Pharmacology Flashcards

1
Q

What is the definition of a receptor?

A

The cellular macromolecule or macromolecular complex with which the drug interacts to elicit a cellular or systemic response

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

What is the definition of potency?

A

Potency is the concentration (EC50) or dose (ED50) of a drug to produce 50% of the drugs maximal effect

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

What is the definition of relative potency?

A

A variant where instead of using units to describe the dose required to reach a certain end point, one ends up using a ratio of equivalent doses

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

What is the definition of efficacy?

A

E(max) is the maximum effect which can be expected from the drug. (When this magnitude is reached, increasing the dose will not increase the magnitude of the effect)

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

What is the definition of intrinsic activity/maximal agonist effect of a drug?

A

The maximal efficacy as a fraction of the maximal efficacy produced by a full agonist of the same type acting through the same receptors in the same condition

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

What actually is a ligand?

A

Usually a small molecule, but they range from ions and small peptides to dissolved proteins

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

What actually is a receptor?

A

A large protein with a 3D structure

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

What does it mean that a ligand and a receptor have molecular complementarity?

A

The shape and chemical properties of their binding sites are matching to permit high-affinity selective binding

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

What are the chemical bonds that make up ligand and receptor binding and give four examples?

A
  1. Van de Waals forces - monoclonal antibodies and their targets
  2. Hydrophobic attraction - suggamadex and rocuronium
  3. Hydrogen bonding - local anaesthetic to a voltage gated sodium channel
  4. Electrostatic attraction - acetylcholine and its receptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is a pharmacophore?

A

The ensemble of steric and electronic features that is necessary to ensure the optimal supramolecular interactions with a specific biological target

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

Think systematically

List the different drug-receptor interactions and give some examples

A

Extracellular
- Soluble extracellular enzymes (dabigatran, perindopril)
Cell Surface
- Cell surface molecules (abciximab)
- Transmembrane non-enzymes (cytokines, interferon gamma)
- Transmembrane proteins with active domains eg receptor kinases (insulin)
- Ligand-gated ion channels (nicotine, suxamethonium)
- Voltage-gated ion channels (lignocaine, verapamil)
- G-protein coupled receptors (dobutamine, metoprolol)
Intracellular
- Soluble intracellular enzymes (GTN)
- Nuclear receptors (corticosteroids, thyroxine)
- Nucliec acids (azithromycin)

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

What are ion channels?

A

They are pore-like transmembrane proteins that alter the local permeability of the cell membrane to ions. Typically, these are fairly selective to which ion they open for

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

How do ligand-gated ion channels work?

A

The binding of a ligand opens the pore, and without the ligand the channel is closed.

Classic examples are acetylcholine and suxamethonium.
Endogenous ligands include serotonin, GABA, glycine and glutamate

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

How do voltage-gated ion channels work?

A

The channels are closed and undergo a conformational change when the transmembrane voltage difference reaches some threshold voltage.

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

How do G-protein coupled receptors work?

A

The G-protein coupled receptor is bound to a GTP-ase protein, which hydrolyses GTP into GDP. When bound to GTP, these proteins become activated, which then allows them to regulate the activity of second messenger systems and amplify the signal of receptor activation

A G-protein is a receptor with seven transmembrane regions, which have their extracellular domain as the receptor. They are made of seven helix domains that stretch back and forth across the membrane

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

How do nuclear receptors work?

A

These receptors, when activated, will bind directly to some sort of ‘response elements’ in the promoter regions of their specific genes. Once it binds to the ligand, the receptor will usually undergo a conformational change which recruits other proteins into a huge multimeric complex, or instead destabilises and deactivates such a complex

These are usually hormone receptors, and their role is to regulate gene transcription. They work slowly.

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

What are the two different types of nuclear receptors and how do they work?

A

Steroid
Generally found in extranuclear cytoplasm, when bound to an agonist they move intranuclear and do their work
Non-steroid
Often intranuclear, found in a heterodimer (bound to other intranuclear receptors and transcription factors)

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

What is an agonist?

A

A ligand that binds to a receptor and alters the receptor state resulting in a biological response

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

What are the 3 different types of agonist?

A

Full agonist
reaches the maximal response capability of the system
Partial agonist
does not reach the maximal response capability of the system even at a full receptor occupancy
Inverse agonist
a ligand that by binding to the receptors, reduces the fraction of them in an active conformation

Note: A partial agonist acts as an antagonist in the presence of a full agonist (if they compete for the same receptors)

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

What does an allosteric modulator do?

A

Increases or decreases the action of a primary agonist whilst having no effect on its own

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

What are spare receptors?

A

They are receptors that exist wherever a full agonist can cause a maximal response when occupying only a fraction of the total receptor population

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

What is an antagonist?

A

A drug that reduced the action of another drug

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

What are the 5 different types of antagonist?

A

Competitive antagonist
competes for the same binding site with an agonist, their binding is mutually exclusive
Non-competitive antagonist
can prevent the action of an agonist without any effect of the binding of the agonist to the receptor
Insurmountable antagonist
can reduce the maximum effect of the agonist, and this inhibitory effect is not affected by increasing agonist concentration
Irreversible antagonist
is insurmountable but it does not have to be non-competitive
Physiological antagonist
non-competitive but it does not have to be insurmountable e.g. something that depresses what the agonist is trying to illicit

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

How does a competitive antagonist effect efficacy and potency?

A

The efficacy is not affected but the potency is increased

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

What is the Schild equation used for?

A

It has some relevance to quantifying the affect of the competitive antagonist on the agonist’s potency. If you plot in as the Schild plot on a graph it can help determine whether one drug is acting as a competitive antagonist against another one

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

What is the Schild equation?

A

C’/C - 1 = [B]/Kb
C’ is concentration of the agonist in the prescence of competitive antagonist
C is concentration of the agonist in the absence of competitive antagonist
[B] is the concentration of the competitive antagonist
Kb is the equilibrium dissociation constant describing the combination of the competitive antagonist with the receptor

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

What is a second messenger?

A

An intermediate molcule for an intracellular signal transduction cascade, which is used to transmit and amplify the signal between an extracellular stimulus and an intracellular effector

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

What are the characteristic features of a second messenger?

A
  • The drug receptor or receptor-ligand interaction often does not result in the direct action of the intracellular effector
  • The intermediate molecule created is synthesised or released in response to the receptor-ligand interaction, and then degraded afterwards
  • The rate of synthesis or degradation of this molecule is tightly regulated to control the magnitude of response to receptor activation, and this regulation can be used to amplify or dampen the response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Where can a second messenger act?

A

It can act locally or it can diffuse distally to convey the signal to a multitude of targets; and multiple secondary messenger systems can interact to produce complex responses to receptor-ligand binding

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

What are the 5 broad types of second messengers?

A
  • Hydrophobic molecules such as DAG and phosphatidylinosotols which do most of their work from the intermembrane space
  • Hydrophilic molecules such as cAMP, cGMP and IP3 which diffuse freely into the cytosol
  • Ions such as ionised potassium, calcium, and sodium
  • Gases such as nitric oxide and carbon monoxide which diffuse easily through lipid and water
  • Soluble proteins such as JAK/STAT, NF-kB
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is cAMP?

A

Cyclic adenosine monophosphate is a cyclic nucleotide secondary messenger

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

How is cAMP produced and degraded?

A

It is produced when G-protein activates adenylyl cyclase.
It is degraded by phosphodiesterases

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

What is cAMPs main targets?

A

Its main downstream targets include protein kinase A (PKA), EPAC and cyclic nucleotide-gated ion channels

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

What is the main action of cAMP?

A

It plays an important role in mediating the response to catecholamines, glycogenelysis, insulin secretion, vision and olfactory sense

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

What are nucleotides made of?

A
  • a phosphate group (or two or three)
  • sugar (classically a pentose sugar such as ribose)
  • a nitrogenous nucleobase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What are the observable effects of cAMP?

A
  • mobilisation of stored energy eg. glycogenolysis
  • vasopressin-mediated water retention
  • parathyroid hormone mediated calcium homeostasis
  • response to catecholamines (beta-adrenergic)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is cGMP?

A

Cyclic guanosine monophospate is a cyclic nucleotide secondary messenger

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

How is cGMP produced and degraded?

A
  • cGMP is produced when guanylyl cyclase is activated by nitric oxide or by a naturetic peptide and this make cGMP from GTP
  • cGMP is degraded by phosphodiesterases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are the main downstream targets of cGMP and briefly tell me their effects?

A

Protein kinase G (PKG) activation
* smooth muscle relaxation by decreased intracellular calcium availability
* negative inotropic effect by reduction of myofilament calcium responsiveness
* increased angiogenesis
cGNP-gated ion channels
* mainly unselective cation channels in retinal and olfactory neuroepithelium and in nephrons
cGMP-modulated phosphodiesterase
* cGMP can bind to phosphodiesterases which increase their activity against both cGMP and cAMP, resulting in the inhibition of both secondary messengers

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

What does PKG do and how does that affect the net effect of cGMP?

A
  • PKG decreases IP3 activity, densisitises myofibrils to calcium, and decreases intracellular calcium availability by several other mechanisms
  • The net effect of cGMP is smooth muscle relaxation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the clinically relevant effects of cGMP?

A

Mainly related to its activation by nitric oxide, leading to increased calcium ion uptake into sarcoplasmic reticulum and a decrease in intracellular calcium, and therefore smooth muscle relaxation

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

Describe the points on the graph including the axis labels, what each line represents and what the dotted lines represent

A

The y-axis is response (% of maximal)
The x-axis is drug concentration (log)
The red line is a full agonist
The blue line is a partial agonist
The outer dotted line is Emax

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

What do the lines on this graph represent?

A

The blue one is a full agonist
The green one is a full agonist with a non-competitive antagonist
The red one is a full agonist with a competitive antagonist

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

What is the definition of volume distribution?

A

The apparent volume into which a drug disperses in order to produce the observed plasma concentration
It is the parameter relating the concentration of a drug in the plasma to the total amount of drug in the body

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

What is volume distribution used for?

A

To calculate loading doses, much as clearance is used to calculate maintenance dose

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

How do you work out the volume distribution?

A

Dose / Plasma concentration

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

What are the units of volume distribution?

A

It can be expressed as Litres (L), or indexed to body mass L/kg

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

What is V-initial/Vc? What affects V-initial?

A

V-initial - Vd of the central compartment (from the rapid distribution phase)
Is often affecting by the degree of protein binding - highly protein-bound drugs with have a high V-initial

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

What is V-extrap? What is it used for?

A

V-extrap - Vd of the tissue compartment (from the elimination phase)
Not used for much!

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

What is V-area?

A

V-area - Vd extrapolated from the area under the curve of the concentration curve

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

How do you calculate Varea?

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

What is V-ss? How do you work it out?

A

V-ss - Vd in a steady state model, often the most useful in calculating the loading dose
Vss = amount of drug in the body in equilibrium conditions/steady state plasma concentrations

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

What are the main molecule factors that influence the volume distribution?

A

Major determinants are drug properties which affect protein binding and tissue binding:
* molecule size
* charge
* pKa
* the lipid/water partition coefficient

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

What are the main patient features that affect volume distribution?

A
  • age
  • gender
  • body muscle/fat proportion
  • level of hydration
  • water distribution (oedema, ascites, APO, pregnancy)
  • extracorpeal sites of distribution (circuit, filters, oxygenation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

How do these things affect Vd?
* Molecule size
* Molecule charge
* pKa
* Lipid solubility
* Water solubility

A
  • Molecule size - large the molecule, harder it is to move, lower Vd
  • Molecule charge - highly ionised molecules, higher water solubility, less likely to move, lower Vd
  • pKa - determines ionisation and lipid solubility
  • Lipid-solubility - highly lipid soluble molecules have a high Vd due to low fat content of the blood stream
  • Water-solubility - difficult to penetrate lipid bilayer, smaller Vd
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What is the definition of half life?

A

(t 1/2) is the time required to reduced the concentration of a drug by a half

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

What is the equation for half life?

A

t 1/2 = 0.693 x Vd/CL
0.693 is the logarithm of 2, it represents the exponential rate of elimination

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

How is half life related to Vd and clearance?

A

An increase in Vd causes an increase in half life
A decrease in the clearance causes an increase in half life

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

How many half lives does it take for a drug to be roughly 97% eliminated?

A

5!
(50% -> 75% -> 87.5% -> 93.75% -> 96.875)

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

How would doubling the dose of a drug affect the half life?

A

It will usually increase its duration of action by one half-life (because it’s clearance is a logarithm function)

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

How does first or zero order kinetics affect half life?

A

First order kinetics drugs have a constant half-life regardless of concentration
With zero order kinetics drugs, the term becomes meaningless, one instead refers to a dose or concentration removed over time

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

What is first order kinetics vs zero order kinetics?

A

First order elimination kinetics - a constant proportion (e.g. percentage) of a drug is eliminated per unit time
Zero order elimination kinetics - a constant amount (e.g. milligrams) of a drug is eliminated per unit time

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

How does concentration affect first order and zero order kinetics?

A

First order kinetics is a concentration dependent process (the higher the concentration, the faster the clearance)
Zero order elimination rate is independent from concentration

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

What is Michaelis-Menten kinetics?

A

It describes enzymatic reactions where a maximum rate of reaction is reached when drug concentration achieves 100% enzyme saturation

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

What is non-linear elimination kinetics?

A

It describes drug clearance by Michaelis-Menten processes, where a drug an low concentration is cleared via first-order kinetics and at high concentratons via zero-order kinetics (phenytoin or ethanol)

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

What is the pharmacology of first order kinetics?

A

This is a logarithmic function. All enzymes and clearance mechanisms are working at well below their maximum capacity, and the rate of drug elimination is directly proportional to drug concentration

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

What does this graph show?

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

What equation can you use to estimate the concentration of the drug at any given time using the semi-logarithmic concentration/time graph?

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

What is the pharmacology mechanism behind Michaelis Menten elimination kinetics?

A

There is a limit on how much enzyme activity there can be before the system becomes saturated.
At low concentrations, the more substrate you give, the faster the reaction. At higher concentrations, the rate of the reaction stays the same because all the enzyme molecules are busy

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

What is the equation of Michaelis Menten kinetics?

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

How does Michaelis Menten elimination kinetics affect the doses we give to patients?

A

When receiving relatively high doses of drugs, a small change in dose will create a disproportionately large change in concentration

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

How is Michealis Menden elimination kinetics relevant to therapeutic index? At what value does it become a narrow index?

A

The drugs at higher doses will have a narrow therapeutic index.
If the drug concentration required to have a useful effect is above 50% of Vmax, the drug will have a narrow therapeutic index

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

How do you calculate the dose rate?

A

Dose rate (mg/hr) = dose (mg) / dosing interval (hrs)

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

How do you calculate the maintenance dose rate?

A

Maintenance dose rate (mg/hr) = desired peak concentration (mg/L) / clearance (L/hr)

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

How do you calculate the loading dose?

A

Loading dose (mg) = desired peak concentration (mg/L) x volume of distribution (L)

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

How do you achieve maintenance dosing in continuous infusions? How long does this take?

A

The drug accumulates gradually. Steady state is achieved when the dose rate and clearance rate are equal.
This takes 3-5 half lives.

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

How do you achieve maintenance dosing in regular dosing? How long does it take?

A

Steady state is achieved in steps, but eventually the dose rate and clearance are equal.
It takes about 5 half-lives

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

Why do you need loading dose?

A

A loading dose rapidly achieves the peak concentration nessecary to equal the clearance, so the desired effect is achieved and maintained sooner.

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

How do you calculate the loading dose?

A

By multiplying the desired peak concentration by the volume distribution.
If the dosing interval is the same as the half-life, the loading dose should be twice the maintenance dose.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

How does oral vs IV administration affect dosing?

A
  • All the loading and maintenace doses have to be adjusted to bioavailability (higher if low bioavailability)
  • The slower instestinal absorption of the drug has a ‘smoothing’ effect on the peaks of concentration, which decreases the concentration-dependent adverse effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are the factors that affect dosing intervals?

A
  • Elimination half life - you often don’t wait til this point as you want a certain level of drug in the blood
  • Therapeutic index
  • Convenience - you wouldn’t expect a patient to take a drug every 10 minutes forever
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is the bioavailability?

A

The fraction of the dose which reaches systemic circulation intact

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

What is absolute vs relative bioavailability?

A

Absolute bioavailability compares one non-IV route with IV administration
Relative bioavailability compares one non-IV route with another non-IV route

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

How is bioavailability measured?

A

Bioavailability is measured using the area under the concentration-time curve (Dost’s Law). The ratio of AUCs is the bioavailability value.
Bioavailability (F) = AUC (oral) / AUC (IV)

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

What does bioequivalent mean?

A

Drugs are considered bioequivalent if the extents and absorption of drugs are so similar that there is likely no clinical difference between their effects

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

What factors affect bioavailability?

A

Generic influences on drug bioavailability
* Drug concentration at side of administration
* Surface area of the absorptive site
* Drug pKa
* Drug molecule size
* pH of the surrounding fluid
Factors affecting GI absorption
* Gastric and intestinal motility
* Tablet disintegration
* Intestinal, bile and bile salt content
* Metabolism by gut wall and by bacteria
* First pass metabolism
Factors affecting first pass metabolism
* Drug absorption and metabolism from the gut
* Metabolism in the gut wall and the bloodstream
* Hepatic blood flow and enzyme activity
Bioavailability from transdermal and mucosal routes
* Mucosal blood flow
* Drug lipophilicity
* Factors affecting membrane penetration
* pH of the mucosal fluid

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

What is morphine’s bioavailiability?

A

Morphine is a drug that has poor bioavailability

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

How is phenytoin’s volume distribution affected and by what?

A

Phenytoin is highly protein-bound, so is highly affected by the low plasma albumin in critically unwell patients

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

How is gentamicin cleared and how does this affect its administration?

A

Gentamicin is rapidly cleared from the kidneys, so is cleared slowly with poor renal function. It is an example of how the doses aren’t changed but the dosing intervals can be extended

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

How is Vancomycin effected by sepsis and why?

A

It has increased renal clearance in the context of hyperdynamic circulatory states, for example in early sepsis so it’s doses need to be closely monitored

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

What sort of molecules are easily renally excreted? What happens to drugs that aren’t?

A

Those that have small molecular volumes, or possess polar characteristics.
These can be readily re-absorbed by the glomerular filtrate.
If they can not be renally excreted, they are often metabolised into molecules that can be

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

Biotransformation reactions can be split into two phases. Phase I and Phase II. What are they?

A

Phase I reactions convert the drug to a more polar environment via oxidation, reduction or hydrolyses

If it is not polar enough, phase II reactions take place, in which an endogenous substrate such as glucuronic acid, sulphuric acid or an amino acid combine with the drug to form a higher polar conjugate

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

Where do biotransformations occur?

A

The principal organ is the liver. Other organs include GI tract, the lungs, the kidneys, the brain and the skin.

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

What is the first pass effect?

A

After oral administration, many drugs are absorbed intact from the small intestine and trasported first via the portal system to the liver, where they undergo extensive metabolism

This is called the first pass effect

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

What cellular organelles house a lot of drug metabolising enzymes?

A

Many drug-metabolising enzymes are housed in the lipophilic endoplasmic reticulum membranes of the liver and other tissues

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

How are microsomes formed and what are their characteristic properties?

A

When the lamellar membranes containing drug enzymes are isolated by homogenisation and fractionation of the cell, they re-form into vesicles called microsomes

Microsomes retain most of the characteristics of intact membranes including a smooth and rough endoplasmic reticulum

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

What two key enzymes are important for oxidative drug metabolism? What do they need to do it?

A

NADPH-cytochrome P450 oxidoreductase
and
Cytochrome P450

Microsomal oxidations require P450, P450 reductase, a reducing agent (NADPH) and an oxygen molecule

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

What happens in the oxidation reaction with cytochrome P450?

A
  1. Oxidised (Fe3+) P450 combines with a drug substrate to form a complex
  2. NADPH donates an electron to the flavoprotein P450 reductase, which reduces the oxidised P450-drug complex
  3. A second electron is donated from NADPH via the same P450 reductase, which reduces oxygen, forming an ‘activated oxygen’-P450-drug complex
  4. This complex transfers activated oxygen to the drug substrate to form the oxidised product
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

There are numerous P450 isoforms, which is the most important one?

A

CYP3A4 metabolises over 50% of prescribed drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What happens if P450 enzyme is induced?

A

Some of the chemically dissimilar P450 substrate drugs, induce P450 expression by enhacing the rate of its synthesis or reducing its rate of degradation

Induction results in accelerated substrate metabolism and usually a decrease in the pharmacological action of the inducer and also of co-administered drugs

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

What are some classic P450 drug substrates?

A

Beta blockers
Warfarin
Oral contraceptives
Statins
Theophylline
Amiodarone
SSRIs
Opioids

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

List some P450 inhibitors

A

INHIBITORS
Sulphonamides
Isoniazid
Cemetidine
K etoconazole
Fluconazole
Alcohol
Ciprofloxacin
Erythromycin
Sodium valproate
.
Chloramphenicol
Omeprazole
Metronidazole**

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

List some P450 Inducers

A

Inducers
Sulphonylureas
Carbamazepine
Rifampicin
Alcohol - chronic
Phenytoin

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

What are the phases of clinical trials?

A
  • Phase 1 - test a new intervention for the first time on a small number of people (20-80) to determine a safe dosage range and identify any side effects
  • Phase 2 - use a small group of volunteers with the disease (several hundred) to determine efficacy and further evaluate safety
  • Phase 3 - use large groups of people (hundreds - thousands) to compare the intervention to other standard interventions
  • Phase 4 - after approval, continue to monitor effectiveness or widely spread adverse effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What is a cardiac arrythmia?

A

Arrythmias represent electrical activity that deviates from the normal synchronous and haemodynamically effective electrical function of the heart as a result of an abnormality in impulse initiation and/or propagation

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

What are some factors that can precipitate or exacerbate arrhythmias?

A
  • ischemia or hypoxia
  • acidosis or alkalosis
  • electrolyte abnormalities
  • excessive catecholamine exposure
  • autonomic influences
  • drug toxicity
  • overstretching of cardiac fibres
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What is the classification of anti-arrhythmics called? What are it’s groups? Give examples for each

A

Vaughn Williams Classification
1. Sodium channel blockers - a) quinine - b) IV lidocaine - c) flecainide
2. Beta blockers - metoprolol, esmolol, bisoprolol
3. K+ channel blockers - amiodarone, sotalol
4. Ca+ channel blockers - verapamil, diltiazem

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

What is a use-dependent or state-dependent drug?

A

It is a drug action that describes when the channels being used more frequently, or are in an inactivated state, are more susceptible to being blocked

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

How do class 1 antiarrhythmics work? What are the subclasses of class 1? - Give an example for each

A
  • Class 1 action is a sodium channel blocker
  • Subclasses of this action reflect effects on the action potential duration (APD).
  • Class 1a drugs prolong the APD and dissociate from the channel with intermediate kinetics - quinine
  • Class 1b drugs shorten the APD in some tissues of the heart and dissociate from the channel with rapid kinetics - IV lidocaine
  • Class 1c drugs have minimal effects on the APD and dissociate from the channel with slow kinetics - flecainide
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

How do class 2 anti-arrhythmics work? Give an example

A

Class 2 action is sympatholytic. Drugs with this action reduce beta-adrenergic activity in the heart - beta blockers - metoprolol, esmolol, bisoprolol

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

How do class 3 anti-arrhythmics work? Give an example

A

Potassium channel blockers manifest as prolongation of the APD. Most drugs with this action block the rapid component of the delayed rectified potassium current. An example is amiodarone

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

How do class 4 anti-arrhythmics work?

A

Calcium channel blockers slow conduction in regions where the action potential upstroke is calcium dependent, e.g. the SA and AV nodes. Examples include verapamil and diltiazem

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

What are the cardiac effects of amiodarone?

A
  • It markedly prolongs the action potential duration (and the QT interval on ECG) by blocking the potassium channels.
  • The APD is prolonged uniformly over a wide range of heart rates, amiodarone does not have a reverse use-dependent action
  • It also significantly blocks inactivated sodium channels. Its action potential prolonging action reinforces this effect.
  • Amiodarone also has weak adrenergic and calcium-channel blocking actions.
  • Consequences of these actions include slowing the heart rate and AV node conduction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What are the extracardiac effects of Amiodarone?

A

Amiodarone causes peripheral vasodilation. This action is prominent after IV administration and may be related to the action of the vehicle

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

What are the toxic effects of amiodarone?

A
  • Symptomatic bradycardia
  • Heart block in patients with known sinus or AV disease
  • Pulmonary fibrosis
  • Abnormal LFTs due to hypersensitivity hepatitis
  • Skin deposits result in photodermatitis and a gray-blue skin discolouration
  • Asymptomatic corneal microdeposits occur in almost all patients after a few weeks
  • Rarely optic neuritis, leading to blindness
  • Hypo or hyperthyroidism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is the pharmacokinetics of amiodarone?

A
  • It is variably absorbed with a bioavailability of 35-60%
  • It undergoes hepatic metabolism, and the major metabolite desethylamiodarone, is bioactive.
  • The elimination half-life is complex with a rapid component of 3-10 days (50% of the drug) and a slower component of several weeks
  • After discontinuation of the drugs, effects are maintained by 1-3 months
  • It is a substrate for liver cytochrome CYP3A4 and inhibits several cytochrome P450 enzymes and may result in high levels of many drugs, including statins, digoxin and warfarin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What are the cardiac effects of verapamil?

A
  • Verapamil blocks both activated and inactivated L-type calcium channels. Thus, it’s effect is more marked in tissues that fire frequently, those that are less completely polarised at rest, and those in which activation depends exclusively on calcium current, such as the SA and AV node
  • It slows the SA node by direct action, but it’s hypotensive action may cause a brief reflex tachycardia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What are the extracardiac effects of verapamil?

A

It causes peripheral vasodilation, which may be helpful in hypertension and peripheral vasospastic disorders

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

What are the effects of verapamil toxicity?

A
  • Verapamil’s cardiotoxic effects are usually dose-related and easily avoided.
  • It can induce AV block when used in large doses or in patients with AV disease
  • When used in VT, hypotension and VF can occur
  • Extracardiac effects include constipation, anxiety and peripheral oedema
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What are the pharmacokinetics of verapamil?

A
  • The half-life of verapamil is approx 4-7 hours
  • It is extensively metabolised by the liver, after oral administration, it’s bioavailability is only about 20%
  • Verapamil dosage is an initial bolus of 5mg over 2-5 minutes, followed by a second bolus if required or a continuous infusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are some other anti-arrhythmics that don’t fit into the conventional classification?

