• 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

1/506

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;

506 Cards in this Set

  • Front
  • Back
What are the three components of the ANS? What can they be divided into? What are the neurotransmitters? What are the major regulators (if applicable?)
1. The sympathetic system ("fight or flight"). The main neurotransmitters are adrenaline and noradrenaline.
2. The parasympathetic system ("rest and digest"). The main neurotransmitter is acetylcholine.
3. The enteric system--a collection of nerve fibers innervating the GI tract, pancreas, and gall bladder--specifically, their smooth muscle, secretory cells, endocrine cells, and blood vessels. The enteric nervous system is regulated by a balance between the sympathetic and parasympathetic systems.
Major regulators of the enteric nervous system are:
a. Substance P
b. VIP
c. 5-hydroxytryptamine
In the ANS, how are efferent messages carried down? To where?
Efferent messages from the autonomic nervous system get carried down a pre-ganglionic neuron, which extends from the brain, down the spinal cord, and synapses on a ganglion. Then, a post-ganglionic neuron transmits the information from the ganglion to the target organ.
How do the ganglia in the parasympathetic and sypathetic systems differ?
In the parasympathetic system, the ganglia tend to be close to the target organ.

In the sympathetic system, the ganglia form two chains on either side of the spinal cord, far from their target organs.
What is the established structure of regulation of the ANS? (Integrates and transmits afferent information that feeds into the ANS)
Cerebral cortex (frontal lobe)

Limbic lobe (emotional input)

Hypothalamus (overall integration of ANS - "the boss")

Reticular formation of brain stem (regulation of pupil size, respiration, heart, BP, swallowing, etc)

Spinal cord (urination, defecation, erection, ejaculation)
Which branch of the NS dominates the arteriole?
Sympathetic (adrenergic)
Which branch of the NS dominates the veins?
Symp (adrenergic)
Which branch of the NS dominates the heart?
Parasymp
Which branch of the NS dominates the radial muscle of he iris?
Symp (alpha andregenic)
Which branch of the NS dominates the sphincter of the iris
Parasymp
Which branch of the NS dominates the ciliary muscle of the eye?
Parasymp
Which branch of the NS dominates Muler's muscle
Symp (a-andregenic)
Which branch of the NS dominates the bronchial smooth muscle?
Parasymp
Which branch of the NS dominates the GI tract?
Parasymp
Which branch of the NS dominates the urinary tract?
Parasymp
Which branch of the NS dominates the salivary glands?
Parasymp
Which branch of the NS dominates the eccrine sweat glands?
Symp (cholinergeic)
Which branch of the NS dominates the apocrine sweat glands?
Symp (a-andregenic)
Which branch of the NS dominates the pilomotor muscles?
Symp (a-andregenic)
What are the parasympathetic (1) and sympathetic (2) actions for the eye?
1. Contraction of pupil for near vision, increased secretion from lacrimal glands

2.Dilation of pupil for ar vsion
What are the parasympathetic (1) and sympathetic (2) actions for the trachea and bronchioles?
1. Constiction, increased secretion

2. Dilation
What are the parasympathetic (1) and sympathetic (2) actions for the adrenal medulla?
1. none

2. Secretion of epinephrine and norepinephrine
What are the parasympathetic (1) and sympathetic (2) actions for the kidney?
1. none

2. Secretion of renin
What are the parasympathetic (1) and sympathetic (2) actions for the ureters and blader?
1. Contraction of detrusor, relaxation of trigone a sphincter

2. Relaxation of detrusor, contraction of trigone and sphincter
What are the parasympathetic (1) and sympathetic (2) actions for the male genitalia?
1. Erection

2. Ejaculation
What are the parasympathetic (1) and sympathetic (2) actions for the female genitalia?
1. None

2. Relaxation of the uterus
What are the parasympathetic (1) and sympathetic (2) actions for the lacrimal glands?
1. Tears

2. None
What are the parasympathetic (1) and sympathetic (2) actions for the salivary glands?
1. Thick, viscous secretion

2. Copious, watery secretion
What are the parasympathetic (1) and sympathetic (2) actions for the heart?
1. Inreased rate and contractility

2. Decreased rate and contractility
What are the parasympathetic (1) and sympathetic (2) actions for the GI tract?
1. Increased motility and muscle one. Increased secretion.

2. Decreased motility and muscle tone
What are the parasympathetic (1) and sympathetic (2) actions for the blood vessels in skeletal muscle?
1. Dilation

2. None
What are the parasympathetic (1) and sympathetic (2) actions for the blood vessels in skin, mucous membranes, and splanchnic areas?
1. None

2. Consticton
What are the organs that only have sympathetic innveration?
Adrenal medula
Kidneys
Pilomotor muscles
Sweat glands

BP also mainly controlled by symp innervatiion.
What are andregenic receptors? What are cholinergenic receptors? Both of these receptors can be involved in the sympathetic NS.
1. Receptors on which epinephrine and norepinephrine act

2. Receptors on which ACh acts
The parasympathetic NS only utlizes what type of receptors?
Cholinergenic
What are the steps in cholinergenic neurotransmission?
1. Ach synth
2. Storage of Ach
3. Release of Ach
4. Receptor binding
5. Degreadation of Ach
6. Choline reuptake
Describe ACh synthesis. What drug can block this step?
1. i. A Na/choline co-transporter brings choline (a charged molecule, derived from a phospholipid called lecithin) from the ECF into the cell. Choline cannot diffuse through the membrane on its own because it has a positively charged N group. This is a slow process, and the rate limiting step in choline synthesis.

Can be blocked by hemichollinium.

Choline reacts with acetyl CoA to form ACh. This reaction is catalyzed by acetyltransferase.
Describe storage of ACh. What drug can block this step?
a proton-driven active transport system moves choline in vesicles for storage, along with ATP and proteoglycan.

Vesamicol blocks the uptake of ACH into the vesicles
Describe release of Ach. What drug can block this step?
an action potential triggers the opening of voltage-gated Ca channels in the pre-synaptic membrane. Ca floods the cell and triggers the ACH vesicles to fuse with the cell membrane and release ACH into the synaptic cleft.

i. The botulinum toxin blocks ACH release
ii. Black widow spider venom causes uncontrolled ACH release
Describe receptor binding. What drug can block this step?
ACH diffuses across the synaptic cleft and binds to a receptor on the post-synaptic cell. There are two different classes of receptors that ACH can bind to:
i. Muscarinic
ii. Nicotinic
Describe degradation of ACh. What drug can block this step?
acetylcholine esterase cleaves ACH into choline and acetate.
Butyrylcholine esterase, found in the plasma, does not play a significant role in signal termination.
Describe choline reuptake?
a Na-coupled transporter moves choline back into the pre-synaptic cell, for use in forming new ACH. ACH can also be recycled directly, via ACH autoreceptor proteins on the membrane of the presynaptic cell.
What is the process of neurotransmitter release? What triggers?
1. Docking

2. Priming

3. Fusion with plasma membrane

Fusion of the vesicle with the plasma membrane is triggered by calcium.
What are the major proteins involved in the docking and fusion process?
1. Synaptobrevin and synaptotagmin

2. SNAP-25 and syntaxin
Describe the docking and fusion process.
1. Synaptobrevin tangles with SNAP-25 and syntaxin

2. Fusion occurs when Ca binds to synaptotamin, on the vesicular membrane, signaling it to fuse with the plasma membrane.
How does botulinum work? What is it? What are the subgroups?
Botulinum is a proteaste that inhibits ACh release by degrading these proteins.

A and E degrade SNAP-25
C1 degrades syntaxin
B, D, F, and G degrade synaptobrevin
How is ACh is degraded?
by ACh esterase into choline and acetate.
What is the structure of ACh esterase? Where does cleavage occur? How does it work out?
1. Anionic binding site

2. Eseratic binding site

Cleavage happens first at the anionic site, then at the esteratic site, releasing choline and acetate.
Blocking ACH esterase will initially increase ACH concentration in the synaptic cleft. However, eventually, ACH concentration will go back down, since a feedback loop will signal the presynaptic neuron to stop synthesizing ACH.
What is BChE, and where is it found?
similar to ACH esterase, but is present in higher concentrations in the circulation, and in lower concentrations in neurons and the NMJ.
What are the two types of cholinergenic receptors?
1. Nicotinic (ligand-gated)

2. Muscarinic (GPCRs)
Where are nicotinic receptors? What is their structure? How do they function? What are they inhibited by?
ligand-gated ion channels, composed of 5 subunits, that open when ACH binds to the α subunit and allow Na into the cell and K out of the cell, resulting in depolarization.

Nicotinic receptors are present in the CNS, autonomic ganglia, adrenal medulla, and NMJ.
Nicotinic receptors in neurons are called Nn receptors. They are inhibited by hexamethonium.

Nicotinic receptors in the NMJ are called Nm receptors. They are inhibited by tubocurarine.
What are the effects of nicotine?
1. Blood pressure increase (mediated by the sympathetic system)

2. Increase in peristalsis (mediated by the parasympathetic system)

3. NA and adrenalin release (sympathetic system, due to effects of nicotine on adrenal gland)
What characterizes the odd-numbered M receptors?
excitatory. ACH binding triggers increased concentrations of IP3 and DAG as second messengers.
Where are the locations of the M1 receptors?
CNS and autonomic ganglia
Where are the locations of the M3 receptors?
smooth muscle of the colon, bladder, and bronchi (triggers contraction)
Where are the locations of the M5 receptors?
nerves in the CNS (participate in positive, presynaptic feedback in the CNS)
wher are the locations of the M4 receptors?
Nerves in the CNS
What is the general action of the M3 receptor?
Trigger contraction of smooth muscle.

GPCR that activates PLC to cleave PIP2 in the cell membrane into IP3 and DAG.

IP3 triggers the release of Ca from the SR. Calcium then activates myosin light chain kinase which phosphorylates myosin and triggers muscle contraction.
Go through the process of muscle contraction.
Go through the process of muscle contraction.
What effect do M3 channels have on endothelial cells? Why? How?
smooth muscle relaxation, rather than smooth muscle contraction. This is due to the presence of nitric oxide.
1. In endothelial cells, two mechanisms provide for an increase in intracellular Ca:
A. Shear (mechanical) stress on the cells
B. Binding of ACH to the M3 channels, as previously described (ad nauseum)
2. Then, Ca activates an enzyme called nitric oxide synthase, that catalyzes the degradation of L-arg into NO and citrulline
3. NO diffuses into the smooth muscle cells, where it activates guanylate cyclase
4. Guanylate cyclase converts GTP to cGMP, stimulating smooth muscle relaxation
Describe the process of smooth muscle relaxation
What are NANCs? What is their purpose?
Another source of NO to stimulate blood vessel dilatation are NANCs (non-adrenergic non-cholinergic neurons), which secrete NO directly into the smooth muscle cells.
What are the two mechanisms in which NO concentration is increased in the penis?
1. M3 activation, leading to activation of NO synthase, and NO production from L-arg
2. NANCs
In terms of NO release in the penis, what does increased NO concentration result in? How is it inhibited?
Increased NO concentration results in increased levels of cGMP, which causes smooth muscle relaxation.
This process can be stopped by an enzyme called phosphodiesterase 5, which degrades cGMP into GMP.
Viagra works by inhibiting phosphodiesterase 5. This prevents the degradation of cGMP and increases arteriolar relaxation, causing prolonged erection
What two types of nicotinic receptors exists? What is the difference?
1. Nn
2. Nm
Both are ligand gated Na/K channels, letting Na out and K into the cell. They are composed of five subunits, the first four of which are the same. However, the Nn channels have a γ subunit, while the Nm channels have an ε subunit.
Describe Nm receptors and their role in muscle contraction.
The entry of Na into the muscle cell causes the membrane to depolarize.
Entry of Na into the cell triggers the release of Ca from the SR, activating contraction. Then, a voltage-gated receptor called the DHPR receptor, and binds to a Ca channel on the SR called the ryanodine receptor. The ryanodine receptor then opens, flooding the cell with Ca, which triggers muscle contraction.
What are the co-transmitters of cholinergenic sypases?
1. ATP--activates a purinergic receptor called the P2Y receptor at the edge of the cholinergic nerve, and inhibits more ACH release from the presynaptic nerve
2. VIP--also released with ACH, VIP causes relaxation of smooth muscle
i. In the salivary glands, VIP relaxes the blood vessels while ACH increases the secretion of saliva
What are the general efects of cholinergenic muscarinic agonists?
1. On internal organs--the same effects sympathetic innervation would have, i.e. increased muscle contractility and secretion from glands

2. Blood vessels--relaxation and vasodilatation, due to activation of NO synthase via the M3 channels

3. Sweat glands--increased secretion
What are the effects of specific muscarinic agonists on M1 of the ELC (in stomach)?
Increased histamine secretion, causes increased acid secretion.
What are the effects of specific muscarinic agonists on M3 in the eye?
Contracts the pupil for near vision, increases secretion from the lacrimal glands
What are the effects of specific muscarinic agonists on M3 in the salivary glands?
Increased secretion
What are the effects of specific muscarinic agonists on M3 in the bronchiolar SM?
Contraction
What are the effects of specific muscarinic agonists on M3 in the staomch?
Contraction of SM, increased gastrin and acid secretion (Which are also increased indrectly, by histamine secreted from ECL cels, which bin to H2 receptors on parietal cells)
What are the effects of specific muscarinic agonists on M3 in the colon?
Increased peristalsis
What are the effects of specific muscarinic agonists on M3 in bladder?
Contraction of detrusor to release uine.

Simultaneously, NO relaxes the urethra.
What are the effects of M3 agonists on the eye?
1. Iris--contracts, to shrink pupil (miosis)
2. Ciliary muscle--contracts to relax lens and enable near vision
3. Lacrimal glands--increased secretion
What are M3 agonsts used for in the eye?
To enhance near vision and treat glaucoma.
What are direct M3 muscarinic agonists? What are they used for?
Muscarinic agonists mimic the effects of ACH on muscarinic receptors. They are used to treat GI tract and genitourinary system disorders. They are also used for post-operative atony and neurogenic bladders.
What are examples of M3 agonist drugs? What are they used for?
1. Bethanechol -- promote Gi and urinary tract motility

2. Pilocarpine -- mitotic agent and sialogogue in treatment of xerostomia

3. Methacholine -- diagnosis of asthma
What is Bethanechol?
a derivative of acetylcholine, in which the acetate is replaced by carbamate, and the choline is methylated. This methylation protects bethanecol from degradation by ACH esterase.
What s pilocarpine?
has a similar structure to muscarine.

Because pilocarpine is lipophilic, it can cross the BB. It's primary use is in treating eye disorders, such as glaucoma. It produces rapid miosis and contraction of the ciliary muscle. Because it stimulates secretion from the lacrimal gland, it is very useful in lowering intraocular pressure. Because it is not specific to a particular type of muscarinic receptor, it is given topically, in small doses.
What is methacholine?
has the same structure as ACH, but with an additional methyl group, to prevent degradation by ACH esterase.
What are the systemic side effects of muscarinic antagonists?
1. Dry mouth, swallowing difficulties
2. Blurred vision
3. Urinary retention
4. Constipation
5. Dry, reddish skin (dry, due to decreased secretions, red, due to unexplained vasodilation of blood vessels in the skin)
6. Increased heart rate
7. Reduced gastric acid secretion
8. "Atropine fever," increased body temperature, due to increased metabolic rate, especially in children
9. Hallucination and confusion
What are the clinical uses for muscarinic antagonists?
1. Diagnostic purposes in eye exams (Atropine/Atrospan)
2. GI disorders such as IBS and colon spasms (Atropine/Spasmalgin)
3. Asthma, to relax bronchiolar smooth muscle (Ipratropium bromide/Aerovent)
4. Bradycardia, especially during surgery (Atropine)
5. Incontinence (tolterodine/Dertozitol)
6. Nerve gas toxicity (Atropine, with TMB-4, or Toxogonine, Atropine, and Benactizine; "TAB")
What are examples of direct muscarinic antagonists?
1. Atropine
2. Scopolamine
How do atropine and scopolamine work> What so they do?
In the eye, atropine relaxes the sphincter muscle of the iris to causes mydriasis (dilated pupil). The ciliary muscles contract, flattening the lens and enhancing far vision. Atropine should NOT be given to patients with glaucoma.
In the GI tract, atropine reduces motility and is used as an antispasmodic. It also can treat hypermotility of the bladder and excess secretions, especially in the salivary glands.
In the heart, atropine causes:
-bradycardia at low doses
-increased heart rate at higher doses
Another direct muscarinic antagonist, scopolamine, produces similar effects to atropine, but has a longer duration, and a greater impact on the CNS. It is used to treat motion sickness.
What are examples of indirect cholinergic agonists? What do they do?
Inhibitors of ACHE and BCHE
ACH esterase (and BCH esterase) prevent degradation of ACH in the synaptic cleft, thereby increasing its concentration.

