dissociation of G-protein trimer, releasing a and by subunits (active forms)

Turned off:
- GTP is hydrolyzed to GDP through GTPase activity of alpha subunit. a-GDP molecule then dissociates from effector and reunites with By subunit.
What is cAMP inactivated by? What are some drugs that inhibit the action of these inhibitors?
Phosphodiesterases. They cause hydrolysis to 5'-AMP.

PDE inhibitors:
Caffeine (general)
Rolipram [asthma] (PDE4 selective)
Sildenafil (Viagra) (PDE5 selective)
Describe breifly the phosphatidylinositol cycle
GPCR activation leads to activation of PLCb which cleaves PIP2 into DAG and IP3. DAG activates protein kinase C and IP3 causes the release of intracellular Ca. DAG is then phosphorylated into phosphatidic acid which combines with inositol (from IP3 recycle) to form PI which is then again phosph. to PIP2. (cycle complete) . IP3 can be converted into IP4 in cells or it can be dephosphorylated into IP by phosphatases and then it is convereted into inositol by Ionsitol 1-phosphatase (this inositol combines with phosphatidic acid)
Which plant compounds can cause PKC activation?
Phorbol esters
On what basis does Lithium work?
It inhibits inositol 1-phosphatase which prevents IP3 recycling.
What is the Rho/Rho kinase system?
It is a pathway which is also activated by GPCRs among others. The free G-alpha unit interacts with guanosine nucleotide exchange factor which forms Rho-GTP (active) from Rho-GDP. This activated Rho kinase which is involved in numerous things. *May be important in pathogenesis of pulmonary hypertension
How does GPCR desensitisation occur?
By:

Receptor phosphorylation
Receptor internalization
What are the types of kinase-linked receptors?
- Receptor tyrosine kinase ( receptors for growth factors, and insulin receptor belongs to this class)
- Serine/Thr kinase (ex. TGF receptor)
- Cytokine (lack intrinsic enzymatic activity so, when activated, they activate a cytosolic tyrosine kinase)
- Guanylyl cyclase-linked receptors - example ANF receptor
How can supersensitivity occur ?
1 - Nerve denervation
2 - Pharmacological block

The end organs become supersensitive to small amounts of transmitter
What are heterotropic interactions?
When one neurotransmitter affects the release of another for example. Homotropic would be when the transmitter binds to the presynaptic autoreceptors and affects the nerve.
How is ACh removed from cholinergic synapses ?
Acetylcholinesterase. Most other cases of synapses, there is active reuptake into the presynaptic nerve or into supporting cells
Which aspect of synthesis/storage/release of amine/aa transmitters cannot be influenced by drugs?
Transmitter diffusion
What are the two actions of acetylcholine?
Muscarinic and Nicotinic.

Muscarinic actions can be reporduced by injection of muscarine and can be abolished by small doses of atropine. These actions include: vasodilation (via NO) and other parasympathetic actions.

Nictonic receptors provide effects similar to those of nicotine. These include: stimulation of all autonomic ganglia, stimulation of voluntary muscle, secretion of adrenaline from the adrenal medulla.
What 3 classes of nictonic ACh receptors are there? General structure?
Muscle
Ganglionic
CNS

Pentameric structure functioning as a ligand-gated ion channels. There are two binding sites for ACh (both need to be bound for activation) located at the extracellular domain of each alpha subunit.
What are muscarinic receptors? Subtypes?
G-protein-coupled receptors. Subtypes: M1-M5.

Odd numbers: 1,3,5 couple with Gq -> inositol phosphate

Even #: 2,4 couple with Gi to inhibit adenylyl cyclase

4 and 5 are mainly in the CNS
Where can M1 receptors be found? M2?
M1 (neural) = Mainly on CNS and peripheral neurons but also on gastric parietal cells. They mediate excitatory effects (mainly by decreasing K conductance)

M2 (cardiac) = occur in the heart and presynaptic terminals of peripheral and central neurons. They exert inhibitory effects (mainly by increasing K conductance and inhibiting Ca channels)

M3 (smooth m.) are found in glandular/smooth muscle and produces mainly excitatory effects (i.e secretions and contraction of visceral smooth muscle). M3 can also cause relaxation of smooth muscle (mainly vascular) which results from release of NO.
Which drug inhibits the ACh carrier (carrier which puts ACh into vesicles)
Vesamicol
What is tubocurarine
A drug that blocks postsynaptic ACh receptors. It is a non-depolarising blocking agent.
What are the types of postsynaptic potentials? Include time for transmission for each
Slow IPSP (I is for inhibitory) are aka hyperpolarising and last 2-5s. M2 receptors play main role buth other transmitters do too.

Slow EPSP last for 10 seconds and is produced by ACh acting on M1 receptors.

Late slow EPSP lasts for 1-2 minutes. Also M1 receptors.
What is depolarization block?
Occur when excitatory nAChRs are persistently activated because of a decrease in the electrical excitability of the postsynatpic cell
Application of nicotine to a sympathathetic ganglion has what effect?
Depolarizes the cell which at first causes an action potential but after a few minutes the transmission is blocked.
What does Hemicholinium do?
Blocks the choline carrier
Neostigmine is a drug which?
Blocks AChEsterase
Which drugs are cholinergic agonists?
Bethanechol *
Carbachol
Pilocarpine *
Methacholine
Nictoine
ACh

* Ones used clinically. They act on both mAChR and nAChR but on the previous one more potently.
Which receptor is responsible for the parasympathetic reflex necessary to accommodate the eye for near vision?
mAChR -> contraction of ciliary body-> relaxing tension on
What is the main use of muscarinic agonists clinically?
Treatment of glaucoma in the form of eye drops. Pilocarpine is the most effective to do this.

(Remember sympathetic innervation of constrictor pupillae leads to dilation which causes blockage of Schlemm canal)
Although used rarely, Bethanechol is also used as a drug. Do you know what for?
Assist bladder emptying or to stimulate GI motility. It acts on M3 receptors
What are the pharmacological properties of atropine? Clinical uses? Side effects?
Non-selective muscarinic antagonist. Well absorbed orally.

Adjunct for anesthesia, bradycardia, Anticholinesterase poisoning

Side Fx: urinary retention, dry mouth, blurred vision
Which drug has similar actions and uses to atropine?
Scopolamine. It is used for the same clinical uses as atropine + treatment for motion sickness
Apart from atropine and scopolamine , what are some other muscarinic antagonists?
1) Atropine methonitrate (lacks CNS effects of atropine) used for GI hypermotility

2) Tropicamide. Opthalmic use to produce mydriasis and cycloplegia

3) Pirenzepine - Selective for M1 - treatment of peptic ulcer

4) Darifenacin - Selective for M3 - treatmnt of urinary incontinence
What effect does atropine have on the CNS? Poisoning?
Mainly excitatory. Atropine poisoning causes excitement and irritability with a rise in body temperature and loss of sweating. This all results from blocking the mAChR in the brain.
Which drugs would one use to treat atropine poisoning?
Anticholinesterases such as physostigmine
Which drugs affect the autonomic ganglia preferntially ?
1) Nicotine
2) Lobeline
3) Dimethylphenypiperazinium (DMPP)

Only Nicotine is used clinically to help people stop smoking.
What was the first antihypertensive treatment?
Hexamethonium. It is no longer in use.
What is the only ganglion-blocking drug currently in clinical use?
Trimetaphan
What is a ganglion-blocking drug? How do they work?
A drug that inhibits post synaptic transmission in ganglia by binding the nicotinic receptors of both para and sympathetic ganglia.

Any one/combo:
- Interference with ACh release (botulinum toxin, hemicholinium)
- Prolonged depolarization (Nicotine and ACh(if cholinesterase is inhibited))
- Interference with postsynaptic action of ACh (blocking neuronal nAChRs)
Which drugs undergo zero-order elimination?
PEA

Phenytoin
Ethanol
Aspirin
What is acetylcholine used for ?
To achieve miosis during opthalmic surgery.
What are the side effects of all cholinergic agonists (incl. acetylcholine) ?
DUMBBELSS

Diarrhea, Urination, Miosis, Bronchospasm, Bradycardia, Excitation of skeletal muscle and CNS, Lacrimation, Sweating, Salivation
Neostigmine

Does it enter the CNS?
Indications
Adverse effects
No it does not.

Myasthenia gravis, urinary retention, postoperative/neurogenic ileus, antidote for non-depolarising neuromuscular junction blockade.

Same as excessive chol. stim.
What does Carbachol do?
Rarely used but it is used for glaucoma and to stimulate miosis during surgery.
What are the effects of pilocarpine? Which receptors does it work on? Adverse effects?
Causes miosis and contraction of ciliary muscle + other regular parasympathetic activities. It is resistant to cleavage by AChE.

Primarily M receptors.

Adverse FX:
Unline the other direct agonists in this category it is able to enter the brain and cause CNS disturbances (hallucinations, convulsions)
What is methacholine used for? Receptors? Adverse effects?
Diagnosis of asthma and hyperactivity in the bronchus

M receptors

Like those of general cholinergic stimulation
What are the common indirect cholinergic agonists (6) ? (anticholinesterases)
PEEPIN

Isoflurophate
Echothiphate
Parathion
Edrophonium
Physostigmine
Neostigmine
Which indirect cholinergic agonist can irreversibly inhibit AChE?
Organophosphates. They bind covalently.

(isoflurophate, echothipate..)
What are isoflurophate and echothiophate used for?
Occasionally for glaucoma
How is cholinesterase inhibitor poisoning treated?
Atropine with pralidoxime ( chemical antagonist used to regenerate active cholinesterase )
Physostigmine

Indications
Adverse effects
Glaucoma ( second choice after pilocarpine) . It enters the CNS.

Overdoses of atropine, phenothiazines, tricyclic antidepressents

Adverse:
- Convulsions
- Muscle paralysis from overstim.
- Cataracts
+ all other symptoms of excessive ACh.
Edrophonium

Clinical use
Adverse fx
Used in the diagnosis of myasthenia gravis and its differentiation from cholinergic crisis. Short acting (5-15min)

Same as excessive chol. stim.
Pyridostigmine

Clinical use
Adverse FX
Long term treatment of myasthenia gravis

Same as excessive chol...
What are 6 muscarinic blockers?
Atropine
Scopolamine
Homatropine
Tropicamide
Pirenzepine
Which drugs that are not specifically muscarinic antagonists possess antimuscarinic properties ?
1) Benzotropine (treatment of Parkinsons)
2) Ipratropium (anti-asthmatics)
3) Thorazine (anti depressent)
Atropine

Mechanism
Actions
Uses
Contraindications
Toxic effects
Antidote
Reversible, nonselective blockade of muscarinic receptors

CNS- hallucinations/restlessness at high doeses.Does not cross BBB.

CV - Low dose - reduces HR . High dose- blocks muscarinic in heart and causes tachycardia

Suppresses sweating.

Uses:
- Bradycardia
- Eye drops ( mydriasis, cyclopleg.)