A
  • Adenosine
  • Digoxin
  • Magnesium
  • Potassium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What is the mechanism of action of adenosine?

A
  • Activation of an inward rectified K+ current and inhibition of calcium current.
  • The results of these actions are marked hyperpolarisation and suppression of calcium-dependent action potentials.
  • When given as a bolus, it directly inhibits AV nodal conduction and increases the AV nodal refractory period but has lesser effects on the SA node.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is the half-life of adenosine and what are the usual doses?

A

It’s half-life in the blood is <10 seconds
It is usually given as a bolus of 6mg, followed by 12mg twice if required

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

What are the toxic effects of adenosine?

A
  • SOB or chest burning
  • Feeling like a sense of ‘impending doom’
  • Induction of high-grade AV block
  • AF
  • Headache, hypotension, nausea and paraesthesia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What are the pharmacokinetics of digoxin?

A
  • 65-80% of digoxin is absorbed after oral administration
  • It is not extensively metabolised, almost two thirds is excreted unchanged by the kidney. It’s renal clearance is proportional to creatinine clearance.
  • The half-life is 36-40 hours in patients with normal renal function.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What are the pharmacodynamics of digoxin?

A
  • It inhibits the Na/K+ATPase pump in the cell membranes (that transports sodium)
  • Mechanical effects - increases cardiac contraction (positively inotropic) by increasing the free calcium concentration. It does this by: 1) increasing intracellular sodium concentration by inhibiting the Na/K+ATPase pump and 2) relatively reducing calcium expulsion from the cell by the sodium calcium exchanger caused by the increased intracellular sodium.
  • Electrical effects - causes an early, brief prolongation of the action potential, followed by a shortening. This is probably from the increased potassium conductance caused by high intracellular calcium. At low doses, is has cardioselective parasympathetic effects by sensitising the baroreceptors, causing central vagus stimulation, and facilitation of muscarinic transmission at the nerve ending-myocyte synapse
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What happens during digoxin toxicity?

A
  • Resting membrane potential is reduced as a result of inhibition of the sodium pump and reduced intracellular potassium. As toxicity progresses, oscillatory depolarising afterpotentials appear following normally evoked action potentials.
  • When afterpotentials reach threshold, they elicit action potentials (premature depolarisations ‘ectopic beats’) that are coupled to the preceding normal action potentials.
  • If afterpotentials happen regularly in the Purkinje system, bigeminy will be seen
  • With further toxification, after each of these action potentials there will be a suprathreshold afterpotential, and a self-sustaining tachycardia, which may go onto develop AF or VF.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What extracardiac effects does digoxin have?

A
  • GI effects - anorexia, nausea, vomiting and diarrhoea
  • CNS effects - vagal and chemoreceptor trigger zone stimulation - disorientation and hallucinations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

How do potassium and digoxin interact?

A

They inhibit each others binding to the NA+/K+ATPase pump.
* Hyperkalaemia reduces the effect of digoxin
* Hypokalaemia increases the effect of digoxin
* Increase cardiac automaticity is inhibited by hyperkalaemia, moderately increased potassium therefore reduces the effects of digoxin

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

How do calcium and magnesium interact with digoxin?

A
  • Hypercalcaemia increases the risk of digoxin toxicity
  • Hypermagnesaemia reduces the effects of digoxin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What receptors does adrenaline bind to? What effect does this have?

A
  • It is an agonist for both alpha and beta receptors. It is therefore a very potent vasoconstrictor and cardiac stimulant.
  • It is positively inotropic and chronotropic (predominantly beta 1 receptors) and causes vasoconstriction in many vascular beds (alpha receptors).
  • It also activates beta 2 receptors in some vessels, leading to their vasodilation and therefore decreasing peripheral vascular resistance, explaining the transient hypotension that can follow administration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What receptors does noradrenaline bind to? What effect does this have?

A
  • Noradrenaline is an agonist for both alpha 1 and 2 receptors and also activates beta 1 with a similar potency to adrenaline but has relatively little effect on beta 2 receptors.
  • Consequently, noradrenaline increases peripheral resistance and both systolic and diastolic blood pressures.
  • Compensatory baroreceptor activation tends to overcome the direct positive chronotropic effects of noradrenaline but the positive inotropic effects on the heart are maintained
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What is dopamine and what are the effects of using it?

A
  • It is the immediate precursor in the synthesis of noradrenaline.
  • It’s use promotes vasodilation in the renal, splanchnic, coronary, cerebral via activation of D1 receptors
  • Dopamine activates beta 1 receptors in the heart.
  • High doses of dopamine may mimic the effects of noradrenaline
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What are the tissues where alpha 1 receptors are and the actions they produce?

A
  • Most innervated vascular smooth muscle - contraction
  • Pupillary dilator muscles - contraction
  • Pilomotor smooth muscle - contraction (erects hair)
  • Prostate - contraction
  • Heart - increases contraction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What are the tissues where alpha 2 receptors are and the actions they produce?

A
  • Post-synaptic neurons - probably multiple actions
  • Platelet - aggregation
  • Adrenergic and cholinergic nerve terminals - inhibits transmitter release
  • Some vascular smooth muscle - contraction
  • Fat cells - increase lipolysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What are the tissues where beta 1 receptors are and the actions they produce?

A
  • Heart - inotropic and chronotropic
  • Juxtuglomerular cells - increases renin release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What are the tissues where beta 2 receptors are and the actions they produce?

A
  • Respiratory - promotes smooth muscle relaxation
  • Uterine - promotes smooth muscle relaxation
  • Vascular smooth muscle - promotes smooth muscle relaxation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What are the tissues where beta 3 receptors are and the actions they produce?

A

Bladder - relaxes detrusor muscle

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

What are the tissues where delta 1 and 2 receptors are and the actions they produce?

A
  • Delta 1 - smooth muscle - dilates renal blood vessels
  • Delta 2 - nerve endings - modulates transmitter release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are the three drug groups traditionally used in angina?

A

Organic nitrates
Calcium channel blockers
Beta blockers

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

What are the pharmacokinetics of nitrates?

A
  • The liver contains a high-capacity nitrate reductase that ultimately inactivates the drug, therefore oral bioavailability is low (<10-20%)
  • The sublingual route is preferred as it avoids the first-pass effect. Both nitroglycerin and isosorbide dinitrate reach therapeutic blood levels within a few minutes via this route
  • The duration of effect is 15-30 minutes.
  • Other routes include oral, transdermal and buccal absorption.
  • Once absorbed, the half-life is only 2-8 minutes
  • The bioavailability of isosorbide mononitrate is 100% and it is metabolised to isosorbide dinitrate
  • Excretion is largely by the kidney
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What is the pharmacodynamics of nitrates mechanism of action in smooth muscle?

A
  • The drug must be bioactivated with the release of nitric oxide
  • Nitroglycerin can be denitrated by glutathione S-transferase in smooth muscle and other cells
  • A mitochondrial enzyme, aldehyde dehydrogenase isoform 2 (ALDH2) and ALDH3 appears to be key in the activation and release of nitric oxide from nitroglycerin and pentarythritol tetranitrate.
  • Free nitrate ion is released, which is then converted to NO. NO combines with the heme group of soluble guanylyl cyclase, activating it and causing an increase in cGMP.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What are the pharmacodynamics of vascular effects of nitrates?

A

All segments of the vascular system from large arteries through to large veins relax in response to nitroglycerin. With veins responsing at the lowest concentrations and arteries at slightly higher ones.
The epicardial coronary arteries are sensitive, but concentric atheromas can present significant dilation.
On the other hand, eccentric lesions permit an increase in flow when nitrates relax the smooth muscle on the side away from the lesion.

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

How do nitrates affect the veins and how does this effect cardiac output?

A

There is marked relaxation of veins, with increased venous capitance and decreased ventricular preload. Pulmonary vascular pressures and heart size are significantly reduced. In the absense of heart failure, cardiac output is reduced. In heart failure, preload is often abnormally high; by reducing preload, it may have a beneficial effect on cardiac output in this condition
Because venous capitance is increased, orthostatic hypotension may be marked and syncope can result

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

What are the indirect effects of nitrates and what does this lead to?

A

The indirect effects of nitroglycerin consist of those compensatory responses evoked by baroreceptors and hormonal mechanisms responsing to decreased arterial pressure, this often results in tachycardia and increased cardiac contractility. Retention of salt and water may also be significant, especially with intermediate and long acting nitrates.

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

How do nitrates affect the arteries and how does this add to side effects?

A

Dilation of large epicardial coronary arteries may improve oxygen delivery in the presence of eccentric atheromas or collateral vessels.
Temporal artery pulsations and a throbbing headache associated with meningeal artery pulsations are common effects of GTN.

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

How do nitrates affect other smooth muscle organs?

A

Relaxation of smooth muscle of the bronchi, GI tract and GU tract have been demonstrated experimentally but have limited clinical effect due to the short half-life. But, recently nitrates have been used to enhance erections. The resulting increase in cGMP causes relaxation in the erectile tissue.

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

What are the effects of nitrates on the action of platelets?

A

NO released from the nitroglycerin stimulates guanylyl cyclase in platelets as in smooth muscle. The increase in cGMP that results is responsible for a decrease in platelet aggregation.

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

What are the acute adverse effects of nitrates?

A

Orthostatic hypotension
Tachycardia
Throbbing headache

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

What are the contraindications to nitrates?

A

Elevated intracranial pressure
Rarely, transdermal GTN patches have ignited during DCR so they should be removed during shocks

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

How does tolerance of nitrates occur?

A

With continuous exposure to nitrates, isolated smooth muscle may develop complete tolerance progressively with long-acting preparations or continuous infusions.
Diminished release of NO resulting from reduced bioactivation may be partly responsible.
Systemic compensation also may play a role

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

What are the nitrate effects in angina of effort?

A
  • Decreased venous return to the heart and resulting reduction of intracardiac volume are important beneficial haemodynamic effects of nitrates
  • Arterial pressure also decreases.
  • Decreased intraventricular pressure and left ventricular volume are associated with decreased wall tension and decreased myocardial oxygen requirement.
  • IV or sublingual nitrate administration consistently increases the caliber of the large epicardial coronary arteries except where blocked by concentric atheromas.
  • Coronary arteriolar resistance tends to decrease, though to a lesser extent
  • The reduction in oxygen demand is the major mechanism for the relief of effort angina
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What are the nitrate effects in variant angina?

A

Relaxing the smooth muscle of the epicardial coronary arteries and relieving coronary artery spasm

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

What are the nitrate effects in unstable angina?

A

Both by dilating the epicardiac coronary arteries and simultaneously reducing myocardial oxygen demand.
Also by decreasing platelet aggregation

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

What are the different ways of administrating nitrates?

A

Short acting Sublingual
Long acting oral
Long acting transdermal
Slow release buccal
Slow release sublingual

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

What is the pharmacokinetics of calcium channel blockers?

A

They are orally active agents and are characterised by high first-pass effect, high plasma protein binding and extensive metabolism
Verapamil and Diltiazem are also used by the IV route

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

What is the mechanism of action of calcium channel blockers?

A

The voltage-gated L type calcium channel blocked in cardiac and smooth muscle consists of α1, α2, β, γ, and δ subunits.
Nifedipine and other dihydropyridines, verapamil and diltiazem all bind to the α1 unit.
The drugs act on the inner side of the membrane and bind more effectively to open channels and inactivated channels.
Binding of the drug reduces the frequency of opening in response to depolarisation. The result is a marked decrease in transmembrane calcium current, which in smooth muscle results in long-lasting relaxation and in cardiac muscle results in reduction in contractility thoughout the heart and decreases in sinus node pacemaker rate and AV node conduction velocity.

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

What are the smooth muscle effects of calcium channel blockers?

A

The cells are relaxed by the calcium channel blockers. Vascular smooth muscle appears to be the most sensitive, but similar relaxation can be shown to bronchiolar, GI and uterine smooth muscle.
Blood pressure is reduced.
The reduction in peripheral vascular resistance is one mechanism by which these agents may benefit the patient with angina

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

Name some calcium channel blockers

A

Dihydropyridines
* Amlodipine
* Nifedipine

Miscellanous
* Diltiazem
* Verapamil

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

How do dihydropyridines and other calcium channel blockers affect smooth muscle differently?

A

In general, the dihydropyridines have a greater ratio of vascular smooth muscle effects relative to cardiac effects than diltiazem and verapamil.

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

How do calcium channel blockers affect cardiac muscle?

A

Excitation-contraction coupling in all cardiac cells requires calcium influx, so these drugs reduce cardiac contractility in a dose-dependent fashion.
In some cases, cardiac output may also decrease. This reduction in cardiac mechanical function is a way in which the calcium channel blockers can reduce the oxygen requirement in patients with angina.
Impulse generation in the SA node and conduction in the AV node may be reduced or blocked by calcium channel blockers

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

How do the different calcium channel blockers differ with regards to their actions on the heart?

A

Verapamil and diltiazem block tachycardias in calcium-dependent cells (the AV node) more selectively than the dihydropyridines.
On the other hand, the dihydropyridines appear to block smooth muscle calcium channels more than diltiazem and verepamil so are therefore less depressant on the heart

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

Why is skeletal muscle not affected by calcium channel blockers?

A

Because it uses intracellular pools of calcium to support excitation-contraction coupling and does not require as much transmembrane calcium influx.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What are the toxic effects for calcium channel blockers?

A

Serious but rare: cardiac depression including bradycardia, AV block, cardiac arrest and heart failure.
Troublesome but not harmful - flushing, dizziness, nausea, constipation and peripheral oedema

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What are the half-lives of the following calcium channel blockers?
* Amlodipine
* Nifedipine
* Diltiazem
* Verapamil

A

Amlodipine - 30-50 hours
Nifedipine - 4 hours
Diltiazem - 3-4 hours
Verapamil - 6 hours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What are the mechanisms of the clinical effects of calcium channel blockers?

A
  • CCBs decrease myocardial contracile force, which reduces myocardial oxygen requirements
  • Calcium channel block in arterial smooth muscle decreases arterial and intraventricular pressure
  • Peripheral vasodilation causes a decline in left ventricular wall stress, which reduces myocardial oxygen requirements
  • Decreased heart rate decreases oxygen requirements
  • Verapamil and diltiazem decrease AV node conduction, often being effective in the management of SVT and in decreasing the ventricular rate in AF or A flutter
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What is the effect of CCBs in overt heart failure?

A

They can cause worsening of failure as a result of their negative inotropic effects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What is the mechanism of action of beta blockers in angina?

A

They decrease heart rate, blood pressure and contractility which decreases myocardial oxygen requirements at rest and during exercise.
Lower heart rate is also associated with an increase in diastolic perfusion time that may increase coronary perfusion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

What are the contraindications to the use of beta blockers?

A

Asthma and other bronchospastic conditions, severe bradycardia, AV block, bradycardia-tachycardia syndrome and unstable left ventricular failure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What are some adverse effects of beta blockers?

A

Fatigue
Impaired exercise toleracne
Insomnia
Unpleasant dreams
Worsening of claudication
Erectile dysfunction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

What is the mechanism and site of action of propranolol?

A
  • It is a non-selective β blocker.
  • Decreases BP primarily as a result of a decrease in cardiac output.
  • Inhibits the stimulation of renin production by catecholamines (mediated by β1 receptors)
  • May also act on peripheral presynaptic β adrenoreceptors to reduce sympathetic vasoconstrictor nerve activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

What are the pharmacodynamics of propranolol?

A
  • Bioavailibility - 25%
  • Half-life - 3-5 hours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

How does the mechanism of metoprolol compare to that of propranolol?

A

Metoprolol is approximately equipotent to propranolol in inhibiting stimulation of β1 adrenoreceptors in the heart but 50-100-fold less potent than propranolol in blocking β2 receptors because it in a β1 selective beta blocker, therefore is better for patients with asthma or peripheral vascular disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

What are the pharmacodynamics of metoprolol?

A
  • It is extensively metabolised by CYP2D6 with high first-pass metabolism.
  • Has a relatively short half-life (4-6 hours) but the extended release preparation can be dosed once daily
  • Dose is not affected by renal function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What are the pharmacodynamics of atenolol?

A
  • It is β1 selective
  • It is not extensively metabolised and is excreted primarily in the urine with a half-life of 6 hours
  • It is usually dosed once daily - it is reported to be less effective than metoprolol
  • Patients with reduced renal function should recieve lower doses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What are the pharmacodynamics of nadolol and carteolol?

A
  • Non-selective β-receptors antagonists
  • Not appreciably metabolised and are excreted to a considerable extent in the urine
  • Patients with reduced renal function should receive lower doses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What are the pharmacokinetics and pharmacodynamics of betaxolol and bisoprolol?

A
  • β1-selective blockers
  • Primarily metabolised in the liver
  • Have long half-lives
  • Can be administered once daiy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What are the mechanisms of action of labetalol, carvedilol and nebivolol?

A

These drugs have both β-blocking and vasodilating effects.
* Labetalol is a β and α blocker. It has a 3:1 ratio of β:α antagonism after oral dosing. BP is lowered by reduction of systemic vascular resistance (α blockade) without significant alteration in HR or cardiac output. It is usually given OD or IV as required
* Carvedilol is a non-selective β-adrenoceptor blocker and α blocker. The average half life is 7-10 hours and is usually given BD. It is useful in heart failure and HTN together
* Nebivolol is a highly β1-selective blocker with vasodilating properties that are not mediated by α blockage. It causes a decrease in peripheral vascular resistance, causing vasodilation. It is extensively metabolised and has active metabolites. The half-life is 10-12 hours but it can be given OD. It is thought to have less adverse effects that other antihypertensives

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What are the pharmacodynamics of esmolol?

A
  • It is a β1-selective blocker that is rapidly metabolised
  • It has a short half-life of 9-10 minutes and is given IV, often as a loading dose then continuous infusion
  • It is often used intra-operatively
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What is a ‘diuretic’ vs a ‘natiuretic’ vs an ‘aquaretic’?

A

A diuretic is an agrent that increases urine volume.
A natiuretic causes an increase in renal sodium excretion
A aquaretic increases excretion of solute-free water

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

Where is carbonic anhydrase present in the kidney and what is its function?

A
  • It is present in many nephron sites, but the predominant location is the epithelial cells of the proximal convoluted tububle.
  • It catalyses the dehydration of H2CO3 to CO2 at the luminal membrane and rehydration of CO2 to H2CO3 in the cytoplasm.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What are some examples of carbonic anhydrase inhibitors and how do they cause diuresis?

A
  • The prototypical carbonic anhydrase inhibitor is acetazolamide.
  • By blocking carbonic anhydrase, inhibitors blunt NaHCO3 reabsorption and cause diuresis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

What are the pharmacokinetics of carbonic anhydrase inhibitors?

A
  • They are well absorbed after oral administration.
  • An increase in urine pH from the HCO3 diuresis is apparent within 30 minutes, maximal at 2 hours and persists for 12 hours after a single dose.
  • Excretion of the drug is by secretion in the proximal tubule S2 segment, therefore dosing must be reduced in renal insufficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What are the pharmacodynamics of carbonic anhydrase inhibitors?

A
  • Inhibition of carbonic anhydrase activity profoundly depresses HCO3 reabsorption in the PCT. At maximal safe inhibitor dosage, 85% of the HCO3 reabsorptive capacity of the superficial PCT is inhibited.
  • Therefore, it causes significant HCO3 losses and hyperchloremic metabolic acidosis.
  • Because of reduced HCO3 in the glomerular filtrate and the fact that HCO3 depletion leads to enhanced NaCl reabsorption by the remainder of the nephron, the diuretic efficacy decreases significantly with use over several days.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What are some of the non-diuretic clinical indications for carbonic anhydrase inhibitors?

A
  • Glaucoma - the reduction of aqueous humor formation by carbonic anhydrase inhibitors decreases the intraocular pressure
  • Urinary alkalisation - uric acid and cystine may form stones in acidic urine, it can be used in the first few days
  • Metabolic acidosis - when the alkalosis is due to excessive use of diuretics with HF patients, IV replacement is contraindicated and acetazolamide can be used to rapidly correct the metabolis alkalosis and correction of volume overload
  • Acute mountain sickness - in serious cases with cerebral oedema, carbonic anhydrase inhibitors can decrease CSF formation and by decreasing the pH of the CSF and brain, it can increase ventilation and diminish symptoms.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What are the adverse effects of carbonic anhydrase inhibitors?

A
  • Hyperchloremic metabolic acidosis - predictably from the chronic reduction of body HCO3 stores
  • Renal stones - calcium phosphate salts are relatively insoluble in alkalike pH, which means the potential for renal stone formation is higher
  • Renal potassium wasting occurs because the increased Na+ presented to the collecting tubule (with HCO3) is partially reabsorbed, increasing the lumen-negative electrical potential in that segment and enhacing K+ secretion.
  • Drowsiness and paresthesias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What are the pharmacodynamics of SGLT2 inhibitors?

A

In the normal individual, the PCT reabsorbs almost all of the glucose filtered by the glomeruli. 90% of the glucose reabsorption occurs through SGLT2, but inhibiting this transporter using the currently available drugs will result in glucose excretion of only 30-50% of the amount filtered

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

How does Angiotensin II affect SGLT2 inhibitors?

A

Angiotensin II has been shown to induce SGLT2 production via the AT1 receptor. Thus, blockade of the RAA axis may result in lower SGLT2 availability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What are the pharmacokinetics of SGLT2 inhibitors?

A
  • They are rapidly absorbed by the GI tract.
  • The elimination half-life of dapagliflozin is 10-12 hours
  • Up to 70% of the give dose is excreted in the urine in the form of 3-O-glucuronide
  • The drugs are not recommended in patients with more severe renal failure or advanced liver disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

Give some examples of SGLT2 inhibitors?

A

Dapagliflozin
Canagliflozin
Empagliflozin
Ipragliflozin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

On a very basic level, how do loop diuretics work?

A

Loop diuretics selectively inhibit NaCl reabsorption in the thick ascending loop.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What are the pharmacokinetics of loop diuretics?

A
  • The are rapidly absorped.
  • They are eliminated by the kindey by glomerular filtration and tubular secretion.
  • Absorption of oral torsemide is more rapid (1 hour) that than of furosemide (2-3 hours) and is nearly as complete with IV administration.
  • The duration of effect for furosemide is usually 2-3 hours. The effects of torsemide lasts 4-6 hours.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What are the pharmacodynamics of loop diuretics?

A
  • Loop diuretics inhibit NKCC2, the luminal Na/K/2Cl transporter in the thick ascending loop of Henle.
  • Thus, the loop diuretics reduce the absorption of NaCL and also diminish the lumen-positive potential that comes from K+ recycling.
  • This positive potential normally drives divalent cation reabsorption in the TAL, and by reducing this potention, loop diuretics cause an increase in Mg and Ca excretion.
  • Loop diuretics have also been shown to induce the expression of COX-2, increasing PGE2, which inhibits salt transport in the TAL.
  • They increase renal blood flow via prostaglandin actions on kidney vasculature
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

How can loop diuretics be used in hypercalcaemia?

A

Loop diuretics can cause an increase in Mg and Ca excretion.
In disorders that cause hypercalcaemia, Ca excretion can be enhanced by treatment with loop diurectics combined with saline infusion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

How can NSAIDs interfere with loop diuretics? What group of patients does this effect?

A

Loop diuretics induce the expression of COX-2.
NSAIDs blunt COX activity, which can interfere with the actions of loop diuretics by reducing prostaglandin synthesis in the kidney.
This interference is minimal is healthy patients but may be significant in patients with nephrotic syndrome of hepatic cirrhosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

How do loop diuretics affect pulmonary congestion and left ventricular filling?

A
  • Loop agents have direct effects on blood flow through several vascular beds via prostaglandin actions.
  • They reduce pulmonary congestion and left ventricular filling pressures in heart failure before a measurable increase in urinary output occurs.
  • Their effects on peripheral vascular tone are also due to release of renal prostaglandins that are induced by the diuretics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

Apart from acute pulmonary oedema and other oedematous conditions, what are the other clinical indications of furosemide?

A
  • Hyperkalaemia - loop diuretics can significantly enhance urinary excretion of K+.
  • AKI - loop agents can increase the rate of urine flow and enhance K+ excretion in acute renal failure.
  • Anion overdose - they are useful in treating toxic ingestions of bromide, fluoride and iodide, which are reabsorbed in the TAL. Saline solution must be administered to replace urinary losses of Na and to provide CL, so as to avoid extracellular fluid volume depletion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

What are the adverse effects of loop diuretics?

A
  • Hypokalaemic metabolic alkalosis - by inhibiting salt reabsorption in the TAL, loop diuretics increase Na delivery to the collecting duct. Increase Na delivery leads to increased secretion of K+ and H+ by the duct, causing hypokalaemic metabolic alkalosis.
  • Ototoxicity - occasionally cause dose-related hearing loss that is usually reversible
  • Hyperuricaemia and precipitated attacks of gout, cause by hypovolaemia-associated enhancement of uric acid reabsorption in the proximal tubule.
  • Hypomagnesaemia is a predictable consequence of the chronic use of loop agents
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What are the pharmacokinetics of thiazides?

A

All thiazides can be administered orally, but there are differences in their metabolism:
* Chlorothiazide, the parent of the group, is not very lipid-soluble and must be given in relatively large doses. It is the only thiazide available for parenteral administration.
* Hydrochlorothiazide is considerably more potent and should be used in much lower doses

All thiazides are secreted by the organic acid secretory system in the proximal tubule and compete with the secretion of uric acid by that system. As a result, thiazide may blunt uric acid secretion and elevate serum uric acid level.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

What are the pharmacodynamics of thiazides?

A
  • Thiazides inhibit NaCl reabsorption from the luminal side of epithelial cells in the DCT by blocking Na/Cl transporter (NCC).
  • Thiazides enhance Ca reabsorption, likely due 1) in the proximal tubule, thiazide-induced volume depletion leads to enhanced Na+ and passive Ca2+ reabsorption; 2) in the DCT, lowering of intracellular Na+ by thiazide-induced blockade of Na+ entry enhances Na/Ca exchange in the basolateral membrane and increases overall reabsorption of Ca2+.
  • The action of thiazides depends in part of renal prostaglandin production, like loop diurectics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What are the main clinical indications for thiazide diuretics?

A

HTN
HF
Nephrolithiasis due to idiopathy hypercalciuria
Nephrogenic diabetes insipidus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What are the adverse effects of thiazides?