Endrophonium
Physostigmine
Pyridostigine

ACHE inhibitors bind to ACH esterase and inactivate it. Their action is reversible. The half life of the drug depends on its affinity for ACHE.
What do indirect cholinergic agonist affect?
1. Sympathetic and parasympathetic ganglia
2. Target organs innervated by parasympathetic fibers or sympathetic cholinergic fibers
3. The NMJ
4. The CNS (if the agent crosses the BB)
What are the parasympathetic effects of indirect cholinergenic agonists?
Decreased heart rate
Bronchiolar contraction
Increased colon motility and secretion
Increased activity and secretion in the bladder
What are the sympathetic effects of indrect cholinergenic agoniss?
sweating
What are the NMJ efects of indirect cholinergenic agonists?
Muscle contraction
How does diagnosis and treatment of myasthenia gravis utilize indirect cholinergeic agonists?
Endrophonium, a short-acting indirect cholinergic agonist, is used for diagnosis

Pyridostigmine, a long-acting indirect cholinergic agonist, is used for treatment
What are organophosphate toxins?
irreversible ACHE inhibitors of ACHE, due to their extremely high affinity for ACHE. Organophosphate toxins are present in nerve gases such as Soman, Sarin, Tabun, and Vx. Nerve gas binds to the esteratic and anionic sites of ACHE, incapacitating its activity.
What are the effects of nerve gas? Sympathetic, Sympthatic, Skeletal Muscle, CNS effects.
1. Parasympathetic effects--
i. Decreased heart rate
ii. Contraction of bronchiolar SM
iii. Increase in saliva secretion
iv. Diarrhea
v. Increased urination
2. Sympathetic effects
i. Sweating
ii. Hypotension
3. Skeletal muscle effects
i. Paralysis, due to desensitization of nicotinic receptors in the NMJ
4. CNS effects (because it can cross the BB)
i. Spasms
ii. Loss of concentration
iii. Inhibition of the respiratory system
How can defense of nerve gas be made?
Organophosphate toxins are present in nerve gases such as Soman, Sarin, Tabun, and Vx. Nerve gas binds to the esteratic and anionic sites of ACHE, incapacitating its activity.
These compounds will fall off the ACHE if another compound is injected that binds to the nerve gas with a higher affinity than ACHE (ex: oxime). Such compounds are given to soldiers in injections, combined with atropine, to combat the cholinergic effects, as a defense against nerve gas. The combined injection is called TMB-4. Quick IV administration is need in order to be effective. High doses are needed, but an overdose will inhibit ACHE, due to the atropine included in the injection.
What are examples of nicotinic receptor agonists?
1. Nicotine (present in tobacco)
2. Lobeline (present in tobacco)
3. DMPP (does not cross the BB)
What are the effects of nicotinic agonists?
1. Parasympathetic
i. Contracts colon, bladder, and bronchioles
2. Sympathetic
i. Contracts blood vessels
ii. Tachycardia
3. Skeletal muscle
i. Increased contraction
4. CNS
i. Increased alertness
ii. Stimulation of the respiratory system
What does nicotinic toxicity result in?
1. Hypertension
2. Arrhythmias
3. Convulsions
4. Respiratory interruptions

Nicotinic receptor drugs work on both the sympathetic and parasympathetic systems, and have widespread effects.
What are specific nicotinic agonists?
Specific nicotinic antagonists (that are not in clinical use) include:
1. Hexamethonium
2. Trimethaphan
Their effects are exactly opposite to the effects of nicotinic agonists.
What are NMJ blockers? Examples?
NMJ blockers block ACH from acting on nicotinic receptors in the NMJ. They are structural analogues of ACH, and can be either antagonists or agonists.

1. Tuocurarine
2. Sucinylcholine
What is tubocurarine?
NMJ blocker that competitively inhibits the binding of ACH to nicotinic receptors on the muscle cell. It also triggers the release of histamine from mast cells (which in turn can cause bronchiolar constriction). Tubocurarine may be used in surgery to relax skeletal muscle.
What is succinylcholine?
depolarizing blocker, which will bind to the nicotinic receptors and cause a transient muscle twitch, but then not fall off, prevent further action potentials from being transmitted to the muscle.
Eventually, in the presence of a depolarizing blocker, the receptors will become desensitized and the cell will gradually repolarize, producing flaccid paralysis.
What are clinical uses of NMJ blockers?
1. Easing intubations
2. Relaxing skeletal muscles during surgery
3. Easing invasive tests
4. During electroshock therapy to depressive tests, to ease convulsions
What are examples of AChE(irreversible)?
ISOFLURATE
Echothiophate
Parathion

Chronic treatment of open-angle glaucoma
Nerve gas
Atropine used to treat organophosphate poisoning
What are examples of AChE reactivation drugs? What do they do?
Pralidoxime

reactivate inhibited acetylcholinesterase before aging occurs
What are examples of ganglionic blockers? What are they used for?
NICOTINE
HEXAMETHONIUM
Mecamylamine
Trimethaphan

Trimethaphan- emergency lowering of BP
What are examples of antimuscarinic drugs? For what are they used?
ATROPINE
Ipratropium
Scopolamine
Propantheline
Benztropine

- Atropine- mydriatic + cycloplegic effects
- antidote for organophosphate overdose
- Scopalamine- prevention of motion sickness
- Ipratropium- second-line therapy in Asthma, COPD
- Benztropine- treatment of Parkinson’s disease
-Toxic effects- “Dry as a bone, Hot as a hare, Red as a beet, Blind as a bat, Mad as a hatter”- Dry mouth, Inhibition of sweating, Tachycardia and cutaneous vasodilation, Blurring of vision, Hallucinations and delirium
What are NMJ blockers (nondepolarizing) used for?
adjuvant drugs for anesthesia
actions can be reversed by edrophonium or neostigmine

Drugs that end in "curonium"
What are NMJ blockers (depolarizing)? What are they used for?
Succinylcholine
Decamethonium

rapid hydrolosis by plasma cholinersterase
Phase I blockade- continuous depolarization; Phase II blockade- neuromuscular blockade (muscle resistant to further depolarization
Dantrolene- treats malignant hyperthermia caused by administration of halothane/succinylcholine
- blocks release of Ca++ from sarcoplasmic reticulum
What are the neurotransmitters that act in the sympathetic NS?
adrenaline (technically, a hormone)and noradrenaline (a neurotransmitter). ACH still plays a role in the sympathetic system because it is the neurotransmitter that acts on the sympathetic ganglia. It also stimulates secretion of adrenaline from the adrenal medulla. Dopamine is the neurotransmitter that acts on renal vascular smooth muscle.
How is noradrenaline converted to adrenaline?
Noradrenaline is converted to adrenaline mainly in the adrenal medulla, by the addition of a methyl group to the amine, catalyzed by N-methyl transferase.
What is the process of the adrenergic synapse?
1. Dopamine synthesis
2. NA synth and storage
3. NA secreted from vescles
4. NA can bind to receptors, dffuse into blood, or undergo reuptake then enter the post-ganglionic cell where it is degraded.
Describe dopamine synthesis in the adrenergic synapse? What is the rate limiting step?
i. Tyrosine (from which all catecholamines are derived) enters the cell via a Na co-transporter
ii. Tyrosine hydroxylase converts tyrosine to DOPA in the cytoplasm (rate limiting step)
iii. DOPA is decarboxylated to form dopamine (DA)
Describe NA synth and storage in the adreneric synaspse.What is it inhibited by?
i. DA is transported into the vesicles by an amine transporter system (inhibited by reserpine)
ii. In the vesicles, DA is converted into NA by dopamine hydroxylase
Describe secretion of NA from vesicles in the adrenergic synapse/ What is it inhibited by?
3. NA is secreted from vesicles, in response to a calcium influx (inhibited by guanethidine and bretylium)
What happens when NA is finally inside the adrenergic synaptic cleft Is there any inhibition? Degradation? How?
i. Bind to receptors and continue the signal transduction
ii. Diffuse into the blood
iii. Undergo reuptake by adrenal receptors on the pre-synaptic cell (inhibited by cocaine and imipramine)
1. Once back inside the cell, it can either be degraded by monoamine oxidase (MAO), an enzyme present on the outer membrane of the mitochondria, or transported back into the vesicles.
Inside the cell, NA has no effect. Its only activity comes from binding to adrenoreceptors.
4. Enter the post-ganglionic cell, where it is degraded by a cytoplasmic enzyme called COMT.
Describe the synthesis of NA. What happens in the brain? In storage vesicles?
Tyrosine (or phenylalanine) is converted to DOPA by the addition of a hydroxyl group to the benzene ring, via tyrosine hydroxylase.
DOPA is converted to DA by removal of the carboxyl group, via DOPA decarboxylase.
In the brain, the pathway ends with the step, leaving DA as the end product.
In the storage vesicles, DA is converted to NA by the addition of a hydroxyl group, via DA β-hydroxylase
Describe the synthesis of NA. What happens in the adrenal glands? In dopaminergic neurons?
In the adrenal glands, phenylethanolamine reacts with NA to form adrenaline, via the addition of a methyl group. This reaction is catalyzed by phenylethanolamine N-methyl transferase.

In dopaminergic neurons, tyrosine is converted to DA, and does not continue on to become NA.
What regulates adrenaline storage? How is it transported? How is it formed?
regulated by alterations in pH. The pH in the cytoplasm is 7.1. the vesicles are more acidic, with a pH of 5.5, which is maintained by an ATP-driven proton pump. To get into the vesicles, DA (or NA) is transported by a non-specific catecholamine/proton exchanger. Therefore, the entry of DA into the vesicles is accompanied by a loss of protons, hence, a rise in pH.
At a concentration of .3-1 M, NA combines with 4 ATPs and chromogranin A to form a complex. However, as the pH in the vesicle rises, driven by an influx of NA, this complex dissociates, freeing the NA to be active, upon release.
What are MAO and COMT? VMA?
In the pre or post-synaptic cell, NA is degraded either by MAO or COMT
MAO is an enzyme found on the outer mitochondrial membrane in adrenergic nerves and epithelial cells of the liver and colon
COMT is found in the cytoplasm of post-synaptic cells and in epithelial cells of the liver and colon
NA should not be given per os, because it will be degraded in the liver by COMT before it reaches its target cells.
The final product of NA metabolism is VMA (vanilyl mandelic acid). Because VMA is excreted by the kidneys, it can be detected in the urine, and is used for diagnostic purposes.
What are agents that decrease NA concentration in the adregenic synaptic cleft? For what are they use? How do they work?
1. α-methyltyrosine--inhibits tyrosine hydroxylase, which ultimately inhibits DA and NA formation. Not in clinical use.
2. Reserpine--inhibits the storage of NA in vesicles by inhibiting the NA transporter. This results in the accumulation of NA in the cytoplasm, where it is metabolized by MAO. Reserpine was previously used to treat hypertension, but was discontinued because it causes depression.
What are agents that increase NA concentration in the andergenic synaptic cleft? For what are they used? How do they work?
1. Cocaine--inhibits the reuptake of NA. It also acts as a local anesthetic, by blocking Na channels. Cocaine has a short half life, and is degraded by BuCHE.
2. Indirect sympatomimetic agents--increase the release of catecholamines from vesicles and competitively inhibit their reuptake.
What are examples of indirect synaptomimetic agents?
i. Amphetamine, methamphetamine
ii. MDMA (street name, ecstasy)
iii. Ephedrine-
iv. Tyramine-
v. Ritalin (methylphenidate)
What are amphetamines used for?
used to treat ADHD, narcolepsy, and obesity. Chronic abuse causes paranoia, hallucination, and other psychiatric symptom.
How does MDMA work?
uses the Na-coupled tyr transporter to get into the cell, and from there, enters the vesicles. In the vesicles, MDMA binds to protons, thereby raising the pH. This allows NA to dissociate from the ATP/chromogranin complex, leave the vesicles via the H+ coupled NA transporter, and exit the cell through non-voltage-gated NA channels. Thus, in the presence of MDMA, NA can exit the cell without being triggered by an action potential.
How does ephedrine work? What is it used for?
a partial adrenergic agonist that has a long half life, because it is a poor substrate for MAO and COMT. Can cross the BBB. Stimulates the CNS and raises heart rate and blood pressure. Sometimes used for asthma and runny noses (because it causes blood vessels to constrict).
What is tyramine?
a normal byproduct of tyrosine metabolism, normally oxidized by MAO. Not widely used clinically, and found in fermented foods.
What is ritalin? How does it ork?
)--similar to amphetamines, Ritalin increases the concentration of DA and NA. It is given to hyperactive children to increase focus and concentration, and is sometimes abused as a stimulating drug.
(Amphetamine, ephedrine, tyramine, and Ritalin all cross the BBB.)
What are the effects of agents that increase catecholamines? Talk about the effects on the CNS, in the perphery, and in the cases of OD.
1. In the CNS
i. Wakefulness, concentration, loss of appetite, mental and motor stimulation
2. In the periphery
i. Tachycardia
ii. Increase in blood pressure
3. In cases of an OD
i. Cardiac arrhythmias
ii. Critical hypertension
iii. CNS convulsions
What are the metabolites of catecholamines?
COMT degrades compounds that have two hydroxyl groups.
MAO degrades compounds that have a methyl group on the alpha position
Tyramine, amphetamine, and ephedrine are all degraded by MAO, but not by COMT
Why should patients on MAO inhibitors avoid tyramine in their diets?
Foods rich in tyramine, such as aged cheese and red wine should be avoided by patients on MAO inhibitors because tyramine is degraded only by MAO, and not COMT. Therefore, tyramine will build up, and lead to hypertension and tachycardia.
Where are the a1 adrenergic receptors work? What are their mechanisms?
Smooth muscle in blood vessels

Increases IP3 and DAG, which raises Ca concentrations and contractility
Where are the a2 adrenergic receptors work? What are their mechanisms?
Adrenergic nerves, vasomotor center

Decreases cAMP, which opens K channels, hyperpolarizing the cell
Where are the b1 adrenergic receptors work? What are their mechanisms?
Cardiac muscle

Increases cAMP, leading to an increase in Ca and contractility
Where are the b2 adrenergic receptors work? What are their mechanisms?
Smooth muscle on internal organs

Increases cAMP
Where are the b3 adrenergic receptors work? What are their mechanisms?
Fat cells

Increases cAMP
What happens when NA binds to an a1 receptor in smooth muscle?
When NA binds to α1 receptors in smooth muscle cells in blood vessels, it increases intracellular calcium, resulting in contraction.

ATP acts as a co-transporter for the α1 receptor.
What are the effects of a1 receptors?
1. Raise blood pressure, due to contraction of smooth muscle on most blood vessels
2. Contract the bladder sphincter to retain urine
3. Dilate pupils (mydriasis) by contracting radial muscle of iris
4. Contract pilomotor muscles on hair follicles
What are the effects of a2 receptors? Example of an agonist? Side effects?
1. Inhibit noradrenaline release from presynaptic cells
2. Decrease sympathetic activity
3. Cause contraction of some blood vessels
And example of an α2 agonists is Clonidine (α-methyldopa), which is used to treat hypertension and drug withdrawal (for addicts). Side effects include platelet aggregation, inhibition of fat decomposition, inhibition of insulin release, and sweating.
What are the effects of b1 receptors?
1. Increase heart rate and contractility
2. Increase renin release in the kidneys
What are the effects of b2 receptors?
1. Stimulate relaxation of the smooth muscle in the blood vessels, bladder, bronchi, and uterus
2. Increase glycogenolysis and K+ uptake in smooth muscle
i. Overactivity may result in a tremor
3. Stimulate glycogenolysis and gluconeogenesis in the liver
4. Stimulate insulin secretion
What are the effects of b3 rceptors?
Stimulate fat decomposition in adipose cells
How is ATP used as a co-transmitter with NA?
1. Activates ligand-gated Na channels called P2x channels in blood vessel smooth muscle. This activation produces the depolarization that triggers the influx of Ca.
2. Activates channels called P2y channels in adrenergic nerves that inhibit the release of noradrenaline
How is Nueropeptide Y used as a c-transmitter with NA?
1. Acts as an additional neurotransmitter in sympathetic neurons innervating blood vessels
2. Activates receptors in adrenergic neurons to inhibit the release of noradrenaline
3. Activates postsynaptic receptors on blood vessels that cause prolonged contraction
What are the different responses of ATP, NA, and NPY in blood vessel contraction?
ATP has a short-term effect
Noradrenaline has a medium range effect
NPY has a long-term effect
Wat are the effects of NA on the b1 receptor in the heart? SA node vs pacemaker.
In the SA node, noradrenaline has the opposite effects to ACH; it binds to a β1 receptor which leads to increased cAMP-->PKA function, and increased heart rate.