Contraindicated in people with with narrow-angle glaucoma

Toxic: Dry as a bone, (xerostomia), Hot as a hare (inhibiton of sweating), Red as a beet ( tachycardia , cutaneous vasodilation), Blind as a bat (blurring), Mad as a hatter (hallucinations)

Antidote = high [ ] of ACh or agonist
Scopolamine

Mechanism
Use
Route
Nonselective competitive blockade

Motion sickness
Transdermally given
Homatropine and Tropicamide

Use
Given to cause mydriasis and cycloplegia
Name four nondepolarizing blockin g agents (subgroup of neuromuscular blocking agents)

Mechanism
Use
Route
Adverse effects
Antidote
Tubocurarine
Pancuronium
Atracurium
Vecuronium

Competitvely block cholinergic transmission at the nicotinic receptors

Adjuvants for anesthesia - promote muscle relaxation

- IV

- Bronchoconstriction and hypotension (histamine release)

- Edrophonium or neostigmine
Name the only depolarizing neuromuscular blocking agent in use in the USA?
Succinylcholine
Succinylcholine

Mechanism
Metabolism
Use
Adv. Effects
Phase 1 - Succinylcholine binds to nAChR, opens Na channels -> depolarization -> transient fasiculations leading to prolonged depolarization ( AChE resistant)
Phase 2 - Membrane partially repolarizes but receptor is now desensitized to ACh = no more action potentials

- Plasma cholinesterase

- Adjuvant to general anesthesia and for rapid intubation

- Adv. effects: Bronchoconstrict. (histamine release), Hypotension/bradycardia, Arrythmias (due to potassium release), Apnea, malignant hyperthermia , Increased intraocular pressure, prolonged paralysis
Do neuromuscular blocking agents block autonomic ganglia too?
No. They use different nicotinic receptors. Tubocurarine may produce a small amount of ganglionic blockade.
Name four ganglionic blockers
Mechanism
Effects
Nicotine
Hexamethonium
Mecamylamine
Trimethaphan

Compete with ACh to bind with nicotinic receptors of both para and symp. ganglia. Nicotine stimulates the ganglia and then blocks them because of persistent depolarization. Drugs like hexameth. mecamyl. and trimetaphan block the ganglia without any prior stimulation.

Heart - Tachycardia ( parasymp is normally dominant)
Vascular - Vasodilation
Eye - Cyclopegia, mydriasis
GI - Reduced motility ; decreased secretions.
Urinary - retention
Sweat - reduced sweating
When is neuromuscular block used?
As an adjunct to anesthesia. It is not a therapeutic intervention.
What is tetanic fade?
a term used to describe the failure of muscle tension to be maintained during a brief period of nerve stimulation at a frequency high enough to produce a fused tetanus. It occurs mainly due to the block of presynaptic nAChRs. It does not occur with a depolarising block.

It i increased by non-depolarising blocking drugs compared to normal.
Characteristics of non-depolarising and depolarising neuromuscular-blocking drugs
-non-depolarising is reversible by anticholinesterase drugs , depolarising is not
- depolarising block produces initial fasciculations and often postoperative pain
What is malignant hyperthermia? Which drugs are implicated? Treatment?
Rare inherited condition, due to a mutation of the ryanodine receptor ( Ca release channel of SR). Results in intense muscle spasm and dramatic rise in body temperature when certain drugs are given.

Succinylcholine and halothane

Treatment by administration of dantrolene (prevents Ca release from SR)
Which agents inhibit Ca entry into the presynpatic neuron?
Mg2+

Aminoglycoside antibiotics (streptomycin and neomycin) -> sometimes cause muscle paralysis clinically
What two toxins inhibit ACh release? Mechanisms? Treatments?
Botulinum toxin
- Contains peptidases that cleave proteins involved in exocytosis.
- Treatment involves antitoxin but must be given before symptoms
- Used to treat blepharospasms as well as other spasms. Wrinkles 2.

B-bungarotoxin
- Contains a phospholipase rather than a peptidase
What are the two types of cholinesterase? Locations? Specificity?
Acetylcholinesterase and butyrylcholinesterase.

Soluble form of AChE is found in CSF and in cholinergic nerve terminals ( monitors free ACh? ). BuChE is found in plasma in soluble form. Bound form of AChE is found in cholinergic synapses (hydrolysis activity)

AChE is quite specific for ACh and methacholine

BuChE is widespread (not only in cholinergic synapses). Function is unclear. Hydrolyses a broader substrates ( butrylylcholine, procaine, succinylcholine, propanidid) [plasma form is important for this]
Which is the only important short acting anticholinesterase? Use?
Edrophonium. Used for diagnostic purposes ( improvement of muscle strength using anticholinesterase is characteristic of myasthenia gravis)
List the medium-duration anticholinesterases
Neostigmine
Pyridostigmine
Physostigmine
List some irreversible anticholinesterases
Dyflos
Parathion
Ecothiopate
What are the effects of anticholinesterase drugs
Affects autonomic cholinergic synapses and neuromuscular junctions. ( just think )

CNS effects - convulsions , depression
Donepezil is an example of what kind of drug
Anticholinesterase used to treat Alzheimer's disease
What are ecothiopate eye drops?
Anticholinesterase drops used to treat glaucoma
Which drugs enhance cholinergic transmission?
Tetraethylammonium
Aminopyridines..

They are not selective enough to be used clinically.
Name the important alpha-selective direct-acting agonists (4)
1) Phenylephrine (a1 receptor)
2) Methoxamine (a1 receptor)
3) Oxymetazoline (a1 receptor)
3) Clonidine (a2 receptor)
4) Methyldopa (a2 receptor)
5) a-methylnoradrenaline (a2)
What are the major B selective direct-acting agonists ?
Dobutamine (b1)
Isoproterenol (both b1 and b2)
Orciprenaline (b2)
Terbutaline (b2)
Salbutamol (b2)
Salmetrol (b2)
What are the major alpha and beta direct-acting agonists?
Epinephrine
Norepinephrine
Dopamine
Which direct-acting agonists are considered catecholamines ?
Epinephrine
NE
Dopamine
Isoproterenol
Dobutamine
Name two indirect-acting adrenergic agonists
Tyramine and amphetamine
Two mixed (direct and indirect) agonists
Ephedrine
Metaraminol
Where are alpha-1 and alpha-2 receptors located?
alpha 1 = postsynaptic membrane
alpha 2 = predominantly on presynaptic membrane (a2 can be found postsynaptically in CNS and blood vessels)
What are the responses to alpha-1 and -2 stimulation?
alpha-1 = release of intracellular calcium via IP3 (Gq protein**) -> sympathetic effects you know (vasoconstriciton etc.).

alpha-2 = cAMP production inhibition (Gi protein). Actions:
1) NE in synapses can bind to the presynpatic receptors and inhibit further release (negative feedback)
2) Suppressed insulin secretion
3) Increased vagal tone
4) Platelet aggregation
Phenylephrine

Action
Uses
Adverse effects
Primarily vasoconstriction.

Nasal decongestant (primary)
Hypotension treatment
To terminate episodes of paroxysmal atrial tachycardia

Rebound mucosal swelling and hypertensive headache
Methoxamine

Use
Adverse effects
Treatment of hypotension and paroxysmal atrial tachycardia

Similar to phenylephrine
Clonidine

Use
Adverse effects
- Treatment of hypertension
- Withdrawal from benzos and opiates
- Treatment of diarrhea in diabetic patients

1) Sedation
2) Dry mouth
3) Sexual dysfunction
4) Orthostatic hypotension
Where are B-receptors found? What occurs from their activation?
B1 = postsynaptic membrane
B2 = pre and postsynaptic

B1 ->Gs-> open Ca channels -> cardiac stimulation and increased lipolysis

B2 ->Gs -> " " -> bronchial smooth muscle + skeletal muscle vasculature dilation, glucagon release is increased, detrusor muscles are relaxed and uterus too.
What do both B1 and B2 receptors produce?
Decreased intestinal tone and motility (alpha receptors cause this too)
Dobutamine

What is it?
Receptors
Effects
Use
Adverse effects
Dopamine analogue

Primarily B1 but some action on B2

Increases HR and contractility (b1) and smooth muscle relaxation (b2)

Treatment of unstable CHF and shock

Arryhthmias, Headache, HTN, Palpitations, Angina, Nausea
Isoproterenol

Receptors
Actions
Use
Adverse effects
B1 and B2 receptors

Increases CV inotropic and chronotropic effect (B1), lowers peripheral vascular resistance (B2), Relaxes smooth muscles (B2)

HR stimulation in heart block and bradycardia

Arrhytmias
Palpitations
Tachycardia
Headache
Albuterol, Metaproterenol, Terbutaline

Receptors
Actions
Uses
Adverse Effects
B2 receptors but can stimulate B1 at higher doses

Stimulation of smooth muscle dilatation

- Treatment of bronchospasm
- Treatment of COPD
- Treatment of bronchitis
- Terbutaline + ritodrine can be used to relax uterus during premature labor.

Arrhythmias
Tachycardia
Headache
Nausea
Epinephrine

Receptors
Action
Use
Adverse
All 4 receptors. Low dose -> B receptors and at high doses -> A receptors.

- CV - Increased HR and contractility
- Resp. - Bronchodilation (B2)
- Metabolic - Increased glycogenolysis and release of glucagon

a) Bronchospasm secondary to asthma
b) Anaphylaxis and cardiac arrest
c) Prolongation of drug effects via vasoconstriction
d) to achieve hemostasis

Arrhythmia
HTN
Palpitation
Tremor
Dizziness
MI
PE
What is epinephrine reversal ?
When EPI is given alone it causes an increase in BP through alpha receptors. When it is given with an alpha-blocker it will cause a decrease in BP because of its B2 activity.
Which receptors does norepinephrine bind to?
A1,A2,B1 receptors. NE has a stronger affinity for alpha-receptors than Beta r.
Norepinephrine

Action
Use
Adverse effects
Vasoconstriction, reflex bradycardia

Shock (last-line agent)

- Tissue hypoxia ( due to vasoconstriction being strong)
- Decreased kidney perfusion
- Arrhythmias
Dopamine

Where is it found
Receptors
Use
Adverse effects
Synthesized in CNS, sympathetic ganglia and adrenal medulla

A1,B1,B2, D1,D2
(D receptors at low dose, B receptors at moderate dose, A1 receptors at higher doses)

Treatment of shock (raises BP via b1) , acute renal failure (increased BF), acute CHF.

- Arrhytmia, Tachycardia, HTN
Tyramine

Action
Use
Adverse
by-product of tyrosine metabolism

Tyramine is taken up by sympathtic neurons -> release of catecholamines

No therapeutic use

Hypertensive emergency when given to patients taking MAO inhibitors
Amphetamine

Actions
Use
Adverse effects
Releases store of NE and dopamine

Treats ADHD and narcolepsy and appetite suppression

Dependence
Psychosis
and so on..
Ephedrine

Action
Use
Adverse
Stimulates release of NE from nerve terminals. Also acts as a direct adrenergic agonist.