A
  • Hypokalaemic metabolic alkalosis - similar to loop diuretics
  • Impaired carbohydrate tolerance - due to both impaired pancreatic release of insulin and diminished tissue utilisation of glucose.
  • Hyperlipidaemia
  • Hyponatraemia due to a combo of hypovolaemia-induced elevation of ADH, reduction in the diluting capacity of the kidney, and incresed thirst
  • Impaired uric acid metabolism & gout
  • Serious but rare: haemolytic anaemia, thrombocytopaenia, acute necrotising pancreatitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

Name some thiazide diuretics

A

Bendroflumethiazide
Hydrochlorothizaide
Indapamide
Chlorothiazide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

Name some potassium-sparing diuretics

A

Spironolactone
Amiloride
Eplerenone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

Briefly, how do potassium-sparing diuretics work?

A

The prevent K+ secretion by antagonising the effects of aldosterone in collecting tubules.

  • Inhibition may occur by direct pharmacologic antagonism of mineralocorticoid receptors (spironolactone, eplerenone)
    OR
  • By inhibition of Na+ influx through ion channels in the luminal membrane (amiloride, triamterene).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

What are the pharmacokinetics of spironolactone?

A

Spironolactone is a synthetic steroid that acts as a competitive antagonist to aldosterone.

  • Onset and duration of its action are determined substantially by the active metabolites canrenone and 7-α-spironolactone, which are produced in the liver and have long half-lives (12-20 hours and approx 14 hours, respectively).
  • Spironolactone binds with high affinity and potently inhibits the androgen receptor, which is an important side effect in males (gynaecomastia and decreased libido)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What are the pharmacokinetics of eplerenone and how are these different from spironolactone?

A

It is a spironolactone analog with much greater selectivity for the mineralocorticoid receptor.
It is several hundredfold less active on androgen and progesterone receptors that spironolactone and therefore, eplerenone has considerably fewer adverse effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What are the pharmacodynamics of potassium-sparing diuretics?

A

They reduce sodium absorption in the collecting tubules and ducts. Potassium absorption (and K+ secretion) at this site is regulated by aldosterone.

  • Spironolactone and eplerenone bind to mineralocorticoid receptors and blunt aldosterone activity.
  • Amiloride and triamterene do not block aldosterone but instead directly interfere with Na+ entry through the epithelial Na+ channels in the apical membrane of the collecting tubule.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What are the clinical indications of K+ sparing diuretics?

A
  • Potassium-sparing diuretics are most useful in states of mineralocorticoid excess or hyperaldosteronism due to primary hypersecretion (Conn’s syndrome, ectopic ACTH production) or secondary hyperaldosteronism (evoked by heart failure, hepatic cirrhosis, nephrotic syndrome)
  • Eplerenone may interfere with some of the fibrotic and inflammatory effects of aldosterone. Thus, it can slow the progression of albuminuria in diabetic patients.
  • Eplerenone has been found to reduce myocardial perfusion defects after MI
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

What are the adverse effects of K+ sparing diuretics?

A
  • Hyperkalaemia - they reduce urinary excretion of K+, the risk of this is greatly increased by renal disease or by use of other drugs that reduce or inhibit renin (beta blockers, NSAIDs) or angiotensin II activity (ACE inhibitors, angiotensin receptor inhibitors
  • Hyperchloraemic metabolic acidosis - by inhibiting H+ secretion in parallel with K+ secretion
  • Gynaecomastia - synthetic steroids may cause endocrine abnormalities by actions on other steroid receptors.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

What are some contraindications to potassium sparing diuretics?

A

Chronic renal infufficiency due to risk of K+
Patient with liver disease may have impaired metabolism of triamterene and spironolactone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

How do osmotic diuretics work?

A

The proximal tubule and descending limb of Henle’s loop are freely permeable to water. Any osmotically active agent that is filtered by the glomerulus but not reabsorbed causes water to be retained in these segments and promotes a water diuresis.
Such agents can be used to reduce intracranial pressure and to promote prompt removal of renal toxins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

What is an example of an osmotic diuretic?

A

Mannitol

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

What are the pharmacokinetics of mannitol?

A
  • Mannitol is poorly absorbed by the GI tract, and when administered orally, it causes osmotic diarrhoea rather than diuresis. It must be given IV
  • Mannitol is not metabolised and is excreted by glomerular filtration within 30-60 minutes, without any important tubular reabsorption or secretion
  • It must be used cautiously in patients with even mild renal insufficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

What are the pharmacodynamics of mannitol?

A
  • Osmotic diuretics have their major effect in the proximal tubule and the descending limb of Henle’s loop.
  • Through osmotic effects, they also oppose the action of ADH in the collecting tubule.
  • The presence of a non-reabsorbable solute such as mannitol prevents the normal absorption of water by interposing a countervailing osmotic force. As a result, urine volume increases.
  • The increase in urine flow decreases the contact time between fluid and the tubular epithelium, thus reducing Na+ as well as water reabsorption.
  • The resulting natuiresis is of lesser magnitude than the water diuresis, leading eventually to excessive water loss and hypernatraemia.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

What are the clinical indications of mannitol?

A

Osmotics diuretics alter Starling forces so that water leaves cells and reduces intracellular volume. This effect is used to reduce intracranial pressure in neurologic conditions and to reduced intraocular pressure before ophthalmologic procedures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

What is the dose of mannitol in reducing ICP and how quick does it work?

A

A dose of 1-2g/kg mannitol is administered IV.
ICP should fall in 60-90 minutes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

When could mannitol be using in dialysis?

A

At times, the rapid lowering of serum osmolality at initiation of dialysis results in symptoms. Many nephrologists also use mannitol to prevent adverse reactions when first stating patients on haemodialysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

What are the adverse effects of mannitol?

A
  • Extracellular volume expansion - mannitol is rapidly distributed in the extracellular compartment and extracts water from cells. Prior to the diuresis, this leads to expansion of the extracellular volume and hyponatraemia. This may complicate heart failure and may produce florid pulmonary oedema. Headache, nausea and vomiting are commonly observed
  • Dehydration, hyerkalaemia and hypernatreamia - excessive use of mannitol without adequate water replacement can ultimately lead to severe dehydration, free water losses, and hypernatraemia. As water is exctracted from cells, intracellular K+ concentration rises, leading to cellular losses and hyperkalaemia.
  • Hyponatraemia - when used in patients with severe renal impariment, parenterally administered mannitol cannot be excreted and is retained in the blood. This causes osmotic extraction of water from cells, leading to hyponatraemia without a decrease in serum osmolality
  • Acute renal failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

What are indirect thrombin inhibitors? Name some. How do they work?

A

The indirect thrombin inhibitors are called this because their antithrombotic effect is exerted by their interaction with a separate protein, antithrombin. Examples include:
* Unfractionated heparin (also known as high molecular weight heparin)
* LMWH
* Fondaparinux

They bind to antithrombin and enhance its inactivation of factor Xa.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

What is the chemical make up of heparin? What is it’s mechanism of action

A

It is a heterogenous mixture of sulfated mucopolysaccharides.

It binds to endothelial cell surfaces and a variety of plasma proteins. Its biological acitivity is dependent upon the endogenous anticoagulant antithrombin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

How does antithrombin work? How does heparin effect it?

A
  • Antithrombin inhibits clotting factor proteases, especially thrombin (IIa), IXa, Xa, by forming equimolar stable complexes with them.
  • In the absence of heparin, these reactions are slow; in the presence of heparin, they are accelerated 1000-fold.
  • The active heparin molecules bind tightly to antithrombin and cause a conformational change in this inhibitor
  • The conformational change of antithrombin exposes its active site for more rapid interaction with the proteases (activated clotting factors)
  • Herpain functions as a cofactor for the reaction wihtout being consumed. Once the complex is formed, heparin is released intact for renewed binding to more antithrombin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

How is HMWH different from LMWH?

A
  • HMWH with high affinity for antithrombin markedly inhibit blood coagulation by inhibiting all three factors, especially thrombin and factor Xa
  • LMWH inhibits activated factor X but has less effect on thrombin than the HMWH.
  • LMWH have equal efficacy, increased bioavailability from the subcut site of injection, and less frequent dosing requirements
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

How do you monitor heparin effect?

A
  • Close monitoring of PTT or anti-Xa units is necessary in patients received unfractionated heparin.
  • Weight-based dosing of LMWH results in predictable pharmacokinetics so the levels are not generally measured except in renal insufficiency, obesity and pregnancy, where it can be determined by anti-Xa levels.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What are the adverse effects of heparin?

A
  • Bleeding risk, osteoporosis, alopecia, mineralocorticoid deficiency
  • HIT - a systemic hypercoagulable state that occurs in 1-4% of individuals treated with UFH.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

What are the contraindications to heparin?

A
  • HIT
  • hypersensitivity to the drug
  • active bleeding
  • haemophilia
  • thrombocytopaenia
  • HTN
  • ICH
  • infective endocarditis
  • TB
  • abortion
  • advanced hepatic or renal disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

What is the dosing for continuous IV administration of heparin?
Whats the ideal anti-Xa level?

A

After an inital bolus of 80-100 units/kg, a continuous infusion of about 15-22 units/kg per hour is required to maintain the anti-Xa activitiy in the range of 0.3-0.7units/ml

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

What is the prophylactic dose of enoxaparin?
What is treatment dose of enoxaparin? What level of anti-factor Xa does this aim for?

A

Prophylatic dose - 30mg BD or 40mg OD subcut
Treatment dose - 1mg/kg subcut every 12 hours for an anti-Xa level of 0.5-1 unit/ml.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q

What is the prophylactic and treatment dose of dalteparin?

A
  • Prophylactic dose 5000 units subcut OD
  • Treatment dose 200 units/kg OD for venous disease or 120 units/kg BD for ACS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

What are the pharmacokinetics of fondaparinux?

A
  • It actively bind antithrombin with high specific activity, resulting in efficient inactivation of factor Xa.
  • It has a long half-life of 15 hours, allowing for OD dosing subcut
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

What are the pharmacokinetics of warfarin?

A
  • It is generally administered orally and has 100% oral bioavailability
  • Over 99% of racemic warfarin is bound to plasma albumin, which may contribute to its small volume of distribution, its long half-life in plasma (36 hours) and lack of unirary excretion of unchanged drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What is the mechanism of action of warfarin?

A
  • It blocks the γ-carboxylation of several glutamate residues in prothrombin and factors VII, IX, and X as well as the endogenous anticoagulant proteins C and S.
  • The blockade result in incomplete coagulation factor molecules that are biologically inactive.
  • The protein carboxylation reaction is coupled to the oxidation of vitamin K. The vitamin must then be reduced to reactivate it.
  • Warfarin prevents reductive metabolism of the inactive vitamin K epoxide back to its active hydroquinone form.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

How long does warfarin take to work? Why?

A
  • There is an 8 to 12 hour delay in the action of warfarin.
  • Its anticoagulant effect results from a balance between partially inhibited synthesis and unaltered degradation of the four vitamin K-dependent clotting factors.
  • The resulting inhibition of coagulation is dependent on their degradation half-lives in the circulation. These half-lives are 6, 24, 40 and 60 hours for factors VII, IX, X and II, respectively.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

Why is warfarin not used to treat patient in an active hypercoagulable state? i.e. bridging

A

Protein C and factor VII have a relatively short half life (6 hours). Thus, the immediate effect of warfarin is to deplete the procoagulant factor VII and anticoagulant protein C, which can paradoxically create a transient hypercoagulable state due to residual activity of the longer half-life procoagulants in the face of protein C depletion..
For this reason, warfarin cannot be used in active hypercoagulability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
236
Q

What are the adverse effects of warfarin?

A

Warfarin crosses the placenta readily and can cause a haemorrhagic disorder in the fetus.
Cutaneous necrosis with reduced activity of protein C sometimes occurs during the first weeks of therapy in patients who have inherited deficiency of protein C.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

What is the administration and dosage of warfarin?

A

It shoukd be initiated with standard doses of 5-10mg. The initial adjustment of PT takes about 1 week, which usually results in a maintenance dose of 5-7mg/d.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

What is the INR?

A

It is the prothrombin time ratio (patient prothrombin time/mean of normal prothrombin time for lab)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

What is the recommended INR in warfarin?

A
  • For prophylaxis and treatment of thrombotic disease - 2-3
  • Artificial valves - 2.5-3.5
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

Occasionally patients exhibit warfarin resistance, defined as progression or recurrence of a thrombotic event whilst in target range. We may raise their INR target or swtich anticoagulation. What diseases is this seen in?

A

Patients with advanced cancers, typically of GI origin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

What are the pharmacokinetic mechanisms for drug interactions with warfarin? Which drugs interact with the pharmacokinetics?

A

They mainly involve cytochrome P450 CYP2C9 enzyme induction, enzyme inhibitiona nd reduced plasma protein binding.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
242
Q

What are the pharmacodynamic mechanisms for drug interactions with warfarin? Which drugs interact with the pharmacodynamics?

A

Pharmacodynamic mechanisms for interactions with warfarin are synergism (impaired haemotasis, reduced clotting factor synthesis), competitive antagonism (vitamin K), and an altered physiological control loop for vitamin K (hereditary resistance to oral anticoagulants)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

What is the chemical make-up of warfarin?

A

Warfarin used clinically is a racemic mixture composed of equal amounts of two molecules.
S-warfarin is four times more potent that R-warfarin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

How do metronidazole, fluconazole and trimethoprim and sulfamethoxazole interact with warfarin?

A

They stereoselectively inhibit the metabolic transformation of S-warfarin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

How do amiodarone, disulfiram and cimetidine interact with warfarin?

A

They inbibit metabolsim of both R-warfarin and S-warfarin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

How do aspirin, hepatic disease and hyperthyroidism augment warfarin’s effects?

A

Aspirin by its effect on platelet function. Hepatic disease and hyperthyroidism by increasing the turnover rate of clotting factors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

How do third-egenration cephalosporins (cefixime, ceftazadine, cefotaxime) interact with warfarin?

A

They eliminate the bacteria in the intestinal tract that produce vitamin K and, like warfarin, also directly inhibit vitamin K epoxide reductase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

How do barbiturates and rifampicin interact with warfarin?

A

They cause a marked decrease of the anticoagulant effect by induction of the hepatic enzymes that transform racemic warfarin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

How does cholesytramine interact with warfarin?

A

It binds warfarin in the intestine and reduced its absorption and bioavailability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
250
Q

What things cause pharcodynamic reductions of anticoagulant effects of warfarin?

A
  • Increased vitamin K - increased synthesis of clotting factors
  • Diuretics chlorthalidone and spironolactone - affecting clotting factor concentration
  • Hereditary resistance - mutation of vitamin K reactivation cycle molecules
  • Hypothyroidism - decreased turnover rate of clotting factors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

What are some drugs with no significant effect of anticoagulant effect of warfarin?

A
  • Ethanol
  • Phenothiazines
  • Benzodiazepines
  • Acetaminophen
  • Opioids
  • Indemethacin
  • Most antibiotics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

How can excessive anticoagulant effect of warfarin by reversed?

A

By:
* stopping the drug
* administering oral or parenteral vitamin K1 (phytonadione)
* FFP
* Prothrombin complex concentrates
* Recombinant factor VIIa

It is important to note that due to the long half-life of warfarin, a single dose of vitamin K or rFVIIa may not be sufficient

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

What are some oral direct factor Xa inhibitors (DOACs)?

A

Rivaroxaban, apixaban, edoxaban.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

How do DOACs work?

A

They inhibit factor Xa, in the final common pathway of clotting.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

What are the pharmacokinetics of rivaroxaban?

A
  • High oral bioavailability when taken with food
  • Following an oral dose, the peak plasma level is achieved within 2-4 hours
  • The drug is extensively protein-bound
  • It is a substrate for the cytochrome P450 system and the P-glycoprotein transporter. Drugs inhibiting these result in increased rivaroxaban effect.
  • 1/3 of the drug is ecreted in the urine and feces.
  • The drug half-life is 5-9 hours in patients aged 20-45 and is increased in the elderly and in those with impaired renal or hepatic function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

What are the pharmacokinetics of apixaban?

A
  • Oral bioavailability of 50% and prolonged absorption, resulting in a half-life of 12 hours with repeat dosing
  • The drug is a substrate of the cytochrome P450 and P-glycoprotein, therefore drugs that inhibit both of these, result in increased drug effect
  • It is excreted in the urine and feces
  • Drug effect is increased in both renal or hepatic function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

What are the pharmacokinetics of edoxaban?

A
  • It is a once-daily Xa inhibitor with a 62% oral bioavailability
  • Peak drug concentrations occur 1-2 hours after dosage and are not affected by food.
  • The drug half-life is 10-14 hours.
  • Edoxaban does not induce CYP450 enzymes
  • No dose reduction is required with concurrent use of P-glycoprotein inhibitors.
  • It is primarily excreted unchanged in the urine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

What are the clinical indications for DOACs?

A
  • Prevention of embolic stroke in patients with AF without valvular heart disease
  • VTE prophylaxis following hip or knee surgery
  • Treatment of VTE
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

What are the dosing regimens for rivaroxaban?

A
  • Prophylactic dose is 10mg PO OD for 35 days for for hip replacement or 12 days for knee replacement
  • Treatment dose of DVT/PE is 15mg BD for three weeks then 20mg OD for 3-6 months depending on risk factors and clinical presentation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

What are the dosing regimens for apixaban?

A
  • AF - 5mg BD
  • VTE - 10mg BD for the first week then 5mg BD from then on
  • Prophylactic dose following hip or knee surgery is 2.5mg BD
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

How can you reverse direct factor Xa inhibitors?

A

Andexanet alfa is a factor Xa ‘decoy’ molecule without procoagulant activities that competes for binding to anti-Xa drugs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
262
Q

What are the pharmacodynamics and pharmacokinetics of dabigatran?

A
  • Dabigatran and its metabolites are direct thrombin inhibitors.
  • Following oral administration, dabigatran etexilate mesylate is converted to dabigatran
  • The oral bioavailability is 3-7% in normal volunteers
  • The drug is a substrate for the P-glycoprotein efflux pump; therefore P-glycoprotein inhibitors should be avoided in patients with impaired renal function
  • The half-life of the drug is 12-17 hours
  • Renal impairment results in prolonged drug clearance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

What are the clinical indications and doses for Dabigatran?

A

Prevention of stroke and systemic embolism in non-valvular atrial fibrillation - 150mg BD, halved if CrCl <30.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
264
Q

What is the reversal of dabigatran?

A

idarucizumab is a humanised monoclonal antibody Fab fragment that binds to dabigatran and reverses the anticoagulant effect. - 5MG IV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
265
Q

How do fibrinolytic drugs work?

A
  • They rapidly lysae thrombi by catalysing the formation of the serine protease plasmn from its precursor zymogen, plasminogen.
  • These drugs create a generalised lytic state when administered IV
  • Thus, both protective haemostatic thombi and target thromboemboli are broken down
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

What is the chemistry of streptokinase?

A
  • It is a protein synthesisted by streptococci that combines with the proactivator plasminogen.
  • This enzymatic complex catalyses the conversion of inactive plasminogen to active plasmin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
267
Q

What is the chemistry of urokinase?

A

It is a human enzyme synthesised by the kidney that directly converts plasminogen to active plasmin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
268
Q

How do tissue plasminogen activators (t-PAs) work? Name some and tell me about them

A
  • These activators preferentially activate plasminogen that is bound to fibrin, which confines fibrinolysis to the formed thrombus and avoids systemic activation
  • Recombinant human t-PA is manufactured as alteplase.
  • Reteplase is another recombinant human t-PA from which several amino acid sequences have been deleted
  • Tenecteplase is a mutant form of t-PA that has a longer half0life, it can be given as an IV bolus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q

What are the indications of fibrinolytic drugs?

A
  • PE with haemodynamic instability
  • Severe DVT such as superior vena caval syndrome
  • Ascending thrombophlebitis of the iliofemoral vein
  • Sometimes AMI
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q

What is the dosing regimen for streptokinase?

A
  • IV loading dose of 250,000 units
  • Maintenance dose of 100,000units/hour for 24-72 hours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

What is the dosing regimen for urokinase?

A
  • Loading dose of 300,000 units given over 10 minutes
  • Maintenance dose of 300,000 units/hour for 12 hours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

What is the dosing regimen for alteplase (t-PA)?

A
  • Loading bolus 15mg
  • 0.75mg/kg over 30 minutes
  • 0.5mg/kg over 60 minutes
273
Q

What is the dosing regimen for Tenecteplase?

A

It is given as a single IV bolus from 30-50mg depending on body weight.

274
Q

Platelet function is regulated by three categories of substances. Name them and tell me whats in each one

A

1) Agents generated outside the platelet that interact with platelet membrane receptors e.g. catecholamines, collagen, thrombin, prostacyclin
2) Agents generated within the platelet that interact with membrane receptors e.g. ADP, prostaglandin D2, prostaglandin E2, and serotonin
3) Agents generated within the platelet that act within the platelet e.g. prostaglandin endoperoxides and thromboxane A2, the cyclic nucleotides cAMP and cGMP and calcium ion.

275
Q

How do the different antiplatelets work?

A
  • Aspirin - inhibition of prostaglandin synthesis
  • Clopidogrel, prasugrel, ticlopidine inhibition of ADP-induced platelet aggregation
  • Abciximab, tirofiban, eptifibatide block glycoprotein IIb/IIIa receptors on platelets
276
Q

What are the pharmacodynamics of aspirin?

A
  • The prostaglandin thromboxane A2 causes platelets to change shape, release their granules and aggregate.
  • Aspirin inhibits the synthesis of thromboxane A2 by irreversible acetylation of the COX enzyme.
277
Q
A
278
Q

What are the adverse effects of ticlopidine?

A
  • Nausea, dyspepsia and diarrhoea in up to 20% of patients
  • Haemorrhage in 5%
  • Leukopaenia in 1%
279
Q

What are the clinical indications and dosing regimens for clopidogrel?

A

NSTEMI - 300mg loading dose followed by 75mg daily
STEMI - 75mg daily
Recent MI, stroke or PVD - 75mg daily

280
Q

How long does clopidogrel take to work and how long is it effective for?

A
  • The antithrombotic effects are dose dependentl within 5 hours after the loading dose, 80% of platelet activity will be inhibited
  • The duration of the antiplatelet effect is 7-10 days
281
Q

What does vitamin K do?

A

It confers biologic activity upon prothrombin and factors VII, IX, and X by participating in their postribosomal modification

282
Q

Where can people get vitamin K from? What type of vitamin is it?

A

It is a fat-soluble substance found primarily in leafy green vegetables.
The dietary requirement is low because the vitamin is additionally synthesised by bacteria that colonise the human intestine.

283
Q

What are the two natural forms of vitamin K and where are they found?

A

Vitamins K1 and K2.
K1 (phytonadione) is found in food.
K2 is found in human tissues and is synthesisted by intestinal bacteria

284
Q

What are the pharmacokinetics of vitamin K?

A
  • It is available clinically in oral and parenteral forms
  • Onset of effect is delayed for 6 hours but the effect is complete by 24 hours when treating depression of prothrombin activity caused by excess warfarin.
285
Q

IV administration of vitamin K should be slow. Why?

A

It can produce dyspnoea, chest and back pain and even death

286
Q

What are the two different subtypes of the anti-cholinergics?

A

Muscarinic and nicotinic subgroups

287
Q

Where do anti-nicotinics have their effect in the CNS?

A

Ganglion blockers and neuromuscular junction blockers make up the antinicotinic drugs

288
Q

How many different subtypes of muscarinic receptors are there? How are they classified? Where are they?

A
  • Five types of muscarinic receptors have been identified.
  • M1 - M5.
  • M1 receptor is on CNS neurons, autonomic postganglionic cell bodies, and many presynaptic sites
  • M2 is in the myocardium, smooth muscle organs, and some neuronal sites
  • M3 receptors are most common on effector cell membranes, especially glandular and smooth muscle cells
  • M4 and M5 are less prominent and play a greater role in the CNS than in the periphery
289
Q

What is an example of an antimuscarinic drug?

A

Atropine
Glycopyrrolate
Hyoscine

290
Q

What is the source and chemistry of atropine?

A
  • Atropine is a tertiary amine alkaloid ester of tropic acid.
  • It is found in the plant Atropa belladonna
  • The commercial atropina is racemin d,l-hyoscyamine. The l(-) isomers of both alkaloids are at least 100 times more potent than the d(+) isomers
291
Q

What is the absorption of antimuscarinics?

A
  • Natural alkaloids and msot tertiary antimuscarinic drugs are well absorbed from the gut and subconjunctival membranes. (benztropine, atropine)
  • In contrast, only 10-30% of a dose of quaternary antimuscarinic drug is absorbed after oral administration, reflecting the decreased lipid solubility of the charged molecule (ipratropium, tiotropium, glycopyrrolate)
292
Q

What is the distribution of antimuscarinics?

A
  • Atropine and other tertiary agents are widely distributed in the body. Significant levels are achieved in the CNS within 30 minutes to 1 hour, and this can limit the dose tolerated when the drug is taken for its peripheral effects.
  • In contrast, quaternary derivatives are poorly taken up by the brain and therefore are relatively free - at low doses- of CNS effects
293
Q

What is the metabolism and excretion of atropine?

A
  • After administration, the elimination of atropine from the blood occurs in two phases: the half-life (t1/2) of the rapid phase is 2 hours and that of the slow phase is approximately 13 hours.
  • About 50% of the dose is excreted unchanged in the urine. Most of the rest appears in the urine as hydrolysis and conjugation products.
  • The drug’s effects on parasymathetic function declines rapidly in all organs except the eye. Effects on the iris and ciliary muscle persists for >72 hours.
294
Q

What is the mechanism of action of atropine?

A
  • It causes reversible blockade of cholinomimetic actions at muscarinic receptors.
  • When atropine binds to the muscarinic receptor, it prevents actions such as the release of inositol triphosphate (IP3) and the inibition of adenylyl cyclase that are caused by by muscarinic agonists.
  • It blocks the actions of acetylcholine as an inverse agonist.
295
Q

The effectiveness of antimuscarinic drugs varies with the tissue and with the source of agonist. What tissues are most sensitive to atropine? What about the source of the agonist?

A
  • The salivary, bronchial, and sweat glands.
  • Secretion of acid by the gastric parietal cells is the least sensitive.
  • In most tissues, antimuscarinic agents block exogenously administered cholinoceptor agonists more effectively than endogenously released acetylcholine
296
Q

How does atropine work on muscarinic receptors compared to nicotinic receptors?

A

Atropine is highly selective for muscarinic receptors.
Its potency at nicotinic receptors is much lower, and actiona at non-muscarinic receptors are generally undetectable clinically

297
Q

Does atropine distingiush between M1, M2 and M3 receptors?

A

No.

298
Q

What are the dominant effectors systems of M1, M2 and M3 receptors? What are some anatagonists that are specific for those receptors?

A
  • M1 - increase IP3 and DAG - Pirenzepine, dicyclomine
  • M2 - decrease cAMP, increase potassium channel current - Gallamine
  • M3 - increase IP3 and DAG 0 oxybutynin, tolterodine, solifenacin
299
Q

What are some of the organ system effects of anti-muscarinics?