In pacemaker cells, NA also binds to β1 channels, which increase cAMP concentrations (via adenylate cyclase). This opens Na channels, allowing the cells to depolarize and fire, increasing contractility and heart rate. NA also stimulates the entry of Ca through L-type channels by increasing cAMP and PKA concentrations. PKA then phosphorylates the L-type channels, increasing their activity, and further depolarizing the cells.
What, again, are the effects of ACh during vagal stimulation of muscarinic channel?
cAMP and PKA concentrations are lowered, thereby downregulating the activity of the If and L-type Ca channels.
What are the effects of NA on the b1 receptors in heart? Cardiac muscle cells.
, NA binds to β1 receptors, increasing cAMP and PKA levels, as usual. Then, PKA opens Ca channels in the cell membrane and the SR, allowing Ca to enter the cell and promote contraction.
What are the Effects of NA on The β2 Receptor in Smooth Muscle?
When NA binds to β2 receptors on smooth muscle cells, it increases cAMP--> PKA concentrations. PKA inactivates the MLCK (myosin light chain kinase) by phosphorylating it, thereby preventing muscle contraction.
What are the effects of NA on the bladder?
NA has different effects on different parts of the bladder:
The detrusor muscle contains β2 receptors. When stimulated by NA, it relaxes.
The sphincter contains α1 receptors. When stimulated by NA, it contracts.
The net effect is urine retention (Mnemonic: type A is tense, type B is relaxed)
Describe fluctuating blood pressure in relation to the NSs and NA.
When baroreceptors in the carotid sinus sense increased blood pressure, the vasomotor center in the brain responds by increasing parasympathetic activity and decreasing sympathetic activity.
A neuron in the vasomotor center uses NA to signal inhibitory α2 receptors. This downregulates adrenergic neurons going to the blood vessels and heart. At the same time, parasympathetic innervation of the heart increases. Peripheral vascular resistance and heart rate decrease.
What are the general names for drugs that affect the sympathetic NS?
Sympathomimetics--direct or indirect sympathetic agonists
Sympatholytics/sympathoplegics--sympathetic antagonists
What are examples of drugs that affect the sympathetic system? What receptors do they affect? What are their clinical uses?
What are Adrenergic Agonists' Effects on Blood Pressure?
α1 Vasoconstriction TPR and BP go up
β1 Heart rate and stroke volume increase Cardiac output and BP go up
β2 vasodilatation TPR and BP go down
What are examples of adrenergic agonists and their effects on blood pressure?
1. Noradrenaline--acts on α1 and β1 receptors, causing blood pressure to rise.
2. Adrenaline--acts on α1, β1, and β2 receptors, causing BP to go up slightly
3. Isoprenaline--acts on β1, and β2 receptors, causing TPR to fall and BP to remain basically the same
In the nervous system, the transfer of information in the brain involved what? What is the main factor in information transfer?
Presynaptic spikes
Transmitter release
Postsynaptic response
Postsynaptic spikes

It is unclear whether the main factor in information transfer is the number of voltage spikes, or their temporal patter (i.e. their frequency).
What are the different types of information loops in which neurons can participate?
1. Feeback
2. Feed-forward
3. Lateral inhibition
How can excitatory synapses be distinguished from inhibitory ones?
Excitatory synapses can be distinguished from inhibitory synapses, because they have small structures called spines on the dendrites of the post-synaptic neurons that have high receptor concentrations.
A single dendrite will receive what kind of signals?
A single dendrite will receive both excitatory and inhibitory signals. The ratio of excitatory:inhibitory synapses across a neuron is roughly 3:1.
Excitatory neurons depolarize.
Inhibitory neurons hyperpolarize.
What are potential drug drug targets during neurotransmitter synthesis, storage, and release?
1. Uptake of precursors
2. Synthesis of the neurotransmitter (can happen either in neurons or in glial cells)
3. Storage of neurotransmitters in synaptic vesicles (ex: amphetamine inhibits vesicular transporters)
4. Degradation of the neurotransmitter in the vesicles or neurons (ex: MAO inhibitors)
5. Depolarization, via the action potential (ex: Phenytoin)
6. Influx of Ca2-
7. Exocytosis of neurotransmitter vesicles
8. Diffusion of the neurotransmitter in the synaptic cleft
9. Interaction of the neurotransmitter with postsynaptic receptors (ex: Diazepam, Memantine)
10. Interaction of the neurotransmitter with presynaptic receptors
11. Inactivation of the neurotransmitter (ex: Tacrine)
12. Reuptake of the neurotransmitter (Prozac, cocaine)
What determines the number of neurotransmitters? Where is concentration the highest?
1. NT concentration
2. Vesicular size
3. Vesicular transport

The concentration of NT is high in the synaptic cleft, since a single action potential can trigger the release of multiple vesicles.
What factors have effect on receptors? What is horizontal alignment?
The kinetics of vesicle release also affect the concentration of NT in the synaptic cleft (e.g. full fusion v. kiss and run mechanism). The geometry of the cleft (note the curves near the regions of NT release) is another factor that helps concentrate NT in regions where there is a high concentration of postsynaptic receptors. Between curves, there is spillover of neurotransmitter between neighboring synapses. Because the geometry of the synapse is so important, misalignment of the synaptic cleft can greatly reduce the efficacy of neurotransmitter release. Horizontal alignment is important for keeping concentration effective.
What are the factors that affect receptor activation?
1. Amount of NT released
2. Kinetics of vesicle release
3. Concentration of NT near the surface of postsynaptic receptors
What are the major types of neurotransmitters?
1. Amino Acids
2. Catecholamines
3. ACh
4. Neuropeptides
5. Other
What are the amino acid NT?
Excitatory:
Aspartate
Glutamate
(NDMA, AMPA, Kainate, AP-4)

Inhibitory:
GABA
Glycine
(Muscimol, Baclofen)
What are the catecholamine NT?
1. Noradrenaline
2. Dopamine
3. Serotonin (5-HT)
What are the "other" NT?
1. NO
2. Histamine
3. Purines
What are the three mechanisms of NT action? What are the major difference b/w these three pathways?
1. Direct activation -- the NT causes an ion channel to open or close

2. Indirect membrane activation of a channel -- through GPCR/Gβγ signaling --all the participants are membrane proteins.

3. Indirect activation of a channel through diffusion of a second messenger -- classical GPCR pathway

The major difference b/w these pathways is the time frame.
What is glutamate's role in the brain? What are its pathways for its synthesis?
Glutamate is the major excitatory NT in the brain. There are two possible pathways for its synthesis:
1. Transamination of alpha-ketoglutarate (a substrate of the Kreb cycle) in neurons.
2. Production from glutamine in glial cells, which is then transported to the presynaptic terminal and converted to glutamate by the enzyme glutaminase.
What are the categories of glutamate receptors? What are some examples?
1. Ionotropic -- form ion channel pores
- AMPA
- NMDA
- Kainate

2. Metabotropic -- do not form ion channel pores and are usually located on the periphery of the synapse (extrasynaptic receptors)
- mGluR (of which there are 8 known subtypes)
What are AMPA receptors? Where are they located ? What are the different subunits? What is the significance? How are they activated? What is the process of desensitization?
AMPA receptors are widespread in the CNS. They have 4 subunits, and are cation-selective channels, permeable to both Na+ and K+. Receptors that contain the Glu-R2 subunit are impermeable to Ca. Receptors that don't contain the Glu-R2 subunit are permeable to Ca, and act as inward rectifiers.
Activation of AMPA receptors is determined by phosphorylation. One glutamate can bind to each subunit of the receptor. These receptors are easily desensitized, and mediate fast, excitatory currents.
AMPA receptors play a role in learning and memory.
Where do AMPA receptors play the biggest role? How are AMPA receptors related to age?
AMPA receptors are the major receptors in nerve terminals, because they are responsible for transmitting the depolarization. A synapse with no AMPA receptors is called "silenced."
Young animals have many silenced synapses, that lack AMPA receptors. As they learn, they develop more AMPA receptors, indicating that the insertion of AMPA receptors in synapses is a possible mechanism for learning and memory. This has been demonstrated in animal models, but not yet in people.
How are kainate receptors structured?
Kainate receptors are similar to AMPA receptors, also composed of 4 subunits (GluR5-7 and Ka1-2). They are permeable to Na+ and K+, and some subunits to Ca+.
(NB: kainate is a synthetic glutamate analogue.)
What are NDMDA receptors? What are they involved in? What is the significance of the various subunits? What are the most crucial subunits?
NMDA receptors are also composed of 4 subunits (two NR1 and two NR2 subunits), and involved in learning and memory. The type of subunits determine decay kinetics of the current, and are developmentally regulated--the more NR2b subunits the receptors have, the greater capacity for learning and memory they will confer. The NR2 subunits are the most crucial, and the receptor is not functional without them. The major function of NMDA receptors is to allow for Ca influx.
What are the requirements for NMDA receptor activation?
1. Glutamate binding
2. Glycine or D-serine binding as a co-agonist (glycine is released by astrocytes)
3. Removal of a Mg2+ block by depolarization (caused by the opening of AMPA receptors)
How do NMDA receptors change over time? Where are they located? What are their affinities? What does this mean?
During early development, most NMDA receptor subunits are NR2b. Later in life, the NR2a subunits become prominent. The NR2b subunits are "extrasynaptic," further from the active zone of the synapse, and have half the glutamate affinity as the NR2a subunits, as well as a much lower affinity for glycine. This lowered affinity slows the current that runs the NMDA channels with the NR2b subunits. Because the current is slower, it lasts longer.
How is glycine needed?
Glycine is an obligatory agonist for the NMDA receptors. It is not obligatory for the kainate receptors.
What are the different types of metabotropic glutamate receptors? Where are they found? What type of feedback to they engage in? How do they differ?
Metabotropic autoreceptors participate in a negative feedback mechanism. When glutamate is released, it binds to presynaptic metabotrobic receptors, and inhibits further glutamate release (i.e. they inhibit their own neurotransmitter).

Metabotropic heteroreceptors are present on GABA-releasing neurons. They bind glutamate spillover from other synapses, and inhibit future GABA release (i.e. they inhibit another neurotransmitter). Heteroreceptors have a much higher glutamate affinity than autoreceptors, so are able to sense spillover glutamate, even at very low concentrations.
How can pre-synaptic inhibition be achieved? What does inhibition depend on?
1. Tonic--inhibition from ambient neurotransmitters that is always active

2. Evoked--inhibition comes in response to action potentials. Evoked inhibition is dependent on the frequency of the spikes--it is high when the action potentials are at a low frequency, and low when the action potentials are at a high frequency. Thus, such inhibition works selectively for low frequency stimulation only (this mechanism is called a "high pass filter"). For unknown reasons, the high pass filter effect works only when the metabotropic receptor is on the postsynaptic cell. It does not work when the metabotropic receptor is on the presynaptic cell.
What are the three subtypes of metabotropic receptors? Where are they found? What are their functions?
Group I--found on postsynaptic cells. Their function is to increase Ca release.
Groups II and III--their major function is presynaptic inhibition. They act to decreases Ca concentration.

Anti-epileptic drugs that are group I antagonists and group II agonists are currently in development.
What is special about cyclothiazine?
removes desensitization of AMPA receptors, acts as an AMPA agonist.
What are the functional aspects of rugs that act on glutamatergic receptors? What are examples?
1. Long term synaptic plasticity--learning and memory
2. Anesthetics (NMDA antagonists, such as ketamine)
3. Neurodegenerative diseases
Ex: memantine, a drug used to slow the onset of Alzheimer's, is a voltage-dependent NMDA antagonist
What is Alzheimer's? What is memantine?
Alzheimer's is the most common late-life dementia. It is characterized by the accumulation of amyloid-β plaques in the brain, due to increased concentrations of amyloid-β. It is thought that loss of synapses plays a role in the cognitive impairment of Alzheimer's, since a major change in Alzheimer's patients' brains is a loss of functional synapses, including a decrease in the transmission, plasticity, and number of synapses. Amyloid-β, in some unknown way, ends up triggering endocytosis of NMDA receptors, leading to spine loss.
Memantine acts by binding to NMDA receptors and inhibiting their endocytosis. It slows the onset of Alzheimer's, but does not stop or reverse it.
What is the basic mechanism for generation of long term memory?
long term potentiation of synaptic strength--i.e. a persistent increase in synaptic strength, following some stimulus from the external environment.
What are the two phases to long-term potentiation?
1. Induction
2. Expression
What is induction?
results in an increase or decrease in synaptic strength. The mechanism of induction is an influx of Ca through NMDA receptors.
NMDA receptors open in response to what stimuli?
1. Glutamate binding
2. The presence of glycine as a co-transmitter, released from surrounding glial cells
3. Removal of the Mg block by depolarization, produced by AMPA receptors
Depolarization of one receptor is not enough to open NMDA receptors; rather, many receptors need to open simultaneously.
What is expression?
an increase in synaptic strength, due to
i. An increase in the number of active AMPA receptors on the post-synaptic cell
ii. An increase in the possibility of NT release (i.e. more glutamate)
iii. An increase in the number of functional synapses
What governs short-term memory? What are its phases?
short-term alteration in synapse strength (lasting seconds to milliseconds), and is composed of the same two phases as LTP:
1. Induction
2. Expression --in most cases of short term potentiation, expression involves a change in the probability of NT release and a depletion in the number of vesicles in the presynaptic cell.
What is synaptic facilitaion? depression?
each pulse needed to get an AP requires less depolarization

each pulse needed to get an AP gets higher. The depression is less pronounced in the presence of ACH.
What is a major inhibitory NT in the brain?
GABA
Describe the synthesis and metabolism of GABA?
1. The synthesis of GABA from glutamate is catalyzed by an enzyme called GAD.
Vitamin B6 is required as a co-factor for GAD.
2. After synthesis, GABA transporters package the GABA into the vesicles.
3. Upon signaling, GABA is released into the synaptic cleft.
4. Transporters on glial cells are responsible for GABA reuptake, clearing it from the synaptic cleft.
- A mitochondrial enzyme called GABA transaminase catalyzes the conversion of GABA into succinic semialdehyde, which eventually enters the Kreb cycle
GABA transaminase can also regenerate glutamate (to be converted into GABA) from α-ketoglutarate
Drugs that inhibit GABA transaminase (ex: Vigabatrin) block the breakdown of GABA, therefore increasing GABA concentration.
What are the two types of GABA receptors?
1. Ionotropic--GABA(A) receptors
2. Metabotropic--GABA(B) receptors
Wha are the GABA(A) receptors? What is their structure?
Major receptors in inhibitory synapses. Pentameric transmembrane proteins with four membrane-spanning domains. Expression of different subunits leads to different GABA(A) receptor subtypes. They bind to two active sites on GABA, with their α and β subunits.

Activation triggers opening of an ionic pore. The GABAA receptor also has a regulatory site called the benzodiazepine site (BZD site), to which benzodiazepines (which are allosteric modulators of GABA receptors) can bind.
What are the drugs that act on GABA receptors?
1. Benzos
2. Barbiturates
3. Neurosteroids
What are benzos? What do they do? What do they treat?
1. Increase the affinity of the receptor for GABA
2. Increase the probability of the GABAA channel opening
Benzos alone do not have any effect on the receptor--their effects are only relevant in the presence of GABA.

treat anxiety disorders (short term), insomnia, status epilepticus, acute mania, and physical dependence. They are also used as a short term anesthetics and muscle relaxants.
What are the adverse effects of benzos? What can be used to treat overdoses?
drowsiness, sedation, ataxia, rebound insomnia, amnesia in the elderly, depression of respiration, and CNS depression when taken in combination with alcohol.

Flumazenil--a competitive antagonist for benzos that is used to treat overdoses, and to aid in recovery from benzos used as anesthetics.
How effects do barbituates have?
1. At low doses, they prolong GABA-induced opening of Cl channels
2. At high doses, they act like GABA and open chloride channels even in the absence of GABA
Barbiturates are rarely used, due to side effects.
What are GABA(B) receptors? What are the differnt subtypes? Where are they expressed? What are the implications?
Metabotropic GPCRs, slower than GABAA.

The specific GPCR of GABAB receptors is GPCRc, which is an obligatory dimer, composed of two subunits GB1 and GB2.