Urinary incontinence, bronchospasm, hypotension

Arrhythmias, Palp., Insomnia , HTN
Metaraminol

Actions
Use
Adverse
Indirectly releases NE. Stimulates alpha receptors too

Treatment of hypotension and PAT episodes

Similar to those of norepinephrine
List 6 alpha blockers
1) Prazosin - a1 adrenergic
2) Doxazosin- a1
3) Terazosin- a1
4) Phenoxybenzamine - nonselective,*irreversible* only one!!
5) Yohimbine- a2
6) Phentolamine - nonselective
Prazosin, Terazosin, Doxazosin

Action
Effect
Use
Adverse FX
Competitively blocks a1 receptors

In vascular sm it inhibits constricton of arterioles and venules -> decreased TPR and lower BP. In bladder, there is relaxation and decreased resistance to urine flow

Treatment of HTN and prevention of urinary retention in patients with benign prostatic hypertrophy

GI hypermotility , Orthostatic HTN (after initial dose especially), sexual dysfunction, dry mouth, dizziness
Phenoxybenzamine

Action
Effects
Duration
Use
Adverse FX
Contraindications
Noncompetitively blocks a1 postsynaptic receptors and a2 presynaptic receptors.

Blocks peripheral vasoconstriction and induces reflex tachycardia

Very long. Covalently binds to receptor

a) Treatment of patients with pheochromocytoma-indcued HTN
b) Treatment of patient with benign prostatic hypertrophy. It reduces the size of the prostate
c) Treament of spinal cord injury induced hyperreflexia leading to high bp
d) Treatment of Raynaud's disease

Orthostatic hypotension, Reflex tachycardia, Inhibition of ejaculation

Patients with CAD
What is the use of Yohimbine?
Treat impotency via penile injection
Phentolamine

Mechanism
Use
Adverse effects
Reversibly blocks a1 and a2

Short term control of pheochromocytoma HTN

Orthostatic hypotension, GI stimulation which may lead to peptic ulcers, tachycardia, MI or arrhythmias
What are B blockers? How can they be subclassified?
Competitive antagonists

1. Selectivity of receptor blockade
2. Possession of intrinsic sympathomimetic activity
3. Capacity to block a-adrenergic receptors
Name four selective B1 blockers. Are they really fully selective of B1?
Atenolol
Esmolol
Acebutolol
Metoprolol

* B-blockers starting with A or M are usually cardioselective

No. At high doses these drugs will block B2 receptors
What is the advantage of B1 selectivity
They are sometimes called cardioselective because they lack unwanted bronchoconstrictor and hypoglycemic effects of nonselective blockers
For the four B1 blockers mentioned in another slide, what is the use of each one?
Atenolol - for HTN, MI
Esmolol - For immediate B blockade
Acebutolol - HTN
Metoprolol - HTN, angina pain, MI
What is the prototpe non-selective B-blocker?
Propranolol
What are the actions of nonselective B-blockers?
Decreased CO and BP

Reduction of sinus rate and conduction through atria

- Peripheral vasoconstriction
- Bronchoconstriction
- Decreased glycogenolysis and glucagon secretion
- Increased VLDL and decreased HDL
Propranolol

Site of metabolism
Absorption
Liver

Compeletly absorbed after oral but only 25% reaches blood (first-pass metabolism)
Indications for nonselective B-blockers
HTN
Angina, tachycardia
Arrhythmia
Thyroid storm
Migraines
Timolol and Nadolol are what kinds of drugs? Use?
nonselective b-adrenergic antagonists

Treatment of glaucoma (decrease production of aqueous humor production)
Adverse effects of nonselective B-blockers
Bradycardia
Bronchoconstriction
Hypoglycemia
Fatigue
Depression
Sexual dysfunction
Acebutolol and pindolol are examples of what kind of drugs? Use? Advantages?
B blockers with intrinsic sympathmimtic activity. They are considered partial agonists as they very mildly stimulate both B1 and B2 receptors.

Treatment of HTN in patients prone to bradycardia

They do not induce bradycardia like full antagonists do and produce bronchoconstriction only at very high doses.
Labetalol

Classification
Mechanism
Clinical use
Adverse effects
Nonselective B-blocer with a1 adrenergic selective blockade

By blocking both those receptors, peripheral vasodilation rather than vasoconstriciton occurs (unlike other b-blockers)

Treatment of HTN and atrial fib.

Orthostatic hypotension and dizziness
Any drugs similar to Labetalol? Use? Action? Contraindications?
Carvedilol (B blocker with a1 blocking properties)

Treatment of HTN
Treatment of chronic CHF (reduce sympathetic activity)

- Reduction of sympathetic activity
- Improvement of diastolic dysfunction bu prolonging filling time

- Contraindicated in ACUTE CHF.
Name one B2 selective blocker
Butoxamine

It is not used clinically
Name two indirect adrenergic antagonists. How do they work?
Guanethidine and Reserpine.

They block the release of NE from nerve endings - in effect, antagonizing the effects of the sympathetic system
Guanethidine

Mechanism
Use
Adverse effects
Enters peripheral adrenergic nerves by a reuptake mechanism for NE and binds to storage vesicles

Treatment of HTN

Orthostatic hypotension and sexual dysfunction
Reserpine

Mechanism
Use
Adverse effects
Blocks NE transport from cytoplasm into intracellular storage vesicles.

Treating HTN (rarely used)

CNS depression and bradycardia
Where is most of the serotonin found in the body?
1) In the wall of the intestine in enterochromaffin cells (>90%)

2) In blood ( in platelets)

3) In CNS (transmitter)
What is the precursor of dopamine?
Tryptophan
Tryptophan is converted into 5-HT in which areas?
In neurons and chromaffin cells but NOT platelets. Platelets contain a 5-HT uptake mechanism which allows platelets to get loaded as they pass through the intestinal circulation
How is 5-HT degraded?
MAO
Mechanism of serotonin release in GI tract? Physiological inhibition?
5-HT is released from enterochromaffin cells into the lamina propria, where it stimulates receptors on enteric neurons

Gut contains serotonin uptake transporterwhich removes 5-HT.
5-HT1 receptors

Location, Action
Location : Mainly in the brain

They function mainly as inhibitory presynaptic receptors and are linked to inhibition of adenylate cyclase
What is special about the 5-HT1D subtype? Drug affecting this receptor?
It is expressed in cerebral blood vessels and it is believed to be important in migraine.

Sumatriptan (agonist used to treat acute attacks)
5-HT1 receptors affect cerebral vessels in an unusual way. Why?
They cause vasoconstriction where in most vessels.. 5-HT2 receptors are responsible for this.
5-HT2 receptors

1) Where are they important
2) Effects
3) When is it important
1) In the periphery
2) Effects on smooth muscle and platelets (5-HT2A)
3) Minor role physiologically but prominent in pathology (asthma etc.)
What is the only 5-HT receptor that is not a GPCR?
5-HT3. This is a ligand-gated ion channel
5-HT3

Where? Action
Mainly in peripheral , particularly on nociceptive sensory neurons.

Strongly excitatory
5-HT4

Action
Main role in GI tract producing neuronal excitation and mediating the 5-HT effect of stimulating peristalsis
What are the important actions of 5-HT?
- Increased GI motility (direct action on smooth muscle and indirect via enteric neurons)

- Contraction of other sm (bronchi.etc)

- Mix of vasoconstriction and dilatation

-Platelet aggregation
- Stimulation of peripheral nociceptive nerve endings
- Excitation/inhibition of CNS neurons
What are the role of 5-HT in the periphery?
Peristalsis, vomiting, inflammatory mediator, nociception, platelet aggregation
What does disturbed 5-HT function give rise to?
Migraine
Carcinoid syndrome
Mood disorders
Anxiety
What are some 5-HT2 antagonists? (3)

Uses
1) Ergotamine
2) Methysergide
3) Cyproheptadine

1) Migraine treatment
2) Migraine prophylaxis +carcinoid syndrome
3) Control symptoms of carcinoid
tumors
5-HT3 receptor antagonist (3)
1) Use
Ondansetron
Granisetron
Tropisetron

1) Used as antiemetic drugs
5-HT4 receptor agonists include (2)

Action
Metoclopramide
Tegaserod

Stimulate coordinated peristaltic activity. Used from treating GI disorders.
What drugs are used for acute migraine attacks?
- Simple analgesics (ex. aspirin)
- Ergotamine (5-HT1D partial agonist)
- Sumatriptan, zolmitriptan (5-HT1D agonist)
Which drugs are used for the prophylaxis of acute migraines?
- B-adrenceptor antagonists (propanolol, metoprolol)
- Pizotifen (5-HT2 receptor antagonist)
- Cyproheptadine (5-HT2 antag.)
- Methysergide (5-HT2 antag)
- Tricyclic antidepressants
- Clonidine (a2 agonist)
- Calcium antagonists
What is carcinoid syndrome?
Rare disorder associated with malignant tumors of enterochromaffin cells. They release 5-HT and other mediators.
P1 and P2 receptors are responsible for what?
P1 respond to adenosine
-subtypes: A-1,2,3
P2 respond to ATP and/or ADP
- subtypes: P2x (ATP) and P2y (ATP,ADP)
ATP

Function
ATP receptor antagonist
Primary neurotransmitter and/or a cotransmitter in noradrenergic nerve terminals.

Suramin
What occurs once ATP is released from the cell?
It is rapidly converted into ADP and adenosine
What is adenosine uptake blocked by?
Dipyridamole
Effects of adenosine
- Vasodilatation (A2)
- Inhibition of platelet aggreg (A2)
- Blocks cardiac AV conduction (A1)
- Bronchoconstriction(A1)
- Release of mediators from mast cells (A3)
- Inhibitor of transmitter release at many peripheral and central synapses (A1)
What may adenosine be administered for?
To terminate supraventricular tachycardias. It is safer than B-blockers and verapamil
Which drugs inhibit A1 and A2 adenosine receptors?
Xanthines such as theophylline
NSAIDS

1) Therapeutic effects
2) Side effects
1) Anti-inflammatory , Analgesic , Antipyretic

2) Gastric irritation (ulcers), RBF effected in compromised kidney, prolong bleeding through inhibition of platelet function, skin rashes
Most traditional NSAIDs inhibit which enzymes? Effect?
COX-1 and COX-2

Inhibition of COX-2 is believed to cause the anti-inflammatory action

Inhibition of COX-1 is believed to be responsible for the side effects experienced
What is the mechanism behind NSAIDs ability to be anti-inflammatory, analgesic and antipyretic?
Anti-inflammatory - decrease in PGE2 and PGI2 reduces vasodilation and edema

Analgesic - Decreased PG generation -> less sensitisation of nociceptive nerve endings to inflammatory mediators like bradykinin and 5-HT

Antipyretic - IL-1 release PG in the CNS where they elevate the set point for temp. NSAIDs prevent this.
List some important NSAIDs
Aspirin
Ibuprofen
Naproxen
Indometacin
Piroxicam
What type of inhibitors are most NSAIDs? Which reaction of the COX enzyme do most inhibit? Mechanism?
Competitive reversible inhibitors (exception: Aspirin = irreversible acetylation of serine)