A
  • CNS - reduce the tremor of Parkinson’s disease; reduce vestibular disturbances, especially motion sickness; slow long-lasting sedative effects on the brain - can caue excitement, agitation, hallucinations and coma
  • Eye - mydriases due to blockage of pupillary muscle contraction; cycloplegia, inability to focus and accomodate due to weakened contraction of ciliary muscle; dry eyes due to reduced lacrimal secretion
  • CVS - tachycardia by blocking vagal slowing
  • Respiratory - bronchodilation and reduce secretion, can reduce larygospasm
  • GI tract - dry mouth due to reduced salivary secretion, gastric secretion is blocked, prolonged gastric emptying time
  • GU tract - relaxes smooth muscle of the uerters and bladder wall and slows voiding
  • Sweat glands - supresses thermoregulatory sweating, blocks innervation of eccrine sweat glands and can cause fever
300
Q

Right, more on atropines effect on the heart. How does it cause the tachycardia? Does this happen straight away? How does it affect the ventricles and the atrium?

A
  • The SA node is very sensitive to muscarinic receptor blockage. Moderate to high therpeutic doses of atropine cause tachycardia in the innervated and spontaneously beating heart by blockage of vagal slowing
  • However, lower doses often result in initial bradycardia before the effects of peripheral vagal block become manifest. This slowing may be due to block of prejunctional M1 receptors on vagal postganglionic fibres that normally limit acetylcholine release in the sinus node.
  • The same mechanisms operate in the AV node, in the presence of high vagal tone, atropine can significantly reduce the PR interval on the ECG by blocking muscarinic receptors in the AV node.
  • Muscarinic effects on atrial muscle are similarly blocked, but these effects are of no clinical significance except in atrial flutter and fibrillation. The ventricles are less affected by antimuscarinic drugs at therapeutic levels of a lesser degree of vagal control.
301
Q

How do antimuscarinics act on bloods vessels?

A

Most blood vessels, except those in thoracic and abdominal viscera, receive no direct innervation from the parasymapthetic system. However, parasympathetic nerve stimulation dilates coronary arteries, and sympathetic cholinergic nerves cause vasodilation in the skeletal muscle vascular bed.
Atropine can block this vasodilation. Furthermore, almost all vessels contain endothelial muscarinic receptors that mediate vasodilation.

302
Q
A
303
Q

What are some therapeutic applications of antimuscarinics?

A
  • Parkinsons disease - in conjunction with levodopa
  • Motion sickness - certain vestibular disorders respond to antimuscaricin drugs, often scopolamine
  • Ophthalmologic disorders - mydriasis greatly faclititates ophthalmoscopic examination of the retina
  • Respiratory disorders - can be used preanaesthetic to reduce laryngospasm; patient with COPD benefit from bronchodilators, especially antimuscarinic agents - ipratropium, tiotropium, aclidinium and umeclidinium. They are used an inhalation drugs along or in combination with a long acting β-adrenoceptor agonist.
  • Cardiovascular disorders - bradycardia due to marked reflex vagal discharge.
  • GI disorders - can provide relief from traveller’s diarrhoea
  • Urinary disorders - provide symptomatic relief from urinary urgency caused by minor inflammatory bladder disorders
304
Q

What are some antimuscarinic drugs used in ophthalmology, their half lives and usual concentration?

A
  • Atropine - duration 5-6 days - 0.5-1% concentration
  • Scopolamine - duration 3-7 days - 0.25% concentration
  • Homatropine - duration 12-24 hours - 2-5% concentration
  • Cyclopentolate - duration 3-6 hours - 0.5-2% concentration
  • Tropicamide - duration 15-60 minutes - 0.1-1% concentration
305
Q

What is the half-life of tiotropium, umeclidinium and aclidinium and dose intervals?

A

Tiotropium (t1/2 - 25 hours) and umeclidiniium (t1/2 - 11 hours) have a longer bronchodilator action than ipratropium (t1/2 - 2 hours) and can be given once daily because they dissociate slowly from M3 receptors?
Aclidinium (t1/2 - 6 hours) is administered twice daily

306
Q

Which antimuscarinics are used in overactive bladder? What is their mechanism of action?

A
  • Receptors for acetylcholine on the urothelium and on afferent nerves as well as the detrusor muscle provide a broad basis for the action of antimucsarinic drugs in the treatment of overactive bladder.
  • Oxybutynin, is somewhat selective for M3 receptors, is used to relieve bladder spasm ater urologic surgery
  • Darifenacin and solifenacin are antagonists that have greater selectivity for M3 receptors than oxybutynin or trospium, so have OD dosing
  • Tolterodine and fesoterodine, M3 selective antimuscarinics, are available for use in adults with urinary incontinence.
307
Q

What routes can be used to give oxybutynin?

A

Oral oxybutynin or instillation of the drug by catheter into the bladder in such patients appears to improve bladder capacity and continence to reduced infection and renal damage.
Transdermally applied oxybutynin or its oral extended-release formation reduced the need for multiple daily doses.

308
Q

How can ati-muscarinics be used to treated BPH?

A
  • They have an adjunct role in therapy of BPH when bladder symptoms (increased urinary frequency) occur.
  • Treatment with α-adrenoceptor antagonist combined with a muscarinic antagonist reduced urinary frequency
309
Q

Mushroom poisoning has traditionally been divided into rapid-onset and delayed-onset types. Tell me about these

A
  • The rapid-onset type is usually apparent within 30 minutes to 2 hours after ingestion of the mushrooms and be caused by a variety of toxins, some produce upset stomach, some hallucinations, some produce signs of muscarinic excess. Atropine can help
  • Delayed-onset mushrooms poisoning mainfests its first symptoms 6-12 hours after ingestion. Although the initial symptoms usually include nausea and vomiting, the major toxicity involved hepatic and renal cellular injury by amatoxins that inhibit RNA polymerase. Atropine is of no value here.
310
Q

What are some adverse effects of atropine?

A
  • Dry mouth - dry as a bone
  • Mydriasis - blind as a bat
  • Tachycardia, hot and flushed skin - red as a beet
  • Agitation and delirium - mad as a hatter
311
Q

How do you treat an overdose of atropine?

A
  • Generally treated symptomatically.You can use physostigmine, but small doses are giving very slowly via IV.
  • Symptomatic treatment may require temperature control with cooling blankets and seizure control with diazepam
312
Q

What are the symptoms of and how would you treat an overdose of quaternary antimuscarinic drugs?

A
  • It is associated with all of the peripheral signs of parasympathetic blockade but few or none of the CNS effects of atropine.
  • These more polar drugs may cause significant ganglionic blockade, however, with marked orthostatic hypertension.
  • Treatment, if required, can be carried out with a quarternary cholinesterase inhibitor such as neostigmine.
313
Q

What are some contraindications of antimuscarinic drugs?

A
  • They are relative not absolute
  • Angle-closure glaucoma
  • In elderly men, antimuscarinic should always be used with caution and should be avoided in those with a history of prostatic hyperplasia
  • Because the antimuscarinic drugs slow gastric emptying, they may increase symptoms in patients with gastric ulcer
314
Q

How do ganglion-blocking drugs work? Why are they not really used?

A
  • Ganglion-blocking agents competitively block the action of acetylcholine and similar agonists at neuronal nicotinic receptors of both parasympathetic and sympathetic autonomic ganglion
  • Their lack of selectivity conders sucha broad range of undesirable effects that they have limited clinical use
315
Q

The neurophysiological state produced by general anaesthetics is characterised by five primary effects. What are they?

A
  • Unconsciousness
  • Amnesia
  • Analgesia
  • Inhibition of autonomic reflexes
  • Skeletal muscle relaxation
316
Q

Where in the neurons do anaesthetics have their effects? What effects do they have?

A
  • Anaesthetics affect neurons at various cellular locations, but the primary focus has been on teh synapse.
  • A presynaptic action may alter the release of neurotransmitters, where a postsynaptic effect may change the frequency or amplitude of impulses exiting the synapse.
  • The cumulative effect of these actions may produce strengthened inhibition or diminished excitation within key areas of the CNS.
317
Q

Regarding inhaled anathestics, a clear distinction should be made between volatile and gaseous anathestics. What is the different and give me some examples?

A
  • Volatile anaesthetics (halothane, isoflurane, desflurane, sevoflurane) have low vapor pressures and thus high bioling points so that they are liquid at room temperature (20°C) and sea-level ambient pressure
  • Gaseous anaesthetics (nitrous oxide, xenon) have high vapor pressures and low boiling points they they are in gas form at room temperature. The characteristics of volatile anaesthetics make it necessary that they be administered using precision vaporisers.
318
Q

What are the principle molecular targets of anaesthetic agents?

A

The main targets studied are neuronal ion channel that mediate impulse conduction in the CNS:
* Chloride channels and potassium channels remain the primary inhibitory ion channels considered legitimate candidates of anaesthetic action
* Excitatory ion channel targets include those activated by acetylcholine, by glutamate, or by serotonin.

319
Q

What are some factors that control uptake and distribution of inhaled anaesthetics?

A
  1. Inspired concentration and ventilation
  2. Solubility
  3. Cardiac output
  4. Alveolar-venous partial pressure difference
320
Q

The driving force for uptake of an inhaled anaesthetic into the body involved inspired concentration and alveolar concentration. How? and how can we alter it?

A
  • The ratio between inspired concentration (partial pressure) and alveolar concentration is the driving force. We can only change the partial pressure
  • The partial pressure of anaesthetic in the inspired gas mixture determines the macimum partial pressure that can be achieved in the alveoli as well as the rate of rise of the partial pressure in the alevoli
321
Q

How can we change induction of inhaled anaesthetic agents? How does this change alveolar concentration?

A
  • To accelerate induction, the anaesthetist increases the inspired anaesthetic partial pressure to create a steeper gradient between inspired and alveolar partial pressure
  • This fractional rise in anaesthetic partial rpessure during induction is usually expressed as a ratio of alveolar concentration (FA) over inspired concentration (FI); the faster FA/FI approaches 1 (representing inspired-to-alveolar equilibrium), the faster anaesthesia onset will be during an inhaled induction
322
Q

Apart from partial pressure, what is another parameter under control of the anaesthetist that directly determines the rate of rise of FA/FI? How? Is this different for different anaesthetics?

A
  • The alveolar ventilation, which can be changed increased by increasing the tidal volume and resp rate to deliver larger amounts of anaesthetic agent faster.
  • The magnitude of the effect is much greater for inhaled anaesthetics with high blood solubility than for those with low blood solubility.
323
Q

What is determined by the blood:gas partition co-efficient?

A
  • The tendency for a given inhaled anaesthetic to pass from the gas phase of the alveolus into the pulmonary capillary blood is determined by the glood:gas partition coefficient.
324
Q

How is the effect of increased ventilation affected by the blood:gas partition coefficient?

A
  • An increased ventilation supplies more anaesthetic molecules to the alveolus, a more soluble anaesthetic (blood:gas partition coefficient >1) will traverse the alveolar capillary membrane more readily, preventing a rise in its alveolar partial pressure.
  • Thus, increased ventilation will replenish the alveolar anaesthetic concentration for a highly soluble anaesthetic but it is not necessary for an anaesthetic with low solubility.
  • Therefore, an increase in ventilation produces only a small change in alveolar partial pressure of an anaesthetic with low blood solubility, but can significantly increase the partial pressure of agents with moderate to high blood solubility such as halothane
325
Q

How does solubility affect the uptake of inhaled anaesthetic?

A
  • As you already know, the blood:gas coefficient is a useful index of solubility and defines the relative affinity of an anesthetic for the blood comapred to the affinity for inspired gas.
  • When an anesthetic with low blood solubility partitions between gas in the lung and pulmonary capillary blood, equilibrium is quickly established and the blood concentration rises rapidly.
  • Conversely, for anaesthetics with greater solubility, more molecules dissolve in the blood before partial pressure change significantly, and arterial concentration of the gas increases less rapidly.
326
Q

The blood:gas coefficient for nitrous oxide in 0.47, what does this mean? what would change with a larger blood:gas coefficient?

A
  • The blood:gas coefficient os 0.47 means that at equilibrium, the concentration in blood is less than half the concentration in the alveolar space (gas).
  • A larger blood:gas partition coefficient causes a greater uptake of anaesthetic into the pulmonary blood flow and therefore increases the time required for FA/FI to approach equilibruim
327
Q

How does cardiac output affect the uptake of inhaled anaesthetic?

A
  • Changes in the flow rate of blood through the lungs also affect the uptake of anaesthetic gases from the alveolar space.
  • An increase in pulmonary blood flow (ie, increased cardiac output) will increase the uptake of anaesthetic, thereby slowing the rate by which FA/FI rises and decreasing the rate of induction of anaesthesia.
328
Q

How does cardiac output effect the distribution of inhaled anaesthetic?

A

The increased uptake of anaesthetic into the blood caused by increased cardiac output will be distributed to all tissues. Since cerebral blood flow is well regulated, the increased anaesthesia uptake caused by increased cardiac output will predominantly be distributed to tissues that are not involved in the site of action of the anaesthetic

329
Q

How does the alveolar-venous partial pressure difference affect distribution of inhaled anaesthetic agents?

A
  • The anaesthetic partial pressure difference between alveolar and mixed venous blood is dependent mainly on uptake of the anaesthetic by the tissues, including non-neuronal tissues.
  • Depending on the rate and extent of tissue uptake, venous blood returning to the lungs may contain significantly less anaesthetic than arterial blood.
  • Anaesthetic uptake into tissues is influenced by factors similar to those that determine transfer of the anaesthetic from the lung to the intravascular space, including tissue:blood partition coefficients, rates of blood flow to the tissues, and concentration gradients.
  • The greater this difference in anaesthetic gas concentrations, the more time it will take to achieve equilibrium with brain tissue
330
Q

During the induction phase of anaesthesia (and the initial phase of the maintenance period), the tissues that exert greatest influence on the arteriovenous anaesthetic concentration gradient are which? why?

A

Those that are highly perfused (e.g. brain, heart, liver, kidneys, and splanchnic bed).
Combined these tissues receive over 75% of the resting cardiac output. In the case of volatile anaesthetics with relatively high solutbility in highly perfused tissues, venous blood concentration initially is very low, and equilibrium with the alveolar space is achieved slowly

331
Q

How does the drug distribution change when going from the induction phase to the maintenance phase of inhaled anaesthetics?

A
  • During maintenance, the drug cotninues to be transferred between various tissues at rates dependent on the solubility of the agent, the concentration gradient between the blood and the respective issue, and the tissue blood flow.
  • Although muscle and skin constitute 50% of the total body mass, anaesthetics accumulate more slowly in these tissues that in highly perfused tissues (e.g. brain) because they receive less blood.
  • Although most anaesthetic agents are highly soluble in adipose tissues, the relatively low blood perfusion to these tissues delays accumulation, and equilibrium is unlikely to occur with most anaesthetics during a typical 1-3 hour operation.
332
Q

What is the MAC?

A

Minimal alveolar concentration (MAC) (%)
It is the anasthetic concentration that produces immobility in 50% of patients exposed to a noxious stimulus

333
Q

The combination of what determines the rate of rise of FA/FI characteristic of each drug?

A

The combined effect of ventilation, solubility in the different tissues, cardiac output, and blood flow distribution determines the rate of tise of FA/FI

334
Q

When is the anaesthetic state achieved using inhaled anaesthetics?

A

When the partial pressure of the anaesthetic in the brain reaches a threshold concentration determined by its potency.

335
Q

How does the alveolar - venous partial pressure difference change with different anaesthetics and why?

A
  • For an insoluble agent like desflurane, the alveolar partial pressure can quickly equilibrate through the blood and brain compartments to reach anaesthetising concentrations.
  • However, for an agent like halothane, its greater solubility in blood and other tissue compartments (higher partition coefficients) produces a steeper decline in the concentration gradient from lung to brain, causing a delayed onset of anaesthesia.
  • Therefore administering a larger concentration of halothane and increased alveolar ventilation are the two strategies that can by used by anaesthetists to speed up the rate of induction with halothane.
336
Q

What is the MAC, blood:gas and brain:blood partition coefficient of nitrous oxide?

A
  • MAC - >100%
  • Blood:Gas partition coefficient - 0.47
  • Brain:Blood partition coefficient - 1.1
337
Q

What is the metabolism, onset and recovery of nitrous oxide?

A
  • No metabolism
  • Incomplete anaesthetic
  • Rapid onset and recovery
338
Q

What is the MAC, blood:gas and brain:blood partition coefficient of desflurane?

A
  • MAC - 6-7%
  • Blood:gas coefficient - 0.42
  • Brain:blood coefficient - 1.3
339
Q

What is the metabolism, volatility and recovery of desflurane?

A
  • Metabolism <0.05%
  • Low volatility
  • Poor induction agent (pungent)
  • Rapid recovery
340
Q

What is the MAC, blood:gas and brain:blood partition coefficient of sevoflurane?

A
  • MAC 2.0%
  • Blood:gas partition coefficient 0.69
  • Brain:blood coeffecient 1.7
341
Q

What is the metabolism, onset and recovery of sevoflurane?

A
  • Metabolism - 2-5% (flouride)
  • Rapid onset and resovery
  • Unstable in soda-lime
342
Q

What is the MAC, blood:gas and brain:blood partition coefficient of isoflurane?

A
  • MAC - 1.40%
  • Blood:gas partition coefficient - 1.40
  • Brain:blood partition coefficient - 2.6
343
Q

What is the metabolism, onset and recovery of isoflurane?

A
  • Metabolism <2%
  • Medium rate of onset and recovery
344
Q

What is the MAC, blood:gas and brain:blood partition coefficient of enflurane?

A
  • MAC - 1.7%
  • Blood:gas partition coefficient - 1.80
  • Brain:blood coefficient - 1.4
345
Q

What is the metabolism, onset and recovery on enflurane?

A
  • Metabolism - 8%
  • Medium rate of onset and recovery
346
Q

What is the MAC, blood:gas and brain:blood partition coefficient of halothane?

A
  • MAC 0.75%
  • Blood:gas partition coefficient - 2.30
  • Brain:blood partition coefficient - 2.9
347
Q

What is the metabolism, rate of onset and recovery of halothane?

A
  • Metabolism >40%
  • Medium rate of onset and recovery
348
Q

What does the time to recovery from inhalation anaesthesia depend on?

A

The rate of elimination of the anaesthetic from the brain, which is determined by similar things to induction: blood:gas partition coefficient, pulmonary blood flow and tissue solubility of the anaesthetic

349
Q

One of the most important factors governing the rate of recovery is the blood:gas partition coefficient of the anaesthetic agent. Why?

A

When the anaesthetists discontinues the administration of the anaesthetic agent to the lung, the alveolar concentration falls rapidly. Insoluble anaesthetics that prefer the gas phase over blood will then rapidly diffuse into the alveolus and be removed from the body by the process of lung ventilation.

350
Q

Two features differentiate the recovery phase from the induction phase. What are they?

A
  • First, transfer of an anaesthetic from the lungs to blood during induction can be enhanced by increasing its concentration in inspired air, but the reverse transfer process cannot be enhanced by because the concentration in the lungs cannot be reduced below zero.
  • Second, at the beginning of the recovery phase, the anaesthetic gas tension in different tissues throughout the body may be quite variable, depending on the specific agent and the duration of anaesthesia. In contrast, at the start of induction of anaesthesia, the initial anaesthetic tension is zero in all tissues.
351
Q

How does the solubility of inahled anaesthetics effect their elimination?

A

Inhaled anaesthetics that are relatively insoluble in blood (i.e. possess low blood:gas partition coefficients) and brain are eliminated faster than the more soluble anaesthetics

352
Q

What is the speed of elimination of the different inhaled anaesthetics? Why?

A
  • The washout of nitrous oxide, desflurane, and sevoflurane occurs at a rapid rate, leading to a more rapid recovery from their anaesthetic effects compared with halothane and isoflurane.
  • Halothane is approximately twice as soluble in brain tissue and five times more soluble in blood than nitrous oxide and desflurane; its elimination therefore takes place more slowly, and recovery from halothane- and isoflurane-based anaesthesia is predictably less rapid.
353
Q

What are the patient and procedure factors that can effect rate of elimination of inhaled anaesthetics?

A
  • The duration of exposure to the anaesthetic can have a significant effect on the speed of emergence from anaesthesia, especially in the case of the more soluble anaesthetics.
  • Accumulation of anaesthetics in muscle, skin, and fat increases with prolonged exposure (especially in obese patients), and blood concentration may decline slowly after discontinuation as the anaesthetic is slowly eliminated from these tissues.
  • Although recover after a short exposure to anasethesia may be rapid even with the more soluble agents, recovery is slow after prolonged administration of halothane or isoflurane
354
Q

How can ventilation change the rate of elimination of inhaled anaesthetic?

A

Since the concentration of anaesthetic in the inspired gas cannot be reduced below zero, hyperventilation is the only way to speed recovery

355
Q

How does anaesthetic metobolism effect the rate of elimination of inhaled anaesthetics? Give an example

A
  • Modern inhaled anaesthetics are eliminated mainly by ventilation and are only metabolised to a very small extent, thus, metabolism of these drugs does not play a significant role in their elimination
  • Hepatic metabolism may also contribute to the elimination of and recovery from some older volatile anaesthetics. For example, halothane is eliminated more rapidly that enflurane because over 40% of inspired halothane is metabolised during an average anaesthetic procedure, whereas less than 10% of enflurane is metabolised over the same period.
356
Q

In terms of the extent of hepatic metabolism of inhaled anaesthetics, what is the rank oder?

A

Halothane > enflurane > sevoflurane > isoflurane > desflurane > nitrous oxide
Nitrous oxide is not metabolised by human tissues

357
Q
A
358
Q

How do inhaled and IV anaesthetics affect the metabolic activity of the brain? What effect does this have?

A
  • Inhaled and IV anaesthetics decrease the metabolic activity of the brain.
  • A decreased cerebral metabolic rate (CMR) generally causes a reduction in blood flow within the brain.
  • However, volatile anaetshetics may also produce cerebral vasodilation, which can increase cerebral blood flow.
  • The net effect on cerebral flow (increase, decrease, or not change) depends on the concentration of anaesthetic delivered.
359
Q

How does the MAC affect the cerebral blood flow?

A
  • At 0.5 MAC, the reduction in cerebral metabolic rate (CMR) is greater than the vasodilation caused by anaesthetics, so cerebral blood flow is decreased.
  • Conversely, at 1.5 MAC, vasodilation by the anaesthetic is greater than the reduction in CMR, so cerebral blood flow is increased.
  • At 1.0 MAC, the effects are balanced and cerebral blood flow is unchanged.
360
Q

When is an increase in cerebral blood flow undesirable? What can you do about this?

A
  • An increase in cerebral blood flow is clinically undesirable in patients who have increased ICP because of brain tumour, ICH or head injury.
  • Therefore, administration of high concentrations of volatile anaesthetics is best avoided in patients with increased ICP.
  • Hyperventilation can be used to attenuate this reesponse; decreasing the PaCO2 through hyperventilation causes cerebral vasoconstriction. If the patient is hyperventilated before the volatile agent is started, the increase in ICP can be minimised
361
Q

How can nitrous oxide effect cerebral blood flow and ICP? How?

A
  • Nitrous oxide can increase cerebral blood flow and cause raised ICP. This effect is most likely caused by activation of the sympathetic nervous sytem.
  • Therefore, nitrous oxide may be combined with other agents (IV anaesthetics) or techniques (hyperventilation) that reduced cerebral blood flow in patients with increased ICP.
362
Q

Traditionally, anaesthetic effects on the brain produce four stages or levels of increasing depth of CNS depression (Guedel’s signs). What are they?

A
  • Stage 1 - anaesthesia: the patient initially experiences analgesia without amnesia
  • Stage 2 - excitement: the patient appears delirious and may vocalise but is completely amnesic. Respiration is rapid, and heart rate and blood pressure increase. Duration and severity of this light stage of anaesthesia are shortened by rapidly increasing the concentration of the agent.
  • Stage 3 - surgical anaesthesia: begins with slowing of respiration and HR and extends to complete cessation of spontaneous respiration (apnea). Four planes of stage III are described based on changes in ocular movements, eye reflexes, and pupil size, indicating increasing depth of anaesthesia
  • Stage 3 - medullary depression - represents severe depression of the CNS, including the vasomotor center in the medulla and respiratory centre in the brainstem. Without circulatory and respiratory support, death would rapidly ensue in stage IV
363
Q

How do volatile inhaled anaesthetics effect cardiac contractility? What effect does this have? How do they affect vasular resistance? Why is this relevant?

A
  • They depress normal cardiac contractility. As a result, all volatile agents tend to decrease mean arterial pressure in direct proportion to their alveolar concentration.
  • With halothane and enflurane, the reduced arterial pressure is caused primarily by myocardial depression (reduced cardiac output) and there is little change in systemic vascular resistance.
  • In contrast, isoflurane, desflurane and sevoflurane produce greater vasodilation with minimal effect on cardiac output.
  • These differences may have important implications for patients with heart failure. Because isoflurane, desflurane, and sevoflurane better preserve cardiac output as well as reduce preload (filling), and afterload (resistance), these agents may be better choices for patients with impaired myocardial function.
364
Q

How does nitrous oxide effect myocardial function?

A
  • Nitrous oxide also depresses myocardial function in a concentration-dependent manner.
  • This depression may be significantly offset by a concomitant activation of the sympathetic nervous system resulting in preservation of cardiac output.
  • Therefore, administration of nitrous oxide in combination with the more potent volatile anaesthetics can minimise circulatory depressant effects by both anaesthetic-sparing and sympathetic-activating actions
365
Q

How do inhaled anaesthetics effect heart rate?

A
  • Because all inhaled anaesthetics produce a dose-dependent decrease in arterial blood pressure, activation of autonomic nevous system reflexes may trigger an increase in heart rate
  • However, halothane, enflurane, and sevoflurane have little effect on heart rate, probably because they attenuate baroreceptor input into the autonomic nervous system
  • Desflurane and isoflurane significantly increase heart rate because they cause less depression of the baroreceptor reflex.
  • In addition, desflurane can trigger transient sympathetic activation - with elevated catecholamine levels - to cause marked increases in heart rate and blood pressure during administration of high desflurane concentrations or when desflurane concentrations are changed rapidly.
366
Q

How do inhaled anaesthetics effect myocardial oxygen consumption?