Alternative splicing of the GB1: GB1a--expressed at excitatory presynaptic terminals. lack = severe problems with learning and memory

2. GB1b--expressed in spines on postsynaptic cells. lack has not been shown to have learning and memory problems
Lack of either subunit renders the receptors non-functional.
What effects do GABA(B) have on presynaptic and postsynaptic cells?
On presynaptic cells, induce reduction of cAMP (via a Gio protein) and inhibit pre-synaptic Ca channels, thereby inhibiting pre-synaptic NT release.

On postsynaptic cells, activate GIRK channels (GPCRs that are inward rectifying K channels) to produce hyperpolarization , resulting in a decreased firing rate.
What effect does a high firing frequency have on GABA?
At high firing frequency, GABA will have a less pronounced inhibitory effect than at low firing frequency, due to short-term plasticity (this is almost always the case with regards to GPCRs on the pre-synaptic cell--the effects of an agonist or an antagonist to a NT will be more pronounced at low firing frequency).
What effects do glycine receptors
produce post-synaptic inhibition, just like the GABAB receptors
What are drugs that act on GABA receptors?
1. Sedatives
2. Anesthetics
3. Anti-epileptics
What are anti-epileptic drugs? What are examples and their mechanisms?
Anti-epileptic drugs are often designed to increase GABAergic transmission in a variety of ways, thus increasing the inhibitory signals in the brain (since GABA is an inhibitory NT)
They include:
1. Vigabatrin--inhibits GABA-T
2. Tiagabine--inhibits GABA reuptake
3. Benzos, barbiturates, Felbamate, topiramate, zonisamide--positive regulates of GABA receptors
Where is ACh synthesied? That are the three major pathways?
interpeduncular nucleus, and can also be synthesized in the striatal interneurons. There are three major ACH pathways in the brain:
1. Nucleus basalis
2. Septohippocampal pathway
3. Pontomesencephalon pathway
What are the two types of ACh receptors? What are the subtypes in the brain?
1. Ionotropic (nicotinic)
2. Metabotropic (muscarinic)--most CNS receptors. The major M subtypes in the brain are--
i. M2--mostly at presynaptic terminals.
ii. M1--mostly at postsynaptic terminals.
The major function of muscarinic receptors is inhibition
How is ACh synthesized?
ACH is synthesized from choline by choline acetyltransferase, and transported into storage vesicles by the vesicular ACH transporter (VAChT). In the synaptic cleft, ACH is degraded by ACH esterase.
What is ACh functionally involved in?
1. Arousal
2. Learning and memory
3. Neurodegenerative diseases (Parkinson's, treated with some anti-cholinergic drugs, and Alzheimer's, treated with some ACHE inhibitors and positive allosteric modulators)
What are the two catecholamines? How are they synthesized?
1. Noradrenalin/norepinephrine
2. Dopamine

synthesized from tyrosine, which is obtained from the diet and converted into DOPA by tyrosine hydroxylase. DOPA is then converted into DA. In the dopaminergic neuros, the pathway ends with the synthesis of DA. In adrenergic neurons, DA is converted into NA.
With what must NA be released with in order to work properly?
NA needs to be released with ATP as a co-factor, in order to work properly. In the extracellular space, NA can bind to either presynaptic or postsynaptic receptors.
What is the negative feedback mechanism that inhibits NT release?
Activation of α2 adrenergic receptors inhibits adenylate cyclase, thereby reducing cAMP levels and Ca influx. This comprises a negative feedback mechanism that inhibits further NT release.
Where is NA synthesized? What are the pathways it is involved in?
In the CNS, NA is synthesized in the locus ceruleus, which is connected by several pathways to the:
1. Limbic system (emotion, mood)
2. Frontal cortex (two pathways)
3. Cerebellum
What is the result of an NE deficiency in the CNS?
1. Impaired attention span
2. Impaired concentration
3. Impaired working memory
4. Slowed information processing
5. Depression
6. Psychomotor retardation
7. Fatigue
How is DA metabolized?
1. MAO--found on the mitochondrial membrane
2. COMT--found in the cytoplasm
What are the DA pathways in the brain?
1. Mesocortical
2. Nigrostriatal
3. Mesolimbic
4. Tubero-hypophyseal
What are the two major types of DA receptors? Where are they expressed?
1. D1--mainly postsynaptic (inhibitory) and function to increase cAMP
2. D2--presynaptic or postsynaptic (inhibitory or stimulatory) and function to either decrease cAMP or increase IP3.

DA has a higher potency for the D2 receptors than for the D1 receptors.
In general, drugs for Parkinson's target the D2 receptors.
How is DA used?
1. Antipsychotics--antagonists of D2 receptors
2. Antiparkinson's--drugs that increase DA activity
- Levodopa--can cross the BBB, and is then metabolized to DOPA
- Amantadin--increases DA release
- MAOB inhibitors--the MAOB subtype only acts on DA, not on other neurotransmitters
- COMT inhibitors
3. Anti-nausea
4. Neuroendocrine functions (e.g. used to treat hyperprolactinemia)
How are DA neurons involved in Parkinson's?
neurons going from the substantia nigra to the striatum inhibit GABAergic output from the striatum. This results in less GABA-mediated inhibitory signals, which results in the characteristic symptoms of Parkinson's. Dopamine agonists are being explored as treatment options.
How is serotonin (5-HT) synthesized and metabolism?
Serotonin is synthesized from tryptophan by tryptophan hydroxylase and then by DOPA decarboxylase.
After release, serotonin is degraded by MAO and aldehyde dehydrogenase, into 5-HIAA.
Where is serotonin made? What are its various pathways?
Raphe nucleus and sent over various pathways to the:
1. Frontal cortex
2. Basal ganglia
3. Limbic system
4. Brainstem
5. Hypothalamus
A serotonin deficiency results in what?
1. Depression
2. Anxiety
3. Panic, phobia
4. OCD
5. Food cravings, bulimia
What are the three major hypothesis about depression?
1. The major cause of depression is a reduction in serotonin release (could be caused by impaired metabolism, storage, release, etc.)
2. The major cause of depression is impaired upregulation of serotonin receptors in response to low serotonin concentrations
3. The gene expression hypothesis claims that 5-HT triggers activation of GPCRs that ultimately triggers expression of a gene called BDNF (brain derived neurotropic factor) that's involved in brain development and neuronal plasticity.
What are the target sites for antidepressants?
1. 5-HT reuptake inhibitors
2. Monoamine pump reuptake inhibitors
3. Blockage of presynaptic α2 receptors
4. MAO inhibition
What are the three major categories of antidepressants?
1. Tricyclics
2. Selective serotonin reuptake inhibitors (SSRIs)
3. MAO inhibitors
What are tricyclics?
5-HT and NE reuptake, are also negative allosteric modulators of NT reuptake. They also all block α1, adrenergic, histaminergic, M1 cholinergic receptors, and Na channels leading to lots and lots of nasty side effects (ex: cardiac arrhythmias from the impaired Na channels).
What are SSRIs?
don't bind to all the other receptors that tricyclics bind to, so they have fewer side effects. Their exact mechanism is unknown. Prozac is an SSRI.
What are MAO inhibitors?
there are two types of MAO inhibitors--
i. MAOA--selective for 5-HT and NE
ii. MAOB--protect neurons by metabolizing certain amines. Also degrade dopamine, and are therefore used for Parkinson's.
What are the challenges of drug delivery?
1. Protecting the drug from degradation by proteases in the plasma
2. Getting the drug across biological membranes
3. Getting the drug across cell membranes
What contributes to tumor formation?
Tumor cells
Angiogenesis
Growth factors
What is sequence of events in tumor angiogenesis?
1. Tumor cells secrete angiogenic factors
2. Blood vessels become hyperpermeable, due to VEGF. Plasma proteins extravasate and form a temporary fibrin scaffold.
3. Proteolytic destruction of the ECM by a protein called MMP
4. Endothelial cells migrate to the location of the sprouting capillary, and begin to proliferate
5. The new tumor vasculature is stabilized by pericytes and SMCs. Anastamosis is formed between the old vessel and the new vasculature.
Compare normal cells and cancerous cells.
Normal cells are relatively homogenous with little mitosis, regulated division, lack angiogenesis, and have contact inhibition.
Cancerous cells are heterogenous with lots of mitosis, secrete a lot of GF, express oncogenes but not tumor suppressor genes, have angiogenesis, and lack contact inhibition.
For tumors to grow, what genetic changes need to accumulate? Examples?
1. Primary genetic change--leads to an increase in cell proliferation and apoptotic cell death
Ex: loss of pRb function, overexpression of c-myc
2. Secondary genetic change--leads to a decrease in apoptosis
Ex: dysfunction of p53, overexpression of bcl-2
3. Subsequent genetic changes--lead to further phenotypic alterations
Ex: invasiveness, metastasis
Describe tumor cell growth.
Tumor cells grow at a logarithmic rate, which is important in deciding therapy doses. The doubling time is unique to each tumor type. Aggressive tumors have faster doubling times.
30 doublings will produce about a billion cells, enough for a lump to appear.
40 doublings will produce about a trillion cells, enough for a significant mass to manifest.

Typical doubling times:
Time For some…
24 hours Lymphomas
2 weeks Leukemias
3 months Breast cancer
Describe tumor growth kinetics.
It takes about 10 generations of dividing for a tumor to divide from a 1 gram mass to a 1 kilogram mass.

Since tumors grow with first order kinetics, a given dose will kill a constant percent of the cancer cells, not a constant number (i.e. the same dose will reduce the number of cells from 106 to 103 as from 103 to 100).

Chemotherapy, unlike most drugs, has a very narrow therapeutic index--the toxic does is very close to the therapeutic dose.
How does chemotherapy work?
Cancerous cells decline with each dose and recover between doses. Recover less with each dose, while the normal body cells recover equally between each dose. Therefore, the overall number of cancer cells should decline, while the number of normal cells should fluctuate within set limits.

If the tumor mass is too large or too aggressive to be treated with surgery and/or drugs, it will decline between drug doses, but not enough, as the mass continues to grow. In such cases, palliative care becomes the primary concern.
What are the types of chemotherapies? When are they given?
Curative chemotherapy--given in order to cure the disease

Adjuvant chemotherapy--given to patients in remission with no evidence of residual disease for a certain time period after signs of the disease have disappeared, to protect against recurrence.

Neoadjuvant chemotherapy--given prior to surgery or irradiation, in order to increase chances of success. Neoadjuvant therapy is given mainly in cases where the tumor is hard to access, or has the potential to damage crucial organs (e.g. lung tumors that are very near the heart). Surgery can then be performed after the tumor has shrunk from the chemo.

Palliative chemotherapy--given to improve symptoms.
70% of chemotherapies are given with palliative intent.
What are the effects of anti-cancer drugs?
1. Tumorigenic stress
2. Cell-cycle arrest
- reversible
- irreversible
3. Cell death
How does the Rb protein involved in the cell cycle?
Activated CDKs in these complexes drive the cell cycle by phosphorylating proteins that are critical for cell cycle transitions. One such protein is the retinoblastoma susceptibility (RB) protein, which normally prevents cells from replicating by forming a tight, inactive complex with the transcription factor E2F. Phosphorylation of RB causes its release, which activates E2F and allows it to stimulate transcription of genes whose products drive cells through the cycle.
What are CDK inhibitors?
Embedded in the cell cycle are surveillance mechanisms that are geared primarily at sensing damage to DNA and chromosomes. These quality control checks are called checkpoints; they ensure that cells with damaged DNA or chromosomes do not complete replication.[53] The G1/S checkpoint monitors the integrity of DNA before replication, whereas the G2/M checkpoint checks DNA after replication and monitors whether the cell can safely enter mitosis. When cells sense DNA damage, checkpoint activation delays the cell cycle and triggers DNA repair mechanisms. If DNA damage is too severe to be repaired, the cells are eliminated by apoptosis, or enter a nonreplicative state called senescence, primarily through p53-dependent mechanisms. Checkpoint defects that allow cells with DNA strand breaks and chromosome abnormalities to divide produce mutations in daughter cells that may lead to neoplasia
What are growth factors? How do they work?
The proliferation of many cell types is driven by polypeptides known as growth factors. These factors, which can have restricted or multiple cell targets, may also promote cell survival, locomotion, contractility, differentiation, and angiogenesis, activities that may be as important as their growth-promoting effects. All growth factors function as ligands that bind to specific receptors, which deliver signals to the target cells. These signals stimulate the transcription of genes that may be silent in resting cells, including genes that control cell cycle entry and progression.
What advantage does the frog egg have for studying cell cycle?
They divide quickly and the stages are easy to follow
In a malignant tumor, what 2 stages of the cell cycle have a larger percentage of cells?
G2 and S
Discuss the second two phases of the Interphase period of the cell cycle.
The S phase (synthetic phase) lasts 8-12 hours in most cells. DNA is replicated and proteins are synthesized, resulting in duplication of hte chromosomes. Centrosomes are also duplicated.

The G2 phase (gap two phase) lasts 2-4 hours. This phase follows the S phase and extends to mitosis. The cell prepares to divide. The centrioles grow to maturity. Energy required for the completion of mitosis is stored and RNA and proteins necessary for mitosis are synthesized, including tubulin for the spindle apparatus.
Please discuss control factors that have been identified which initiate and/or induce progression through the cell cycle.
Several control factors, including a category of proteins known as cyclins as well as cyclin-dependent kinases (CDKs) have been identified.

During G1 phase, cyclins D and E bind to their respective CDKs. These complexes enable the cell to enter and advance through the S phase.

Cyclin A binds to its CDKs, thus enabling the cell to leave the S phase and enter the G2 phase, and also to manufacture cyclin B.

Cyclin B binds to its CDK, inducing the cell to leave the G2 phase and enter the M phase.
Which cell cycles are targeted in certain chemotherapies?
The main phase targeted is the S phase. Others ()(ex:tubulin inhibitors) interfere with the M phase.
Drugs that bind covalently to the DNA may kill cells in the S phase or may hang out in the DNA until the cell reaches the S phase and then kill it. These drugs are considered non-phase specific (ex: alkylating agents).
Name S phase-specific drugs?
cytosine arabinoside
hydroxyurea
Name S phase-specific, self-limiting drugs.
6 mercaptopurine
methotrexate
Name M phase-specific drugs.
vincristine
vinblastine
paclitaxel
Name phase-nonspecific drugs.
alkylating drugs
nitrosoureas
antitumor antibiotics
procarbazine
cisplatin
dacarbazine
What regulates apoptosis?
Apoptosis is regulated by bcl-2, and induced by proteins called caspaces.

p53, and important mediator of the cellular response to DNA damage, is mutated in a lot of cancers but is hard to use as a target, because it is not stable. Proteins involved in the p53 pathway make more practical targets.
What types of cancer exist?
1. Hematological malignancies
i. Leukemia--proliferation of immature WBC precursors. Non-localized and disseminated
ii. Lymphomas--a disease of the lymph nodes. Localized and disseminated
A. Hodgkin's
B. Non-Hodgkins
2. Solid tumors--localized
i. Carcinomas--originate in epithelial cells
ii. Sarcomas--originate in mesenchymal cells (embryonic connective tissue) and grow in soft tissue
What is the difference between malignant and benign cancers?
Malignant cancers are invasive, non-encapsulated, and metastatic.
Benign cancers are encapsulated and non-metastatic.
Pic: an encapsulated tumor (bottom) compared to a non-encapsulated tumor. In the non-encapsulated tumor, muscle cells and tumor cells can be seen mixed together.
What are cancer etiologies?
1. Environmental factors
2. Radiation
3. Oncogenic viruses
4. Iatrogenic--e.g. chemotherapies that work by damaging the DNA, hormone replacement therapy, immunosuppressive drugs
5. Contributing factors--age, sex, race, genetics, geography, lifestyle, exposure levels
What are ways of evaluating response to therapy?
1. Physical exam
2. Imaging
3. Tumor markers (a big area of investigation today, for both diagnosis and evaluation of treatment)
i. CEA, CA-125, β-HCG, α-fetoprotein, PSA
4. Repeated biopsy
What are the classes of anti-cancer drugs?
1. Alkylating agents--DNA damaging, mutagenic agents
2. Anti-metabolites--imitate endogenous compounds in the body
3. Antibiotics--DNA damaging, mutagenic
4. Antimitotics
5. Hormones and antagonists
6. Molecularly-targeted therapy
7. Anti-angiogenics
What are examples of anti-metabolites?
i. Purine analogs (6-MP, 6-TG)
ii. Pyrimidine analogs (5-FU, Ara-CTP)
iii. Antifolates (MTX)
What are examples of antimitotics? Examples?
i. Microtubule inhibitors
1. Vinca alakloids
2. Paclitaxel (Taxol), Docetaxel
ii. Chromatin function inhibitors
1. Podophyllotoxins (etoposide, teniposide)
2. Camptothecin
What is an isobologram?
An isobologram is a graph designed to evaluate drug interactions. Down the middle of the graph will be a line called the line of additivity. Anything to the left of the line is synergistic. Anything to the right of the line is antagonistic. Anything on the line is additive.
What is the mechanism of action for akylating agents? What do they target?
Alkylating agents are strong electrophiles that act by covalently binding to nucleophilic groups on the DNA, usually the N7 of guanine. They can be either--
1. Monofunctional--bind to one DNA strand at a time
2. Bifunctional--bind to two DNA strands at a time, and are more effective
They are non-cell cycle specific, very toxic, mutagenic, and carcinogenic (so they can actually induce the reappearance of a new cancer years after treatment).