The dioxygenation reaction

NSAIDs enter the hydrophobic channel of COX enzymes and prevent substrate FA from entering
NSAIDs' antiplatelet activity is due to what?
Decreased production of thromboxane. This effect lasts 7 days ( life span of platelet)

Low-dose, long-term aspirin use irreversibly blocks the formation of thromboxane A2 in platelets, producing an inhibitory effect on platelet aggregation
What are six groups of NSAIDs and give examples of each
Salicylates:
- Aspirin
- Diflunisal
- Salsalate
Pyrazolones:
- Phenylbutazone (very toxic)
Indoleacetic acids:
- Indomethacin
- Sulindac
- Etodolac
- Ketorolac
Propionic Acids:
- Naprosen
- Ketoprofen
- Ibuprofen
Oxicams:
- Piroxicam
Fenamates:
- Mefenamic acid
- Meclofenamic acid
What serious renal effect does chronic NSAID consuption/abuse cause?
Analgesic nephropathy characterised by chronic nephritis and renal papillary necrosis
Give examples of NSAIDs that would be given clinically for short-term pain releif and long-term
Short term: aspirin, ibuprofen, paracetamol

Chronic pain: difunisal, naproxen, piroxicam
Where does absorption of aspirin occur?
It is is protonated (making it neutral) in the stomach, thus facilitating passage across the mucosa. However, most absorption occurs in the ileum.
What unwanted effects are specific in aspirin and other salicylates?
Salicylism characterized by tinnitus, vertigo, decreased hearing , and vomiting and nausea

Reye's syndrome, disorder of children characterized by hepatic encephalopathy followed by an acute viral illness.
What are some important interactions of aspirin ? Contraindicated?
Increases the effect of warfarin
Interferes with the effect of uricosuric agents

Should not be used in gout (reduces urate excretion)
What usually occurs at toxic doses of aspirin (like suicide attempts)?
Uncompensated respiratory and metabolic acidosis .

Compare with large doses which cause salicylism and with therapeutic doses usually gastric distress
What are the unwanted effects of paracetamol (acetaminophen)
Therapeutic dose - few effects and uncommon

Toxic doses - Nausea + vomiting at first and then fatal hepatotoxicity caused by build up of metabolite N-acetyl-p-benzoquinone imine .


Agents that increase glutathione ( used to make a conjugate for drugs) can prevent liver damage if given early
Which drugs are COX-2 selective inhibitors?
The coxibs. Very rarely used because of CV risks.
What does the word antihistamine refer to?
H1 receptor antagonist
Action of antihistamines
Decrease histamine-mediated contraction of sm of bronchi, intestine and uterus.
H1 receptor antagonists show affinity for what type of other receptor (Not H1)
Muscarinic, although lower than for histamine receptor. They act as muscarinic antagonists.
What sort of action does cyproheptadine have?
5-HT antagonist as well as an H1 receptor antagonist.
What are the clinical uses of H1 receptor antagonists?
Allergic reactions
- non-sedating drugs (fexofenadine, cetirizine) for rhinitin (hay fever) and urticaria
- topical preps for insect bites
- injectable formulas for anaphyl.

Antiemetics
- prevention of motion sickness

Sedation
- Some H1 antagonist (promethazine ( or SYZZURP) are fairly strong sedatives
What are the clinical uses of immunosuppressants
1) Suppress rejection of transplants
2) Suppress graft vs host in bone marrow transplants
3) Treat conditions with an autoimmune component
Name some autoimmune diseases which can be treated using immunosuppression drugs?
- Idiopathic thrombocytopenic purpura
- Hemolytic anemia (some forms)
- Glomerulonephritis (some forms)
- Myasthenia gravis
- SLE
- Rheumatoid arthritis
- Psoriasis
- Ulcerative colitis
List the different classes of immunosuppressive drugs. Example of each?
1) Drugs that inhibit IL-2 production/action (ciclosporin, tacrolimus)

2) Drugs that inhibit cytokine gene expression (corticosteroids)

3) Drugs that inhibit pure and pyrimidine synthesis (azathioprine , mycophenolate mofetil)

4) Drugs that block the T-cell surface molecules involved in signalling
Ciclosporin

What is it
Actions
Mechanism
It is a potent immunosuppressive with no effect on the acute inflammatory reaction. It does not involve cytotoxicity.

a) Decreased clonal proliferation of T cells by inhibiting IL-2 synthesis and possibly also decreasingexpression of IL-2 receptors
b) Reduced induction and clonal proliferation of cytotoxic T cells (CD8)
c) Reduced functon of effector T cells
d) reduction of T cell-dependent B cell responses

Ciclosporin binds to cyclophili ( an immunophilin) in the cytosol of Th cells. This complex then binds and inhibits calcineurin(phosphatase) that activates transcription factors initation IL-2 transcription.
What is the most common and most serious adverse effect of ciclosporin? Other adverse effects?

Does ciclosporin depress the bone marrow?
Nephrotoxicity

Hepatotoxicity and HTN, anorexia, lethargy, hirsutism, parasethsia , gum hypertrophy

No it does not.
Tacrolimus -Compare to ciclosporin

Pimecrolimus and sirolimus - What are they and use
More potent than ciclosporin. It does not bind to cyclophilin like ciclosporin but instead to a different immunophilin named FKBP . This complex inhibits clacineurin. More severe side effects with the incidence of nephrotoxicity and neurotroxicity being higher. GI and metabolic disturbances (hyperglycemia) also occur as side effects.

Pimecrolimus (used for atopic eczema) and sirolimus ( used to prevent organ rejection) are immunosuppressant with similar properties to tacrolimus
Glucorticoids

Purpose in immunosuppression
Action
Immunosuppression is mediated by both effects of glucorticoids (effects on immune response and anti-inflammatory response). They are immunosuppressant chiefly.

Decrease transcription of IL-2 but also of many other ones (incl. TNF-a, IFN-y, IL-1 etc.) in both the induction and effector phases of the immune response. This does this throguh inhibiting the action of transcription factors (such as activator protein-1 and NF kB)
Azathioprine

Action
Use
Mechanism
Which immune reactions are depressed by this drug?
Unwanted effects?
Intereferes with purine synthesis and is cytotoxic

Used particuarly for control of autoimmune disease and to prevent tissue rejection.

The drug is metabolised to give mercaptopurine which is a purine analogue that inhibits DNA syntheis.

Both cell and antibody-mediated because it inhibits clonal proliferation during the induction phase.

Depression of bone marrow (main unwanted effect), anausea, vomiting etc.
Mycophenolate Mofetil

Mechanism
Use
It is converted into mycophenolic acid which restrains T and B lymphocyte proliferation and reduces the production of CD8 T cells by inhibiting inosine monophosphate dehydrogenase (crucial for de novo purine biosynthesis in both T and B cells). This is the only way these cells can make them so the drug is fairly selective ( other cells can use other pathways)

To curtail transplant rejection
What are the principal eicosanoids
PGs , thromboxanes, leukotrienes
Besides arachidonic acid, PLA2 also generates something else. Do you know what?
lysoglyceryl-phosphorylcholine . This is the precursor of PAF.
Apart from PLA2 , what other enzyme can release arachidonate ?
Two step process involving PLC and DAG lipase. It releases glycerol and arachidonate
What is the main eicosanoid produced by platelets? vascular endothelium?
TXA2 and PGI2 , respectively.
What receptors do PGD2, PGF2a, PGI2, PGE2 and TXA2 bind to?
DP,FP,IP,EP,TP respectively.
PGD2

Actions
Vasodilation
Inhibition of platelet aggregation
Bronchoconstrictor
Relaxes GI and uterine muscle
PGF2a

Actions
Myometrial contraction
PGI2

Actions
Vasodilation
Inhibition of platelet aggregation
Renin release and natriuresis
TXA2

Actions
Vasoconstriction
Platelet aggregation
Bronchoconstriction
PGE2

Actions
Contraction of bronchial and GI sm (EP1)

Pyrogenic (inducing fever) - final mediator of raising the temperature

Bronchodilation, vasodilation, GI relaxation (EP2)

Contraction of intestinal sm, inhibition of gastric acid secretion, increased gastric mucus secretion, inhibition of lipolysis and autonomic neurotransmitter release and stimulates the contraction of the pregnant uterus (EP3)
Which prostaglandins are the most important in acute inflammation ? Chronic?
PGE2 (predominates) and PGI2 (local tissue and vessels) and PGD2 from mast cells.

PGE2 and TXA2 from monocytes/macrophages
How do prostanoids (PG and TXA2) produce vasodilation, increased permeability and pain
Really they don't do it directly. They synergize with other vasodilators like histamine and bradykinin. (potentiate). They potentiate the effect of bradykinin by sensitizing afferent C fibers to the effects of noxious stimuli.
What are some clinical uses of prostanoids?
OB/GYN
- Termination of pregnancy (gemeprost, misoprostol)
- Induction of labour (dinoprostone or misoprostol)
- Postpartum hemorrhage (carboprost)

GI
- Prevent ulcers associated with NSAID use (misoprostol)

CV
- maintain patency of ductus arteriosus until surgical correction (alprostadil - PGE1)
- Inhibit platelet aggregation (epoprostenol-PGI2)
- Primary pulmonary HTN (epoprostenol)

Ophthalmic
- Latanoprost for glaucoma
What are leukotrienes made by?
White cells - leuko.
Leukotrienes

Receptors
Actions of types
Cysteinyl-leukotrienes are of particular important in what type of inflammation?
BLT (LTB4) and CysLT (rest)

LTB4 - potent chemotactic agent for neutrophils and macrophages, proliferation of macro and lympho and cytokine release

Cysteinyl-leukotrienes act on the respiratory system (bronchoconstriction, mucus secretion (+) and the CV system (vasodilation, constriction of coronary vessels)
LTD4 increases nasal blood flow and local vascular permeability

Asthma (LTB4 is important in all types)
Source of PAF? Actions?
Platelets stimulated with thrombin and most inflammatory cells

Vasodilation, Increased vascular permeability, chemotaxis and activation of leukocytes, activation of platelets and smooth muscle contraction
Which CysLT-receptor antagonists are in use for the treatment of asthma?
Zafirlukast and montelukast
Which NSAID stimulates the synthesis of lipoxins?
Aspirin
What is PAF implicated in? (diseases)
Bronchial hyperresponsiveness and delayed phase of asthma
What are some lipooxygenase inhibitors ?
Azelastine
Diethylcarbamazine
Nordihydroguaiaretic acid
zileuton
What kind of receptors do most cytokines act on? Exception?
Kinase-linked receptors (such as Jak/Stat)

Chemokines act on G-protein coupled receptors.
Anti-inflammatory cytokines
TGF-B, IL-4, IL-10, IL-13
Drugs affecting gout act in which ways?
Inhibit uric acid synthesis (allopurinol)

Increasing uric acid excretion (uricosuric agents - probenecid, sulfinpyrazone)

Inhibiting leukocyte migration to the joint (colchicine)

General anti-inflammatory and analgesic effect (NSAID)
Allopurinol

Mechanism
Adverse effects
When not to treat
Interactions
Competitive inhibition of xanthine oxidase reducing the synthesis of uric acid. Its action is due to alloxanthine (non-competitive metabolite inhibitor of allopurinol from xanthine oxidase.