A
  • Inhaled anaesthetics tend to reduce myocardial oxygen consumption, which reflects depression of normal cardiac contractility and decreased arterial blood pressure.
  • In addition, inhaled anaesthetics produce coronary vasodilation. The net effect of decreased oxygen demand and increased coronary flow (oxygen supply) is improved myocardial oxygenation.
  • However, other factors such as surgical stimulation, intravascular volume status, blood oxygen levels, and withdrawal of perioperative β blockers, may tilt the oxygen supply-demand balance toward myocardial ischaemia.
367
Q

Why might some volatile anaesthetics cause arrhythmias?

A
  • Halothane and, to a lesser extent, other volatile anaesthetics sensitise the myocardium to epinephrine and circulating catecholaemines.
  • Ventricular arrhythmias may occur when patients under anaesthetia with halothane are given sympathomimetic drugs or have high circulating levels of endogenous catecholamines (e.g anxious patients, administration of epinephrine-containing local anaesthetics, inadequate intraoperative anaesthesia or analgesia, patients with pheuchromocytomas).
  • This effect is less marked for isoflurane, sevoflurane, and desflurane.
368
Q

How do inhaled anaesthetics affect bronchodilation and airways? What effect does this have on asthmatic patients?

A
  • All volatile anaesthetics possess varying degrees of bronchodilating properties, an effect of value in patients with active wheezing and in status asthmaticus.
  • However, airway irritation, which may provoke coughing or breath-holding, is induced by the pungency of some volatile anaesthetics.
  • The pungency of isoflurane and desflurane makes these agents less suitable for induction of anaesthesia in patients with active bronchospasm. These reactions rarely occur with halothane and sevoflurane, which are considered nonpungent.
  • Therefore, the bronchodilating action of halothane and sevoflurane makes them the agents of choice in patients with underlying airway problems.
  • Nitrous oxide is also nonpungent and can facilitate inhalation induction of anaesthesia in a patient with bronchospasm.
369
Q

How do inhaled anaesthetics affect resp rate and tidal volume?

A
  • The control of breathing is significantly affected by inhaled anaesthetics.
  • With the exception of nitrous oxide, all inhaled anaesthetics cause a dose-dependent decrease in tidal volume and an increase in resp rate, resulting in a rapid, shallow breathing pattern.
  • However, an increase in respiratory rate varies among agents and does not fully compensate for the decrease in tidal volume, resulting in a decrease in alveolar ventilation.
  • In addition, all volatile anaesthetics are respiratory depressants, as defined by a reduced ventilatory response to increased levels of CO2 in the blood.
  • By this hypoventilation mechanism, all volatile anaesthetics increase the resting level of PaCO2 in spontaneously breathing patients.
370
Q

How do volatile anaesthetics affect the apneic threshold? How can you change this?

A
  • Volatile anaesthetics raise the apneic threshold (PaCO2 level below which apnea occurs through lack of CO2-driven respiratory stimulation) and decrease the ventilatory response to hypoxia.
  • Clinically, the respiratory depressant effects of anaesthetics are overcome by assisting (controlling) ventilation mechanically.
  • The ventilatory depression produced by inhaled anaesthetics may be counteracted by surgical stimulation; however, low, subanaesthetic concentrations of volatile anaesthetic present after surgery in the early recovery period can continue to depress the compensatory increase in ventilation normally caused by hypoxia.
371
Q

How do inhaled anaesthetics effect mucociliary function? What are the effects of this?

A

Inhaled anaesthetics also depress mucocilliary function in the airway. During prolonged exposure to inhaled anaesthetics, mucus pooling and plugging may result in atelectasis and the development of postoperative respiratory complications, including hypoxameia and respiratory infections.

372
Q

What are the renal effects of inhaled anaesthetics?

A
  • Inhaled anaesthetics tend to decrease GFR and urine flow.
  • Renal blood flow may also be decreased by some agents, but filtration fraction is increased, implying that autoregulatory control of efferent arteriole tone helps compensate and limits the reduction in GFR.
  • In general these anaesthetic effects are minor compared with the stress of surgery itself and usually reversible after discontinuation of the anaesthetic
373
Q

What are the hepatic effects of inhaled anaesthetics?

A
  • Volatile anaesthetics cause a concentration-dependent decrease in portal vein blood flow that parallels the decline in cardiac output produced by these agents.
  • However, total hepatic blood flow may be relatively preserved as hepatic artery blood flow to the liver may increase or stay the same.
  • Although transient changes in liver function tests may occur following exposure to volatile anaesthetics, persistent elevation in liver enzymes is rare except following repeated exposures to halothane
374
Q

How do inhaled anaesthetics effect uterine smooth muscle?

A
  • Nitrous oxide appears to have little effect of uterine musculature
  • However, the halogenated anaesthetics are potent uterine muscle relaxants and produce this effect in a concentration dependent fashion.
  • This pharmacological effect can be helpful when profound uterine relaxation is required for intrauterine fetal manipulation or manual extraction of a retained placenta during delivery.
  • However, it can also lead to increased uterine bleeding after delivery when uterine contraction is desired
375
Q

Do inhaled anaesthetic agents have nephrotoxicity?

A
  • Metabolism of enflurane and sevoflurane may generate compounds that are potentially nephrotoxic. although their metabolism can liberate nephrotoxic fluoride ions, significant renal injury has been reported only for enflurane with prolonged exposure.
  • The insolubility and rapid elimination of sevoflurane may prevent toxicity. This drug may be degraded by CO2 absorbents in anaesthesia machines to form a nephrotoxic vinyl ether compound termed “compound A” which, in high concentrations, has caused proximal tubular necrosis in rats, but no reports of renal injury in humans has been reported
376
Q

Does nitrous oxide have haematotoxicity?

A
  • Prolonged exposure to nitrous oxide decreases methionine synthase activity, which theoretically could cause megaloblastic anaemia.
  • Megaloblastic bone marrow changes have been observed in patients after 12-hour exposure to 50% nitrous oxide.
  • Chronic exposure of dental personnel to nitrous oxide in inadequately ventilated dental operating suites is a potential occupational hazard.
377
Q

How are inhaled anaesthetics related to carbon monoxide?

A
  • All inhaled anaesthetics can produce some carbon monoxide (CO) from their interaction with strong bases in dry carbon dioxide absorbers.
  • CO binds to haemoglobin with high affinity, reduced oxygen delivery to tissues. Desflurane produces the most CO, and intraoperative formation of CO has been reported.
  • CO production can be avoided simply by using fresh carbon dioxide absorbent and by preventing its complete desiccation.
378
Q

What is malignant hyperthermia? What causes it?

A

Malignant hyperthermia is a heritable genetic disorder of skeletal muscle that occurs in susceptible individuals exposed to volatile anaesthetics while undergoing general anaesthesia. The depolarising muscle relaxant succinylcholine may also trigger malignant hyperthermia.

379
Q

What are the clinical characteristics of malignant hyperthermia?

A

The malignant hyperthermia syndrome consists of muscle rigidity, hyperthermia, rapid onset of tachycardia and hypercapnia, hyperkalaemia, and metabolic acidosis following exposure to one or more triggering agents.

380
Q

What is the underlying cellular basis of malignant hyperthermia?

A

A specific biochemical abnormality - an increase in free cytosolic calcium concentration in skeletal muscle cells - may be the underlying cellular basis of malignant hyperthermia.

381
Q
A
382
Q

What are the characteristics of malignant hyperthermia?

A
  • Malignant hyperthermia susceptibility is characterised by genetic heterogeneity, and several predisposing clinical myopathies have been identified
  • It has been associated with mutations in the gene coding for the skeletal muscle ryanodine receptor (RyR1, the calcium release channel on the sarcoplasmic reticulum), and mutant alleles of the gene encoding the α1 of the skeletal muscle L-type voltage-dependent calcium channel
383
Q

How can you test for genetic susceptibility of malignant hyperthermia?

A
  • Genetic testing cannot definitely determine malignant hyperthermia susceptibility.
  • Currently, the most reliable test to establish susceptibility is the in vitro caffeine-halothane contracture test using skeletal muscle biopsy samples.
  • Genetic counselling is recommended for family members of a person who has experienced a well-documented malignant hyperthermia reaction in the operating room
384
Q

What is the most likely cause of hepatic dysfunction following surgery and general anaesthsia?

A

Hepatic dysfunction following surgery and general anaesthesia is most likely caused by hypovolaemic shock, infection conferred by blood transfusion, or other surgical stresses rather than by volatile anaesthetic toxicity.
However, a small subset of individuals previously exposed to halothane developed fulminant hepatic failure.
Cases of hepatitis following exposure to other volatile anaesthetics, including enflurane, isoflurane, and desflurane, have rarely been reported

385
Q

What is the incidence of halothane hepatitis

A

1 in 20,000-35,0000

386
Q

What do most local anaesthetics consist of? What types have a shorter or longer duration of action?

A
  • A lipophilic group (eg. an aromatic ring) connected by an intermediate chain via an ester or amine to an ionisable group (eg. a tertiary amine)
  • In addition to the general physical properties of the molecules, specific stereochemical configurations are associated with differences in the potency of stereoisomers (eg. levobupivacaine, ropivacaine).
  • Because ester links are more prone to hydrolysis than amide links, esters usually have a shorter duration of action.
387
Q

Are local anaesthetics acids or bases? How are they available clinically? Why?

A

Local anaesthetics are weak bases and are usually made available clinically as salts to increase solubility and stability?

388
Q

How do local anaesthetics exist in the body? How are the proportions of these forms governed?

A
  • In the body, they exist either as the uncharged base or as a cation.
  • The relative proportions of these two forms are governed by their pKa and the pH of the body fluids according to the Henderson-Hasselbach equation, which can be expressed as:
389
Q

Why can pKa be used as an effective way to consider the tendency for compounds to exist in a charged or uncharged form?

A

If the concentration of base and conjugate acid are equal, the second portion of the Henderson-Hasslebach equation drops out as log 1 = 0, leaving:

pKa = pH (when base concentration = conjugate acid concentration

So the lower the pKa, the greater the proportion of uncharged weak bases at a given pH.

390
Q

What is the pKa of most local anaesthetics? What does this mean for its chemistry? Which one is different?

A
  • Because the pKa of most local anaesthetics is in the range of 7.5-9.0, the charged, cationic form will consitute the larger percentage at physiological pH.
  • A glaring exception is benzocaine, which has a pKa around 3.5, and thus exists solely as the nonionized base under normal physiological conditions
391
Q

Why is the issue of ionisation of local anaesthetics important? Why is this complicated?

A
  • The issue of ionisation is important because the cationic form is the most active at the receptor site.
  • It is complex because the receptor site for local anaesthetics is at the inner vestibule of the sodium channel, and the charged form of the anaesthetic penetrated biologic membranes poorly.
  • Thus, the uncharged form is important for cell penetration. After penetration into the cytoplasm, equilibration leads to formation and binding of the charged cation at the sodium channel, and hence the production of a clinical effect
  • Drugs may also reach the receptor laterally through what has been termed the hydrophobic pathway.
392
Q

What are the chemical and clinical situations that can effect the effectiveness of local anaesthetics?

A
  • Local anaesthetics are less effective when they are injected into infected tissues because the low extracellular pH favors the charged form, with less of the neutral base available for diffusion across the membrane
  • Conversely, adding bicarbonate to a local anaesthetic, will raise the effective concentration of the nonionised form and thus shorten the onset time of a regional block.
393
Q

Systemic absorption if injected local anaesthetic from the site of administration is determined by several factors. What are they?

A

Dosage
Site of injection
Drug-tissue binding
Local tissue blood flow
Use of vasoconstrictor (adrenaline)
Physicochemical properties of the drug itself
* anaesthetics that are more lipid soluble are generally more potent, have a longer duration of action and take longer to achieve their clinical effect
* extensive protein bnding also serves to increase the duration of action

394
Q

How does application of local anaesthetics to a vascular vs non-vascular area effect its absorption?

A

Application of local anaesthetic to a highly vascularised area such as the tracheal mucose or the tissue surrounding intercostal nerrves results in more rapid absorption and thus higher blood levels than if the local anaesthetic in injected into a poorly perfused tissue such as subcutaneous fat.

395
Q

When local anaesthetic is used for major conduction blocks, the peak serum levels will vary as a function of the specific shite of injection. Where is the highest and lowest?

A

The intercostal blocks are among the highest, and sciatic and femoral among the lowest

396
Q

What are the effects of vasoconstrictors being used with the local anaesthetics?

A

When vasoconstrictors are used with local anaesthetics, the resultant reduction in blood flow serves to reduce the rate of systemic absorption and thus diminishes peak serum levels.
This effect is generally most evident with short-acting, less potent, and less lipid-soluble anaesthetics.

397
Q

As local anesthetic is usually injected directly at the site of the target organ, distribution within this compartment plays an essential role with respect to achievement of clinical effect. What factors affect this? How is this shown in CSF?

Describe the solution terms !

A
  • Anaesthetics delivered into the subarachnoid space will be diluted with CSF and the pattern of distribution will be dependent upon a host of factors, among the most critical being the specific gravity relative to that of CSF and the patients position.
  • Solutions are termed hyperbaric, isobaric, and hypobaric, and respectively descend, remain relatively static, or ascend, within the subarachnoid space due to gravity when the patient sits upright.
398
Q

The peak blood levels achieved during major conduction anaesthesia will be minimally affected by the concentration of anaesthetic or the speed of injection. The disposition of these agents can be well approximated by a two-compartment model. Describe this…

A
  • The inital alpha phase reflects rapid distribution in blood and highly perfused organs (e.g. brain, liver, heart, kidney), characterised by a steep exponential decline in concentration.
  • This is followed by a slowed declining beta phase reflecting distribution into less well perfused tissue (e.g. muscle, gut), and may assume a nearly linear rate of decline.
399
Q

What is the uptake of local anaesthetic into the lungs? Why?

A

The potential toxicity of the local anaesthetics is affected by the protective effort afforded by uptake by the lungs, which serve to attenuate the arterial concentration, though the time course and magnitude of this effect have not been adequately characterised.

400
Q

Where are local anaesthetics excreted? How does it differ between amide and ester types?

A

They are converted to more water-soluble metabolites in the liver (amide type) or in plasma (ester type), which are excreted in the urine.

401
Q

How are local anaesthetics excreted?

A
  • Since local anaesthetics in the uncharged form diffuse readily through lipid membranes, little or no urinary excretion of the neutral form occurs
  • Acidification of urine promotes ionisation of the tertiary amine base to the more water-soluble charged form, leading to more rapid elimination.
402
Q

How are ester-type local anaesthetics metabolised?

A
  • Ester-type local anaesthetics are hydrolysed very rapidly in the blood by circulating butyrylcholinesterase to inactive metabolites.
  • For example, the half-lives of procaine and chloroprocaine in plasma are less than a minute.
  • However, excessive concentrations may accumulate in patients with reduced or absent plasma hydrolysis secondary to atypical plasma cholinesterase
403
Q

How are the amide local anaesthetics metabolised?

A

The amide local anaesthetics undergo complex biotransformation in the liver, which includes hydroxylation and N-dealkylation by liver microsomal cytochrome P450 isozymes.

404
Q

There is considerable variation in the rate of liver metabolism of individual amide compounds. Tell me the fastest to the slowest…

What does this mean in terms of toxicity?

A

Prilocaine > lidocaine > mepivacaine > ropivacaine, bupivacaine, levobupivacaine (slowest)

Toxicity from amide-type local anaesthetics is more likely to occur in patients with hepatic disease.

405
Q

Decreased hepatic elimination of local anaesthetics would be anticipated in patients with reduced hepatic blood flow. What does this mean about the general anaesthetics used and the elimination of local anaesthetic?

A

The hepatic elimination of local anaesthetics in patients anaesthetised with volatile anaesthetics (which reduce liver blood flow) is slower than in patients anaesthetised with IV anaesthetic.

406
Q

If the sodium current is blocked over a critical length of the nerve, propagation across the blacked area is no longer possible. What is the critical length in myelinated nerves?

A

2 - 3 nodes of Ranvier

407
Q

What is the mechanism of action of local anaesthetic?

A
  • The primary mechanism of action of local anaesthetics is blockade of voltage-gated sodium channels.
  • When progressively increasing concentrations of a local anaesthetic are applied to a nerve fiber, the threshold for excitation increases, impulse conduction slows, the rate of rise of the action potential declines, action potential amplitude decreases, and, finally, the ability to generate an action potential is completely abolished.
  • These progressive effects result from binding of the local anaesthetic to more and more sodium channels.
  • If the sodium current is blocked over a critical length of the nerve, propagation across the blacked area is no longer possible.
408
Q

The blockade of sodium channels by most local anaesthetics is dependent on what?

A

It is both voltage and time dependent:
* Channels in the rested state, which predominate at more negative membrane potentials, have a much lower affinity for local anaesthetics than activated (open state) and inactivated channels, which predominate at more positive membrane potentials. Therefore, the effect of a given drug concentration is more marked in rapidly firing axons than in resting fibers.
* Between successive action potentials, a portion of the sodium channels will recover from the local anaesthetic block. The recovery from drug induced block is 10-1000 times slower than recovery of channels from normal inactivation. As a result, the refractory period is lengthened and the nerve conducts fewer action potentials

409
Q

How does elevated extracellular calcium and potassium affect local anaesthetic?

A
  • Elevated extracellular calcium partially antagonises the action of local anaesthetics owing to the calcium-induced increase in the surface potential on the membrane (which favours the low-affinity rested state)
  • Conversely, increases in extracellular potassium depolarise the membrane potential and favor teh inactivated state, enhancing the effect of local anaesthetics.
410
Q

Local anaesthetics bind to the sodium channel with low affinity and poor specificity, and there are multiple other sites for which their affinity is nearly the same as that for sodium channel binding. What are they?

A
  • At clinically relevant concentrations, local anaesthetics are potentially active at countless other channels (e.g. potassium and calcium), enzymes (eg. adenylyl cyclase) and receptors (NMDA, G protein-coupled, 5-HT3, neurokinin-1 [substance P receptor])
  • Circulating anaesthetics also demostrate antithrombotic effects, having an impact on coagulation, platelet aggregeation, and the microcirculation, as well as modulation of inflammation
411
Q

What is the t1/2 Distribution (min) of:
Bupivacaine
Lidocaine
Mepivacaine
Prilocaine
Ropivacaine

A

Bupivacaine - 28 mins
Lidocaine - 10 mins
Mepivacaine - 7 mins
Prilocaine - 5 mins
Ropivacaine - 23 mins

412
Q

What is the t1/2 Elimination (hours) of:
Bupivacaine
Lidocaine
Mepivacaine
Prilocaine
Ropivacaine

A

Bupivacaine - 3.5 hours
Lidocaine - 1.6 hours
Mepivacaine - 1.9 hours
Prilocaine - 1.5 hours
Ropivacaine - 4.2 hours

413
Q

What is the volume of distribution at steady stae per 70kg body weight (L) of:
Bupivacaine
Lidocaine
Mepivacaine
Prilocaine
Ropivacaine

A

Bupivacaine - 72L
Lidocaine - 91L
Mepivacaine - 84L
Prilocaine - 261 L
Ropivacaine - 47L

414
Q

What is the clearance (L/min) of:
Bupivacaine
Lidocaine
Mepivacaine
Prilocaine
Ropivacaine

A

Bupivacaine - 0.47L/min
Lidocaine - 0.95L/min
Mepivacaine - 0.78L/min
Prilocaine - 2.84L/min
Ropivacaine - 0.44L/min

415
Q

Nerve fibres differ significantly in their susceptibility to local anaesthetic blockade. Tell me how?

A
  • It has been traditionally taught that local anaesthetics preferentially block smaller diameter fibers first because the distance over which such fibers can passively propagate an electrical impulse is shorter. However, a variable proportion of large fibers are blocked prior to the disappearance of the small fiber component of the compound action potential.
  • Most notably, myelinated nerves tend to be blocked before unmyelinated nerves of the same diameter.
  • Blockade by local anaesthetics is more marked at higher frequencies of depolarisation due to their state-dependent mechanism.
416
Q

Are sensory or motor fibers more susceptible to local anaesthetic blockade? Why? Give some examples

A
  • Sensory fibers have a high firing rate and relatively long action potential duration.
  • Motor fibers fire at a slower rate and have a shorter action potential duration.
  • As type A delta and C fibers participate in high-frequency pain transmission, this characteristic may favour blockade of these fibers earlier and with lower concentrations of local anaesthetics
417
Q

In addition to the effect of intrinsic vulnerability to local anaesthetic block, the anatomic organisation of the peripheral nerve bundle may impact the onset and suscepibility of its components. How?

A

Anesthetic placed outside the nerve bundle will reach and anesthetise the proximal fibers located at the outer portion of the bundle first, and sensory block will occur in sequence from proximal and distal.

418
Q

What are the usual routes of administration of local anaesthetics?

A
  • Topical application (nasal mucose, wound margins)
  • Injection in the vicinity of peripheral nerve endings (perineural infiltration)
  • Major nerve trunks (blocks), and injection into the epidural and subarachnoid spaces surrounding the spinal cord.
419
Q

In clinical practice, there is generally an orderly evolution of block components. What is it? Give an example

A
  • It begins with sympathetic transmission and progressing to temperature, pain, light touch, and finally motor block.
  • This is most readily appreciated during onset of spinal anaesthesia, where a spatial discrepancy can be detected, the most vulnerable components achieving greater dermatomal spread. Thus, loss of sensation of cold will be roughly two segments above the analgesic level for pinprick, which is turn will be roughly two segments rostral to loss of light touch recognition.
420
Q

Several benefits may be derived from addition of vasoconstrictor to a local anaesthetic in a block. Tell me some:

A

1) Localised neuronal uptake is enhanced because of higher sustained local tissue concentrations that can translate clinically into a longer duration block. This may enable more prolonged procedures, extended duration of post op pain control, and lower total anaesthetic requirement
2) Peak blood levels will be lowered as absorption is more closely matched to metabolism and elimination, and the risk of systemic toxic effects is reduced.

421
Q

When incorporated into a spinal anaesthetic, adrenaline may not only contribute to prolongation of the local anaesthetic but also has a direct analgesic effect. How?

A

It exerts a direct analgesic effect mediated by postsynaptic α2 adrenoreceptors within the spinal cord.

422
Q

What are the untoward effects of inclusion of adrenaline in blocks?

A
  • It can potentiate the neurotoxicity of local anaesthetics used for peripheral nerve blocks or spinal anaesthesia.
  • Further, the use of a vasoconstricting agent in an area that lacks adequate collateral flow (e.g. digital block) is generally avoided.
423
Q

Local anaesthetics are sometimes used systemically in chronic pain. How does this work?

A

Esclating doses of anaesthetic appear to exert the following systemic actions:
1) low concentrations may preferentially suppress ectopic impulse generation in chronically injured peripheral nerves
2) moderate concentrations may suppress central sensitisation, which would explain therapeutic benefit that may extend beyond the anaesthetic exposure
3) higher concentrations will produce general anaesthetic effects and may culminate in serious toxicity

424
Q

Local anaesthetic toxicity derives from two distinct processes. What are they?

A

1) systemic effects following inadvertent intravascular injection or absorption of the local anaesthetic from the site of administration
2) neurotoxicity resulting from local effects produced by direct contact with neural elements

425
Q

What are the systems involved local anaesthetic toxicity?

A
  • CNS toxicity
  • Cardiotoxicity
426
Q

What are the CNS toxicity clinical features of local anaesthetic?

A
  • All local anaesthetics have the ability to produce sedation, light-headedness, visual and auditory disturbances, and restlessness when high plasma concentrations result from rapid absorption or inadvertent intravascular administration.
  • An early symptom is circumoral and tongue numbness and metallic taste
  • At higher concentrations, nystagmus and muscular twitching occur, following by tonic-clonic convulsions.
427
Q

How do local anaesthetics cause their CNS toxic effects? What is the classic pattern they follow?

A
  • Local anaesthetics apparently cause depression of cortical inhibitory pathways, thereby allowing unopposed activity of excitatory neuronal pathways.
  • This transitional stage of unbalanced excitation (ie. seizure activity) is then followed by generalised CNS depression
428
Q

When large doses of a local anaesthetic are required, what can you do to provide some prophylaxis against local anaesthetic-induced CNS toxicity?

A

Premedication with a parenteral benzodiazpeine (eg. diazepam or midazolam) will provide prophylaxis against local anaesthetic-induced CNS toxicity but little, if any, effect on cardiovascular toxicity, potentially delaying recognition of a life-threatening overdose

429
Q

It seizures do occur from local anaesthetic toxicity, what is critical to prevent and how will you do that?

A
  • It is critical to prevent hypoxaemia and acidosis, which potentiate anaesthetic toxicity.
  • Rapid tracheal intubation can facilitate adequate ventilation and oxygenation, and is essential to prevent pulmonary aspiration of gastric contents in patients at risk.
430
Q

Seizures induced by local anaesthetics should be rapidly controlled to prevent patient harm and exaceration of acidosis. What do you use to control them?

A

Benzodiazepine are advocated as first line drugs - 0.03-0.06mg/kg Midazolam - because of their haemodynamic stability, but small doses of propofol (0.25-0.5mg/kg) were considered acceptable alternatives, as they are often more immediately available in the setting of local anasethetic administration.

431
Q

Which local anaesthetic has the highest risk of cardiotoxicity?

A

Bupivacaine

432
Q

What do you use to reverse bupivocaine toxicity? How does that work?

A

A relatively simple, practical, and apparently effective therapy for resistant bupivacaine cardiotoxicity is the use of IV lipid emulsion.
Some of its effect is related to its ability to extract a lipophilic drug from aqueous plasma, thus reducing its effective concentration at tissue targets, a mechanism termed “lipid sink”

433
Q

How do local anaesthetics cause localised neural injury?

A

The studies have demonstrated myriad deleterious effects including conduction failure, membrane damage, enzyme leakage, cytoskeletal disruption, accumulation of intracellular calcium, disruption of axonal transport, growth cone collapse and apoptosis.
It is not to do with voltage-gated sodium channel blockade so the clinical effect and toxicity are not tightly linked.

434
Q

What are some transient neurological symptoms that can occur with neuraxial administration of local anaesthetic?

A

A syndrome of transient pain or dysesthesia, or both, have been linked to lidocaine for spinal anaesthetics.

435
Q

What is bupivacaine used for?

A
  • Based on concerns for cardiotoxicity, bupivacaine is often avoided for techniques that demand high volumes of concentrated anaesthetic.
  • In contracts, relatively low concentrations (<0.25%) are frequently used to achieve prolonged peripheral anasethetic and analgesia for post-op pain control. It if often the agent of choice for epidural infusions used for post-op pain control and for labor analgesia.
436
Q

What is the risk of prilocaine with system toxicity? Why?

A

Prilocaine has the highest clearance of the amino-amide anasethetics, imparting reduced risk of systemic toxicity.

437
Q

What does EMLA stand for? What is it made of?

