Pharmacokinetics Flashcards

1
Q

What does pharmacodynamics describe

A

the interaction of drugs and their receptors (including affinity and efficacy) and the tissue distribution of the receptor

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

What does pharmacokinetics describe

A

the factors that determine the concentration of the drug at the target receptor

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

What does relationship between administered dose and concentration at the target site depend upon

A

absorption,
distribution,
metabolism
excretion

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

How does plasma concentration of a drug vary over time for:

a) a single iv bolus dose
b) oral dose

A

a) initially high then decreases

b) initially increases to a peak as drug is absorbed, before decreasing as metabolism and excretion increase

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

What is the range the plasma conc of a drug must be within

A

therapeutic window - higher than minimum effective conc. but lower than the minimum toxic conc

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

Do you want the therapeutic window to be wide

A

yes this is ideal

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

How is the therapeutic window reflected in dosing strategies

A

An effective dosing strategy will maintain the plasma concentration within
the therapeutic window for as long as necessary

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

What kind of chemicals are local anaesthetics

Where do they act? What does this mean?

A

weak bases

intracellular side of the Nav channels
They must cross the plasma membrane to access the intracellular compartment.

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

How do local anaesthetics cross into the intracellular compartment

A

in their un-ionised form (D)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Why does the chemistry of inflammation affect the transport of local anaesthetics

A

Local inflammation decreases tissue pH (higher H+
concentration).

This shifts the equilibrium to favour DH+

Local anaesthesia is delayed or even prevented

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

Which form of a weak base/acid is more likely to cross the PM

A

un-ionised form is usually more lipid soluble so diffuses readily, whereas
the ionised form cannot cross the membrane

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

Name 2 subgroups in the SLC superfamily

A

organic anion transporters (OATs)

organic
cation transporters (OCTs).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Name a SRC that is a drug target itself

A

SERT (transports serotonin)

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

Name members of the SRC superfamily that are a) highly selective and b) less selective

A

a) SERT

b) OCT, OAT - can transport a wide range of structurally diverse molecules

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

Describe the ABC superfamily

Give an example

A

ATP-binding cassette superfamily use ATP to drive drug efflux from cells.

MDR1 (P-glycoprotein, P-gp) is a particular example. They may be important drug resistance.

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

What are SLC and ABC superfamilies particularly important for in pharmacokinetics

A

absorption from the small intestine, excretion into the bile or urine, and transport across
the blood-brain barrier

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

What is the only time a drug does not have to cross a barrier to reach the plasma

A

iv

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

What is the fastest and most certain route of drug administration

When would this be useful

A

iv

if the drug has a small therapeutic window

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

Why might you use a slow iv administration of a drug over a fast administration

A

A rapid bolus injection can produce a very high plasma
concentration, initially in the right side of the heart and pulmonary circulation. Slow i.v.
infusion avoids this high peak plasma concentration while still avoiding uncertainly inherent
in absorption from other sites

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

Disadvantages of iv drug administration (3)

A

injection or infusion requires a

skilled practitioner, and inconvenience to the patient. There is also risk of infection

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

What are disadvantages of subcut. or intramuscular injections

A

rate of absorption can be unpredictable and inconsistent. These
injections can also be painful.

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

What is the rate of absorption dependant on for subcutaneous/ intramuscular injections

A

diffusion through the tissue and removal by local

blood flow, as long as the drug can cross the capillary endothelium

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

How does absorption differ between intramuscular and subcit injections

A

Blood flow to
the muscle is higher than to the skin, so absorption from intramuscular is often more rapid
than for subcutaneous. Exercise increases muscle blood flow, increasing absorption.

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

Why is oral administration the most common route for drug administration

A

cheap and easy to administer (self administering)

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

How are oral drugs absorbed

A

via gut epithelium

lipophilic molecules will diffuse passively

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

What does rate of diffusion of oral drug across gut epithelium depend upon (4)

A

rate of diffusion is highest for highly lipophilic drugs, whereas
charged or highly polar drugs diffuse poorly. The rate of diffusion also depends on the
concentration gradient and the surface area of membrane.

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

Where does most oral drug absorption occur in the gut

A

small intestine (large SA)

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

Are drugs absorbed in the stomach

A

e stomach has a relatively low surface area and a thick mucosa, so little drug absorption
occurs. An exception are the tetracyclines, which are soluble at acid pH but insoluble at
neutral pH.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Why are tetracyclines absorbed in the stomach

what if they are not absorbed in the stomach

A

soluble at acid pH but insoluble at
neutral pH.