GI disturbances and allergic reactions. Stevens-Johnson syndrome and toxic epidermal necrolysis can occur as well

During an acute attack of gout

Increases the effect of mercaptopurine and azathioprine and cyclophosphamide. ( These are all anti-cancer drugs). The effect of warfarin is also increased because its metabolism is inhibited.
Treatment of gout with uricosuric drugs and allopurinol is initiated with what accompanying drug?
an NSAID.
When are uricosuric agents used?
Prophylaxis for patients with gout who have severe adverse reactions to allopurinol
Colchicine

Mechanism
Use
Adverse
Prevents migration of neutrophils into the joint by binding to tubulin and reducing cell motility by depolymerisation of microtubules.

To prevent and relieve acute attacks

GI problems : nausea , abdominal pain, diarrhea
Drugs available for treating acute attack of gout?
Indomethacin , Phenylbutazone, Colchicine
Aspirin is an NSAID - can it be used in treatment of gout?
NO! being a salicylate makes it inhibit uric acid secretion into the renal tubules . Contraindicated !!
Which drugs are used to treat chronic gout?
Allopurinol and Uricosuric agents
Which drugs are most frequently used in the treatment of rheumatoid arthritis ?
DMARDs and NSAIDs
Besides DMARDs and NSAIDs what other drugs are used?
Some immunosuppressants like azathioprine and ciclosporin and glucocorticoids
What drugs fall under DMARDs?
Methotrexate , Sulfasalazine, gold compounds, penicillamine, chloroquine
Methotrexate

Mechanism
Folic acid antagonist with cytotoxic and immunosuppressant activity
What are the three isoforms of NOS? Characteristics
iNOS (NOS-2) , eNOS (NOS-3), nNOS (NOS-1)

* eNOS isn't restricted to the endothelium (found also in cardiocytes, bone cells etc..)

* iNOS is also present in endothelial cells

nNOS and iNOS are soluble cytosolic enzymes
What are the substrates for NOS?
L-arginine and O2
How is NO inactivated?
By combination with heme of hemoglobin or by oxidation to nitrite and nitrate -> excreted.
Which secondary messanger does NO use?
cGMP
The effects of cGMP are terminated by which enzymes?
Phosphodiesterases
Which drugs are inhibitors of phosphodiesterase type V ? What are they used for?
Sildenafil and tadalafil. Treating erectile dysfunction because they potentiate NO action.
Effects of NO
- Autocrine and Paracrine action activating cGMP (by binding to heme of guanylate cyclase) which leads to inhibition of Ca -induced smooth muscle contraction (Vasodilation). NO also hyperpolarizes vascular smooth muscle

NANC neurotransmitter in many tissues. Important in upper airways, GI and control of penile erection. It is implicated in the neuronal development of synaptic plasticity in the CNS as well. (Synaptic effect in PNS and CNS)

Host defense (cytotoxic peroxynitrite when bound with superoxide anion)

Cytoprotection
What do high concentrations of NO cause? What is the main action of inhaled NO? Use?
Pulmonary edema and methemoglobinemia

Pulmonary vasodilation

Reduce pulmonary HTN and to improve O2 delivery in patients with ARDS
Which drug inhibit the production of NO?
Glucorticoids inhibit biosynthesis of iNOS

Synthetic arginine analogues

Endogenous NOS inhibitors (ADMA and PIN)
What is a NO donor?
glyceryl trinitrate
What does NO and sepsis have to do with anything? What
NO benefits host defence by killing organism but excessive NO causes harmful hypotension.
Some other diseases NO may be implicated in?
Underproduction of neuronal NO is seen in babies with hypertrophic pyloric stenosis.

NO production by the endothelium is reduced in patients with hypercholesterolemia and some other risks factors for atherosclerosis

Overproduction may be important in neurodegenerative disease (and sepsis :) )
What are some nitric oxide donors?
Nitroprusside and organic nitrovasodilators
Failure to increase endogenous NO biosynthesis normally during pregnancy is thought to cause?
Eclampsia, a hypertensive disorder that accounts for many maternal deaths
Excessive NDMA receptor activation causes?
Increases NO synthesis and contributes to several forms of brain damage
Endothelial dysfunction in diabetic patients can cause..
erectile dysfunction due to blunted relaxation to ACh despite preserved responses to nitroprusside
Which prostaglandins *only ones* that decay rapidly without prostaglandin-specific enzymes?
PGI2 and TXA2
Formula for calculating bioavailability (F)?
F=AUC(route)/AUC(IV)
The elimination of drugs that follow first-order kinetics can be characterized by a proportionately constant. What is it called, and what is it defined as?
Clearance, Cl. Clearance is defined as: Cl=rate of elimination/plasma concentration
Elimination half-life can be derived from graphs of plasma concentration v time, or it can be obtained by a calculation. What is it?
t1/2=0.693*Vd/Cl
The loading dose must fill the volume of distribution of the drug (Vd) to achieve the target plasma concentration (Cp). The formula is
Loading dose=(Vd*Cp)/F
Maintenance dosage must replace the drug that is being eliminated by the body over time to maintain a steady Cp, and thus it involves Cl. The formula for maintenance dosage is:
Maintenance dosage=(Cl*Cp)/F
Drug receptors can be divided into five groups. What are the groups, and give some examples of each group
1. Steroid: Estrogen, corticosteroids, thyroid hormone
2. Ion channel: Acetylcholine (on nicotinic AChR)
3. Transmembrane tyrosine kinase: Insulin
4. JAK-STAT: Cytokines
5.GPCR: Norepinephrine, ACh (on muscarinic receptors)
In the US, scheduled drugs are drugs that are considered to have significant potential for illicit use. They are ranked according to their perceived social danger. The different ranks are
I - Banned from prescription or anything other than research use
II - Strongly addicting drugs that nevertheless have important medical uses
III, IV and V - Progressively less addicting
In the sympathetic nervous system, the neurotrannsmitter at post-ganglionic nerve endings varies depending on the tissue being innervated. The neurotransmitters used are ACh and Norepinephrine. Give some examples where they are used
1. ACh is used for the adrenal medulla, thermoregulatory sweat glands and some vasodilator fibers going to skeletal muscle blood vessels
2. For (most) everything else, there's norepinephrine
A drug that can inhibit hydroxylation of tyrosine in the production of norepinephrine
metyrosine
Monoamine oxidases (MAOs) are present where?
On the mitochondria in nerve endings
Why aren't NE-synthesizing neurons inhibited by botulin toxin?
They don't contain the endocytoic mechanism for taking up botulin toxin. Therefore they are not inhibited.
Drug that blocks the reuptake of choline in nerve endings? (3)
1. hemicholinium-3, or hemicholine
2. blocks the high-affinity transporter ChT, this is the rate-limiting step in ACh synthesis
3. classified as an indirect acetylcholine anatgonist
Drug that blocks the uptake of ACh.
1. vesamicol
2. it is an experimental drug, and it blocks the uptake of ACh into storage vesicles
3. classified as a physiological cholinergic antagonist.
Give a few examples of drugs working on presynaptic, noradrenergic nerve endings (4 [5])
1. metyrosine - inhibits Tyrosine Hydroxylase; decreases catecholamine synthesis
2. reserpine - blocks vesicular monoamine transporter (VMAT); uptake of dopamine into storage vesicles.
3. guanethidine - competes with norepinephrine uptake into storage vesicles, depleting the nerve ending of NE. In addition it blocks the relase of neurotransmitters in response to an action potential
4. TCA and cocaine - block the Uptake 1 transporter
G protein associated with, effect and second messenger of nicotinic receptors
1. none
2. opens Na-K-channel
3. depolzarizes cell
G protein associated with, effect and second messenger of muscarinic receptors.
1. Gq (smooth muscle, some glands), Gi (cardiac muscle).
2. Increases IP3 and DAG (Gq), decreases cAMP & opens K+ channels (Gi)
G protein associated with, effect and second messenger of α1-receptors.
1. Gq (smooth muscle, some glands)
2. Increases IP3 and DAG
G protein associated with, effect and second messenger of α2-receptors
1. Gi (smooth muscle, preganglionic nerve endings, CNS)
2. Decreases cAMP
G protein associated with, effect and second messenger of beta1, beta2, beta3 receptors
1. Gs (smooth and cardiac muscle, juxtaglomerular apparatus, adipocytes)
2. Increases cAMP.
Effect of sympathetic discharge (and receptor type) on eye
dilates pupil (α)
Effect of sympathetic discharge (and receptor type) on airways
dilates bronchioles (β2)
Effect of sympathetic discharge (and receptor type) on GI tract
slows motility (α, β2)
Effect of sympathetic discharge (and receptor type) on GU tract
contracts sphincters, mediates ejaculation (α)
Effect of sympathetic discharge (and receptor type) on vessels (2)
1. constricts arterioles in skin and splanhcnic vessels (α)
2. dilates skeletal muscle vessels (β2)
Effect of sympathetic discharge (and receptor type) on heart
accelerates all pacemaker and AV conduction, increases force of contraction (β1, β2)
Effect of sympathetic discharge (and receptor type) on exocrine glands (2)
1. increases sweating (M)
2. salivation (slight, α)
Effect of sympathetic discharge (and receptor type) on metabolism (4)
Increases:

1. glycogenolysis (β)
2. free fatty acids in blood (β)
3. renin release (β)
4. potassium release and uptake (β)
5. potentiates thyroid effects (β)
Effect of sympathetic discharge (and receptor type) on skeletal muscle
1. increases strength (alpha)
2. causes tremor (beta2)
Effect of parasympathetic discharge (and receptor type) on eye (2)
1. constricts pupil (M)
2. focuses for near vision (M)
Effect of parasympathetic discharge (and receptor type) on airways
constricts bronchioles (M)
Effect of parasympathetic discharge (and receptor type) on GI tract
increases motility, secretion (M)
Effect of parasympathetic discharge (and receptor type) on GU tract
1. contracts wall of bladder (M)
2. mediates erection (M)
Effect of parasympathetic discharge (and receptor type) on vessels
little effect
Effect of parasympathetic discharge (and receptor type) on heart (2)
1. slows sinus rate and AV conduction (M)
2. increases AV refractory period (M)
Effect of parasympathetic discharge (and receptor type) on exocrine glands
1. increases salivation markedly (M)
2. increases lacrimal secretion (M)
3. increases gastric secretion (M)
4. increases duodenal secretion (M)
5. increases pancreatic secretion (M)
Effect of parasympathetic discharge (and receptor type) on metabolism
little effect
Effect of parasympathetic discharge (and receptor type) on skeletal muscle
little effect
Clinical uses of cholinomimetics (6)
1. Treatment of Glaucoma (constrict, reduce intraocular pressure by increasing outflow of aqueous humor)
2. Reduce systemic toxicity (pilocarpine and physostigmine)
3. Treatment of ileus (bethanechol or neostigmine orally or subcutaneously)
4. Treatment of urinary retention
5. Myasthenia gravis (direct-acting cholinomimetics are useless here)
6. Insecticides
Toxicities of direct-acting and indirect acting cholinomimetics are similar, but because cholinesterase inhibitors amplify the nicotinic and the muscarinic actions, more nicotinic manifestations may be observed. These toxicities are best remembered with the mnemonic
D - diarrhea, defecation (M)
U - urination (M)
M - miosis (muscarinic)
B - bronchospasm (M)
B - bradycardia (M))
E - excitation (of CNS and skeletal muscle receptors)
L - lacrimation (M)
S - salivation (M)
S - sweating (M)
Muscarinic receptor blockers are what kind of blockers?
They are competitive pharmacologic antagonists, shifting the graded dose-response curve for agonists to the right
Nicotinic receptor blockers are what kind of blockers?
Include two separate groups of drugs that selectively block ganglia or the neuromuscular junction in skeletal muscle.
1. ganglion blockers are rarely used; neuromuscular blockers are very important in anesthesiology.
2. both categories are competitive pharmacological antagonists.
Actions and toxicities of muscarinic blockers on the CNS (5)
Therapeutic doses:

1. sedation
2. some reduction of motion sickness
3. reduction of Parkinsonian tremor.