Their targets include amino acids, carboxyl, sulfhydryl and imidazole groups, and nucleic acids.
When alkylating agents bind to the DNA, they form cross-links, leading to the impairment of replication and RNA transcription
Describe the chemistry of alkylating agents.
alkylating agents have highly electrophilic chloroethylamine groups (boxed in red). Some have one chlorodiethylamine groups (monofunctional), and others have two (bifunctional). The R group will differ, depending on the drug. The alkylating agent will work by electrophilic attack on N, O, and S on the DNA (preferably, the N7 of guanine).

Bifunctional agents can cause either intrastrand linking, or cross-linking between the two strands.
What does DNA alkylation lead to?
DNA alkylation will result in cross-linking, mutations, disruption of base pairing, depurination, and strand breaks. A variety of biochemical events in nucleic acid and protein synthesis will be disrupted.
Alkylating agents are highly cytotoxic, and specific to all rapidly dividing cells.
What are the mechanisms of tumor resistance? How?
1. Decreased drug uptake or increased drug removal
Cells can eliminate drugs via the P-glycoprotein pump ("P" for permeable) which uses ATP to remove harmful substances. Upregulation of this pump can help confer resistance to many drugs.
2. Increased repair of the defect caused by the drug
One mechanism of resistance to alkylating agents is high expression of DNA excision agents, enzymes which cut out the cross-linked DNA.
Because these enzymes will make the cells even more prone to mutation, alkylating drugs can, in the long run, make cancer cells even more dangerous. Years after treatment, patients can develop secondary malignancies that are the product of the treatment of the first cancer.
3. Cross-resistance to drugs of the same family

The faster the doubling time of the tumor, the more effective alkylating agents will be, since they act on rapidly dividing cells. Dormant cells in G0 can repair the damage they cause.
What are examples of nitrogen mustards?
1. Mechlorethamine
2. Chlorambucil
3. Melphalan
4. Cyclophosphamide
What are examples of nitroureas?
1. Streptozocin
2. Carmustine (BCNU)
3. Lomustine (CCNU)
4. Semustine (methyl-CCNU)
What are examples of methylating agents?
1. Procarbazine
2. Dacarbazine
3. Temodal
Mechlorethamine
the first anti-cancer drug in clinical use. Side effects include nausea, vomiting, CNS stimulation (because it can cross the BBB), and bone marrow suppression (BMS). Latent viral infections may appear, due to immunosuppression.

Chlorambucil, melphalan also have BMS
Cyclophosphamide
most commonly used nitrogen mustard, with wide application. Cyclophosphamide is given as a pro-drug which is metabolized by the P450 system in the liver into its active form (a bifunctional alkylating agent that is very toxic). Because cyclophosphmide is not activated until it gets to the liver, it can be administered per os.
How is cyclophophamide activated? How is it metabolized?
Cyclophosphamide depends on liver enzymes for its activation. Therefore, it may not be effective in patients with liver disease.
Metabolism of cyclophosphamide in the liver yields two products:
i. The active metabolite, phosphoramide mustard
ii. Acrolein, a substance which is highly toxic to normal cells, but can be neutralized by another drug, called Mesna. Therefore, cyclophosphamide is given in conjunction with Mesna.
How do nitrosoureas work? What is special about them? What are their side effects? What are they used for?
produce interstrand DNA cross-links and protein carbamoylation. They are lipophilic, and can cross the BBB which makes them a popular drug for brain cancer. They are non-cross reactive, meaning that if a cancer develops resistance to one nitrosourea, it will not develop resistance to other alkylating agents.
This is because nitrosoureas are more vulnerable to the P-glycoprotein pump than other alkylating agents.
Side effects include nausea, vomiting, and BMS
Clinically, they are used for primary and metastatic CNS tumors. They are also effective in lung cancer.
Streptozocin
a nitrosourea that is toxic to the β cells of the pancreas, used to treat insulinomas. It has a low bone marrow toxicity.
Dacarbazine
a purine intermediate analog. Needs to be transformed to an active metabolite called methylhydrazine. Nausea, vomiting, etc.
Temodal
mostly used for brain tumors
Busalfan
an oral agent used against chronic granulocytic leukemia. Myelosuppressive
Ethyleneimines (Aziridines)
have three member ethyleneimmonium rings, and are more effective at low pH. Used to treat bladder cancer.
Cisplatin
cisplatin is an alkylating agent with its electrophilic groups attached to platinum. Its major side effect is renal toxicity, which can be countered by giving lots of fluids, to help clear the kidneys. Other side effects include vomiting and neuropathies.
How can cisplatin toxicity be limited?
i. Secluding the drug within a liposome
ii. Conjugating the drug to a macromolecule. Because tumor blood vessels tend to be new and leaky, the enlarged version of Cisplatin will still be able to exit the blood vessels into the tumor and affect the cancer cells. However, it will not be able to pass through normal blood
What are antimetabolite? How are they used? In what cell cycle are they most effective?
Antimetabolites are analogs of normal compounds within the cell that generally interfere with purine or pyrimidine availability by:
i. Competitively inhibiting them
Ex: agonists of purines and pyrimidines
ii. Inhibiting their synthesis
Ex: inhibitors of thymidylate synthase and DNA polymerase
Antimetabolites are cell cycle specific, and are cytotoxic during the S phase. They are most effective against rapidly proliferating cells.
What is most effective method of using antimetabolites?
The dose response curve of an antimetabolite is non-linear (i.e. increasing the dose only increases the effect up to a certain threshold), since once all the enzyme the drug is blocking is blocked, increasing the dose won't help. Increasing the length of treatment time is more effective than increasing the dose.
What are the four major antimetabolites? How are they used?
1. Methotrexate--a folic acid analog that inhibits dihydrofolate reductase
2. 5-FU--a uracil analog that inhibits thymidylate synthase and interferes with RNA synthesis and action
3. Gemcitabine--a sugar similar to deoxyribose, that inhibits DNA polymerase
4. Ara-C--similar to Gemcitabine
Why is folic acid important?
Folic acid is a vitamin that is essential to cell replication. If deficient, it will prevent DNA and RNA synthesis and the cell's entry into the S phase. It is structurally similar to folic acid.
Inhibition of folic acid is particularly lethal to cancer cells, since they are rapidly dividing.
How is folic acid metabolized?
Folic acid is obtained from the diet or produced by intestinal flora, and reduced to the tetrahydrofolate form by dihydrofolate reductase (DHFR). Then, tetrahydrofolic acid receives a methylene group from vitamin B12 in order to be activated. Then, the activated form (5,10 methylenetetrahydrofolate) reacts with uracil in a reaction catalyzed by thymidylate synthase, to produce thymidine.
What are MTX and 5-FU?
1. Methotrexate--a folic acid analog that inhibits dihydrofolate reductase
2. 5-FU--a uracil analog that inhibits thymidylate synthase and interferes with RNA synthesis and action

5-FU has a similar action to MTX. It is a uracil analog that gets incorporated into the RNA, and blocks thymidine synthase.

MTX blocks tetrahydrofolate production by inhibiting DHFR.
5-FU blocks thymidine production by inhibiting thymidylate synthase.
What is administered in the case of MTX toxicity? How does it work?
A drug called Leucovorin is administered in cases of MTX toxicity. Leucovorin is an exogenous version of 5,10 methylenetetrahydrofolate. Therefore, cells can produce thymidine directly from it, using thymidine synthase, and do not need folic acid and DHFR.
How do cancer cells develop resistance to MTX?
1. Decreasing polyglutamation, which makes MTX less active
2. Increasing DHFR expression
3. Altering DHFR, with a lower affinity for MTX
4. Altering the folate carrier or receptor binding protein
How do cancer cels develop resistance of 5-U?
1. Alterations in thymidylate synthase
2. Decreased intracellular activation
3. Loss of p53 (for unknown reasons)
Capecitabine (Xeloda)
an oral pro-drug of 5-FU, which can sometimes be toxic to the heart. It may also cause redness, itching, and swelling in the wrists and feet ("hand and foot syndrome").
What is Ara-C (Cytosine Arabinoside)?
Ara-C is an analog of cytosine, with an extra hydroxyl group that inhibits DNA polymerase. It acts as a pyrimidine antagonist, and is specific to the S phase.
What is Gemcitabine?
Gemcitabine is another nucleoside analog that gets incorporated into the DNA and blocks synthesis. It is the only available drug that can prolong survival time for pancreatic cancer. Aside from the typical side effects, it causes transient elevations of serum transaminases, proteinuria, and hematuria.
What are antitumor antibiotics?
bacterial or funal derivatives, interfere with cellular processes such as DNA or protein synthesis
Doxorubicin
fungal anthracycline that intercalates within the DNA, causes single and double strand breaks, and inhibits topoisomerase II
Mitomycin C
binds to DNA and form cross-links and DNA adducts
Bleomycin
a small peptide that binds to DNA, which results in single-stranded and double-stranded breaks
What is the mechanism for doxorubicin cytotoxicity?
1. Topo II inhibition (the most important mechanism)
2. DNA intercalation between base pairs
3. Free radicals formation (relevant mainly for cardiac toxicity)
Which drugs target topoisomerase II? How?
Anthracyclines & Epipodopylotoxins
Topo II is a molecular target for epipodophyllotoxins and anthracyclines. These drugs form a covalent topo II-DNA complexes that prevent the enzyme from completing the religation portion of the process (and induce protein-linked breaks in the DNA)

(Topo II inhibition with stabilization of the DNA-Topo II complex and production of DNA double-strand breaks).
How do anthracyclines work in terms of OH radicals?
Anthracyclines undergo chemical reduction through enzymatically catalyzed or iron catalyzed pathways to yield reactive free radical intermediates (hydrogen peroxide and hydroxyl radical) that can cause oxidative damage to cell membrane proteins and DNA.
(in addition, iron-drug complexes are formed, that bind to DNA).
What are signs of doxorubin toxicity?
Myelosuppression (mainly neutropenia)
Cardiomyopathy
Alopecia
Nausea and vomiting
A vesicant
Diarrhea
Etc.
What happens with pegylated liposomal doxorubicin? (Doxil)
Same mechanism of action as doxorubicin
Doxorubicin is encapsulated in Pegylated liposomes (resulting in different P/K profile and different spectrum of toxicity & activity).
What are some doxorubicin analogues? Their effects?
Epirubicin
Mitoxanthrone

Similar activity
Less cardiotoxicity
How does dactinomycin work?
Binds to DNA by DNA intercalation: blocks the ability to serve as a template for RNA (and DNA) synthesis.
What is the mechanism of action for antimitotics? Examples?
natural products that interfere with microtubule synthesis and degradation, leading to inhibition of cell division.

Paclitaxel (Taxol): stabilizes microtubules, inhibit the cell cycle during mitosis.
Vinblastine: plant alkaloid that causes depolymerization of microtubules.
How do vinca alkaloids work?
Inhibiting microtubules assembly
M-phase
How do taxanes work?
Promote microtubules assembly , resistance to de-polymerization
M-phase
What is the mechanism of action for vinca alkaloids?
binding to specific site on tubulin with prevention of polymerization, inhibition of microtubule assembly and mitotic spindle formation (leading to metaphase arrest)
How does resistance come about?
MDR-1 = multi drug resistance gene- Encodes p-Gp (p-glycoprotein) efflux pump
Resistance=over expression of MDR-1
Cross resistance among natural products
Drugs that effectively and safely inhibit the pump are still looked for. (Verapamil…)
mutations in alpha and beta tubulin
What is the mechanism of action for taxanes?
Binding is reversible and stabilize the microtubules against depolymerization (induce tubular polymerization), thereby disrupting normal microtubule dynamics (halts mitosis)(and lead to arrest at G2/M phase).
What are the mechanisms of resistance to taxanes?
P-gp overexpression ;MDR (mediated by the 170-kD p gp pump efflux, encoded by the mdr1 gene)

Mutations of tubulin

p53 mutations (taxanes induce apoptosis, induced by p53)

overexpression of Bcl-2
What are the topo-I inhibitors?
Irinotecan
Topotecan

Failure to repair DNA breakage and interference with transcription and replication
How do topoisomerase I interactive agents work?
Topo I “inhibition”, with production of single-strand DNA breaks:
Topo I breaks DNA to allow strand to uncoil.
the drug stops topo I from repairing the strand in one spot.
Completion of DNA replication is prevented.


Note: the term “Topo I inhibition”, although commonly used, is inaccurate (since the DNA breakage by the enzyme is enhanced). “Topoisomerase I interactive agents” or “poisoning topo I” are more accurate terms.
Irinotecan
Colorectal cancer (in combination with 5FU and leucovorin) and lung cancer
Metabolise in the liver
Clears in the kidney
Toxicity:
Diarrhea
Netropenia
Nausea and vomiting
Abdominal cramps
etc
Topotecan
Ovarian cancer
Metabolise in the liver
30% clears in the kidney
Toxicity:
Myelosuppression
Nausea and vomiting
Diarrhea
Abdominal cramps
Alopecia
fatigue
etc
What are the mechanisms for irinotecan resistance?
Low carboxylesterase (=the enzyme that catalyzes the conversion of irinotecan to SN-38) expression.

Overexpression of UGT (Uridine diphosphate glucuronosyltransferase=the enzyme that inactivates SN-38, a toxic active metabolite).
Filgrastim (Neupogen):
non-glycosylated methionyl G-CSF; produced in culture by bacteria; has an additional amino acid and is not glycosylated (addition of a saccharide residue following the assembly of amino acids).
Lenograstim (Granocyte):
glycosylated G-CSF; produced in culture by mammalian cells; identical to the natural molecule.
Summary of alkylating agents.
CCNS drugs. Form reactive molecular species that alkylate
nucleophilic groups on DNA bases (e.g N7 guanine)- leads to cross linking of bases,
Abnormal base pairing and DNA strand breakage.
Resistance: Increased DNA repair, decreased drug permeability, production of trapping
agents (Thiols).
Examples:
Cyclophasphamide (nitrogen mustard), Cisplatin/carboplatin
Mechanism, Pharmacokinetics (e.g. Hepathic Cytocrome P450- acrolein), clinical use,
toxicity, resistance, rescue (e.g. hydration, mesna).
Summary of antimetabolites.
CCS drugs (S phase- DNA synthesis pathway-see figure).
Structurally similar to endogenous compounds and are antagonists of folic acid,
purines, or pyrimidines. Interfers with nucleic acid and protein metabolism.
Formation of polyglutamate derivatives, DHFR, TS.
Examples:
Methotrexate, Ara-C, Gemcitabine, 5-FU/Xeloda.
Mechanism, PK, clinical use, toxicity, resistance (decreased drug accumulation,
changes in drug sensitivity or DHFR activity, decreased polyglutamation),
rescue (e.g. leucovorin).
Summary of vinca alkaloids.
CCS drugs. (M phase).
Block formation of mitotic spindle by preventing the assembly or disassembly
of tubulin dimers into microtubules.
Examples:
Vinblastine, Vincristine, paclitaxel, docetaxel.
Mechanism, PK, clinical use, toxicity, resistance (increased efflux via membrane drug
transporter), rescue.
Summary of antibiotics.
CCNS drugs.
Intercalation between base pairs, inhibition of topoisomerase II, generation of free
radicals. Block synthesis of RNA and DNA and cause DNA strand scission and
membrane disruption.
Examples:
Anthracyclins: Doxorubicin/Doxil, Mitomycin.
Mechanism, PK, clinical use, toxicity (BM, GI, alopecia, cardiotoxicity), resistance,
rescue (e.g. dexrazoxane-inhibitor of iron-mediated free radical).
How to manage side effects of chemotherapy?
CCNS drugs.
Intercalation between base pairs, inhibition of topoisomerase II, generation of free
radicals. Block synthesis of RNA and DNA and cause DNA strand scission and
membrane disruption.
Examples:
Anthracyclins: Doxorubicin/Doxil, Mitomycin.
Mechanism, PK, clinical use, toxicity (BM, GI, alopecia, cardiotoxicity), resistance,
rescue (e.g. dexrazoxane-inhibitor of iron-mediated free radical).
What are the rules for effective chemotherapy combinations?
Each drug produces at least some response
Different mechanism of action
Different dose limiting toxicity
Trastuzumab (Herceptin)
Herceptin is humanized anti-HER2 MAB (95% human and 5% murine). It binds with the extracellular domain of the HER2 cell-surface receptor, thereby inhibiting the growth of breast tumor cells that over express HER2.
Tumors strongly overexpressing HER2 (IHC 3+) and/or with proven HER2 gene amplification (FISH positive) are most responsive to herceptin.
What are the steps required for angiogenesis?
1. Secretion of angiogenic factors
2. Degradation of ECM
3. EC proliferation and migration
4. Stabilization of new vessels
What is the relationship b/w ErbB receptors and cancer?
Overexpression of ErbB-1 and/or ErbB-2 is associated with poor prognosis and advanced-stage cancers