Any tetracycline that enters the intestines precipitates and is lost in the faeces

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

What happens to the equilibria of weak bases in the stomach

what does this mean for absorption

A

shifted to DH+

poor absorption, may become trapped

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What happens to weak acids in the stomach

A

shifts equilibrium towards DH so absorption is favoured

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

Does the small intestine favour absorption of weak acids or bases

A

upper small intestine favours absorption of weak bases over weak acids. Despite these
effects, the extremely large surface area of the small intestine means that most oral drugs
are absorbed in the small intestine

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

Name a drug that is absorbed by a gut transporter

which transporter

what does it treat

A

levadopa

phenylalanine transporter

Parkinson’s

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

Name 2 things that affects general drug absorption in the gut

A

splanchnic bloodflow

gastric motility

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

How does gastric motility affect drug absorption

A

The rate of
absorption of drugs that are mainly absorbed in the intestines will be increased if gastric
emptying is accelerated and decreased if gastric emptying is slowed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

How does vomiting or diarrhoea affect drug absorption

A

reduces absorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

How does splanchnic blood flow affect drug absorption

A

Increased splanchnic blood
flow after a meal may increase the rate of absorption, whereas decreased splanchnic blood
flow in heart failure may reduce the rate of drug absorption.

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

How can you stop stomach acid degrading oral drugs

A

enteric coating

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

describe enteric coating

A

usually stable at acidic pH but break down at higher pH.

Digestion of peptides and proteins prevents their use as orally-active drugs (e.g. insulin)

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

What is the first pass effect

A

To reach the systemic plasma, the drug must be absorbed through gut epithelium and then
travel through via the portal circulation and the liver. Many drugs are metabolised by
enzymes in the small intestinal wall or the liver

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What is bioavailability

A

The fraction of the delivered dose that reaches the systemic circulation

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

Name some factors that can cause low bioavailability (5)

A

inability to cross the gut epithelium,

transport back into the gut lumen,

metabolism of the drug in the intestinal
wall or liver (or by bacteria),

patient-specific factors such as interactions with other drugs
or food,

altered motility (e.g. vomiting).

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

Which drug administration routes can be taken to avoid stomach or the first pass effect (2)

A

rectal or sublingual

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

What is the series of the aqueous compartments in the body considered in pharmacokinetics

A

plasma |(endothelium) | interstitial fluid | (PM) | intracellular | (specialised barrier eg BBB) | transcellular

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

Describe the vascular endothelium

A

a single layer of cells that lines blood vessels and separates the
plasma compartment from the interstitial compartment. The nature of the endothelial
barrier varies between tissues.

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

What and how can some molecules pass through the vascular endothelium

A

endothelial cells of peripheral capillaries have small gaps between the cells that allow free passage to polar small molecules less than approximately 500-600 daltons. Small, lipophilic molecules can also diffuse through the endothelial cell plasma membrane

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

Give an example of a larger molecule that is restricted to the plasma unless specifically transported

Why is it restricted to the plasma

A

the anti-coagulant, heparin

Unfractionated
heparin is a carbohydrate polymer of variable chain length, with a molecular weight of 3-30
kDa. It cannot permeate the gaps between endothelial cells, and is not specifically
transported

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

Describe the capillary walls supplying the brain

A

have very tight junctions between endothelial cells and are further surrounded by astrocytes

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

which molecules can pass the BBB

A

if they are sufficiently lipophilic or if taken up by transporters/ via transcytosis

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

What can disrupt the BBB

Give an example

A

inflammation

In bacterial meningitis, for example, allowing increased access to hydrophilic
antibiotics such as aminoglycoside antibiotics and β-lactam antibiotics (e.g. penicillin).

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

Name an important BBB transporter

A

Pglycoprotein (P-gp; MDR1)

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

What is ivermectin toxicity associated with

A

a frameshift mutation and

premature stop codon in the mdr1a gene in collie dogs

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

Why can human MDR1 polymorphisms be associated with drug toxicity

What is it also associated with

A

. P-gp reduces the
therapeutic effects of some drugs in the CNS, but also reduces the CNS toxicity of some
drugs

even clinical efficiency of antidepressants, but
this is less well established.

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

How does fetal pH relate to maternal pH

What can this lead to

A

Fetal pH is usually slightly lower

than maternal pH, which can lead to ion trapping

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Name an example which shows the placenta is not a perfect barrier to drugs

What is an ongoing scandal showing it is still a problem

A

thalidomide

The recent sodium valproate scandal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What is Vd and what does it describe

A

volume of distribution (Vd)

often used to describe the distribution of drugs in the body.

the volume that would contain the total amount of drug in the body at a
concentration equal to the plasma concentration.