Toxic doses:

4. hallucinations
5. convulsions
Actions and toxicities of muscarinic blockers on the eyes
Marked mydriasis and cycloplegia.
What is cycloplegia?
loss of accomodation (due to paralysis of the ciliary muscle)
Actions and toxicities of muscarinic blockers on the airways (2)
1. little effect on normal airways
2. used to treat bronchospasm in some patients with asthma.
Actions and toxicities of muscarinic blockers on the GI tract
High therapeutic doses; reduce hypermotility and secretion of gastric acid, but other drugs are preferred. Salivation is markedly reduced by small doses.
Actions and toxicities of muscarinic blockers on the GU tract
moderate doses reduce bladder tone and may precipitate urinary retention (esp. in older men)
Actions and toxicities of muscarinic blockers on the cardiovascular system (3)
at therapeutic doses;

1. no effect on vessels
2. heart rate and AV conduction velocity usually increase due to vagal blockade.

High doses:

3. flushing of the skin
Toxicities of ganglion blockers (5)
1. cycloplegia
2. orthostatic hypotension
3. constipation
4. urinary retention
5. sexual dysfunction
Modes of action of neuromuscular nicotinic blockers (2)
1. competitive pharmacologic antagonism (nondepolarizing blockers)
2. prolonged acetylcholine-like agonist action (depolarizing blockkers)
Mode of action and indications for nondepolarizing blockers
1. Prevent depolarization of the end-plate
2. Used for surgical procedures of medium and long duration
Toxicities of nondepolarizing blockers
1. hypotension (from histamine release and ganglion blockade)
2. excessive neuromuscular block requiring prolonged respiratory support
Six representative antimuscarinic drugs
1. atropine
2. scopolamine
3. benzotropine
4. glycopyrrolate
5. ipratropium
6. oxybutynin
Properties of atropine (5)
1. lipid soluble
2. enters CNS and eye readily
3. duration of action 4-8 hours except eye, >72 hours.
4. reduces airway secretion and AV block
5. causes mydriasis and cycloplegia; an important antidote for cholinomimetic overdose
Properties and uses of scopolamine (3)
1. similar to atropine
2. strong anti-motion sickness effect
3. causes mydriasis and cycloplegia
Properties and uses of benzotropine (3)
1. centrally acting anticholinergic/antihistaminic agent
2. enters CNS readily
3. treat parkinsonism
Properties and uses of glycopyrrolate (3)
1. enters CNS poorly
2. good peripheral muscarinic blockade
3. reduces parasympathetic effects on the GI and GU tracts
Properties and uses of ipratropium (3)
1. anticholinergic (anti-muscarinic)
2. enters CNS poorly
3. short half-life in blood
4. used by inhalation route for asthma
Properties and uses of oxybutynin (2)
1. enters CNS but strong peripheral muscarinic blockade
2. reduces bladder urgency, spasm
Some representative nicotinic-blocking drugs (4)
1. hexamethonium (prototype ganglion blocker)
2. trimethaphan (ganglion blocker)
3. d-Tubocurarine
4. succinylcholine (depolarizing nicotinic blocker)
Properties and uses of hexamethonium (3)
1. prototype ganglion blocker
2. no CNS effects
3. no uses (obsolete antihypertensive)
Properties and uses of trimethaphan
1. very short-acting ganglion blocker, parenteral only
2. used in hypertensive emergencies
Properties and uses of d-tubocurarine
1. long acting (3-60 minutes) nondepolarizing neuromuscular blocker prototype, histamine releaser and weak ganglion blocker; requires good renal function for elimination
2. produces neuromuscular paralysis for surgery; mechanical ventilation is usually required
Properties and uses of succinylcholine (4)
1. An AGONIST
2. depolarizing neuromuscular blocker
3. very short (3-10 minutes) duration of action
4. produces neuromuscular paralysis of very short duration
What is pralidoxime? (3)
1. cholinesterase regenerator
2. antidote in severe cholinesterase inhibitor poisoning caused by organophosphate insecticides (atropine is also used in all cases of cholinesterase inhibitor poisoning)
3. it is an antagonist, has a high affinity for phosphorous, and frees up the cholinesterase
Examples of direct-acting sympathomimetics (4)
1. epinephrine
2. norepinephrine
3. dopamine
4. isoproterenol
Examples of indirect-acting sympathomimetics (4)
1. amphetamines and ephedrine (displace transmitter from its stores)
2. cocaine and TCAs (inhibit reuptake)
Actions of sypathomimetics on the CNS
1. indirect-acting agents produce a dose dependent sequence of stimulant effects, ranging from mildly alerting and reduction of fatigue to a definite elvation of mood and insomnia, and to marked anorexia and euphoria
2. probably more related to dopamine release than to NE release
Actions of sympathomimetics on eyes
α1-activation in the pupillary dilator muscle results in mydriasis
Actions of sympathomimetics on airways
beta2-activation results in bronchodilation
Actions of sympathomimetics on the GI tract
both alpha and beta receptors mediate reduced motility
Actions of sympathomimetics on the GU tract
1. α1 receptors mediate increased sphincter tone in the bladder and prostate
2. β2 receptors mediate uterine relaxation
Actions of sympathomimetics on the heart
1. β-receptors mediate increased myocardial contractility and increased heart rate
2. the net heart rate effects depend on the reflexes evoked by the changes in blood pressure
Other effects of β-receptor activation (3)
1. increased blood insulin and FFA levels
2. hyperkalemia, followed by hypokalemia and leukocytosis
3. β2 agonists cause tremor in skeletal muscles at most doses (α agonists may increase strength at high doses)
Uses of sympathomimetics (CNS)
amphetamines are used to treat hyperkinetic attention deficit disorder and narcolepsy, and to decrease appetite
Uses of sympathomimetics (pulmonary and cardiac) (3)
1. epinephrine is used to treat anaphylactic shock
2. beta2 agonists (by inhalation) for acute asthmatic bronchospasm
3. beta agonists are occasionally used to increase heart rate
Vascular uses of sympathomimetics (3)
1. alpha agonists are used to decrease blood flow (to reduce bleeding and congestion and to prolong local anesthesia)
2. dopamine is used to maintain renal blood flow in shock
3. midodrine is used to treat idiopathic orthostatic hypotension
Genitourinal (GU) tract uses of sympathomimetics
1. α-agonists: reduce urinary incontinence; long-acting indirect oral agents (e.g., ephedrine) are suitable
2. β2 agonists, e.g., ritodrine and terbutaline, are sometimes used to suppress preterm labour (but their value is controversial)
Toxicities of sympathomimetics (4)
1. arrhythmias (all)
2. myocardial infarction (all)
3. amphetamine and cocaine have a high addiction potential, may cause seizures
4. in high systemic concentrations: α-agonists - stroke; high local concentrations - local tissue ischemia and necrosis
Give some examples of representative α-blockers
1. prazosin (α1-selective) - competitive pharmacologic antagonist
2. phentolamine (nonselective) - competitive pharmacologic antagonist
3. phenoxybenzamine (nonselective) - irreversible in mode of action, thus preferred in pheochromocytomas
Action of α-blockers
1. prevent smooth muscle contraction
2. decreased peripheral resistance and blood pressure, often accompanied by reflex tachycardia
3. in the GU tract, α-receptors mediate contraction of prostate smooth muscle; α-blockers are able to reduce urinary obstruction in men with benign prostatic hyperplasia
What is epinephrine reversal phenomenon?
In the presence of high concentration of epinephrine, α-blockers cause an actual reversal of of the blood pressure response to the agonist: the normal hypertensive response (mediated by α-receptors) is converted to a hypotensive response (mediated by β2 receptors)
Norepinephrine binds to which receptors? (3)
1. α1
2. α2
3. β1
Epinephrine binds to which receptors?
1. alpha1
2. alpha2
3. beta1
4. beta2
Isoproterenol binds to which receptors? (2)
1. β1
2. β2
Dopamine binds to which blood vessel and heart receptors?
1. D2
2. (beta)
2. alpha (high concentrations)
Phenylephrine binds to which receptors?
1. α1
2. α2
Albuterol binds to
β2 receptors (it is a sympathomimetic)
Effect on vascular tone, blood pressure and heart rate of norepinephrine
1. skin/viscera: ++
2. skeletal muscle: (+)
3. kidneys: (+)
4. blood pressure: +++
5. heart rate: - - (reflex)
Effect on vascular tone, blood pressure and heart rate of epinephrine
1. skin/viscera: ++
2. skeletal muscle: - -
3. kidneys: (+)
4. blood pressure: ++
5. heart rate: variable
Effect on vascular tone, blood pressure and heart rate of isoproterenol
1. skin/viscera: none
2. skeletal muscle: - -
3. kidneys: none
4. blood pressure: - -
5. heart rate: +++
Effect on vascular tone, blood pressure and heart rate of dopamine
1. skin/viscera: none
2. skeletal muscle: none
3. kidneys: - -
4. blood pressure: +
5. heart rate: variable
Effect on vascular tone, blood pressure and heart rate of phenylephrine
1. skin/viscera: +
2. skeletal muscle: (+)
3. kidneys: +
4. blood pressure: +
5. heart rate: - - (reflex)
Effect on vascular tone, blood pressure and heart rate of albuterol
1. skin/viscera: none
2. skeletal muscle: - -
3. kidneys: none
4. blood pressure: -
5. heart rate: +
Prototype substances that bind to all α-receptors? (3)
1. norepinephrine
2. epinephrine
3. phenylephrine
Prototype substance that bind to α1 receptors, and its properties
1. midodrine
2. activates Gq, increases IP3, DAG
3. smooth muscle contraction
Prototype substance that binds to α2 receptors, and its properties (4)
1. clonidine
2. activates Gi, decreases cAMP
3. inhibits transmitter release
4. smooth muscle contraction
Prototype substances that bind to all beta receptors (2)
1. a. isoproterenol (isoprenaline)
b. epinephrine
2. activates Gs, increases cAMP
3. effects vary on the subfamily
Prototype substance that bind to β1 receptors
1. dobutamine
2. activates Gs, increases cAMP
3. cardiac stimulation
4. increased renin release
Prototype substance that binds to β2 receptors
1. albuterol
2. activates Gs, increases cAMP
3. cardiac stimulation
4. smooth muscle relaxation
5. glycogenolysis
6. tremor
Prototype substance that bind to β3 receptors
1. -
2. activates Gs, increased cAMP
3. lipolysis
Prototype substance that bind to all dopamine receptors
dopamine
Prototype substance that binds to D1 receptors, and its properties
1. fenoldopam
2. activates Gs, increased cAMP
3. vasodilation
Prototype substance that binds to D2 receptors and its properties
1. -
2. activates Gi, decreases cAMP
3. inhibition of transmitter release
Toxicities of α-blockers (3)
1. tachycardia (all) as a reflex (nonselective more so than selective α1 blockers)
2. GI upset (phentolamine and phenoxybenzamine)