A

Eutectic Mixture of Local Anaesthetics.
This formulation, containing 2.5% lidocaine and 2.5% prilocaine, permits anaesthetic penetration of the keratinised layer of skin, producing localised numbness

438
Q

What are some amide local anasethetics?

A

Lidocaine, bupivacaine, prilocaine, articaine, ropivacaine

439
Q

What are some ester type local anasethetics?

A

Chloroprocaine
Cocaine
Procaine
Tetracaine
Benzocaine

440
Q

Suxamethonium is the only clinically useful depolarising muscle relaxant. It’s mechanism of action is split into two phases. Tell me about the mechanism of action of Phase 1.

A

Phase 1 block (depolarising) - its neuromuscular effects are like those of acetylcholine except that it produced a longer effect at the myoneural junction.
* It reacts with the nicotinic receptor to open the channel and cause depolarisation of the motor end plate, and this in turn spreads to the adjacent membranes, causing transient contractions of muscle motor units.
* Because it is not metabolised effectively at the synapse, the depolarised membranes remain depolarised and unresponsive to subsequent impulses (ie. a state of depolarising blockade).
* Furthermore, because excitation-contraction coupling requires end-plate repolarisation (“repriming”) and repetitive firing to maintain muscle tension, a flaccid paralysis occurs.

441
Q

How do neuromuscular blocking drugs work and what are they often used with and for?

A

They interfere with transmission at the neuromuscular end plate and lack CNS activity.
They are used primary as adjuncts during general anaesthesia to optimise surgical conditions and to facilitate endotracheal intubation in order to ensure adequate ventilation.

442
Q

The mechanism of neuromuscular transmission at the motor end plate is simialar to that described for preganglionic cholinergic nerves. What is the process of it?

A
  • The arrivial of an action potential at the motor nerve terminal causes an influx of calcium and release of the neurotransmitter acetylcholine
  • Acetylcholine then diffuses across the synaptic cleft to activate nicotinic receptors located at the motor end plate, cause the channel to open.
  • The subsequent movement of sodium and potassium through the channel is associated with graded depolarisation of the end plate membrane.
  • The change in voltage is termed the motor end plate potential
443
Q

What is the magnitude of the end plate potential related to? What is the outcome of the different potentials?

A
  • The magnitude of the end plate potential is directly related to the amount of acetylcholine released.
  • If the potential is small, the permeability and the end plate potential return to normal without an impulse being propagated from the end plate region to the rest of the muscle membrane.
  • However, if the end plate potential is large, the adjacent muscle membrane is depolarised, and an action potential will be propagated along the entire muscle fiber. Muscle contraction is then initiated by excitation-contraction coupling
444
Q

At least two additional types of acetylcholine receptors are found within the neuromuscular apparatus. What and where are they? What do they do?

A
  • One type is located on the presynaptic motor axon temrinal, and activation of these receptors mobilises additional transmitted for subsequent release by moving more acetylcholine vesicles towards the synaptic membrane
  • The second type of receptor is found on extrajunctional cells and is not normally involved in neuromusclar transmission. However, under certain conditions (eg, prolonged immobilisation, thermal burns), these receptors may proliferate sufficiently to affect subsequent neuromusclar transmittion.
445
Q

Skeletal muscle relaxation and paralysis can occur from interruption of function at several sites along the pathway from the CNS to myelinated somatic nerves, unmyelinated motor nerve terminals, the motor end place, and the intracellular muscle contractil apparatus itself.
Blockade of end plate function cna be accomplished by two basic mechanisms. What are they?

A
  • First, pharmacological blockade of the physiologic agonist acetylcholine is characteristic of the antagonist neuromuscular blocking drugs (ie, non-depolarising neuromuscular blocking drugs). These drugs prevent access of the transmitted to its receptor and thereby prevent depolarisation. The prototype of this nondepolarising subgroup is d- tubocurarine.
  • The second mechanism can be produced by an excess of depolarising agonist, such as acetylcholine. This seemingly paradoxical effect of acetylcholine also occurs at the ganglionic nicotinic acetylcholine receptor. The prototypical depolarising blocking drug is succinylcholine
446
Q

What is the chemistry of the different types of neuromuscular blocking drugs?

A
  • All of the available neuromuscular blocking drugs bear a structural resemblance to acetylcholine. For example, succinylcholine is two acetylcholine molecules liked end-to-end
  • In contrast to the linear structure of succinylcholine and other depolarising drugs, the non-depolarising agents (e.g. pancuronium) concerl the “double-acetylcholine” structure in one of two types of bulky, semi-rigid ring systems.
  • Another feature common to all currently used neuromuscular blockers is the presence of one of two quarternary nitrogens, which makes them poorly lipid soluble and limits entry into the CNS
447
Q

What are the modes of administration of neuromuscular blocking drugs?

A

They are highly polar compounds and inactive orally; they must be administered parenterally

448
Q

What is the distribution and elimination of non-depolarising relaxant drugs? e.g. pancuronium, rocuronium

A
  • The rate of disappearacne of a nondepolarising neuromuscular blocking drug from the blood is characterised by a rapid initial distribution phase following by a slower elimination phase.
  • Neuromuscular blocking drugs are highly ionised, do not readily cross cell membranes, and are not strongly bound in peripheral tissues. Therefore, their volume of distribution (80-140 mL/kg) is only slightly larger than the blood volume
449
Q

What effects the duration and half-life on non-depolarising relaxant drugs?

Such an pancuronium and rocuronium

A
  • The duration of neuromuscular blockade produced by non-depolarised relaxants is strongly correlated with the elimination half-life
  • Drugs that are excreted by the kidney typically have longer half-lives, leading to longer durations of action (>35 minutes)
  • Drugs eliminated by the liver tend to have shorter half-lives and durations of action.
450
Q

What are some steroid derivative non-depolarising muscle relaxant drugs and how are they metabolised?

A
  • Pancuronium, rocuronium, vecuronium
  • All steroidal muscle relaxants are metabolised to their 3-hydroxy, 17-hydroxy, or 3-17-dihydroxy products in the liver
  • The 3-hydroxy metabolites are usuaully 40-80% as potent as the parent drug
451
Q

What are the intermediate-acting steroid muscle relaxants and how are they eliminated?

A

Vecuronium and Rocuronium tend to be more dependent on biliary excretion or hepatic metabolism for their elimination.

The duration of action of these relaxants may be prolonged significantly by patients with impaired liver function

452
Q

What is atracurium? How is it metabolised and why is it not really used anymore?

A
  • Atracurium is an intermediate-acting isoquinoline nondepolarising muscle relaxant.
  • In addition to hepatic metabolism, atracurium is inactivated by a form of spontaneous breakdown known as Hofmann elimination.
  • The main breakdown products are laudanosine and a related quaternary acid, neither of which possess any neurmuscular blocking properties.
  • Laudanosine readily crosses the blood-brain barrier, and high blood concentrations may cause seizures and an increase in the volatile anaesthetic requirement.
453
Q

What is cisatracurium? How is it different to atracurium?

A

Atracurium has several steroisomers, and the potent isomer cisatracurium is very common. Although it resembles atracurium, it has less dependence on hepatic inactivation, produced less laudanosine, and is much less likely to release histamine. Therefore, it has all the advantages of atracurium with fewer adverse effects.

454
Q

What is the duration of action on succinylcholine/suxamethonium? Why?

A

The extremely short duration of action of suxamethonium (5-10 minutes) is due to its rapid hydrolysis by butyrylcholinesterase and psuedocholinesterase in the liver and plasma, respectively.

455
Q

How is suxamethonium metabolised and eliminated?

A
  • Rapid hydrolysis by butyrylcholinesterase and psuedocholinesterase in the liver and plasma, respectively.
  • Plasma cholinesterase metabolism is the predominant pathway for suxamethonium elimination
  • The primary metabolite of succinylcholines, succinylmonocholine, is rapidly broken down to succinic acid and choline.
  • Because plasma cholinesterase has an enormous capacity to hydrolyse succinylcholine, only a small percentage of the original IV dose ever reaches the neuro-muscular junction
456
Q

How do the circulating levels of plasma cholinesterase influence the duratio of action of succinylcholine?

A
  • Because there is little, if any, plasma cholinesterase at the motor end plate, a succinylcholine-induced blockade is terminated by its diffusion away from the end plate into extracellular fluid
  • Therefore, the circulating levels of plasma cholinesterase influence the duration of action of succinylcholine by determining the amount of the drug that reaches the motor end plate.
457
Q

What is the dibucaine number? What is it used for and how?

A
  • Neuromuscular blockade produced by suxamethonium can be prolonged in patient with an abnormal genetic variant of plasma cholinesterase. The dibucaine number is a measure of the ability of a patient to metabolise suxamethonium and can be used to identify at-risk patients.
  • Under standardised test condition, dibucaine inhibits the normal enzyme by 80% and the abnormal enzyme by only 20%
458
Q

What is the elimination, clearance and approximate duration of action of atracurium?

A
  • Elimination - spontaneous
  • Clearance - 6.6mL/kg/min
  • Duration - 20-35 minutes
459
Q

What is the elimination, clearance and approximate duration of action of Cisatracurium?

A
  • Elimination - mostly spontaneous
  • Clearance - 5-6mL/kg/min
  • Duration - 25-44 minutes
460
Q

What is the elimination, clearance and approximate duration of action of Pancuronium?

A
  • Elimination - kidney (80%)
  • Clearance - 1.7-1.8mL/kg/min
  • Duration - >35 minutes
461
Q

What is the elimination, clearance and approximate duration of action of Rocuronium?

A
  • Elimination - liver (75-90%) and kidney
  • Clearance - 2.9mL/kg/min
  • Duration - 20-35 mins
462
Q

What is the elimination, clearance and approximate duration of action of suxamethonium?

A
  • Elimination - Plasma ChE2 (100%)
  • Clearance - >100mL/kg/min
  • Duration - <8 minutes
463
Q

What is the mechanism of action of non-depolarising muscle relaxants (eg. rocuronium)?

A
  • When small doses are administered, they act predominantly at the nicotinic receptor site by competing with acetylcholine.
  • In larger doses, they can enter the pore of the ion channel to produce a more intense motor blockade. This action further weakens neuromuscular transmission and diminishes the ability of the acetylcholine inhibitors (eg. neostigmine, pydridostigmine) to antagonise the effect of nondepolarising muscle relaxants
  • They can also block prejunctional sodium channels. As a result of this action, msucle realxants interfere with the mobilisation of acetylcholine at the nerve ending and cause fade of evoked nerve twitch contractions.
464
Q

How does the potency of a non-depolarising relaxant effect its onset and duration?

A

The least potent non-depolarising relaxants (e.g. rocuronium) have the fastest onset and the shortest duration of action.

465
Q

What is the principle behind the reversal of residual blockade by cholinesterase inhibitors?

A

One consequence of the surmountable nature of the post-synaptic blockade produced by nondepolarising muscle relaxants is the fact that tetanic stimulation (rapid delivery of electrical stimuli to a peripheral nerve) releases a large quality of acetylcholine and is followed by transient posttetanic facilitation of the twitch strength (ie. relief of blockade)

466
Q

What is the different between the effect of neostigmine on rocuronium vs suxamethonium?

A
  • Rocuronium - antagonistic
  • Suxamethonium
    - phase 1 - augmented
    - phase 2 - antagonistic
467
Q

What is the initial excitatory effect on skeletal muscle of rocuronium vs suxamethonium?

A
  • Rocuronium - none
  • Suxamethonium
    - Phase 1 - fasciculations
    - Phase 2 - none
468
Q

What is the response to a tetenic stimulus in rocuronium vs suxamethonium?

A
  • Rocuronium - unsustained (fade)
  • Suxamethonium
    - Phase 1 - sustained2 (no fade)
    - Phase 2 - unsustained (fade)
469
Q

What is the post-tetanic facilitation of rocuronium vs suxamethonium?

A
  • Rocuronium - yes
  • Suxamethonium
    - Phase 1 - no
    - Phase 2 - yes
470
Q

What is the rate of recovery when using rocuronium vs suxamethonium?

A
  • Rocuronium - 30-60 minutes 3
  • Suxamethonium
    - Phase 1 - 4-8 minutes
    - Phase 2 - >20 minutes 3
471
Q

Suxamethonium is the only clinically useful depolarising muscle relaxant. It’s mechanism of action is split into two phases. Tell me about the mechanism of action of Phase 1.

A

Phase 1 block (depolarising) - its neuromuscular effects are like those of acetylcholine except that it produced a longer effect at the myoneural junction.
* It reacts with the nicotinic receptor to open the channel and cause depolarisation of the motor end plate, and this in turn spreads to the adjacent membranes, causing transient contractions of muscle motor units.
* Because it is not metabolised effectively at the synapse, the depolarised membranes remain depolarised and unresponsive to subsequent impulses (ie. a state of depolarising blockade).
* Furthermore, because excitation-contraction coupling requires end-plate repolarisation (“repriming”) and repetitive firing to maintain muscle tension, a flaccid paralysis occurs.

472
Q

What is phase 2 of the mechanism of action of suxamethonium?

A
  • With prolonged exposure to suxamethonium, the initial end plate depolarisation decreases and the membrane becomes repolarised.
  • Despite this repolarisation, the membrane cannot easily be depolarised again because it is desensitised.
  • The mechanism for this desensitising phase is unclear but some evidence indicates that channel block may become more important than agonist action at the receptor in phase 2.
  • Regardless of the mechanism, the channels behave as if they are in a prolonged closed state.
  • Later in phase 2, the characteristics of the blockade are nearly identical to those of a non-depolarising block (a nonsustained twitch response to a tetanic stimulus), with possible reversal by acetylcholinesterase inhibitors.
473
Q

The standard approach for monitoring the effects of muscle relaxants during surgery uses peripheral nerve stimulation to elicit motor responses. What are the three most commonly used patterns?

A

1) single-twitch stimulation
2) train-of-four (TOF) stimulation
3) tetanic stimulation

474
Q

How is single-twitch stimulation used to monitor the level of muscle relaxant during surgery?

A
  • A single supramaximal electrical stimulus is applied to a peripheral nerve at frequencies from 0.1Hz to 1.0Hz.
  • The higher frequency is often used during induction and reversal to more accurately determine the peak (maximal) drug effect.
475
Q

How is trial of four stimulation used to monitor the level of muscle relaxant during surgery?

A
  • TOF stimulation involved four successive supramaximal stimuli given at intervals of 0.5 seconds (2Hz).
  • Each stimulus in the TOF causes the muscle to contract, and the relative magnitude of the response of the fourth twitch compared with the first twitch is the TOF ratio.
476
Q

How does the trial of four ratio compare in non-depolarising and depolarising muscle relaxants?

A
  • With a depolarising block, all four twitches are reduced in a dose-related fashion.
  • With a nondepolarising block, the TOF ratio decreases (fades) and is inversely proportional to the degree of blockage.
  • During recovery from nondepolarising block, the amount of fade decreases and the TOF ratio approaches 1.0.
477
Q

What TOF ratio is required for resumption of spontaneous ventilation?

A

A TOF ratio greater than 0.7.
However, complete clinical recovery from a nondepolarising block is considered to require a TOF greater than 0.9.

478
Q

What is tetanic stimulation in regards to measuring muscle relaxants?

A

Tetanic stimulation consists of a very rapid (30-100Hz) delivery of electrical stimuli for several seconds.

479
Q

How does tetanic stimulation effect non-depolarising muscle relaxants?

A
  • During a non-depolarising neuromuscular block (and a phase 2 sux block), the response is not sustained and fade of twitch reponses is observed.
  • Fade in response to tetanic stimulation is normally considered a presynaptic event. However, the degree of fade depends primarily on the degree of neuromuscular blockade.
  • During a partial non-depolarising blockade, tetanic nerve stimulation is followed by an increase in the posttetanic twitch reponse, so-called posttetanic facilitation of neuromuscular transmission.
  • During intense neuromuscular blockade, there is no response to either tetanic or posttetanic stimulation.
  • As the intensity of the block diminishes, the response to posttetanic twicth stimulation reappears
480
Q

The reappearance of the first response to twitch stimulation after tetanic stimulation reflects the duration of profound (clinical) neuromuscular blockade. How do you determine the posttetanic count? What does it mean?

A
  • 5 seconds of 50Hz tetany is applied, followed by 3 seconds of rest, followed by 1Hz pulses for about 10 seconds (10 pulses).
  • The counted number of muscle twitches provides an estimation of the depth of blockade.
  • For instance, a post-tetanic cound of 2 suggests no twitch response (by TOF) for about 20-30 minutes, and a post-tetanic count of 5 correlates to a no-twict response (by TOF) of about 10-15 minutes
481
Q

The double-burst stimulation pattern is another mode of electrical nerve stimulation developed with the goal of allowing for manual detction of residual neuromuscle blockade. When is it used? What is it?

A
  • It is used with it is not possible to record the responses to single-twitch, TOF, or tetanic stimulation.
  • In this pattern, three nerve stimuli are delivered at 50Hz followed by a 700ms rest period and then by two or three additional stimuli at 50Hz.
  • It is easier to detect fade in responses to double-burst stimulation than to TOF stimulation.
  • The absense of fade in response to double-burst stimulation implies that clinically significant residual neuromsucular blockade does not exist.
482
Q

What is the order of muscles to be relaxed after administration of non-depolarising relaxant drugs?

A
  • Administration of tubocurarine, 0.1-0.4mg/kg IV, initially causes motor weakness, followed by the skeletal muscles becoming flaccid and inexcitable to electrical stimulation.
  • In general, larger muscles (eg, abdominal, trunk, paraspinous, diaphragm) are more rapidly resistant to neuromusclar blockade and recover more rapidly than smaller muscles (eg, facial, foot, hand).
  • The diaphragm is usually the last muscle to be paralysed and the first one ton recover
483
Q

Of the currently available nondepolarising muscle relaxants, which one has the msot rapid onset time? What is it?

A

Rocuronium has the most rapid onset time (60-120 seconds)

484
Q

What is the dose, pattern and duration of action of suxamethonium?

A
  • Following the administration of suxamethonium (0.75-1.5mg/kg IV), transient muscle fasciculations occur over the chest and abdomen within 30 seconds, although general anaesthesia and the prior administration of a small dose of a nondepolarising muscle relaxant tend to attenuate them.
  • As paralysis develops rapidly (<90 seconds), the arm, neck and leg muscles are initially relaxed followed by the respiratory muscles.
  • As a result of succinycholine’s rapid hydrolysis by cholinesterase in the plasma (and liver), the duration of the neuromuscular block typically lasts less than 10 minutes.
485
Q

What are the cardiovascular effects of non-depolarising muscle relaxants?

A
  • Vecuronium, cisatracurium, and rocuronium have minimal, if any, cardiovascular effects.
  • Pancuronium and atracurium produce cardiovascular effects that are mediated by autonomic of histamine receptors.
  • Tubocurarine and, to a lesser extent, atracurium can produce hypotension as a result of systemic histamine release, and with larger doses, ganglionic blockade may occur with tubocurarine.
  • Pancuronium causes a moderate increase in HR and a smaller increase in CO.
  • Bronchospasm may be produced by neuromuscular blockers that release histamine (eg, atracurium), but after induction of general anaesthesia, insertion of an endotracheal tube is the most common cause of bronchospasm.
486
Q

What are the cardiac effects of suxamethonium?

A
  • It can cause cardiac arrhythmias, especially when administered during halothane anaesthesia.
  • The drug stimulates autonomic cholinoreceptors, including the nicotinic receptors at both sympathetic and parasympathetic ganglia and muscarinic receptors in the heart (eg, sinus node).
  • The negative inotropic and chronotropic responses to succinylcholine can be attenuated by administration of an anticholinergic drug (atropine).
  • At large doses of sux, positive inotropic and chronotropic effects may be observed.
  • On the other hand, bradycardia has been repeatedly oberved when a second dose of sux is given less than 5 minutes after the first dose.
487
Q

What are some adverse effects of suxamethonium?

A
  • Hyperkalaemia - during administration of sux, porassium is released from muscles, likely due to fasciculations. Patients with burns , nerve damage or neuromusclar disease, closed head injury and other trauma may develop proliferation of extrajunctional acetylcholine receptors. If this is great enough, sufficient potassium may be released to result in cardiac arrest
  • It may be associated with the rapid onset of increased intraocular pressure (< 60 seconds), peaking at 2-4 minutes, and declining after 5 minutes.
  • In heavily muscled patient, the fasciulations associated may cause an increase in intragastric pressure, increasing the risk of regurgitation and aspiration.
  • Myalgias are a common post-op complaint of heavily muscled patients and those who receive large doses of sux.
488
Q

What are the drugs that muscle relaxants interact with?

A
  • Anaesthetics
  • Antibiotics
  • Local anaesthetics
  • Anti-arrhythmics
489
Q

What are the interactions between anaesthetics and muscle relaxants? Which ones have the most effect?

A
  • Inhaled (volatile) anaesthetics potentiate the neuromuscular blockade produced by nondepolarising muscle relaxants in a dose-dependent fashion.
  • Inhaled anaesthetics augment the effects of msucle relaxants in the following order: isoflurane (most); sevoflurane, desflurane, halothane, and nitrous oxide (least)
490
Q

What are the important mechanisms of interaction between anaesthetics and muscle relaxants?

A

1) nervous system depression at sites proximal to the neuromuscular junction (CNS)
2) increased muscle blood flow (due to peripheral vasodilation produced by volatile anaesthetics), which allows a larger fraction of the injected muscle relaxant to reach the neuromuscular junction
3) decreased senstitivity of the postjunctional membrane to depolarisation

491
Q

How do antibiotics and muscle relaxants interact?

A
  • There is enhancement of neuromusular blockade by antibiotics (aminoglycosides).
  • Many of the antibiotics have been shown to cause a depression of evoked release of acetylcholine similar to that caused by administering magnesium. mechanism of this prejunction effect appears to be blockade of specific P-type calcium channels in the motor nerve terminal.
492
Q

How do local anaesthetics interact with muscle relaxants?

A
  • In small doses, local anaesthetics can depress post-tetanic potentiation via a prejunctional neural effect.
  • In large doses, local anaesthetics can block neuromuscular transmission. With these higher doses, local anaesthetics block acetylcholine-induced msucle contractions as a result of blockade of the nicotinic receptor ion channels.
493
Q

Several diseases can diminish or augment the neuromuscular blockade produced by non-depolarising muscle relaxants. What are they and how do they effect it?

A
  • Myasthenia gravis enhances the neuromuscular blockade produced by these drugs.
  • Advanced age is assoicated with a prolonged duration of action from non-depolarising relaxants as a result of decreased clearance of the drugs by the liver and kidneys. As a result, the doase of these drugs should be reduced in older patients.
  • Conversely, patients with severe burns and those with upper motor neuron disease are resistant to non-depolarising muscle relaxants. Probably due to proliferation of extrajunctional receptors, which results in an increased dose requirement for the non-depolarising relaxant to block a sufficient number of receptors.
494
Q

How can you reverse non-depolarising muscle relaxants? (not including Sugammadex)

A
  • The cholinesterase inhibitors (neostigmine and pyridostigmine) antagonise them by increasing the availability of acetylcholine at the motor end plate, mainly by inhibition of acetylcholinesterase. To a lesser extent, there cholinesterase inhibitors also increase the release of this transmitted from the motor nerve terminal.
  • In contrast, edrophonium antagonises neuromsucular blockade purely by inhibiting acetylcholinesterase activity. It has a more rapid onset of action but may be less effective than neostigmine in the presence of profound neuromuscular blockade.
495
Q

What is sugammadex? How does it work?

A
  • It is a rapid reversel agent of the steroid neuromuscular blocking agents rocuronium and vecuronium
  • It binds tightly to rocuronium in a 1:1 ratio. By binding to plasma rocuronium, sugammadex decreases the free plasma concentration and establishes a concentration gradient for rocuronium to diffase away from the neuromuscular junction back into the circulation, where it is quickly bound by free sugammadex.
496
Q

Currently, three dose ranges are recommended for sugammadex. What are they and for what?

A
  • 2mg/kg to reverse shallow neuromuscular blackade (spontaneous recovery has reached the second twitch in TOF stimulation)
  • 4mg/kg to reverse deeper blockade (1-2 posttetanic count and no reponse to TOF stimulation)
  • 16mg/kg for immediate reversal following administration of a single dose of 1.2mg/kg of rocuronium.
497
Q

How is sugammadex excreted?

A
  • In patients with normal renal function, the suagmmadex-rocuronium complex is typically excreted unchanged in the urine within 24 hours
  • In patients with renal insufficiency, complete urinary elimination may take much longer.
498
Q

What are the adverse effects and drug interactions of sugammadex?

A
  • Anaphylaxis
  • Hypersensitivity reactions such as nausea, pruritis, and urticaria
  • Marked bradycardia that may progress to cardiac arrest
  • It can decrease the efficacy of the progesterone oral contraceptive and a non-hormonal contractive is recommended for 7 days post-sugammadex use.
499
Q

What is the chemistry of IV anaesthetics that allows then to have rapid onset of action?

A

IV anaesthetics used for induction of general anaetshesia are lipophilic and preferentially partition into highly perfused lipophilic tissues (brain, spinal cord), which accounts for their rapid onset of action

500
Q

What effects the termination of the effect of a single bolus of IV anaesthetics? What does this mean for their duration of action?

A

Regardless of the extent and speed of their metabolism, termination of the effect of a single bolus is determined by redistribution of the drug into less perfused and inactive tissues such as skeletal muscle and fat.
Thus, all drugs used for induction of anaesthesia have a similar duration of action when administered as a single bolus dose despite significant differences in their metabolism.

501
Q

What are some common IV anaesthetics?

A

Thiopental, midazolam, propofol, ketamine

502
Q

Why is propofol a good IV anaesthetic?

A

Because its pharmacokinetic profile allows for continuous infusions, propofol is a good alternative to inhaled anaesthetics for maintenance of anaesthesia and is a common choice for sedation.

503
Q

What can be used to create TIVA (total IV anaesthesia (no gas))?

A

When used during maintenance of anaesthesia, propofol infusion can be supplemented with IV opioids and neuromuscular blockers as needed to completely avoid the use of inhaled anaesthetics.

504
Q

What is the induction dose of:
* diazepam
* ketamine
* midazolam
* propofol
* thiopental

A

Diazepam - 0.3-0.6mg/kg
Ketamine - 1-2mg/kg
Midazolam - 0.1-0.3mg/kg
Propofol - 1-2.5mg/kg
Thiopental - 3-5mg/kg

505
Q

What is the duration of action of:
* diazepam
* ketamine
* midazolam
* propofol
* thiopental

A

Diazepam - 15-30 minutes
Ketamine - 5-10 minutes
Midazolam - 15-20 minutes
Propofol - 3-8 minutes
Thiopental - 5-10 minutes

506
Q

What is the chemistry of propofol?