Overexpressed ErbB proteins still respond to regulation, making them attractive targets for anticancer therapies
What is ErbB-1?
Also known as epidermal growth factor receptor (EGFR)

First family member identified

Involved in normal cell growth and differentiation

Disturbances in ErbB-1 signaling can lead to cell transformation

Ligands specific for ErbB-1 include EGF, transforming growth factor (TGF)-α, and amphiregulin
What is ErbB-2?
Also known as HER2/neu

No known ligands

Activation thought to occur through heterodimerization with other ErbB family members

ErbB-2 is the preferred heterodimerization partner with other family members

Most resistant to intracellular degradation, slower to inactivate compared to other family members

ErbB-2 overexpression in tumors correlates with poor prognosis and decreased survival times
What is ErbB-?
Lacks intrinsic tyrosine kinase activity

Often found in heterodimers with ErbB-2

Cytoplasmic domain contains 6 sites where the p85 subunit of PI3 kinase can bind

Heterodimers containing ErbB-3 (especially ErbB-2/ErbB-3) are thought to be particularly strong activators of the PI3K/Akt pathway that is associated with cell survival and resistance to cancer therapy
What is ErbB-4?
Important in cardiac and brain development

Expression might correlate with presence of hormone receptors in breast tumors, more favorable prognosis

Multiple splice variants, some of which contain binding sites for PI3K

Slow internalization

Ligands include neuregulins, betacellulin, epiregulin, and heparin-binding (HB) EGF
What are the common mechanisms for ErbB activation in tumors?
Protein mutation (ie, EGFRvIII—constitutively active deletion mutant lacking most of the ECD)

Genetic changes resulting in overexpression of either normal or constitutively active receptors

Production of EGF or TGF-α by tumor cells: sets up an autocrine loop leading to constitutive ErbB-1 activation
How are hormones and antagonists used is chemotherapy?
Mechanism of action:
inhibits synthesis or effects of the steroid hormones that are necessary for growth of certain tumors, such as breast and prostate tumors.

Examples:
Tamoxifen: binds to estrogen receptors (ER) as an antagonist inhibitor of estrogen.
Anastrozole: inhibits aromatase, the enzyme that catalyzes the final step in estrogen production.
Fulvestrant
Classified as a “pure antiestrogen”: functions by destroying the estrogen receptors (unlike tamoxifen, does not have an agonist effect).
What is a major effect of aspirin? What can it be used to treat and prevent?
Reduces platelet aggregation

1. Angina
2. Stroke
3. Peripheral vascular disease

Blocking prostanoid synthesis has an anti-inflammatory effect.
Aspirin blocks prostaglandin synthesis by inhibiting COX.
Progestins
Indirect action on the hypothalamus-pituitary axis consisting of inhibition of gonadotropin-releasing hormone release.
Direct action resulting in the inhibition of cellular proliferation.
Examples:
-Medroxyprogesterone acetate (Provera)
-Megestrol acetate (Megace)
What are autocoids? What are examples?
a diverse group of signaling molecules that have local effects and often act on smooth muscle. They include:
1. Eicosanoids (the biggies)
2. Serotonin
3. Endothelin
4. Histamine
5. Bradykinin
6. Angiotensin
What are eicosanoids? How are they synthesized? Where do they exist? What is the most common form? From where are they derived?
Eicosanoids are lipid-derived autacoids synthesized by the oxidation of polyunsaturated long-chain fatty acids, with 20 carbons and more than one double bonds. They exist all over the body, and have diverse functions.
The most common eicosanoid is arachidonic acid (C20:4).

Eicosanoids are derived from the diet as omega-3 and omega-6 fatty acids (found in fish oil).
Aminogluthetimide
Aminogluthetimide is a non selective aromatase inhibitor. Although it inhibits aromatization of androgens to estrogens, its main action is inhibition of adrenal steroidogenesis at early steps (inhibition of the synthesis of aldosterone, cortisol ,and androgenes).
What is the eicosanoid cascade?
1. AA is released from the cell membrane, usually by phospholipase A2 (found in snake venom)
2. Synthesis of eicosanoids
3. Excretion from the cell
4. Binding to membrane receptors on nearby cells
5. Signal transduction, via a GPCR
6. Effect
Ketoconazole
High doses inhibit gonadal and adrenal steroidogenesis by inhibiting the P-450-dependent enzyme system.
What are the two potential pathways for archidonic acid?
1. The lipoxygenase pathway, which produces leukotrienes
2. The cyclooxegenase pathway, which produces prostaglandins, prostacyclins,and thromboxanes
What is the process of prostaglandin synthesis?
In the COX pathway, the addition of an oxygen to AA creates a closed ring structure, which converts AA into prostaglandin G2. Then, PGG2 is peroxidized to produce PGH2.

PGH2 can give rise to many different prostaglandins, prostacyclins, and thromboxanes.
Where do prostacylcins affect?
endothelium
kidney
platelets
brain
Where does thromboxane A2 affect?
platelets
vascular SM cells
macrophages
kidney
Where does prostaglandin D2 affect?
mast cells
brain
airways
Where does prostaglandin E2 affect?
brain
kidney
vascular SM cells
platelets
Where does prostaglandin F2a affect?
uterus
airways
vascular SM cells
eye
Which prostanoids dilate arterial tonus? constrict?
Dilate:
PGE2
PGD2
PGI2

Constrict:
PGF2a
TXA2
C4b2a fxns in:

Possible answers:
a. C3 convertase Alternative Pathway
b. C3 convertase Classic Pathway
c. C5 convertase Alternative Pathway
d. C5 convertase Classic Pathway
e. MAC
is the C3 convertase of the Classical Pathway

(Anything C4 is classical pathway!)
Which prostanoid relaxes the bronchial tree?
PGE2 (EP2)
PGI2
Which prostanoids contract GI peristalsis? Relax?
Contract:
PGE2 (EP1+3)
PGF2a

Relax:
PGE2 (EP2)
PGD2
Which prostanoid decreases stomach acid secretion? Increases muscoal protection?
PGE2 (EP3)
Which prostranoids contract the uterus? relax?
Contract:
PGE2
PGF2a

RelaxL
PGD2
Which prostaglandins mediate swelling?
Increased capillary permeability

PGE2
PGI2
Which prostaglandins mediate redness and heat?
Vasodilation

PGE2
PGI2
PGD2
Which prostaglandins mediate pain? fever?
PGE2
What is COX? How is it shaped? What is its catalytic site? How does aspirin work on it?
COX is a tunnel-shaped enzyme, with a catalytic site inside its tunnel. The catalytic site has a serine residue at position 529.
Aspirin acts by irreversibly binding to the serine in the active site of COX and acetylating it. This acetylation blocks AA from entering the tunnel and reaching the active site.
Is the acetylation of serine reversible? What happens?
Because acetylation of the serine is irreversible, a COX molecule bound to aspirin will never regain functionality, and new COX molecules will need to be synthesized for more prostaglandins to be made.
What is the technique for evaluating platelet function? What happens with the addition of aspirin?
Platelet aggregation can be evaluated using a sample of platelet-rich plasma, shining a light through the sample, and measuring the absorbency. Then, factors that should induce clotting are added to the sample. If the platelets are functioning properly, they will clot, making the sample clearer and allowing more light to pass through it upon a second measurement.


If aspirin (ASA in the graph below) is added to a sample of plasma and then clotting factors are added, light absorption will not go down as much as it would without the aspirin, because clotting is inhibited.
What are the three major actions of NSAIDs?
1. Anti-inflammatory
2. Analgesic
3. Antipyretic
All NSAIDs inhibit COX (usually by reversible, competitive inhibition).
What are the clinical indications for aspirin use?
1. Fever
2. Acute pain, especially if caused by inflammation
i. Menstrual pain, since prostaglandins cause uterine contraction
3. Chronic pain
i. Rheumatological disease
ii. Degenerative bone diseases
4. Anti-platelet effects (only Aspirin is used, not the other NSAIDs)
What are adverse effects of NSAIDS? (6) GI, kidneys, platelets, uterus, etc
1. Erosions of the GI tract, leading to ulcers, perforations, and bleeding, especially in the gastric and duodenal area. May also cause nausea and abdominal discomfort.
2. Water and Na retention in the kidneys, especially in patients with CHF and cirrhosis
3. Decreased renal function and nephrititis
i. Long term NSAID use can cause renal failure, called analgesic neuropathy
4. Decreased platelet function, which may cause bleeding
5. Relaxation of the uterus (can prolong labor, so NSAIDs are not used during childbirth)
6. Hypersensitivity reactions, possibly due to diversion of the arachidonic acid metabolism into the leukotriene pathway, since leukotrienes mediate asthma-like responses.
How can NSAIDS have adverse effects on the kidneys? Particularily on which patient population? Why?
NSAIDs have particularly adverse effects on the kidneys in people with CHF or cirrhosis. This is because both of those conditions will activate the release of renin/angiotensin, which constrict the arterioles of the glomerulus. Normally, prostaglandins have vasodilatory effects on the afferent arteriole. However, if a patient is on NSAIDs, this effect will be eliminated, allowing the vasoconstrictive effects of angiotensin on the afferent arteriole to be unopposed. The afferent arteriole will clamp down, messing up the pressure gradient in the glomerulus.
What is the explanation of adverse of NSAIDS on the GI tract?
NSAIDs inhibit prostaglandins in the gastric mucosa, where prostaglandins normally reduce acid secretion, increase bicarb secretion, and stimulate proliferation of epithelial cells (which helps heal erosions). This results in an increased tendency towards ulcers.
Even very low doses of aspirin can cause damage to the GI tract.
What are the two COX isoforms? What do they express? What are they involved in?
1. COX-1--expressed constitutively, fulfills housekeeping functions (e.g. gastroprotection, clotting)
2. COX-2--is inducible in response to particular signals (ex: stress, cytokines) and mediates pain and inflammation
What happens when COX is selectively inhibited? What are examples of COX-2 inhibitors?
Inhibiting both COX enzymes will eliminate the normal, housekeeping functions of COX-1, resulting in the aforementioned adverse effects of NSAID use. Inhibiting COX-2 only eliminates pain and inflammation, without disturbing other prostaglandin effects.
Glucocorticoids and IL-4 are selective COX-2 inhibitors.
What is the structure of COXs? What is the difference b/w COX 1 & 2? What are examples of drugs/what are the mechanics?
COXs have two catalytic sites, one for cyclooxygenation, and one for peroxygenation.
The difference between COX-1 and COX-2 is in the cyclooxygenation site--COX-1 has a bulky isoleucine reside in the active site and COX-2 has valine in the same position, allowing a larger entrance to the active site of COX-2:

COX-2 selective drugs are designed to fit into the pocket in the cyclooxygenation active site that is only present in COX-2.

Selective COX-2 inhibitors have "coxib" in their names.
What is IC50?
: IC50--the concentration of a drug need to achieve 50% of the effect.
Compare the different COX inhibitors. , COX-2 selective inhibitors are to the right of the line. Drugs that block both COXs equally are on the line. Drugs that block COX-1 better are to the left of the line.
What are the tendencies of COX-2 inhibitors in comparison to COX1 inhib? What is an example that was taken off the market for adverse cardio events?
Selective COX-2 inhibitors do not tend to block platelet function. They also have much lower GI toxicity.
Rofecoxib (Vioxx) is a selective COX-2 inhibitor. It was taken off the market for causing adverse cardiovascular events
How does the body use thromboxanes and prostacyclins? Describe the effects of COX inhibitors here.
vascular endothelium of atherosclerotic arteries experiences an inflammatory reaction. Therefore, such arteries use COX-2 to produce prostcyclin.
Platelets use COX-1 to produce thromboxane.


Selective COX-2 inhibitors block prostacyclin synthesis, but do not affect thromboxane. This results in the unopposed effects of thromboxane, which increase the possibility of clotting, resulting in adverse cardiovascular events.
What is the problem with coxibs?
Coxibs are now sold with a warning. They have approximately a two-fold risk of causing heart attacks. Therefore, they are unlikely to be problematic in people with a low risk for heart attack.
Celecoxib is still on the market in the US but is contraindicated in patients at high risk for adverse cardiovascular event.
What are potential problems with medication overdoses? What are examples?
1. Exaggeration of the desired therapeutic effect
Ex: beta blocker overdoses cause bradycardia
2. Exaggeration of an adverse effect that may be minor at the therapeutic dose
Ex: decreased renal function in NSAID overdose
3. Toxic effects that are not seen at the therapeutic dose
Ex: acetaminophen overdoses cause liver toxicity
What are dose response curves? What is their shape? What do tey express.
Dose response curves chart the effect of a drug as a function of its concentration. They are typically sigmoidal, linear in the middle with plateaus at the edges. In the graph below, the blue line indicates the therapeutic dose and the red line indicates the toxic dose. The concentration difference between the therapeutic dose and the toxic dose is called the margin of safety
What is the median lethal dose? Why does this variable exist?
Because of the variability in response, the median lethal dose of a drug is defined as the LD50--the dose that kills 50% of a population
What does it mean for a medication to have linear pharmacokinetics? How does this relate to toxic doses?
Most medications have linear (first order) pharmacokinetics--meaning that doubling the dose of a drug will not affect its half life. This is not true with regard to toxic doses, which are usually beyond the linear region in the sigmoidal dose response curve.
Therefore, the half lives and effects of toxic doses may be very different--toxicokinetics studies the kinetics of drugs at toxic doses.
What is pill clumping? What is the problem?
Another unique effect of toxic doses is called pill clumping, in which a high dose of tablets will form a clump in the stomach, from which medicine will be dispensed into the body at an unpredictable rate. It may take a while for the effects of the drug to manifest, as long as the drug is locked up in the clump, but once the clump starts to dissolve, the effects will be much more pronounced than usual, due to the high dose.
What steps are needed for treatment of poisoning?
Airway
Breathing
Circulation
Decontamination
Enhanced elimination
Focused therapy
Get toxicological help
What are the patterns of toxic syndrome?
1. Sedative/hypnotic and opoid
2. Cholinergic
3. Anticholinergic
4. Sympathomimetic
5. Serotonergic
What is the clinical presentation of Sedative/Hypnotic and Opoid Toxidrome?
Altered mental state
Slow, shallow breathing (apnea in the sedative toxidrome can be fatal)
Hypotension, and sometimes bradycardia and hypothermia
Decreased bowel activity ("hypoperistalsis")
What are potential culprits of Sedative/Hypnotic and Opoid Toxidrome?
Benzos
Opiates
Antiepileptics
Antipsychotics
Barbiturates
Ethanol
What is clinical presentation of cholinergenic toxidrome? How frequently is this seen?
Salivation
Lacrimation and meiosis
Urination
Diarrhea
GI cramps
Emesis
Sweating, wheezing, respiratory secretions (which may cause suffocation)

The cholinergic toxic syndrome is rare. It will most commonly occur in cases of chemical warfare or pesticide poisoning.
To simplify, the clinical presentation involves lots and lots of fluid loss.
What are potential culprits of cholinergenic toxidrome?
Carbamate
Organophosphate Toxins (ACH esterase inhibitors)
What is the clinical presentation of anticholinergenic toxidrome? How frequently is this seen?
Dry skin, fever, flushing
Psychosis, coma, seizures
Tachycardia, hypertension
Hypoperistalsis
Mydriasis
arrhythmias

(all unopposed, sympathetic activity)