3. postural hypotensive response (some α1-selective agents)
Prototype full antagonist beta blocker
propranolol (stage fright, familial tremor)
Properties of pindolol
1. Nonselective β-blocker
2. partial β-agonist, used as antagonist
Prototype β1-selective antagonist
atenolol
Mechanism of action of carvedilol (2)
1. selective α1 antagonist
2. nonselective β antagonist
Mechanism of action of labetalol
1. selective α1 antagonist
2. nonselective β-antagonist
Mechanism of action of esmolol
1. parenterally given selective β1 antagonist (antiarrhythmic)
2. half life: 9 mins, 4.5 in <16 yo
Define parenteral
by other means than the alimentary tract (e.g., IV or intramuscular)
CNS effects of beta blockers (2)
1. sedation/lethargy
2. reduction of anxiety
Effects of β-blockers on the eyes
1. reduced secretion of aqueous humor
2. no significant effect on pupil or focus
Airway effects of beta blockers
marked bronchospasm in patients with airway disease, especially asthma
Cardiovascular effects of beta blockers (3)
1. slowed heart rate and AV conduction
2. reduced myocardial contractility
3. reduced blood pressure
GI and GU tract effects of β-blockers
little effect
Other effects of β-blockers (4)
1. reduced skeletal muscle tremor
2. reduced glucose release from the liver
3. reduced renin release from the kidney
4. reduced thyroid hormone effects
Uses of β-blockers (5)
1. Treatment of HTN, angina, and arrhythmias
2. reduce mortality and morbidity after MI or HF
3. oral β-blockers reduce familial tremor and stage fright
4. topical β-blockers to treat glaucoma
5. IV and oral β-blockers are useful in treatment of thyrotoxicosis
define: mortality
deathrate: the ratio of deaths in an area to the population of that area; expressed per 1000 per year
define: morbidity
the symptoms and/or disability resulting from a disease
Toxicities of β-blockers (4)
1. symptomatic cardiac depression (bradycardia, AV block, diminished cardiac output)
2. elevated blood glucose, lipids, and uric acid (chronic therapy)
3. severe bronchospasm (in asthma patients)
4. symptoms of hypoglycemia are masked (e.g., from an insulin overdose)
General properties of H1 blocking drugs
1. used in treatment of hayfever and urticaria, and a few IgE-mediated allergies.
2. toxicities reflect their effects on the ANS and CNS
General properties of H2 blocking drugs
1. used in treatment of acid-peptic disease, especially heartburn and peptic ulcer
2. toxicities (almost exclusively cimetidine) include:
a. inhibition of hepatic CYP450
b. antiandrogenic action
Examples of typical H2 blockers
1. cimetidine
2. famotidine
3. nizatidine
4. ranitidine
Examples of H3 blockers
1. clobenpropit
2. abt-239 (nootropic)
3. ciproxifan
4. ciprilisant
In which cells is serotonin stored?
1. platelets
2. neurons
3. enterochromaffin cells
4. cells in the gut
What is the main metabolite of serotonin?
1. hydroxyindoleacetic acid (HIAA)
2. is increased in urine of patients with carcinoid tumor
Effects of increased serotonin (and other substances) produced by carcinoid tumor include
1. diarrhea
2. bronchospasm
3. variable changes in blood pressure
4. flushing of the skin
Serotonin's effects are mediated by a large number of receptors, four of which are of clinical importance. Which ones?
1. 5-HT 1D (GPCR)
2. 5-HT2 (GPCR)
3. 5-HT3 (ion channel)
4. 5-HT4 (GPCR)
Clinical value of serotonin?
Serotonin itself is of no value as a drug, but two more selective agonists are used:
1. 5-HT 1D (e.g., sumatriptan) for migraine. Toxicities include coronary vasospasm
2. 5-HT4 agonists (e.g., tegaserod and cisapride) increase the release of ACh in the gut--> increased motility. Toxicities include tosade de pointes arrhythmia and diarrhea.
What is the standard treatment of migraine headache?
1. Simple analgesics, with sumatriptan or a similar "triptan" drug, or with ergotamine
2. Prophylaxis is partially successful using propranolol, amitriptyline, calcium channel blockers, and others
H1 receptor functions:
1. Gq coupled
2. increased IP3 and DAG, smooth muscle contraction (EXCEPT vessels), increased secretion
3. located in smooth muscle, gland cells, some nerve endings
H2 receptor functions:
1. Gs coupled
2. Increased cAMP, increased acid secretion, cardiac stimulation
3. located in parietal cells (stomach) and heart
H3 receptor functions
1. Gi coupled
2. decreased cAMP, modulates transmitter release
3. presynaptic autoreceptor in nerve endings
Histamine H1 antagonists can be divided into three groups. What are they? And what are their uses?
1. First generation, older - allergy, sleep aid, motion sickness, nausea of chemotherapy
2. First generation, newer - allergy
3. Second generation - allergy
Examples of first generation, older, H1 antagonists
1. Diphenhydramine
2. dimenhydrinate
Examples of first generation, newer, H1 antagonists
1. chlorpheniramine
2. cyclizine
Examples of second generation H1 antagonists
1. fexofenadine
2. loratadine
3. desloratadine
4. cetrizine
Toxicities of first generation, older antihistamines
1. strong alpha and muscarinic block
2. strongly sedative
Toxicities of first generation, newer antihistamines
1. much reduced sedative and ANS effects
Toxicities of second generation antihistamines
negligible CNS and ANS effects
Location and function of 5-HT1D receptors
1. Located in neurons, perhaps other places
2. mediates increased IP3, DAG
Location and function of 5-HT2 receptors
1. smooth muscle, other?
2. increased IP3 and DAG, smooth muscle contraction
Location and function of 5-HT3 receptors
1. chemoreceptors in the central and peripheral nervous system
2. nasuea and vomiting
Location and function of 5-HT4 receptors
1. neurons in the gut
2. increased GI motility
Some notes on 5-HT2 antagonists
5-HT2 antagonists, such as ketanserin, have been developed for the treatment of carcinoid tumors. However, older drugs with overlapping effects (e.g., phenoxybenzamine [5-HT+alpha blocker] or cyproheptadine [5-HT+H1 blocker]) appear to be just as useful. In addition ketanserin has been used to treat HTN in Europe.
Indications for 5-HT3 antagonists?
Extremely useful antinauseants and antiemetics for patients undergoing general anesthesia or chemotherapy
Examples of 5-HT3 antagonists
1. ondansetron
2. granisetron
3. tropisetron
4. dolasetron
Other serotonin related drugs
1. ergot alkaloids (complex partial agonists at 5-Ht, alpha, and dopamine receptors)
2. ergotamine (for migraine) - natural
3. ergonovine (oxytocic agent) - natural
4. bromocriptine (used in hyperprolactinemia and parkinsonism) - semisynthetic
5. pergolide (parkinsonism) - semisynthetic
6. LSD - semisynthetic. no meidcal use, powerful hallucinogenic
Effects and clinical uses of endogenous PGE2
1. vasodilation, bronchodilation, oxytocic
2. oxytocic (as dinoprostone)
Effects and clinical uses of PGE1 analog misoprostol
1. increased bicarbonate and mucus secretion in stomach
2. prevention of NSAID-induced ulcers
Effects and clinical uses of PGE1 analog alprostadil
1. smooth muscle relaxation
2. erectile dysfunction
Effects and clinical uses of endogenous PGF2alpha
1. vasoconstriction, bronchoconstriction, oxytocic
2. no clinical uses
Effects and clinical uses of PGF2alpha analog latanoprost
1. increased aqueous humor drainage
2. chronic glaucoma
Effects and clinical uses of prostacyclin (PGI2)
1. prevents platelet aggregation, vasodilation
2. severe pulmonary hypertension (as epoprostenol)
Effects and clinical uses of thromboxane (TXA2)
1. facilitates platelet aggregation
2. no clinical uses
Effects and clinical uses of LTB4
1. leukocyte chemotaxis
2. no clinical uses
Effects and clinical uses of LTC4
1. bronchoconstriction
2. no clinical uses
Effects and clinical uses of LTD4
1. bronchoconstriction
2. no clinical uses
Difference between NSAIDs and steroids?
Steroids are powerful anti-inflammatory agents, whereas NSAIDs have weaker anti-inflammatory actions, but are more suitable for long-term therapy because they are less toxic
Older NSAIDs include
1. aspirin
2. salicylate
Newer non-COX selective NSAIDs include
1. indomethacin
2. ibuprofen
3. diclofenac
4. indomethacin
5. ketorolac
6. naproxen
7. piroxicam
Selective COX-2 inhibitors include
1. celecoxib
2. valdecoxib
3. other members of this drug
Half-life and comments on aspirin
1. 15 minutes (but active metabolite has a half life of 3-15 hours)
2. Widely used for pain, fever, inflammation, and antiplatelet action
Half-life and comments on celecoxib
1. 11 hours
2. selective COX-2 inhibitor; a sulfonamide
Half-life and comments on diclofenac
1. 1.1 hours
2. general use in inflammation, pain
Half-life and comments on ibuprofen
1. 2 hours
2. general use in inflammation, pain
Half-life and comments on indomethacin
1. 4-5 hours
2. general use in inflammation, pain
Half-life and comments on ketorolac
1. 4-10 hours
2. parenteral use in pain; an opioid substitute
Half-life and comments on naproxen
1. 12 hours
2. longer action
Half-life and comments on piroxicam
1. 20 hours
2. longest action
Half-life and comments on salicylic acid
1. 3-15 hours
2. active metabolite of aspirin, zero order elimination at high concentrations
What sets aspirin apart from all other NSAIDs?
It irreversibly blocks COX, especially in platelets. Its antiplatelet effect is, therefore, longer in duration (24-48 hrs) than its anti-inflammatory and analgesic action (4-6 hours)
Toxicities of the NSAIDs include
1. GI upset
2. peptic ulcers with or without bleeding
3. renal damage
4. diversion of arachidonic acid metabolism to the leukotriene pathway (may rarely present with anaphylaxis)
Pros and cons of COX-2 selective inhibitors
1. Pro: lower risk of GI bleeding
2. Con. increased risk of cardiovascular events (stroke and MI)
Comments on acetaminophen
1. similar analgesic and antiypritic effect to aspirin
2. weak COX inhibitor
3. little or no anti-inflammatory effect
4. metabolites of acetaminophen may cause hepatic necrosis (overdose)
What is gout?
1. chronic metabolic disease characterized by an elevated body load of uric acid.
2. manifested by depositus of urate crystals in soft tissue and joints
3. in the joints, these crystals trigger episodes of acute painful arthritis
Which classes of drugs are used in the treatment of gout?
1. anti-inflammatory agents (mainstay of therapy)
2. uricosuric agents (increases excretion of uric acid)
2. inhibitor of uric acid synthesis (allopurinol)
Which anti-inflammatory drugs are are often used in the treatment of acute joint pain?
1. ibuprofen and indomethacin
2. colcichine (highly selective for gouty arthritis) - but is usually restricted to low-dose prophylactic therapy because of GI and hepatic toxicity
Which uricosuric agents are normally used in the treatment of gout?
1. probenecid
2. sulfinpyrazone