A
  • Propofol is an alkyl phenol with hypnotic properties that is chemically distinct from other groups of IV anaesthetics.
  • Because of its poor solubility in water, it is formulated as an emulsion containing 10% soybean oil, 2.25% glycerol, and 1.2% lecithin, the major component of the egg yolk phosphatide fraction.
  • It has a pH of approx 7, and has a propofol concentration of 1%.
507
Q

What is the mechanism of action of propofol?

A

The presumed mechanism of action of propofol is through potentiation of the chloride current mediated through the GABAA receptor complex

508
Q

What are the pharmacokinetics of propofol?

A
  • It is rapidly metabolised in the liver; the resulting water-soluble compounds are presumed to be inactive and are excreted through the kidneys.
  • Plasma clearance is high and exceeds hepatic blood flow, indicating the important of extrahepatic metabolism, which presumably occurs in the lungs and may account for the eliminaiton of up to 30% of a bolus dose of the drug.
509
Q

What is the recovery of propofol like? Why?

A

The recovery from propofol is more complete, with less “hangover” than that observed with thiopental, likely due to the high plasma clearance.

510
Q

What is the duration of action of propofol and why?

A

The transfer of propofol from the plasma (central) compartment and the associated termination of drug effect after a single bolus dose are mainly the result of redistribution from highly perfused (brain) to less-well-perfused (skeletal muscle) compartments.
Awakening after an induction dose of propofol usually occurs within 8-10 minutes.

511
Q

What is the context-sensitive half-time of a drug? What is it used for?

A

The context-sensitive half-time of a drug describes the elimination half-time after discontinuation of a continuous infusion as a function of the duration of the infusion.
It is an important parameter in assessing the suitability of a drug for use as maintenance anaesthetic.

512
Q

What is the context-sensitive half-life of propofol? What does this mean for its recovery?

A

The context-sensitive half-time of propofol is brief, even after a prolonged infusion, and therefore, recovery occurs relatively promptly.

513
Q

What are the CNS effects of propofol?

A
  • Propofol acts as hypnotic but does not have analgesia properties.
  • Although the drug leads to general suppression of CNS activity, excitatory effects such as twitching or spontanous movement are occasionally observed during induction anaesthesia.
514
Q

How does propofol effect cerebral blood flow and metabolic rate for oxygen, ICP and intraocular pressure? How does this compare to thiopental?

A

Propofol decreases cerebral blood flow and the cerebral metabolic rate for oxygen (CMRO2), which decreases ICP and intraocular pressure; the magnitude of these changes is comparable to thiopental.
Although propofol can produce a desired decrease in ICP, the combination of reduced cerebral blood flow and reduced MAP due to preipheral vasodilation can critically decrease cerebral perfusion pressure.

515
Q

What are the cardiovascular effects of propofol?

A
  • Compared with other induction drugs, propofol produces the most pronounced decrease in systemic blood pressure; this is a result of profound vasodilation in both arterial and venous circulations leading to reductions in preload and afterload.
  • Because the hypotensive effects are further augmented by the inhibition of the normal baroreflex response, the vasodilation only leads to a small increase in heart rate.
516
Q

What effects the effect propofol has on systemic blood pressure?

A

The drop is systemic BP caused by propofol is more pronounced with increased age, in patients with reduced intravascular fluid volume, and with rapid injection.

517
Q

What are the respiratory effects of propofol?

A

Propofol is a potent respiratory depressant and generally produces apnoea afer an induction dose.
A maintenance infusion reduced minute ventilation through reductions in tidal volume and resp rate, with the effect on tidal volume being more pronounced.
In addition, the ventilatory response to hypoxia and hypercapnia is reduced.

518
Q

How does propofol effect upper airway reflexes compared to thiopental? What is this useful for?

A

Propofol causes a greater reduction in upper airway reflexes than thiopental does, which makes it well suited for instrumentation of the airway, such as placement of LMA

519
Q

Is propofol painful to give?

A

Yes, pain on injection is a common complaint and can be reduced by premedication with an opioid or co-administration with lidocaine.
Dilution of propofol and the use of larger veins for injection can also reduce the incidence and severity of injection pain

520
Q

What is the induction dose of propofol? What factors require you to reduce or increase the induction dose of propofol?

A
  • The most common use of propofol is to facilitate induction of general anaesthesia by bolus injection of 1-2.5mg/kg IV.
  • Increasing age, reduced cardiovascular reserve, or premedication with benzodiazepines or opioids reduces the required induction dose
  • Children require high doses (2.5-3.5mg/kg)
521
Q

When used for sedation of mechanically ventilated patients in the ICU or for sedation during procedures, what is the required plasma concentration of propofol and what is the infusion rate required for this?

A

The required plasma concentration is 1-2mcg/ml, which can be achieved with a continuous infusion at 25-75 mcg/kg/min.

522
Q

Give a example of a barbiturate used for induction of general anaesthesia?

A

Thiopental

523
Q

What are the pharmacodynamics of the anaesthetic effect of barbiturates?

A

The anaesthetic effect of barbiturates presumably involves a combination of enhancement of inhibitory transmission and inhibition of excitatory neurotransmission.
Although the effects on inhibitory transmission probably result from activation of the GABAA receptor complex, the effects on excitatory transmission are less well understood

524
Q

Is thiopental metabolised?

A

Yes, it undergoes hepatic metabolism, mostly by oxidation but also by N-dealkylation, desulfuration, and destruction of the bartbituric acid ring structure.

525
Q

What is the elimination half-time and recovery after a single bolus of thiopental?

A

Although thiopental is metabolised slowly and has a long elimination half-life (11 hours), recovery after a single bolus injection is comparable to that of propofol (5-10 minutes) because it depends on redistribution to inactive tissue sites rather than on metabolism.
However, if administered through repeated bolus infection or continuous infusion, recovery will be markedly prolonged because elimination will depend on metabolism under these circumstances.

526
Q

What are the CNS effects of barbiturates (thiopental)?

A

They produce dose-dependent CNS depression ranging from sedation to general anaesthesia when adminitered as bolus injections.
They do not produce analgesia.

527
Q

How do barbiturates effect cerebral blood flow, colume and ICP?

A

They are potent cerebral vasoconstrictors and produce predictable decreases in cerebral blood flow, cerebral blood volume, and ICP. As a result they decrease the cerebral metabolic rate of O2 consumption in a dose-dependent manner up to a dose at which they suppress all EEG activity.

528
Q

The ability of barbiturates to decrease ICP and CMRO2 is useful in the management of which patients?

A

Patients with space-occupying intracranial lesions. They may provide neuroprotection from focal cerebral ischaemia (stroke, surgical retraction, temporary clips during aneurysm surgery), but probably not from global cerebral ischaemia (eg, from cardiac arrest).

529
Q

What are the cardiovascular effects of barbiturates (thiopental)?

A
  • The decrease in systemic blood pressure associated with administration of barbiturates for induction of anaesthetsia is primarily due to peripheral vasodilation and is usually smaller than the blood pressure decrease associated with propofol
  • There are also transient direct negative inotropic effects on the heart
530
Q

The depressant effects on systemic blood pressure caused by barbiturates (thiopental) are increased in what cohort of patients? Why?

A

Patients with hypovolaemia, cardiac tamponade, cardiomyopathy, coronary artery disease, or cardiac valvular disease because such patients are less able to compensate for the effects of peripheral vasodilation.

531
Q

What are the respiratory effects of barbiturates (thiopental)?

A
  • Barbiturates are respiratory depressants, and a usual induction dose of thiopental or methohexital typically produces transient apnea.
  • Barbiturates lead to decreased minute ventilation through reduced tidal volumes and respiratory rate and also decrease the ventilatory responses to hypercapnia and hypoxia.
532
Q

How does suppression of laryngeal reflexes and cough reflexes vary between thiopental and propofol?

A

Suppression of laryngeal and cough reflexes is not as profound as after an equianaesthetic propofol administration, which makes barbiturates an inferior choice for airway intrumentation in the absence of neuromusclar blocking drugs.

533
Q

What is the dosage of thiopental for induction of anaesthesia and how long does it take for the anaesthesia to work?

A

The principal clinical use of thiopental (3-5mg/kg IV) is for induction of anaesthesia, which usually occurs in less than 30 seconds.

534
Q

What are some benzodiazepines commonly used in the perioperative period?

A

Midazolam, lorazepam, and less frequently, diazepam.

535
Q

What are the pharmacokinetics of benzodiazepines in the anaesthesia setting?

A

The highly lipid-soluble benzodiazepines rapidly enter the CNS, which accounts for their rapid onset of action, followed by redistribution to inactive tissues sites and subsequent termination of the drug effect.

536
Q

How is the effect-site equilibration time different between midazolam, propofol and thiopental? What does this mean clinically?

A
  • Despite its prompt passage into the brain, midazolam is considered to have a slower effect-site equilibration time than propofol and thiopental.
  • IV doses of midazolam should be sufficiently spaced to permit the peak clinical effect to be recognised before a repeat dose is considered.
537
Q

How do the benzodiazepines context-sensitive half-time vary? What does this mean clinically?

A

Midazolam has the shortest context-sensitive half-time, which makes it the only one of the three benzodiazepine drugs suitable for continuous infusion.

538
Q

How do benzodiazepines affect the CNS system?

A
  • Benzodiazepines decrease CMRO2 and cerebral blood flow but to a smaller extent that propofol or the baribiturates.
  • Patients with decreased intracranial compliance demonstrate little or no change in ICP after the administration of midazolam.
539
Q

How can the CNS effects of benzodiazepines be terminated?

A

The CNS effects of benzodiazepines can be promptly terminated by administration of the selective benzodiazepine antagonist flumenazil, which improves their safety profile.

540
Q

What are the cardiovascular effects of benzodiazepines? How does it vary between them?

A
  • If used for the induction of anaesthesia, midazolam produces a greater decrease in systemic blood pressure than comparable doses of diazepam.
  • These changes are most likely due to peripheral vasodilation as cardiac output is not changed.
541
Q

What are the respiratory effects of benzodiazepines?

A
  • Benzodiazepines produce minimal depression of ventilation, although transient apnea may follow rapid IV administration of midazolam for induction of anaesthesia.
  • Another problem affecting ventilation is airway obstruction induced by the hypnotic effects of benzodiazepines.
542
Q

What is the dosage and clinical uses of midazolam for anaestatic-based things? In both adults and children

A
  • Midazolam (1-2mg IV) is effective for premedication, sedation during regional anaesthesia, and brief therapeutic procedures.
  • Midazolam is also the most commonly used oral premedication for children; 0.5mg/kg administered orally 30 minutes before induction of anaesthesia provides reliable sedation and anxiolysis in children without producing delayed awakening.
  • General anesthesia can be induced by the administration of midazolam (0.1-0.3mg/kg IV)
543
Q

How does the onset and amnesia effects vary between midazolam and diazepam?

A

Midazolam has a more rapid onset, with greater amnesia and less post-op sedation, than diazepam.

544
Q

How does the onset of unconsciousness and awakening time vary between using midazolam as general anaesthetic and propofol and thiopental?

A
  • The onset of unconsciousness is slower after the administration of midazolam than it is after thiopental, propofol, or etomidate.
  • Delayed awakening is a potential disadvantage, limiting the usefulness for induction of general anaesthesia
545
Q

What is the chemistry of ketamine?

A
  • Ketamine is a partially water-soluble and highly lipid-soluble phencyclidine derivative differing form most other IV anaesthetics in that it produces significant analgesia
  • Of the two stereoisomers, the S(+) form is more potent than the R(-) isomer, but only the racemic mixture of ketamine is usually used.
546
Q

What is the characteristic stat observed after an induction dose of ketamine?

A

It is known as “dissociative anaesthesia”, wherein the patient’s eyes remain open with a slow nystagmic gaze (cataleptic state).

547
Q

What is the mechanism of action of ketamine?

A

Ketamine’s mechanism of action is complex, but the major effect is probably produced through inhibition of the NMDA receptor complex

548
Q

What are the pharmacokinetics of ketamine?

A
  • The high lipid solubility of ketamine ensures a rapid onset of its effect
  • The effect of a single bolus injection is terminated by redistribution to inactive tissue sites.
  • Metabolism occurs primarily in the liver and involves N-demethylation by the cytochrome P450 system.
  • Norketamine, the primary active metabolite, is less potent and is subsequently hydroxylated and conjugated into water-solubl inactive metabolites that are excreted in urine
549
Q

What happens with the airway, eyes, lacrimation and salivation if ketamine is administered as the sole anaesthetic?

A
  • If ketamine is aadministered at the sole anaesthetic, amnesia is not as complete as with the benzodiazepines.
  • Reflexes are often preserved, but it cannot be assumed that patients are able to protect the upper airway
  • Their eyes remain open and the pupils are moderately dilated with a nystagmic gaze.
  • Frequently lacrimation and salivation are increased, and premedication with an anticholinergic drug may be indicated to limit this effect
550
Q

What are the CNS effects of ketamine? When should it not be used?

A
  • In contrast to other IV anaesthetics, ketamine is considered to be a cerebral vasodilator that increases cerebral blood flow, as well as CMRO2.
  • Thus, ketamine has traditionally not been recommended for use in patients with increased ICP.
551
Q

Unpleasant emergence reactions after administration are the main factor limiting ketamine’s use. What are they?

A
  • Such reactions may include vivid colourful dreams, hallucinations, out-of-body experiences, and increased and distorted visual, tactile, and auditory sensitivity.
  • These reactions can be associated with fear and confusion, but a euphoric state may also be induced, which explains the potential for abuse of the drug.
552
Q

What are the cardiovascular effects of ketamine?

A
  • Ketamine can produce transient but significant increases in systemic blood pressure, heart rate, and cardiac output, presumable by centrally mediated sympathetic stimulation.
  • Ketamine is also considered to be a direct myocardial depressant. This is usually masked by its stimulation of the sympathetic nervous system but may become apparent in critically ill patients with limited ability to increase their sympathetic nervous system activity.
553
Q

What are the respiratory effects of ketamine?

A
  • Ketamine is not thought to produce significant respiratory depression.
  • Transient hypoventilation and, in rare cases, a short period of apnea can follow rapid administration of a large IV dose for induction of anaesthesia.
  • Especially in children, the risk of laryngospasm because of increased salivation must be considered; this risk can be reduced by premedication with an anticholinergic drug.
554
Q

What is the dose of ketamine?

A
  • Induction of anaesthesia can be achieved with ketamine, 1-2 mg/kg IV or 4-6 mg/kg IM.
  • General anaesthesia can be achieved with the infusion of ketamine, 15-45mcg/kg/min, plus 50-70% nitrous oxide or by ketamine alone, 30-90 mcg/kg/min
555
Q

What is the metabolism of benzodiazepines?

A
  • Hepatic metabolism accounts for the clearance of all benzodiazepines. The patterns and rates of metabolism depend on the individual drug.
  • Most benzodiazepines undergo microsomal oxidation (phase 1 reactions)catalysed by cytochrome P450 isozymes.
  • the metabolites are subsequently conjugated (phase 2 reactions) to form glucuronides that are excreted in the urine.
556
Q

What is the half-lives of the phase 1 metabolites of benzodiazepines?

A
  • The metabolites are conjugated (phase 2 reactions) to form glucuronides that are excreted in the urine.
  • However, many phase 1 metabolites of benzodiazepines are pharmacologially active, some with long half-lives.
  • For example, desmethyldiazepam, which has an elimination half-life of more than 40 hours, is an active metabolite of chlordiazepoxide, diazepam, prazepam, and clorazepate.
557
Q

What is the time to peak blood levels and the half-lives of major metabolites of alprazolam?

A
  • Time to peak blood levels - 1-2 hours
  • Half life - 12-15 hours
  • Rapid oral absorption
558
Q

What is the time to peak blood levels and the half-lives of major metabolites of diazepam?

A
  • Time to peak blood levels - 1-2 hours
  • Half-lives of diazepam - 20-80 hours
  • It has active metabolites and erratic bioavailability from IM injection
559
Q

What is the time to peak blood levels and the half-lives of major metabolites of lorazepam?

A
  • Time to peak levels - 1-6 hours
  • Half-lives of major metabolites - 10-20 hours
  • No active metabolites
560
Q

What is the time to peak blood levels and the half-lives of major metabolites of temazepam?

A
  • Time to peak blood levels - 2-3 hours
  • Half-life of major metabolites - 10-40 hours
  • Slow oral absorption
561
Q

What molecules do the benzodiazepines, barbiturates and other sedatives bind to?

A

They bind to molecular components of the GABAA receptor in neuronal membranes in the CNS.
This receptor, which functions as a chloride ion channel, is activated by the inhibitory neurotransmitter GABA.

562
Q

What is the structure of the GABAA receptor? Where do benzodiazepines bind to?

A
  • A major inform of the GABAA receptor that is found in many regions of the brain consists of two α1 subunits, two β2 subunits, and one γ2 subunit.
  • In this isoform, the two binding sites for GABA are locate between adjacent α1 and β2 subunits, and the binding pocket for benzodiazepines (the BZ site of the GABAA receptor) is between an α1 and the γ2 subunit.
563
Q

What effect do Benzodiazepines have after they’ve bound to the GABAA receptor?

A
  • Benzodiazepines potentiate GABAergic inhibition at all levels of the neuraxis, including the spinal cord, hypothalamus, hippocampus, substantia nigra, cerebellar cortex and cerebral cortex.
  • They appear to increase the efficiency of GABAergic synaptic inhibition. They do not substitute for GABA but appear to enhance GABA’s effects allosterically without directly activating GABAA receptors or opening the associated chloride channels.
  • The enhancement in chloride ion conductance induced by the interaction on benzodiazepines with GABA takes the form of an increase in the frequency of channel-opening events.
564
Q

How do barbiturates work and how does their mechanism of action different from benzos?

A
  • Barbiturates also facilitate the actions of GABA at multiple sites in the CNS, but - in contrast to benzos - they appear to increase the duration of the GABA-gated chloride channel openings.
  • At high concentrations, the barbiutrates may also be GABA-mimetic, directly activating chloride channels.
  • They are less selective than benzodiazepines, because they also depress the actions of the excitatory neurotransmitted glutamic acid via binding the AMPA receptor.
565
Q

The GABAA receptor-chloride ion channel macromolecule is very versatile. Three types of ligang-benzodiazepine receptor interactions have been reported there. What are they?

A

1) Agonists facilitate GABA actions, and this occurs at multiple BZ binding sites
2) Antagonists are typified by the synthetic benzodiazepine derivative flumazenil, which blocks the actions of benzodiazepines
3) Inverse agonists act as negative allosteric modulators of GABA-receptor function.

566
Q

What are the general effects of benzodiazepines on patterns of normal sleep?

A

1) the latency of sleep onset is decreased (time to fall asleep)
2) the duration of stage 2 NREM (non-rapid eye movement) is increased
3) the duration of REM (rapid eye movement) sleep is decreased
4) the duration of stage 4 NREM slow-wave sleep is decreased

567
Q

What is drug tolerance?

A

Decreased responsiveness to a drug following repeated exposure.

568
Q

What is physiological dependence?

A

An altered physiological state that requires continuous drug administration to prevent an abstinance or withdrawal syndrome.

569
Q

What is the half-life of flumenazil? What is it used for and how does it do that?

A
  • Flumenazil is a competitive antagonist of benzodiazepines.
  • When given IV, it acts rapidly but has a short half-life (0.7-1.3 hours) due to rapid hepatic clearance
570
Q

What are the pharmacokinetics of ethanol?

A
  • Ethanol is a small water-soluble molecule that is absorbed rapidly from the GI tract.
  • After ingestion of alcohol in the fasting state, peak blood alcohol concentrations are reached within 30 minutes.
  • Distribution is rapid, with tissue levels approximating the concentration in blood.
  • Over 90% of alcohol consumed is oxidised in the liver; much of the remainder is excreted through the lungs and in the urine.
571
Q

What is the volume of distribution of ethanol?

A

It approximates total body water (0.5-0.7L/kg).

572
Q

How does ethanol effect men and women differently? Why?

A

After an equivalent oral dose of alcohol, women have a higher peak concentration than men, in part because woen have a lower total body water content and in part because if differences in first-pass metabolism.

573
Q

What kinetics does the rate of alcohol oxidation usually follow?

A

At levels of ethanol usually achieved in blood, the rate of exidation follows zero-order kinetics; that is, it is independent of time and concentration of the drug.

574
Q

What level of alcohol can be metabolised and at what rate?

A

The typical adult can metabolise 7-10g (150-220 mmol) of alcohol per hour, the equivalent of approx one drink.

575
Q

What is the primary pathway for the metabolism of alcohol? What happens during it?

A
  • The primary pathway for alcohol metabolism involves alcohol dehydrogenase (ADH), a family of cytosolic enzymes that catalyse the conversion of alcohol to acetaldehyde.
  • During conversion of ethanol by ADH to acetaldehyde, hydrogen ion is transferred from ethanol to the cofactor nicotinamide adenine dinucleotide (NAD+) to form NADH.
  • As a result, alcohol oxidation generates an excess of reducing equivalents in the liver, chiefly as NADH.
576
Q

Where is ADH located for alcohol metabolism? How does this vary between men and women?

A
  • It is located mainly in the liver, but small amounts are found in other organs such as the brain and stomach.
  • Some metabolism of ethanol by ADH occurs in the stomach in men, but a smaller amount occurs in women, who appear to have lower levels of the gastric enzyme.
577
Q

What system is induced during chronic alcohol consumption? What happens during this?

A
  • During chronic alcohol consumption, the microsomal ethanol-oxidising system (MEOS) activity is induced.
  • The enzyme system uses NADPH as a cofactor in the metabolism of ethanol and consists primarily of cytochrome P450
  • As a result, chronic alcohol consumption results in significant increases not only in ethanol metabolism but also in the clearance of other drugs eliminated by the cytochrome P450s that constitute the MEOS system, and in the generation of the toxic byproducts of cytochrome P450 reactions (toxins, free radicals, H2O2)
578
Q

How is acetaldehyde formed from alcohol metabolised?

A
  • Much of the acetaldehyde formed from alcohol is oxidised in the liver in a reaction catalysed by mitochondrial NAD-dependent aldehyde dehydrogenase (ALDH).
  • The product of this reaction is acetate, which can be further metabolised to CO2 and water, or used to form acetyl-CoA.
579
Q

How does disulfiram work?

A
  • Oxidation of acetaldehyde is inhibited by disulfiram, a drug that has been used to deter drinking by patients with alcohol dependence.
  • When ethanol is consumed in the presence of disulfiram, acetaldehyde accumulates and causes an unpleasant reaction of facial flushing, nausea, vomiting, dizziness, and headache.
580
Q

At what different blood alcohol concentrations do which clinical effects occur?

A

50-100mg/dL - sedation, subjective “high”, slower reaction times
100-200mg/dL - impaired motor function, slurred speech, ataxia
200-300mg/dL - emesis, stupor
300-400mg/dL - coma
>400 mg/dL - respiratory depression, death

581
Q

The CNS is markedly affected by acute alcohol consumption. What symptoms does it cause?

A

Alcohol causes sedation, relief of anxiety and, at higher concentrations, slurred speech, ataxia, impaired judgement, and disinhibited behaviour.

582
Q

How does alcohol have its effects on the CNS?

A
  • Ethanol affects a large number of membrane proteins that participate in signalling pathways, including neurotransmitter receptors for amines, amino acids, opioid and neuropeptides; enzymes such as Na+/K2-ATPase, adenylyl cyclase, and ion channels.
  • Acute ethanol exposure enhances the action of GABA at GABAA receptors.
  • Ethanol inhibits the ability of glutamate to open the cation channel associated with the N-methyl-D-aspartate (NMDA) subtype of glutamate receptors. The NMDA receptor is implicated in many aspects of cognitive function, including learning and memory, which is why blackouts happen
583
Q

How does ethanol effect the heart?

A

Significant depression of myocardial contractility has been observed in individuals who acutely consume moderate amounts of alcohol

584
Q
A
585
Q

How does ethanol effect smooth muscle?

A

Ethanol is a vasodilater, probably as a result of both CNS effects (depression of teh vasomotor center) and direct smooth muscle relaxation caused by its metabolite, acetaldehyde.

586
Q

What molecules cause the tissue damage caused by chronic alcohol ingestion?

A

The tissue damage caused by chronic alcohol ingestion results from a combination of the direct effects of ethanol and acetaldehyde, and the metabolic consequences of processing a heavy load of metabolically active substances.

587
Q

What are the specific mechanisms implicated in tissue damage caused by chronic alcohol consumption?

A

Specific mechanisms implicated in tissue damage include:
* increased oxidative stress coupled with depletion of glutathione
* damage to mitochondria
* growth factor dysregulation
* potentiation of cytokine-induced injury

588
Q

What causes death linked to alcohol consumption?

A
  • liver disease
  • cancer
  • accidents
  • suicide
589
Q

What is the most common medical complication of alcohol abuse? What proportion of chronic drinkers get this?

A

Liver disease
15-30% of chronic heavy drinkers eventually develop severe liver disease.

590
Q

What is the pathogenesis of alcohol liver disease?

A

A multifactorial process involving:
* metabolic repercussions of ethanol oxidation in the liver
* dysregulation of fatty acid oxidation and synthesis
* activation of the innate immune system by a combination of direct effects of ethanol and its metabolites and by bacterial endotoxins that access the liver as a result of ethanol-induced changes in the intestinal tract.
* Tumour necrosis factor-α appears to play a pivotal role in the progression of alcoholic liver disease and may be a fruitful therapeutic target.

591
Q

How does alcohol affect the pancreas?

A
  • Chronic alcohol ingestion is the most common cause of chronic pancreatitis in the Western world.
  • In addition to its direct toxic effect on pancreatic acinar cells, alcohol alters pancreatic epithelial permeability and promots the formation of protein plugs and calcium carbonate-containing stones.
592
Q

Chronic alcohol drinkers, when forced to reduce or discontinue alcohol, experience a withdrawal syndrome, which indicates the existence of physical dependence. What are the symptoms of this? What determines the intensity of the withdrawal syndrome?

A
  • Alcohol withdrawal symptoms usually consist of hyperexcitability in mild cases and seizures, toxic psychosis, and delirium tremens in severe cases.
  • The dose, rate, and duration of alcohol consumption determine the intensity of the withdrawal syndrome.
593
Q

What are the characteristics of psychological dependence on alcohol?

A
  • Psychological dependence on alcohol is characterised by a compulsive desire to experience the rewarding effects of alcohol and, for current drinks, a desire to avoid the negative consequences of withdrawal.
  • People who have recovered from alcoholism and become abstinent still experience periods of intense craving for alcohol that can be triggered by environmental cues.
594
Q

What mechanisms underly the seizures that occur in alcohol withdrawal?

