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

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

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)

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

67
Q

What does this graph show?

A
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
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

70
Q

What is the equation of Michaelis Menten kinetics?

A
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

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

73
Q

How do you calculate the dose rate?

A

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

74
Q

How do you calculate the maintenance dose rate?

A

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

75
Q

How do you calculate the loading dose?

A

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

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.

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

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.

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.

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
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
82
Q

What is the bioavailability?

A

The fraction of the dose which reaches systemic circulation intact

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

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)

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

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

87
Q

What is morphine’s bioavailiability?

A

Morphine is a drug that has poor bioavailability

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

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

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

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

92
Q

Biotransformation reactions can be split into two phases. Phase I and Phase II.

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

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.

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

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

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

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

98
Q

What happens in the oxidation reaction?

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 reduced oxygen, forming an ‘activated oxygen’-P450-drug complex
  4. This complex transfers activated oxygen to the drug substrate to form the oxidised product
99
Q

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

A

CYP3A4 metabolises over 50% of prescribed drugs

100
Q
A
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

102
Q

What are some classic P450 drug substrates?

A

Beta blockers
Warfarin
Oral contraceptives
Statins
Theophylline
Amiodarone
SSRIs
Opioids

103
Q

List some P450 inhibitors

A

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

104
Q

List some P450 Inducers

A

Inducers
Sulphonylureas
Carbamazepine
Rifampicin
Alcohol - chronic
Phenytoin

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
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

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
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

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 suscuptible to being clocked

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
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

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

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

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
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

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
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
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
119
Q

What are the extracardiac effects of verapamil?

A

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

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
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
122
Q

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

A
  • Adenosine
  • Digoxin
  • Magnesium
  • Potassium
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.
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

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
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.
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
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.
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
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

131
Q

How do calcium and magnesium interact with digoxin?

A
  • Hypercalcaemia increases the risk of digoxin toxicity
  • Hypermagnesaemia reduces the effects of digoxin
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
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
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
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
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
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
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
139
Q

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

A

Bladder - relaxes detrusor muscle

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
141
Q

What are the three drug groups traditionally used in angina?

A

Organic nitrates
Calcium channel blockers
Beta blockers

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
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.
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.

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

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.

146
Q

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

A

Dilation of large epicardiacl 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.

147
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.

148
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.

149
Q

What are the acute adverse effects of nitrates?

A

Orthostatic hypotension
Tachycardia
Throbbin heaache

150
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

151
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

152
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
153
Q

What are the nitrate effects in variant angina?

A

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

154
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

155
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