Anticholinergic toxic syndrome is relatively common, since lots of drugs of abuse have anticholinergic side effects.
What are potential culprits of anticholinergentic toxidrome?
Antihistamines
Tricyclic antidepressants
Antipsychotics (especially phenothiazine)
Atropine
Datura (a weed that contains a high concentration of atropine-like chemicals)
What is the clinical presentation of sympathomimetic toxidrome?
Hypertension, tachycardia
Sweating
Mydriasis
Tremor
Seizures
Arrhythmias
What are the potential culprites of cympathomimetic toxidrome? How to differentiate b/w it and anticholinergenic toxidrome?
β-agonists (used to treat hypotension and asthma, found in epipens)
Theophylline
Caffeine (whee!)
Cocaine
Amphetamines
Pseudoephedrine

The sympathomimetic and anticholinergic toxidromes are very similar; however, redness, fever, bladder atony, and hypoperistalsis are more suggestive of an anticholinergic toxin.
What is the clincial presentation of serotonergic toxidrome?
Irritability, nervousness, tremor
Fever
Hypereflexia, clonus (involuntary muscle contraction), myoclonus
Flushing, sweating
Diarrhea
What are the potential culprits of serotonergic toxidrome?
SSRIs (ex: Prozac)
Clomipramine
How to treat a coma due to an unknown cause? What are each of the parts?
1. IV dextrose (to correct for hypoglycemia)
2. Oxygen
3. IV Naloxone (an antidote to opiates)
4. Thiamine (in patients that are suspected of vitamin B1 deficiency, mainly alcoholics)
5. IV flumazenil (used to be used to treat benzo toxicity, but is not widely used today, because if a patient takes psychotropic drugs in conjunction with a benzo sedative, the two drugs will neutralize each other--but if the benzo is neutralized, the psychotropic, pro-epileptic drug will be unopposed, and the patient will go into status epilepticus.)
(pneumonic is DONT)
What is a gastric lavage?
a large orogastric tube is inserted through the mouth, into the stomach. Then, water is pushed through the tube, the tube is attached to a pump, and the contents of the stomach are pumped back out the tube. Then, the whole procedure is repeated. This is rarely used today, because there are more efficient, less unpleasant ways to prevent absorption.
How is activated charcoal used?
charcoal is able to bind to a lot of toxins, and prevent them from being absorbed in the body. One gram of charcoal has a lot of pores, giving it a surface area of 500 m2, which increases is binding capacity.
Because charcoal cannot be absorbed by the body, the toxins it binds to get excreted with it.
The recommended dose is 1 gm charcoal/Kg body weight.
What are drugs that are not absorbed by activated charcoal?
i. Pesticides, potassium
ii. Hydrocarbons
iii. Alcohols
iv. Iron, insectisides
v. Lithium
vi. Solvents
When must activated charcoal be given? How does this change for drugs that enter enterohepatic circulation? What are examples of these drugs?
Activated charcoal needs to be given within about 1 hour of ingestion of the toxin in order to be effective.
For drugs that enter the enterohepatic circulation, repeated doses of activated charcoal are given. Such drugs include:
i. Antimalarials (quinine, theophylline)
ii. Barbiturates
iii. Carbamazepine
iv. Dapsone
What is whole bowel irrigation?
similar to gastric lavage. A nasogastric tube is inserted, and then polyethylene glycol is administered through the tube and flushed through the body.
How can elimination of a toxin be enhanced?
i. Changing the pH of the urine to decrease reabsorption

ii. Hemodialysis (works on small, water soluble compounds such as alcohol, salicylates, lithium, and heavy metal chelates)

iii. Perfusion of the blood with activated charcoal (works on large, protein-bound or lipophilic toxins, such as theopylline, phenytoin, phenobarbital, and carbamazepine)
Name the antidote.

Paracetamol
N-acetylcysteine
Name the antidote.

Opiates
naloxone
Name the antidote.

Benzodiazepines
Flumazenil
Name the antidote.

Methanol, ethylene glycol
Ethanol/fomepizole
Name the antidote.

iron
deferoxamine
Name the antidote.

organophosphates
Atropine+oximes
Name the antidote.

Digoxin
Fab fragments (Digifab)
Name the antidote.

Beta-blocker, CCB
Glucagon
Name the antidote.

Tricyclic antidepressants
Sodium bicarbonate
What is the difference between pharmacogenetics and pharmacogenomics?
Pharmacogenetics--studies drug responses to variations in the DNA sequence only
Pharmacogenomics --studies drug responses to variations in DNA and RNA, so relates to both genes and gene expression.
What are the leading drugs that cause adverse reactions in the US?
Warfarin, acetaminophen (liver toxicity), Aspirin, Ibuprofen, Clopidogrel, and Phenytoin.
What are causes of adverse drug reactions?
1. Poor metabolism of the drug
2. Poor efficacy, possibly caused by
i. Speedy metabolism
ii. Altered drug target
What are prodrugs? What are examples?
Prodrugs, that are given in an inactive form and metabolized in the body to an active form, have poor efficacy when the metabolism is slowed, but will also be less likely to cause an adverse event in such a case.
Examples of prodrugs include codeine, Tamoxifen, and Clopidogrel
How are lipophilic drugs metabolized?
In two steps:
Phase I -- liver converts drug into a more hydrophilic form

Phase II -- conjugation og the drug to another compound that makes it even more hydrophilic
Explain the purpose of the P450 enzymes. What are they responsible for? Where are they expressed?
Most drugs are metabolized in the liver by the P450 enzymes (aka CYPs). There are 57 CYPs in the genome that are responsible for phase I oxidative metabolism of most hydrophobic and aromatic molecules, including:
1. Steroid hormones
2. Xenobiotics
3. Carcinogens

CYPs are mostly expressed in the liver, but can also be expressed in the brain, muscle, intestines, lungs, and placenta.
What drugs are metabolized by CYP3A4/5? What inhibits? How?
Most drugs are metabolized by CYP3A4/5. Smoking, alcohol, and certain drugs (phenytoin, carbamazepine, certain herbal substances) inhibit CYP3A4/5--this will inhibit metabolism of other drugs, preventing either activation of a prodrug, or clearance of a drug from the body.
What drugs are metabolized by CYP2D6? When is it dysfunctional? Deficiencies tend to affect which patient population?
CYP2D6 metabolizes about a quarter of drugs. It is dysfunctional in patients who are poor metabolizers and overactive in patients with gene duplications.
Deficiencies in CYP2D6 are more common in Africans and Caucasians than in Japanese and Chinese.
Tramadol
Codeine

Administration, metabolization. Sensitivity.
1. Tramadol and Codeine--analgesics that are administered as prodrugs and metabolized to active compounds by CYP2D6. Their effects are limited in CYP2D6-deficient patients, so other analgesics (e.g. morphine) need to be given to such patients. People with extra gene copies of CYP2D6 (also pretty common) respond extremely sensitively to these drugs, and need to be treated with lower doses, to avoid the danger of an overdose.
Tamoxifen

Administration, metabolization. Sensitivity.
an estrogen-blocker, used to treat breast cancer in patients with a normal estrogen receptor, and used preventatively in patients at high risk for breast cancer. Tamoxifen is administered as a prodrug and also metabolized to its active form by CYP2D6. Therefore, patients that are deficient in CYP2D6 do not respond to Tamoxifen.
CYP2C19 deficiency affects which patient population? What are some drugs that are metabolized? What do they do?
A CYP2C19 deficiency is rare in Caucasians, and more common in Japan.

CYP2C19-metabolized drugs to remember are:
1. Plavix (Clopidogrel)--inhibits platelet aggregation, given to stroke patients
2. Omeprazole--a proton pump inhibitor, given in combination with Amoxicillin, to treat H. pylori infections. Omeprazole is not given as a prodrug but as an active metabolite, so drug concentrations are much higher in poor metabolizers, and too low in ultra-metabolizers.
Poor Omeprazole metabolizers have a higher cure rate, indicating that perhaps the drug should be administered in higher doses.
How is acetaminophen metabolized? What happens in patients deficient in these enzymes?
Acetaminophen is metabolized both by CYP enzymes and by phase II enzymes, which perform sulfation. Patients deficient in these enzymes will divert acetaminophen metabolism into a toxic pathway, catalyzed by CYP2E1.
What is 5-FU? How is it metabolized? When is it not effective?
5-FU is a non-competitive inhibitor of thymidylate synthase, used to treat breast and colorectal cancer. It is administered as a prodrug and both
1. Converted into an active metabolite inside cells
2. Metabolized into an inactive compound by dihydropyrimidine dehydrogenase (DHD).
5-FU is not effective in tumors that overexpress thymidylate synthase, and can be toxic in DHD-deficient patients, who need to be treated with doses much lower than the recommended dose.
What is deficient in diabetes insipidus? How can it be treated? From where is it released?
Vasopressin (ADH)--released in response to rising plasma tonicity or falling blood pressure. Causes vasoconstriction and water retention in the kidneys. Is deficient in diabetes insipidus, which can be treated with a vasopressin analog called Desmopressin.
Describe prolactin? What is it similar to? What does it do? What inhibits it? What stimulates its release? What can cause high prolactin?
Prolactin similar to GH, that promotes growth and function of the mammary glands. It also may have effects on the immune system and coagulation. Its secretion is inhibited by DA (it is the only AP hormone that is not controlled by a negative feedback mechanism). Its release is stimulated by TRH, oxytocin (secreted during childbirth), and estrogen. Sleep, exercise, sleep.

Hypothyroidism can cause high prolactin, since in primary hypothyroidism, TRH levels will be elevated. Also,
Chronic renal failure
Renal Disease
Drugs (Reserpine, Morphine..)
What are clinical manifestations of hyperprolactinema? How does it differ between women and men?
1. Women
"galactorrhea amenorrea syndrome"
Typically caused by microadenomas
2. Men
Impotence, erectile dysfunction, infertility
Typically caused by macroadenomas
How should hyperprolactinemia be treated? What are some side effects?
1. Bromocriptine
2. Carbegoline--the drug of choice, because it has a longer half life and can therefore be given less frequently, and involves fewer side effects
Most prolactin-secreting adenomas will respond to Bromocriptine and Carbegoline, although microadenomas are easier to treat than microadenomas.

Side effects of DA agonist drugs include:
1. Nausea
2. GI side effects
3. Hypertension
Describe the release of GH, IGF-I?
GH is secreted in a pulsatile function, mostly at night, which should be taken into account clinically, when measuring a patient's GH levels.
IGF-I levels are steady over the course of a day, although they change with age, increasing until about 20, and then decreasing with age.
In terms of measuring GH excess, how can is be measured? Wheat the the clinical steps?
IGF1 levels can be measured to evaluate GH excess. They are elevated if GH is, and decreased if it is decreased.
If a patient has high IGF-I levels, the next clinical step is to perform an OGTT--a patient with elevated GH will not be able to suppress GH in response to an oral glucose load.
If the OGTT is positive, the next step is to take an MRI, to test for a pituitary tumor.
What are the best treatments for acromegaly?
is surgical removal of the pituitary. Other treatment options include:
1. Somatostatin analogs--given to most patients in conjunction with surgery. Somatostatins inhibit secretion of ALL the pituitary hormones, as well as suppress the release of insulin in the GI tract.
Synthetic somatostatin analogs with long half lives, that do not inhibit insulin release are on the market.
2. Pegvisomant--a GH receptor antagonist that is effective in decreasing GH and IGF-I levels, although it may be involved in some carcinogenesis.
3. DA agonists
4. Radiation
What are the goals of acromegaly treatment?
1. Remove the initial tumor
2. Restore GH levels to normal
3. Restore IGF levels to normal
4. Control symptoms, to reduce comortalities
What are examples of somatostatin analogs that treat acromegaly? What are some side effects? What can somatostatin analogs also be used to treat?
1. Octreotide
2. Lanreotide

Side effects:
1. Adverse GI effects (diarrhea, nausea, abdominal discomfort)
2. Gallstones (since they decrease gall bladder motility)
3. Impaired glucose regulation

Aside from acromegaly, somatostatin analogs are also used to treat TSH-secreting tumors and carcinoid tumors.
How can GH deficiency be diagnosed? How can it be treated?
Low IGF-I levels are an indication of a GH deficiency. A GH deficiency diagnosis can be confirmed using two tests that, in a normal person, should stimulate GH secretion:
1. Glucagon test
2. GHRH and arginine

Low GH can be treated by subcutaneous administration of GHRH; side effects include headaches, edema, arthralgia, and possibly carcinogenesis.
What are gonadotropins? What stimulates their release? How is it secreted?
gonadotropins (FSH and LH) are glycoproteins that regulate the steroid sex hormones. Their release is stimulated by GNRH.
What are synthetic GNRH analogs that inhibit GNRH release? What can they be used to treat?
1. Leuprolide
2. Goserelin
3. Nafarelin
4. Histrelin
They can be used to treat prostate cancer, endometriosis, and precocious puberty. Side effects include hypersensitivity, dermatitis, headache, diminished libido, and in women, sweating, depression, and ovarian cysts, and in men, bone pain and gynecomastia.
GNRH analogs are contraindicated in pregnant and breastfeeding patients.
How can infertility be treated? What are some FSH analogs?
Gonadotropins are used to treat infertility. FSH analogs, followed by hCG (an LH agonist) injection, are used to stimulate ovulation in women.
FSH analogs include:
1. Menotropins
2. Urofollitropin
3. Follitropin beta
How does progestin work?
inhibits ovulation by inhibiting LH secretion. Also causes thickening of the cervical mucus, and prevents implantation.
How is the thyroid regulated?
1. The hypothalamus releases TRH (thyrotropin release hormone)
2. TRH stimulates the anterior pituitary to release TSH (thyroid stimulating hormone)
i. The release of TSH is inhibited by T4 and T3.
3. TSH stimulates the release of thyroid hormone from the thyroid gland. There are two major forms of thyroid hormone:
i. T3--the active hormone, that comprises about 20% of thyroid secretions.
ii. T4--an inactive form, that comprises about 80% of thyroid secretions and is converted into T3 in the liver.
Patients with thyroid deficiency are given T4 supplements.
Go through the process or thyroid hormone synthesis?
Binding of TSH to TSH receptors on the thyroid stimulates the uptake of iodide (I-) into the epithelial cells of the thyroid.
1. Iodide enters the cells on the basolateral side via the Na/iodide (NIS) transporter, and is oxidized to iodine (I2) by thryoide peroxidase.
2. Iodine is pumped out of the cells on the apical side, into the follicular lumen.
3. In the lumen, iodine binds to tyrosine residues attached to thyroglobulins, to form either DIT (diiodotyrosine) or MIT (monoiodotyrosine). This step is also catalyzed by thyroid peroxidase.
4. Iodination of thyroglobulin can be inhibited by propylthiouracil methimazole or elevated iodide.
i. DIT can combine with another DIT to form T4
ii. DIT can combine with MIT to form T3
1. Condensation of DIT and MIT to form T4 or T3 can also be inhibited by propylthiouracil methimazole
5. TSH stimulation triggers the thyroglobulin complex to be endocytosed back into the follicular cells, where T3 and T4 are released from the thyroglobulin and sent into the bloodstream.
i. Thyroid hormone release is inhibited by elevated iodide.
How does thyroid hormone circulate in the blood?
Thyroid hormone circulates in the blood bound to thyroid binding protein. T4 is converted into the more active T3 when it enters the cells or in the liver.
Inside the cells, T3 binds to regions on the DNA called thyroid hormone responsive elements, and ultimately activates target genes.
More T4 is present in the bound form than T3.
The T4 half life is 7 days. The T3 half life is one day.
If a patient on T4 replacement therapy forgets to take a pill one day, it's not a big deal, because of the long half life of T4.