They inhibit the uric acid reabsorption transporter in the straight segment of proximal tubules
Mechanism of action of allopurinol
1. inhibits xanthine oxidase
2. reduces concentration of insoluble uric acid
3. increases concentration of uric acid precursors, xanthine and hypoxanthine
Clinical correlations on the treatment of gout
1. acute attacks require strong anti-inflammatory drugs, strong analgesics, or both
2. indomethacin and ketorolac are often used. ketorolac may be used parenterally
3. uricosurics may precipitate an immediate attack and should always be used in conjunction with colcichine or other agents initially
4. allopurinol's benefits are slow in onset and are suitable only for long-term therapy
In addition to the obvious cardiovascular events that hypertriglyceridemia may cause, it may also contribute to an incresed risk of
acute pancreatitis
Drugs used in hypercholesterolemia include
1. statins
2. resins
3. ezetimibe
4. niacin
5. fibrates
Mechanism of action of statins
(e.g., levostatin and atorvastatin)
1. inhibitors of cholesterol synthesis (HMG-CoA reductase inhbitors)
2. reduced cholesterol concentrtion results in increased LDL-binding receptors on liver cells, which increases clearance of LDL from blood
3. most statins only reduce cholesterol, but atorvastatin reduces both cholesterol and triglyceride levels
Mechanism of action of resins
(e.g., cholestyramine and colestipol)
1. nonabsorbable macromolecules, bind to and prevent reabsorption of cholesterol and bile acids
2. resins tend to increase triglycerides
Mechanism of action of exetimibe
new drug that reduces the absorption of cholesterol from the intestines, the exact mechanism is unknown
Mechanism of action of niacin
(nicotinic acid and vitamin b3)
1. the exact mechanism is not fully understood.
2. high doses cause a decrease in secretion of VLDL from the liver into blood, and thus decreased LDL.
3. is also effective in hypertriglyceridemia and may cause increase in HDL
Mechanism of action of the fibrates
(e.g., gemfibrozil and fenofibrate)
1. activate peroxisome proliferator-activator receptor alpha (PPAR-a)
2. increases synthesis of lipoprotein lipase and other enzymes
3. serum triglyceride concentrations are reduced, may be a reduction in LDL
Toxicities of statins
1. may cause serious skeletal muscle or liver damage
2. may interfere with myelination in growing infants.

These drugs are contraindicated in pregnancy
Toxicities of niacin
1. high doses causes a transient uncomfortable flushing with itching (may be reduced with aspirin)
2. may rarely cause elevation of:
a. liver enzymes
b. hyperuricemia
c. glucose intolerance
Toxicities of resins
1. unpleasant gritty taste
2. may cause GI discomfort (including bloating and constipation)
3. impaired fat-soluble vitamin and drug absorption

Should be avoided in hypertriglyceridemia
Toxicities of fibrates
1. nausea
2. skin rash
3. increased effect of antiplatelet drugs
4. increased risk of gall stones
Anticoagulants include
1. warfarin (oral; blocks reactivation of vitamin K epoxide; monitor with PT and INR)
2. heparin (parenterally, binds factor Xa and AT III)
Heparin is available in both HMW and LMW forms. The differences
1. HMW - binds Xa and AT III. AT III in this bound form is activated 1000-fold --> immediate clotting inhibition. The response is variable, must monitor.
b. LMW - inhibit factor Xa, but less effect on AT III. Response is predictable and so does not require monitoring.
Examples of LMW heparins
1. enoxaparin
2. dalteparin
Major clinical application of heparins and warfarin are
in the prevention and treatment of venous clotting, especially DVT
Toxicities of heparins and warfarin
1. bleeding
2. warfarin is a teratogen
3. heparin causes dose-dependent thrombocytopenia, but occasionally causes a more severe loss of platelets
Most important antiplatelet drugs include
1. aspirin
2. clopidogrel - blocks ADP receptor on platelets
3. ticlopidine - blocks ADP receptor on platelets
4. fiban drugs (eptifibatide and tirofiban) - IIb/IIIa blocker
5. abciximab - IIb/IIIa blocker
Most important clinical use of fibrinolytics
intravenous administration for pulmonary embolism and myocardial infarction
Most important fibrinolytic drugs
1. streptokinase - may cause allergic response, bleeding
2. alteplase (tPA), reteplase - bleeding
3. urokinase - bleeding
Anemia results from
1. inadequate dietary iron
2. inadequate vitamin intake
3. inadequate RBC production
4. accelerated RBC breakdown
What is anemia?
a deficiency of red blood cells (RBCs) or hemoglobin
For simple iron deficiency anemia, oral supplementation is effective. In what form is iron given?
1. ferrous sulfate
2. ferrous gluconate

ferric iron salts are poorly absorbed
Iron is irritating to the stomach, annd some patients cannot tolerate the doses required for rapid correction of severe deficiency. What options do these patients have?
1. iron dextran
2. iron sucrose complex
3. iron gluconate complex

all parenteral iron
Manifestations of iron poisoning include
1. vomiting
2. abdominal pain
3. diarrhea
3. necrotizing enteritis, followed by shock, lethargy, metabolic acidosis, death
How do you treat iron poisoning?
Prompt diagnosis and an iron chelator, such as deferoxamine
Growth factor used for anemia
1. epoetin (human recombinant erythropoietin)
2. increases RBC production
3. may cause HTN, thrombotic events
Growth factor used for granulocytopenia (F)
1. filgrastim (human recombinant GM-CSF)
2. increases neutrophil production
3. may cause bone pain
Growth factor used for granulocytopenia (S)
1. sargramostim (human recombinant G-CSF)
2. stimulates most marrow precursors, but increases neutrophil production the most
3. may cause fever, myalgia, peripheral and pulmonary edema
Growth factors used for thrombocytopenia
1. oprelvekin (human recombinant IL-11)
2. stimulates platelet production
3. may cause fatigue, headache, dizziness
Definition of asthma
asthma is a chronic inflammtory airway disease, with superimposed transient episodes of bronchoconstriction
Therapy of asthma consists of
1. bronchodilators - control of symptoms
2. anti-inflammatory drugs - prophylaxis
Bronchoconstriction in asthma is mediated mostly by
1. leukotrienes
2. other smooth muscle activators from mast cells and eosinophils in the airways
Bronchodilators consist of
1. beta2-selective agonists
2. muscarinic blockers
3. theophylline
Drug of choice in acute episodes of asthmatic bronchoconstriction
1. beta2-selective agonists, such as:
a. albuterol
b. metaproterenol
2. they are physiologic leukotriene antagonists (and others increasing cAMP)
3.
Toxicities of beta2-selective agonists
1. tremor
2. tachycardia

However, do not limit the drug during an acute attack: uncontrolled bronchoconstriction can be fatal
The most important muscarinic antagonists used in asthma are atropinelike. Their names are
1. ipratropium
2. tiotropium

*not as effective as SABAs
Toxicities of ipratropium and tiotropium
1. dry mouth
2. tachycardia (rarely)
3. cycloplegia (rarely)
Toxicities of theophylline include
1. GI disturbances
2. tremor
3. arrhythmias
4. convulsions
Third kind of bronchodilator
1. theophylline - is related to caffeine and theobromine, but more effective than either
2. inhibits phosphodiesterase -->increased levels of cAMP
3. not active when inhaled
Which anti-inflammatory drugs are used in asthma?
1. corticosteroids
2. mast cell stabilizers
3. leukotriene antagonists
Most important prophylactic agent for asthma
corticosteroids - given by metered-dose inhaler when possible
Which steroids are most commonly used in asthma?
1. beclomethasone
2. fluticasone
3. uhm... others...
How do corticosteroids help in asthma?
inhibit leukotriene and inflammatory cytokine production and release
Toxicities of inhaled corticosteroids include
1. temporary retardation of growth young children
2. candidal yeast infection of larynx
Mast cell stabilizers used in asthma include
1. cromolyn
2. nedocromil

highly insoluble compounds that can be administered by metered-dose inhalation with practically no systemic toxicity
How do mast cell stabilizers work?
Probably by inhibiting mast cell release of inflammatory and allergic mediators, including leukotrienes, cytokines, and histamine
Leukotriene receptor antagonists used in asthma
1. zafirlukast
2. montelukast

orally active (advantageous in young children and others who can't use inhalers)
Downside of leukotriene receptor antagonists?
1. not as effective as corticosteroids
2. no value in acute bronchospasm
Physostigmine and bethanechol in small doses have similar effects on all of the following
1. salivary glands
2. ureteral tone
3. sweat glands
4. gastric secretion
Insecticidal activity of parathion (4)
1. indirectly acts on acetylcholinesterase; ingested-> oxidized to paraoxon
2. paraoxon is the most potent acetylcholinesterase inhibitory insecticide used
3. about 70% as potent as sarin
4. easily absorbed through skin
Parathion has all of the following characteristics (4)
1. less persistent in the environment than DDT
2. more toxic to humans than malathion
3. very lipid soluble and easily absorbed through skin and lungs
4. its toxicity, if treated early, may be partially reversed by pralidoxime
In the treatment of myasthenia gravis, the best agent for distinguishing between myasthenic crisis (insufficient therapy) and cholinergic crisis (excessive therapy) is
1. edrophonium
2. in myasthenia gravis the patient's muscle strength will increase due to more availible acetylcholine
3. in a cholinergic crisis, the weakness will worsen due to induction of a depolarizing block
The cause of death in nerve gas poisoning is usually
asphyxiation