A

Up-regulation of the NMDA subtype of glutamate receptors and voltage-sensitive Ca2+ channels may underlie the seizures that accompany the alcohol withdrawal syndrome

595
Q

What mechanism is believed to play a significant role in tolerance and withdrawal? Why?

A

GABA neurotransmission because
1) sedative-hypnotic drugs that enhance GABAergic neurotransmission are able to substitute for alcohol during alcohol withdrawal
2) there is evidence of downregulation of GABAA-mediated responses with chronic alcohol exposure

596
Q

What neurotransmitters does ethanol effect? How?

A
  • Ethanol modulates neural activity in the brain’s mesolimbic dopamine reward circuit and increases dopamine release in the nucleus accumbens
  • Alcohol affects local concentrations of serotonin, opioids, and dopamine - neurotransmitters involved in the brain reward system.
597
Q

Consumption of large amounts of alcohol over extended periods (usually years) often leads to neurologic deficits. What is the most common neurologic abnormality?

A

Generalised symmetric peripheral nerve injury, which begins with distal paraesthesias of the hands and feet.
Degenerative changes can also result in gait disturbances and ataxia.

598
Q

What is a relatively uncommon but important neurotoxic effect of chronic alcohol intake? Tell me about it

A

Wernicke-Korsakoff syndrome
* It is characterised by paralysis of the external eye muscles, ataxia, and a confused state that can progress to coma and death.
* It is associated with thiamine deficiency but rarely is seen in the absence of alcoholism.
* Often, the ocular signs, ataxia, and confusion improve promptly upon administration of thiamine.

599
Q

What are the cardiovascular complications of chronic alcohol consumption?

A

1) Cardiomyopathy and heart failure
2) Arrhythmias
3) HTN
4) Coronary heart disease

600
Q

How does alcohol cause cardiomyopathy?

A
  • Heavy alcohol consumption of long duration is associated with a dilated cardiomyopathy with ventricular hypertrophy and fibrosis.
  • It causes cardiac membrane disruption, depressed function of mitochondria and sarcoplasmic reticulum, intracellular accumulation of phospholipids and fatty acids, and up-regularion of voltage-gated calcium channels.
601
Q

How does the prognosis of alcoholic dilated cardiomyopathy differ from idiopathic dilated cardiomyopathy? Why?

A
  • Patients with alcohol-induced dilated cardiomyopathy do significantly worse than patients with idiopathi dilated cardiomyopathy, even though cessation of drinking is associated with a reduction on cardiac size and improved function.
  • The poorer prognosis for patients who continue to drink appears to be in part to interference by ethanol with the beneficial effects of β blockers and ACE inhibitors
602
Q

How does chronic alcohol consumption cause arrythmias?

A
  • Heavy drinking - and especially “binge” drinking - are associated with both atrial and ventricular arrhythmias.
  • Patients undergoing alcohol withdrawal syndrome can develop severe arrhythmias that may reflect abnormalities of potassium and magnesium metabolism as well as enhanced release of catcholamines.
  • Seizures, syncope and sudden death during alcohol withdrawal may be due to these arryhthmias
603
Q

How does chronic alcohol consumption cause HTN?

A

A link between heavier alcohol consumption and HTN has been firmly established.

604
Q

How does moderate alcohol consumption effect coronary heart disease?

A

Although the deleterious effects of excessive alcohol use on the cardiovascular system are well established, there is strong epidemiological evidence that moderate alcohol consumption actually prevents coronary heart disease, ischaemic stroke and peripheral arterial disease.

605
Q

What receptor is potentially linked with the hallucinatory effects in the brain? What do these receptors do?

A

Serotonin receptors, such as 5-HT2A receptor and potentially 5-HT2C.

These receptors module the release of dopamine, noradrenaline, glutamate, GABA, and acetylcholine.

Stimulation of 5-HT2A receptors leads to depolarisation of glutamate neutrons, but also stabilisation of NMDA receptors on postsynaptic neurone.

606
Q

How is the serotonin receptors used in the mechanism of action of second-generation antipsychotic drugs (clozapine)

A

5-HT2A- receptor blockade is a key factor in the mechanism of action of clozapine, risperidone, olanzapine, quetiapine, aripiprazole.

They are inverse agonists of the 5-HT2A receptor.

607
Q

What are some of the classes of antipsychotic drugs and examples for each?

A
  • Penothiazine derivatives - chlorpromazine
  • Thiothixene
  • Butytophenone derivatives - haloperidol
  • Second-generation antipsychotic drugs - clozapine, olanzapine, quetiapine, risperidone, aripiprazole
608
Q

How are antipsychotics absorbed?

A
  • Many are readily but incompletely absorbed
  • Furthermore, many undergo significant first-pass metabolism
  • Oral doses of chlorpromazine have systemic availability of 25-35%, whereas haloperidol, which has less first-pass metabolism, has an average of systemic availability of about 65%
609
Q

What is the general volume of distribution of anti-psychotics? Why? What is their general duration of action? Why?

A
  • Many antipsychotic drugs are highly lipid soluble and protein bound (92-99%)
  • They tend to have large volumes of distribution.
  • They generally have a much longer clinical duration of action than would have been estimated from their plasma half-lives. This is paralleled by prolonged occupancy of D2 dopamine receptors in the brain by typical antipsychotic drugs.
610
Q

How long does it take for chlorpromazine to be excreted?

A
  • Metabolites of chlorpromazine may be excreted in the urine weeks after the last dose of chronically administered drug.
  • Long-acting injective formulations may cause some blockade of D2 receptors 3-6 months after the last injection.
611
Q

What is the average time for relapse in stable patients with schizophrenia who discontinue their drugs? What is the exception to this?

A

Around 6 months!
The exception is clozapine, with which the relapse after discontinuation is usually rapid and severe.

612
Q

How are antipsychotic drugs metabolised?

A

Most antipsychotic drugs are almost completely metabolised by oxidation or demethylation, catalysed by liver microsomal cytochrome P450 enzymes.

613
Q

Five dopaminergic systems or pathways are important for understanding schizophrenia and the mechanism of action of antipsychotic drugs. What are they?

A
  1. The mesolimbic-mesocortical pathway, which projects from cell bodies in the ventral tegmentum in separate bundles of axons to the limbic system and neocortex
  2. The nigrostriatal pathway, which consists of neurone that project from the substantia nigra to the dorsal striatum, it is involved in the coordination of voluntary movement
  3. The tuberoinfundibular system - arising in the arcuate nuclei and periventricular neurone and releasing dopamine into the pituitary portal circulation. Dopamine released by these neuorns physiologically inhibits prolactin secretion from the anterior pituitary.
  4. The medullary-periventricular pathway consists of neurons in the motor nucleus of the vagus, which is involved in eating behaviour.
  5. The incertohypothalamic pathway forms connections from the medial zone incerta to the hypothalamus and amygdala, which appears to regular the anticipatory motivational phase of copulatory behaviour
614
Q

There are five dopamine receptors, consisting of two separate families. What are they and what do they do? How do they relate to antipsychotics?

A
  • The D1-like receptor group (D1, D5), which increases cAMP and is not correlated with the therapeutic potency of antipsychotics
  • The D2-like receptor group (D2, D3, D4), which decreases cAMP
615
Q

What is the mechanism of action of first-generation antipsychotic agents? How many receptors do they need to occupy to be effective?

A

They block D2 receptors stereoselectively for the most part, and their binding affinity is very strongly correlated with clinical antipsychotic and extrapyramidal potency. At least 60% occupancy of stratal D2 receptors is required for efficacy.

616
Q

How does the percentage of D2 receptors occupied to achieve efficacy by first-generation and second-generation antipsychotics differ? Why?

A

Second generation antipsychotic drugs such as clozapine and olanzapine are effective at lower occupancy levels (30-50%) of D2 receptors, most likely because of their concurrent high occupancy of 5HT2A receptors.

617
Q

What are some of the adverse pharmacologic effect of antipsychotics on the autonomic nervous system? Give the manifestation and the mechanism

A

Muscarinic cholinoreceptor blockade - loss of accommodation, dry mouth, difficulty urinating, constipation

α-Adrenoreceptor blockade - orthostatic hypotension, impotence, failure to ejaculate

618
Q

What are some of the adverse pharmacologic effect of antipsychotics on the central nervous system? Give the manifestation and the mechanism

A

** Dopamine-receptor blockade** - Parkinson’s syndrome, akathisia, dystonia

Supersensitivity of dopamine receptors - tardive dyskinesia

Muscarinnic blockade - toxic-confusional state

619
Q

What is an adverse pharmacologic effect of antipsychotics on the endocrine system? Give the manifestation and the mechanism

A

Dopamine-receptor blockade resulting in hyperprolactinaemia - amenorrhoea, galactorrhea, infertility, impotence

620
Q

What are the psychiatric indications of antipsychotics?

A

Schizophrenia, bipolar disorder, psychotic depression and treatment-resistant depression, schizoaffective disorder

621
Q

What type of drug is chlorpromazine?

A

An aliphatic phenothiazine

622
Q

What type of drug is haloperidol? What are its advantages and disadvantages?

A
  • It is a butyrophenone
  • Advantages - parenteral form also available
  • Disadvantages - severe extrapyramidal syndrome
623
Q

What type of drug is clozapine? What are its advantages and disadvantages?

A
  • It is a dibenzodiazepine
  • Advantages - may benefit treatment-resistant patients; little extrapyramidal toxicity
  • Disadvantages - may cause agranulocytosis in up to 2% of patients; dose-related lowering of seizure threshold
624
Q

What type of drug is risperidone? What are its advantages and disadvantages?

A
  • It is a benzisoxazole
  • Advantage - broad efficacy; little to no extrapyramidal system dysfunction at low doses
  • Disadvantage - extrapyramidal system dysfunction and hypotension with higher doses
625
Q

What type of drug is olanzapine? What are its advantages and disadvantages?

A
  • It is a thienobenzodiazepine
  • Advantages - effective against negative as well as positive symptoms; little or no extrapyramidal system dysfunction
  • Disadvantages - weight gain; dose-related lowering of seizure threshold
626
Q

What is the usual range of daily doses for antipsychotic use of haloperidol, clozapine, olanzapine, quetiapine, risperidone and aripiprazole?

A

Haloperidol - 2-60mg daily
Clozapine - 300-600mg daily
Olanzapine - 10-30mg daily
Quetiapine - 150-800mg daily
Risperidone - 4-16mg daily
Aripiprazole - 10-30mg daily

627
Q

Why does tardive dyskinesia happen with antipsychotics? Is it easier to treat early or late?

A

It is caused by a relative cholinergic deficiency secondary to super sensitivity of dopamine receptors in the caudate-putamen.

Early recognition is important, since advanced cases may be difficult to reverse.

628
Q

If agranulocytosis is going to occur secondary to clozapine, when does it usually happen? How do we monitor for it?

A

Usually between the 6th and 18th weeks of therapy.
Because of the risk, patients receiving clozapine must have weekly blood counts for the first 6 months of treatment and every 3 weeks thereafter.

629
Q

What is neuroleptic malignant syndrome? What are the symptoms and pathologies associated with it?

A

This life-threatening disorder occurs in patients who are extremely sensitive to the extrapyramidal effects of antipsychotic agents.
The initial symptom is marked muscle rigidity. If sweating is impaired, as it often is during treatment with anticholinergic drugs, fever may ensue, often reaching dangerous levels.
The stress leukocytosis and high fever associated with the syndrome suggest an infection process.
Autonomic instability, with altered BP and HR is often present.
A severe form of extrapyramidal syndrome follows.

630
Q

What blood test apart from leukocytes is elevated in neuroleptic malignant syndrome?

A

Muscle-type CK levels are usually elevated, reflecting muscle damage.

631
Q

What is the cause of neuroleptic malignant syndrome?

A

It is believed to result from an excessively rapid blockade of postsynaptic dopamine receptors.

632
Q

How do you treat neuroleptic malignant syndrome?

A
  • Early in the course, vigorous treatment of the extrapyramidal syndrome with antiparkinsonism drugs is worthwhile.
  • Muscle relaxants, particularly diazepam, are often useful
  • If fever is present, cooling by physical measures should be tried.
633
Q

What usually happens after an overdose of antipsychotics?

A
  • They are rarely fatal.
  • In general, drowsiness proceeds to come, with an intervening period of agitation.
  • Neuromuscular excitability may be increased and proceed to convulsions.
  • Pupils are biotic, and deep tendon reflexes are decreased.
634
Q

What is the absorption, distribution, metabolism and excretion of lithium?

A

Absorption - virtually complete within 6-8 hours; peak plasma levels in 30 minutes to 2 hours
Distribution - in total body water; slow entry into intracellular compartments; some sequestration in bone
Metabolism - none
Excretion - virtually entirely in urine; lithium clearance about 20% of creatinine; plasma half-life about 20 hours.

635
Q

What is the target plasma concentration and dosage of lithium?

A

Target plasma concentration 0.6-1.4mEq/L

Dosage - 0.5mEq/kg/day in divided doses

636
Q

Lithium directly inhibits its two signal transduction pathways. What are they are what are the consequences of this?

A
  • It both suppresses inositol signalling through depletion of intracellular inositol and inhibits GSK-3, a multifunctional protein kinase.
  • GSK-3 is a component of diverse intracellular signalling pathways. These include signalling via insulin/insulin-like growth factor, brain-derived neurotrophic factor (BDNF) and the Ant pathway.
  • Lithium induced inhibition of GSK-3 results in reduction of phosphorylation of β-catenin, which allows β-catenin to accumulate and translocate to the nucleus. There, β-catenin facilitates transcription of a variety of proteins.
  • The pathways othat are facilitated by the accumulation of β-catenin via GSK-3 inhibition modulate energy metabolism, provide neuroprotection, and increase neuroplasticity.
637
Q

What electrolyte is lithium closely related to? Why?

A
  • Lithium is closely related to sodium in its properties
  • It can substitute for sodium in generating action potentials and in Na+-Na+ exchange across the membrane.
  • At therapeutic concentrations, it does not significantly affect the Na+-Ca2+ exchanger or the Na+/K+-ATPase pump
638
Q

What enzymes are affected by lithium? What are the enzyme functions and the action of lithium on them?

A

Inositol monophosphatase - the rate-limiting enzyme in inositol recycling; it is inhibited by lithium, resulting in depletion of substrate IP3
Inositol polyphosphate 1-phosphatase - another enzyme in isositol recycling; inhibited by lithium, resulting in depletion of substrate for IP3 production
Biphosphate nucleotidase - involved in AMP production; inhibited by lithium; may be target that results in lithium-induced nephrogenic diabetes insipidus

639
Q

When is a lithium level ideally taken?

A

Usually 10-12 hours after the last dose

640
Q

How long after starting lithium should a level be checked?

A
  • An initial determination of serum lithium concentration should be obtained about 5 days after the start of treatment, at which time steady-state conditions should have been attained to determine the desired level and will show whether increases or decreases in the dose are required.
  • The serum concentration attained with the adjusted dosage can be checked after another 5 days.
  • Once the desired concentration has been achieved, levels can be measured at increasing intervals unless the schedule is influence by intercurrent illness.
641
Q

The decision to use lithium as prophylactic treatment for bipolar depends on many factors. What are they?

A

The frequency and severity of previous episodes
A crescendo pattern of appearance
The degree to which the patient is willing to follow a program of indefinite maintenance therapy
Patients with a history or two or more mood cycles or any clearly defined bipolar I diagnosed ones are probably candidates for maintenance treatment.

642
Q

What are some drugs that interact with lithium? How?

A
  • Renal clearance of lithium is reduced about 25% by diuretics (eg, thiazides), and doses may need to by reduced by a similar amount.
  • A similar reduction in lithium clearance is found with several of the newer NSAIDs that block synthesis of prostaglandins
643
Q

What are the adverse effects of lithium? How do you treat them?

A
  • Tremor can be alleviated with propranolol and atenolol
  • Decreased thyroid function is reversible and non-progressive, TFTs should be monitored every 6-12 months
  • Nephrogenic diabetes insipidus - polydipsia and polyuria are common adverse effects due to a loss of responsiveness for ADH, it is resistant to vasopressin but response to amiloride.
  • Oedema is common due to the effect of lithium on sodium retention.
  • Bradycardia-tachycardia “sick sinus” syndrome because the ion further depresses the sinus node.
  • Leukocytosis, transient acneiform eruptions and folliculitis
644
Q

What are the two hypotheses for the pathophysiology of major depression?

A

The monoamine hypothesis - a deficit in function of amount of monoamines is central to the biology of depression
The neurotrophic hypothesis - eutrophic and endocrine factors play a major role

645
Q

What is the neurotrophic hypothesis of depression pathophysiology?

A
  • Nerve growth factors such as brain-derived neurotrophic factor (BDNF) are critical in the regulation of neural plasticity, resilience, and neurogenesis.
  • Depression is associated with the loss of neurotrophic support and antidepressant therapies increase neurogenesis and synaptic connectivity in cortical areas such as the hippocampus.
  • BDNF is thought to exert its influence on neuronal survival and growth effects by activating the tyrosine kinase receptors B in both neurone and glia.
  • Depression appears to be associated with a drop in BDNF levels in the CSF and serum as well as with a decrease in tyrosine kinase receptor B activity and a decrease in the size of the hippocampus.
646
Q

What is the monoamine hypothesis of depression?

A
  • It suggests that depression is related to a deficiency in the amount or function of cortical and limbic serotonin (5-HT), noradrenaline (NE) and dopamine (DA).
  • The excitatory neurotransmitter glutamate appears to be elevated in the CSF of depressed patients and decreased glutamine/glutamate ratio in their plasma.
647
Q

What are some neuroendocrine factors in the pathophysiology of depression?

A
  • It is associated with elevated cortisol levels, non suppression of ACTH release in the dexamethasone suppression test and chronically elevated levels or corticotropin-releasing hormone.
  • Thyroid dysregulation has also been reported in depressed patients. These include a blunting of response to thyrotropin to thyrotropin-releasing hormone and elevations in circulating thyroxine during depressed states.
  • Estrogen deficiency states, which occur in the postpartum and postmenopausal periods, are thought to play a role in the ethology of depression in some women.
648
Q

There are currently six available SSRIs. What are they? Are they lipophilic or lipophobic?

A
  • Fluoxetine, sertraline, citalopram, paroxetine, fluvoxamine, escitalopram
  • They are all highly lipophilic
649
Q

Two classes of antidepressant act as combined serotonin and noradrenaline reuptake inhibitors. What are they?

A

Selective serotonin-noradrenaline reuptake inhibitors and Tricyclic antidepressants

650
Q

Give some SNRI examples. What can they be used for apart from for depression?

A

Venlafaxine, desvenlafaxine and duloxetine.

They have applications in the treatment of pain disorders including neuropathies and fibromyalgia. also GAD, stress urinary incontinence and vasomotor symptoms of menopause.

651
Q

How do the mechanism of actions of SNRIs and TCAs differ and how are they similar?

A
  • All SNRIs bind the serotonin and noradrenaline transporters, as do the TCAs
  • However, unlike the TCAs, the SNRIs do not have much affinity for other receptors.
652
Q

Apart from SSRIs, and SNRIs. What are the other type of antidepressants and give examples of them? What are they used for?

A
  • 5-HT2 Receptor Blockers - trazodone and nefazodone - used for major depression, sedation and hypnosis
  • Tetracyclics - mirtazapine, bupropion - increased release of noradrenaline and 5-HT - used for major depression, sedation
  • Monoamine oxidase inhibitors - selegiline, phenelzine - irreversible selective MAO-B inhibition - used for Parkinson’s and unresponsive depression
653
Q

The antidepressants share several pharmacokinetic features. What are they?

A

Most have fairly rapid oral absorption, achieve peak plasma levels within 2-3 hours, are tightly bound to plasma proteins, undergo hepatic metabolism and are really cleared.

654
Q

What is fluoxetine metabolised into? What is the half-life of this metabolite?

A
  • It is metabolised to an active product, norfluoxetine, which may have plasma concentrations greater than those of fluoxetine.
  • The elimination half-life of norfluoxetine is about three times longer than fluoxetine and contributes to the longest half-life of all the SSRIs
655
Q

What is the bioavailability of citalopram, escitalopram, fluoxetine, paroxetine and sertraline?

A

Citalopram - 80%
Escitalopram - 80%
Fluoxetine - 70%
Paroxetine - 50%
Sertraline - 45%

656
Q

What is the bioavailability of duloxetine and venlafaxine?

A

Duloxetine - 50%
Venlafaxine - 45%

657
Q

What is the bioavailability of amitriptyline, trazadone and mirtazepine?

A

Amitriptyline - 45%
Trazadone - 95%
Mirtazapine - 50%

658
Q

What is the half-lives of venlafaxine and desvenlafaxine?

A

Both have similar half-lives of about 8-11 hours. They have the lowest protein binding of all antidepressants.

659
Q

What are the pharmacokinetics of tricyclic antidepressants?

A
  • They tend to be well absorbed and have long half-lives (around 30 hours).
  • They undergo extensive metabolism via demethylation, aromatic hydroxylation, and glucuronide conjugation.
  • Only about 5% of TCAs are excreted unchanged in the urine.
660
Q

What are the pharmacodynamics of SSRIs?

A
  • The serotonin transporter (SERT) is a glycoprotein embedded in the axon terminal and cell body membranes of serotonergic neurons.
  • When extracellular serotonin binds to receptors on the transport, conformational changes occur in the transport and serotonin, Na+ and Cl- are moved into the cell.
  • Binding of intracellular K+ then results in the release of serotonin inside the cell and return of the transporter to its original confirmation.
  • SSRIs allosterically inhibit the transport by binding the SERT receptor at a site other than the serotonin binding site.
  • At therapeutic doses, about 80% of the activity of the transporter is inhibited
661
Q

What are the pharmacodynamics of SNRIs? How do they differ between themselves?

A
  • SNRIs bind both the serotonin and the noradrenaline transporters.
  • The noradrenaline transporter (NET) is structurally very similar to the 5-HT transporter, it is a domain complex that allosterically binds noradrenaline. The NET also has a moderate affinity for dopamine.
  • Venlafaxine is a weak inhibitor of NET, whereas the other SNRIs are more balanced inhibitors of both SERT and NET.
662
Q

How do SNRIs differ from TCAs? Why does this mean they’re used more?

A

The SNRIs lack the potent antihistamine, α-adrenergic blocking, and anticholinergic effects of the TCAs.
Thus, they tend to by favoured over the TCAs due to their better tolerability.

663
Q

What are the pharmacodynamics of TCAs?

A

They resemble the SNRIs in function, and their antidepressant activity is thought to relate primarily to their inhibition of 5-HT and noradrenaline reuptake.

664
Q

How do the different TCAs effect sertraline transporters and noradrenaline transporters differently?

A
  • Clomipramine has relatively little affinity for NET but potently binds SERT.
  • Nortriptyline is relatively more selective for NET.
  • Imipramine has more serotonin effect initially, but its metabolite, desipramine, then balances this effect with more NET inhibition.
665
Q

Mirtazapine has a complex pharmacokinetic mechanism. Tell me about it

A
  • It is an antagonist of the presynaptic α2 auto receptor and enhances the release of both noradrenaline and 5-HT.
  • In addition, it is an antagonist of 5-HT2 and 5-HT3 receptors.
  • Finally, mirtazepine is a potent H1 antagonist, which is associated with the drug’s sedative effects.
666
Q

What are some of the clinical uses of antidepressants?

A
  • Major depression - both acute and chronic
  • Anxiety disorders including PTSD, OCD, social anxiety disorder, GAD and panic disorder.
  • Pain disorders - particularly TCAs
  • Premenstrual dysphoric disorder - 2/52 every month is as effective as every day
  • Smoking cessation
  • Bulimia but don’t appear to be helpful in anorexia
667
Q

What is the difference between panic disorder and GAD?

A
  • Panic disorder is characterised by recurrent episodes of brief overwhelming anxiety, which often occur without a precipitant
  • GAD is characterised by chronic, free-floating anxiety and undue worry that tends to be chronic in nature
668
Q

What is the usual therapeutic dose for citalopram, escitalopram, fluoxetine, paroxetine and sertraline?

A

Citalopram - 20-60mg/day
Escitalopram - 10-30mg/day
Fluoxetine - 20-60mg/day
Paroxetine - 20-60mg/day
Sertraline - 50-200mg/day

669
Q

What are the usual therapeutic doses for venlafaxine and desvenlafaxine?

A

Venlafaxine - 75-375mg/day
Desvenlafaxine - 50-200mg/day

670
Q

What is the usual therapeutic dose for mirtazepine?

A

15-45mg/day

671
Q

What are the adverse effects of SSRIs?

A
  • Increased serotonergic activity in the gut - nausea, GI upset, diarrhoea - usually tend to improve after the first week
  • Diminished sexual function and interest
  • Increase in headaches and insomnia.
  • Weight gain
672
Q

What are the adverse effects of SNRIs?

A
  • They have many of the serotonergic adverse effects associated with SSRIs.
  • In addition, they also have noradrenergic effects including increased BP and HR, and CNS activation such as insomnia, anxiety and agitation.
673
Q

What are the adverse effects of TCAs?

A
  • The are primarily anticholinergic effects such as dry mouth, constipation, urinary retention, blurred vision and confusion.
  • The α-blocking property often result in orthostatic hypotension.
674
Q

What are the symptoms, level required to achieve suicide and treatment of TCA overdose?

A
  • Overdose can induce lethal arrhythmias, including VF and VT. In addition, BP changes and anticholinergic effects including altered mental state and seizures are someones seen
  • A 1500mg dose (< 7 days supply) is enough to be lethal in many patients
  • Treatment typically involves cardiac monitoring, airway support, and gastric lavage. Sodium bicarb is often administered to displace the TCA from cardiac sodium channels.
675
Q

What are the symptoms of MAOI overdose?

A

Autonomic instability, hyperadrenergic symptoms, psychotic symptoms, confusion, delirium, fever and seizures.

676
Q

What is the clinical presentation of serotonin syndrome?

A

HTN, hyperreflexia, tremor, clonus, hyperthermia, hyperactive bowel sounds, diarrhoea, mydriasis, agitation, coma - onset within hours

677
Q

What are the precipitating drugs for serotonin syndrome?

A

SSRIs
SNRIs
MAOIs
Linezolid
Tramadol
Ondansetron
Sumatriptan
MDMA
LSD

678
Q

How do you treat serotonin syndrome?

A

Sedation (benzodiazepines), paralysis, intubation, and ventilation; consider 5-HT2 block with chlorpromazine

679
Q
A