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

What is the equation to Vd

A

𝑉𝑑 =
𝑡𝑜𝑡𝑎𝑙 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝑑𝑟𝑢𝑔 𝑖𝑛 𝑡ℎ𝑒 𝑏𝑜𝑑𝑦
————————-
𝑝𝑙𝑎𝑠𝑚𝑎 𝑐𝑜𝑛𝑐𝑒𝑛𝑡𝑟𝑎𝑡𝑖𝑜𝑛 (𝐶)

where C=C(free) + C (bound)

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

What is the Vd of heparin similar to

A

plasma volume (restricted to this compartment)

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

What kind of drug is gentamicin

Why is it useful when considering Vd

A

aminoglycoside antibiotic

it is small enough to cross the endothelium between the cells. It
distributes in the plasma and interstitial compartments -
represent the total extracellular fluid

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

What is the Vd of ethanol

A

similar to total body water water (42-45 litres) as it has a broad distribution throughout the body compartments

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

How does tissue binding/ partitioning into fat affect Vd

Why

A

increases Vd

it is the free (unbound) drug that exchanges between
compartments. As more drug binds to tissues in the interstitial, intracellular or fat
compartments, the concentration of free drug there falls. More drug will leave
the plasma and the plasma concentration falls. Therefore, tissue binding or partitioning
means that a greater amount of drug is accommodated with a lower plasma concentration.

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

Where can tissue binding occur

Where does it mostly happen for lipophilic drugs

A

to membranes, extracellular proteins or receptors

more likely to partition into fat

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

Which tissues can heavy metals adsorb to

A

bone

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

Why is the Vd very high for highly lipophilic drugs or heavy metals

A

Heavy metals

can become adsorbed to bone, lipophilic drugs are more likely to partition into fat so more tissue binding has occurred

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

What is the Vd of morphine

A

250 litres!!

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

What must be remembered when considering the Vd of thiopental

A

it is a very lipophilic GA so partitioning in fat is particularly important

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

What can decrease Vd

A

binding to plasma proteins

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

Why will Vd increase as more drug binds to plasma proteins

A

total concentration in plasma will be higher so

total drug in body/ plasma conc

will be smaller as denominator is bigger

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

What does binding to plasma proteins allow in PK

A

allows the plasma to carry more drug

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

Is it free or bound drug that exchanges with other body compartments

A

free

It is the free (unbound) drug that exchanges with other body compartments, and it is the free drug that is cleared by metabolism or renal excretion.

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

What is the major binding protein in plasma

What is the plasma [albumin]

How does this concentration compare with therapeutic concentration of most drugs

A

albumin

0.6mM

much higher than therapeutic conc

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

True or false

Albumin has many binding sites

A

true
including two sites for acidic drugs (e.g. warfarin,
salicylic acid, phenytoin) and other sites for neutral and basic drugs

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

Can warfarin bind to albumin

A

yes warfarin is acidc and albumin has two sites for acidic drugs (e.g. warfarin,
salicylic acid, phenytoin).

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

Give 4 instances when albumin levels can fall

A

liver disease,
old age,
nephrotic disease
major burns

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

What must you consider when using data about oral administration from one species to another (4)

What does this lead to

A

Taste and feeding behaviours (e.g. neophilia vs. neophobia) can affect how best to administer a drug to different species.

Differences in digestive tract physiology (e.g. in ruminants) can affect the rate and extent of drug absorption.

Grooming behaviour can lead to (often unintended) oral administration of topically-applied drugs.

Species-specific differences in drug metabolism are also extensive.

This can lead to poor correlations of bioavailability in humans other animals.

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

Which drugs can bind to α1-glycoprotein

Where is it expressed

A

basic (partitcularly beta blockers and antidepressants)

Although normally expressed at a low level in plasma,
it is an acute phase reactant that is increased during inflammation or stress.

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

True or false
plasma protein binding is saturable

What does this mean for C(free) and C(bound)

Which drugs is tshi knowledge important for

A

true

When a low percentage of binding sites are occupied,
an increase in dose gives a proportional increase in Cfree and Cbound. As the binding sites become saturated, an increase in dose will disproportionately increase in Cfree

drugs that almost saturate albumin in their therapeutic range (e.g.
phenytoin).

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

Give an example of competition for albumin binding

A

bilirubin which is normally eliminated by conjugation by neonates but competes against sulphonamides for binding

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

Why can it be dangerous to give sulphonamides to neonates

A

bilirubin normally
eliminated by conjugation, which is slow in neonates. If it is displaced from plasma proteins
by e.g. sulphonamides, Cfree will increase. This can lead to increased bilirubin in the brain and neurological damage.