Thyroid hormone has two rings; an inner ring and an outer ring. Deiodination of the outer ring converts T4 into the active T3. Deiodination of the inner ring can produce a biologically inert form of T3, call reverse T3 (rT3).
What is the Wolff Chaikoff Effect?
The Wolff-Chaikoff effect refers to a decrease in thyroid hormone synthesis that will occur as the result of excessive iodine intake (ex: use of iodine as contract material in a CT).
High levels of iodine lead to high intracellular iodide, which inhibits thyroglobulin iodination and thyroid hormone release.
In a normal gland, the Wolff-Chaikoff effect will reverse itself if the iodine overload is removed.
What are the effects of thyroid hormone?
1. Increased metabolic rate
2. Increased oxygen consumption
3. Increased glucose uptake
4. CNS development and function

Hypothyroidism is more common than hyperthyroidism.
What is Hashimoto's Thyroiditis?
Hashimoto's is an autoimmune disease that predominates in women between 30-50. In it, antibodies attack thyroid peroxidase and thyroglobulin, leading to decreased production of T3 and T4. It is also associated with increased incidence of other autoimmune disease.
Symptoms of Hashimoto's represent the lowered metabolic rate, including:
1. Fatigue, lethargy, cold intolerance
2. Mental slowness
3. Lessened appetite but increased weight
4. Dry skin and hair
5. Periorbital puffiness
6. Enlarged heart
7. Anemia
8. Hoarseness in severe cases, due to edema of vocal cords
What are the lab values for Hashimoto's?
Test Value
TSH High
T4 Low (no point in testing T3, which will usually be normal)
CPK (derived from muscle) Elevated (for unknown reasons)
Cholesterol High
How can Hashimoto's be treated?
Hashimoto's is treated with T4 (thyroxine) replacement therapy.
T4 can lead to complications in patients with heart problems. Such patients should be started on very low doses, and gradually be titrated up.
What is the difference between clinical and subclinical hyperthyroidism?
In clinical hyperthyroidism, T3 and T4 are elevated.
In subclinical hyperthyroidism, TSH is high, but T4 levels are within a normal range. There is no indication to treat subclinical hyperthyroidism.
What is Grave's Disease?
Grave's disease is an autoimmune condition in which antibodies that stimulate TSH receptors are produced. It is the most common cause of hyperthyroidism.
About 80% of people with Grave's also test positive for the antibodies to thyroid peroxidase, which are also present in Hashimoto's.
What are the symptoms of Grave's?
1. Goiter
2. Ophthalmopathy, caused by the stimulation of fibroblasts by the thyroid stimulating Ig
3. Localized dermopathy
4. Association with a higher risk of another autoimmune disease
What are the lab test values for Grave's? How is it treated?
Test Value
TSH Low
T3 and T4 High (although, sometimes only T3 will be elevated)
Ca and alkaline phosphatase High (due to bone turnover)
Glucose High (due to insulin resistance)
Thyroid antibodies and thyroid stimulating Ig will be detectible in the blood.
Grave's is treated with steroid therapy, and sometimes by surgical removal of the thyroid.
How can hyperthyroidism be treated?
1. Antithyroid drugs
i. Methimazole
ii. Propylthiouracil (PTU)--not a first-choice drug, because it is associated with rash, urticaria, arthralgia, hepatitis, and agranulocytosis, in rare cases. However, because methimazole cannot be given during pregnancy (it has been correlated with birth defects), PTU becomes the drug of choice during pregnancy, since it binds to large proteins that cannot cross the placenta.
iii. Amiodarone--a new drug that induces thyroid dysfunction
2. Radioactive iodide (I131)--contraindicated in pregnant or nursing women, and only works in patients with normal iodine uptake. Restoration of euthyroidism takes about 8 weeks. Initial treatment coincides with an immediate, transitory worsening of the hyperthyroidism, as destroyed thyroid cells release reserves of hormone into the body.
3. Thyroidectomy--used to treat large goiters, pregnant patients, or patients with severe adverse reactions to anti-thyroid drugs.
What can be used to treat the underlying causes of hyperthyroidism?
1. β blockers, to decrease the heart rate until the drugs start to work
Anti-thyroid drugs take a while to kick in, which the reserve of hormone in the body is used up
2. Glucocorticoids in patients with thyroid storm (severe thyrotoxicosis)
3. Inorganic iodid for patients who will have their thyroid removed soon, or in patients with thyroid storm, one hour after administration of antithyroid drugs, to block the transformation of iodide into thyroid hormone.
4. Cholestyramine--blocks absorption of T4 in the GI tract
5. Potassium percholate--inhibits the discharge of thyroid hormones from the gland. However, potassium percholate is not a drug of choice, because it causes neutropenia and nephrotic syndrome.
How to differentiated b/w thyroiditis and Graves?
Initially, thyroid hormone levels will go up, as the destroyed cells release pre-formed hormone into the bloodstream. Then, this hyperthyroidism will be followed by long-term hypothyroidism, as the thyroid cannot synthesize new hormone.
Clinical indications of early thyroiditis are low TSH and high thyroid hormones. It can be differentiated from Grave's, because unlike a patient with Grave's, a scan of a thyroiditis patient will show low uptake of contrast material in the thyroid gland.

Later stages of thyroiditis involve low thyroid hormone levels, after the released hormone gets used up, and TSH will go up, due to the low thyroid hormone levels.
As TSH levels go up, iodide uptake levels will also go up.
What are risk factors for osteroporosis?
1. Caucasian ethnicity
2. Smoking
3. Low body weight
4. Estrogen deficiency
5. Low Ca intake
6. Alcoholism
What are SERMS? What are examples?
Selective estrogen receptor modulators (SERMs)--SERMs are estrogen analogues that can bind to the estrogen receptor and act as agonists in some organs, but antagonists in others. Two SERMs in common use are tamoxifen and raloxifene:
Tamoxifen is given for breast cancer
Raloxifene is given for osteoporosis, although it has been shown to also decrease the risk of braest cancer
What are drugs for osteoporosis?
1. Biphosphanates
2. Teriparatide
3. Denosumab
What are biphosphanates? How to they work? Examples?
1. Biphosphanates--prevent bone resorption by getting incorporated into the bone and triggering apoptosis of osteoclasts. Biphosphanate action is permanent, and they do not dissociated once they incorporate into the bone. They are used to treat osteoporosis, hypercalcemia, and many malignancies (especially multiple myeloma). They need to be taken orally on an empty stomach. Available biphosphanates are:
i. Alendronate
ii. Risendronate
iii. Ibandronate
iv. Zolendronate (given as a once-yearly injection)
What is teriparatide? How is it used?
a PTH analog, given as a daily injection to treat osteoporosis. Side effects include transient hypercalcemia, headaches, and hypercalciuria. Teriparatide should only used up to 24 months, since its effects do not persist over time (and 'cause it causes sarcoma in rats).
What is denosumab? How does it work?
a new drug (approved this year) that acts as a monoclonal antibody to the RANK ligand. The RANK ligand binds to RANK receptors, and activates osteoclasts. Therefore, Denosumab suppresses osteoclast activity. It is given as an injection every six months, and unlike the biphosphanates, its effects are reversible, and disappear once therapy is stopped.
What are the symptoms of hypercalcemia?
1. GI symptoms (constipation, anorexia, nausea, vomiting, dyspepsia, and rarely, pancreatitis)
2. Renal symptoms (polyuria, polydypsia, nephrolithiasis--in longstanding cases--, nephrocalcinosis
3. Neuromuscular depression, confusion, stupor, coma
4. Bradycardia, AV block, hypertension
How to treat hypercalcemia?
The first step in treating hypercalcemia is hydration (IV saline).
Other steps include:
1. Inhibit bone resorption with high dose biphosphanates and/or calcitonin
2. Inhibit Ca reabsorption in the kidneys with loop diuretics like Furosemide
3. Glucocorticoids, if the hypercalcemia is related to vitamin D intoxication (counteract the effect of vitamin D) or a malignancy affected by glucocorticoids
4. Dialysis, in cases of renal failure

Cinecalcet--a calcium sensing receptor agonist. Used to treat patients with secondary hyperparathyroidism, due to renal failure.
What are the causes and treatments for hypocalcemia?
1. Primary hypoparathyroidism--patients are treated with 1,25 hydroxyvitamin D (Calcitriol)
2. Hereditary--pseudohypoparathyroidism, vitamin D resistant Rickets
3. Iatrogenic--e.g. thyroidectomy, which can damage the parathyroid glands

Hypocalcemia is treated with Ca and calcitriol.
What are the rapid-acting insulins?
Lispro (due to lysine, proline switch(
Aspart
Glulisine
What are the long-acting insulins?
Glargine (lower isoelectric point)
Detemir
What is a synthetic amylin analog? What does it do? What is it for?
Pramlintide

Adjunct to mealtime therapy,
Delays gastric emptying,
Decreases postprandial glucagon secretion
What sulfonylureas?
Tolbutamide
Glyburide (safe for pregnancy)

Promote insulin release from beta cells of pancreas
Blocking K+ channels, leading to Ca influx, reducing hepatic glucose
Risk of hypoglycemia
What are meglintides?
Rapid onset, short action.
Overlapping action w/sulfonylueras
Post-prandial effect.

Rare risk of hyoglycemia either at night or with a missed meal.
What are biguanides?
Metformin

Decreases insulin resistance
Inhibits hepatic gluconeogenesis
Reduces LDL

Preferred for Type II.
Monitor renal function.
What are glitazones?
Insulin sensitizer; insulin required
Targets peroxisome proliferator (PPARy), which regulates adipocyte prodcution, secretion of fatty acids

Pio-- no LDL effect
Resi -- increased LDL
What are a-glucosidase inhibitors?
Type II, preprandial, delays carb digestion

Arabose (also inhibs pancreatic a-amylase)
Miglitol
What are DPP-IV inhibitors?
Sitagliptin

Increases glucose-dependent insulin release, descreases secretion of glucagon
Where are mineralocorticoids produced? How is secretion regulated?
Produced by the adrenal in th zona glomerulosa.

Secretion mainly influenced by renin-angiotensin.

Aldosterone promotes reabsorption of sodium from the distal and collecting renal tubules.
What is spironolactone?
Mineralocorticoid antagonist:
Spironolactone: onset of action is slow with effect lasting for 2-3 days.
Androgen antagonist therefore is used as a treatment of hirsutism in women
Diuretic
CHF
Side effect: hyperkalemia, gynecomastia, menstrual abnormalities, sedation, headaches , skin rashes.
What is glucocorticoid therapy? What for?
Glucocorticoids may be administered as replacement therapy in patients with primary or secondary adrenal insufficiency

adrenal suppression therapy in congenital adrenal hyperplasia

as anti-inflammatory or immunosuppressant therapy in a broad range of mostly nonendocrine disorders affecting many different systems
What is Addison's Disease? How to treat?
Adrenal Insufficency

Hypotension
Hyperpigmentation
Hypoglycemia
Hyponatremia and hyperkalemia

Acute -- IV hydrocortisone
Chonric -- low doses of cortisones
What is CAH?
Group of disorders characterized by specific defects in the synthesis of cortisol.
Decreased cortisol synthesis leads to a compensatory ACTH release . The gland becomes hyperplastic and produces abnormal amounts of precursors.
What is ketoconazole? What does it do? Side effect?
Ketoconazole is an imidazole derivative which was originally developed as an oral anti-fungal agent.

An inhibitor of cortisol secretion by 11ß-hydroxylase inhibition (less inhibition of aldosterone and sex steroid through inhibition of other enzymes)

Principal side effect of ketoconazole is hepatotoxicity.
What is mitotane? What does it do? Side effect?
Mitotane (OPDDD), an isomer of the insecticide DDD (belonging to the same family of chemicals as the insecticide DDT).

Inhibits cholesterol side-chain cleavage and 11-ß-hydroxylase in the adrenal gland.

In treatement for adrenal carcinoma it appears to cause tumor regression and improve survival.

Toxic: adrenal insufficiency, gastrointestinal side effects, nerological disturbance, hepatotoxicity, hypercholesterolemia, rash.
What is aminogletethimide? What does it do? Side effect?
Inhibits: the side-chain cleavage of choleterol to pregnenolone.

It thus inhibits not only cholesterol but also estrogen and aldosterone production.

Inhibits 11b-hydroxylase and 18-hydroxylase

A strong aromatase inhibitor
What is metyrapone? What does it do? Side effect?
Metyrapone acts primarily to inhibit the enzyme 11ß-hydroxylase, thus blocking the production of cortisol from 11-deoxycortisol.

Side effect: increase in androgens
What is etomidate? What does it do? Side effect?
Etomidate is an imidazole derived anesthetic agent.
More potent than ketoconazole (work on similar enzymes)
At higher concentratios it also appears to have an effect on choleterol side-chain cleavage.
Has to ge given parenterally
What are the effects of cortisol?
1. Promotion of normal, intermediary metabolism--upregulate gluconeogenesis, increase amino acid uptake in the liver and kidney, and stimulate protein and lipid catabolism.
2. Raise plasma glucose levels to deal with stress
3. Enhance vasoconstrictive effects of adrenergic stimuli on small blood vessels
4. Decrease the concentration of WBCs (except for PMNs) in the blood by causing them to relocate to lymphatic tissue, increase concentration of hemoglobin, RBCs, platelets, and PMNs
i. This effect has made corticosteroids a treatment option for leukemia
5. Anti-inflammatory effects
i. Indirect inhibition of phospholipase A2, thereby inhibiting prostaglandin synthesis
6. Stimulate TSH secretion
What is the difference between fudrocortisone and spinolacterone?
Fudrocortisone is a mineralocorticoid agonist, with a very high affinity for the aldosterone receptor.
Spironolactone is a mineralocorticoid antagonist, used to treat hyperaldosteronism. Side effects include hyperkalemia and hypertension.
How can Addison's be diagnosed? How is it fatal?
Addison's can be potentially fatal, because patients can become dangerously hypotensive, and go into shock.
Addison's is diagnosed by administering a synthetic ACTH, called cosyntropin. If cortisol levels go up, the patient does not have Addison's. If cortisol levels do not go up, the patient probably does have Addison's.
What are drugs that inhibit steroidogenesis?
1. Mitotane--a derivative of an insecticide called DDD that inhibits cortisol secretion by inhibting 11 β-hydroxylase. Mitotane will destroy the adrenal cells, if given for a long time period, so is usually used for cancers of the adrenal glands.
2. Ketoconazole--also given for fungal infections, but at a lower dose. Ketoconazole also inhibits cortisol secretion by inhibiting 11 β-hydroxylase. The major side effect is hepatotoxicity. Ketoconazole also impairs the cytochrome P450 system, so can interact with other drugs. Unlike mitotane, ketoconazole does not destroy the adrenal cells.
3. Aminglutethymide--used to treat adrenal carcinomas
4. Metyrapone
5. Etomidate
What are drugs that inhibit ACTH secretion?
1. Bromocriptine
2. Cypropheptadine
3. Valproic acid
4. Mifepristone--competitive inhibitor of cortisol
Biguanides are contraindicated in which patients?
-renal dysfunction, indicated by high serum creatinine levels
-CHF (since such patients are more prone to lactic acidosis)
-patients who were given iodinated materal for radiological studies
-patients with acute or chronic metabolic acidosis
-allergic patients
When do meglintides stimulate insulin secretion?
Meglitinides stimulate insulin secretion ONLY in the presence of glucose (which makes them less likely than sulfonylureas to cause hypoglycemic episodes), but their effects are short-lived.
Dopamine
a1 agonist at high doses, b- & D1 receptor agonist at low doses
= vasocontriction, increased HR, contractility

promotes renal perfusion

treatment of severe hypotension and shock
Pilocarpine
muscarinic agonist (like bethanecol and charbacol)

can enter brain, causing CNS disturbances
Neostigmine
more polar than physostigmine, does not enter CNS

used for myasthenia gravis
Nicotine
ganglionic blocker
Reserpine
blocks DA transport into vesicle
Cocaine
Imipramine
Prevents NA reuptake.
bladder (a,b)
detrusor -- b2

sphincter -- a1
phenoxybenzamine
treat pheocytochroma and diffuiculties in urination

reflex tachycardia
orthostatic hypertension
doxazosin
hypertension and those who have difficulty urinating

decreases blood pressure
bleomycin
scisson of DNA by oxidaive process

cell-cyle specific, causes cells to acculuate in G2

PULMONARY TOXICITY
Side effects of nitrosureas
BMD, renal toxicity, pulmonary fibrosis
How can the neutropenia due to taxols be reversed?
treatment with G-CSF Filgrastim
What chemotherapeutric drugs do not cause BMS?
bleomycin
vincristine
hormones
most molecularly-target agents
What does drug permeation depend on?
solubility
concentration gradient
surface area, vascularity
volume of distribution
V - Dose/C (t=0)

Vd is low when a high % of drug is bound to plasma protein
What is zero order elimination?
A constant amount of drug is eliminted per unity time; independependent of plasma concentration

no fixed half life
What is first-order elimination
half life is constant
elimination variable
clearance constant

rate of elimination = GFR + active secretion - reabsorption
Maintanence dose
MD = C(ss) Cl t
-------------------------
f
single dose half life
.7 x Vd/Cl
Multiple Dose

Infusion rate
Cl x C(ss)
loading dose
(Vd x Cp)/f
What is rate of infusion?
It determines plasma level at SS (all takes the same amount of time)
What is the leading dose?
Needed to rapidly acheive effective blood levels
What is affinity?
Ability of drug to bind to receptor, shown by proximity to y axis *if curve are parallel)
Which is a cancer therapy given post-op to destroy microscopic cancer forms?
adjuvant therapy
A breast cancer patient was given Paclitaxel as an adjuvant therapy--what toxicity is associated?
Peripheral neuropathy
Not prescribed for epilepsy?
GABA A agonists
What is D-serine?
An allosteric modulator of metabotropic gluatamate receptrors