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

Is a drug evenly distributed once it reaches the plasma

A

Once a drug has been absorbed into the plasma it will be rapidly and evenly distributed
through the blood. However, blood supply to different tissues is not evenly distributed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

Which organs are not well perfused

A

skin, skeletal muscle and

fat

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is a consequence of some drugs being rapidly distributed through well perfused tissues but distribute through poorly-perfused tissues more slowly

A

that the apparent volume of distribution immediately after an i.v. bolus injection may be
much lower than the apparent volume of distribution much later, or when a steady state is
reached during infusions.

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

What are drugs metabolised by

A

the same enzyme systems that defend us from xenobiotics

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

What are phytoalexins

A

toxins against predation

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

What are important sources of xenobiotics

A

plants
environmental pollution, cosmetic products, food additives, agrochemicals, foods processing, as well as pharmaceutical drugs.

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

Why do hydrophilic xenobiotics not have to be metabolised

A

Membranes are a barrier to absorption of hydrophilic

xenobiotics, so can be readily excluded. Hydrophilic xenobiotics that are absorbed can also be readily excreted.

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

What would happen to lipophilic xenobiotics if they were not metabolised

A

not readily excreted , so they would accumulate in fats and phospholipid bilayers

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

What is the general strategy for metabolism of of lipophilic molecules

A

to convert a

lipophilic molecule into a more hydrophilic molecule that can be readily excreted.

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

What is the main site of xenobiotic metabolism

Name 6 other sites

A

liver

small intestine,
nasal mucosa
lungs
skin 
kidneys
blood
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What is the first site for first pass metabolism

A

small intestine

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

How can the process of hepatic drug metabolism be divided

A

into phase 1 (functionalisation) and phase 2 (conjugation)

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

What are the purposes of phases 1 and 2 of hepatic drug metabolism

A

phase 1: makes a more reactive metabolite

phase 2: Makes a less reactive metabolite that is
(usually) more hydrophilic and higher molecular weight

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

What happens in phase 1 of hepatic drug metabolism

A

introduces/ unmasks a functional group to alter biological properties of drug

may activate or inactivate the drug

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

Give an example of a pro-drug

A

codeine - turned on by hepatic drug metabolism

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

What does phase 2 of hepatic drug metabolism do

how does it affect drug activity

A

uses the reactive functional group to add another molecule. This usually increases
the water solubility of the drug.

Phase 2 can also inactive a drug, though even some
conjugated drugs can have activity

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

name a drug that already has an appropriate functional group so does not require phase 1

A

paracetamol

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

Which superfamily is very important in phase 1 of drug metabolism

A

cytochrome P450

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

What does the CYP superfamily comprise

Is it well conserved?

A

18 CYP families with 57 genes (and 58 pseudogenes) have been identified in the human genome.

no: huge diversity between species, in terms of number of genes,
expression of orthologues, tissue distribution patterns, substrate specificities and activities,
and sensitivity to inhibitors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

How many drugs does CYP2D6 metabolised

A

involved in metabolism of up to 25% of current drugs.

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

How does the presence of CYP2D6 vary between individuals

What does this mean for drug research

A

Humans carry only one CYP2D6 gene, which is missing or nonfunctional in 7-8% of Caucasian Americans. Mice have nine different, functional Cyp2d genes and rats have six

This makes it difficult to extrapolate drug metabolism studies from animals to humans

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

Which are the most important CYP families

A

CYP1, 2, 3

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

Which CYP is responsible for metabolism of half of clinical drugs

A

CYP3A4 is responsible for metabolism of approximately 50% of clinical drugs

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

How can you tell a CYP that deals with metabolism of endogenous products from those which metabolise xenobiotics

A

CYPs involved
in metabolism of endogenous substrates (such as cholesterol) have very strict substrate
specificity. Xenobiotic metabolising CYPs cannot afford this selectivity. They must accommodate large structural diversity of substrates with a large and fluid substrate binding
site

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

What does the broad scope of xenobiotic metabolising CYP sacrifice

A

rate - slower than other CYP

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

How does the wide structural diversity of of potential substrates affect drug-drug interactions

A

makes drugdrug interactions more likely

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

Where are CYPs located

A

ER membrane

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

Where is a site of lipophilic drug accumulation in the cell

A

ER membrane where CYPs are located

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Which molecules do CYPs use to metabolise drugs

A

O2 and haem.

Haem binds O2 into CYP active site.

CYPs also use H+
from NADPH, supplied by NADPH-cytochrome P450 oxidoreductase

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

What is a potentially harmful outcome of CYP activity

How is this mitigated

A

Often, more O2 is
consumed than needed, so superoxide is produced.

Superoxide dismutase converts this safely into water

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

How can you inhibit CYPs

A

Since many drugs can be metabolised by the same CYP isoform, drugs may compete

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is Prozac a substrate for

A
Fluoxetine (Prozac) is a substrate of CYP2D6, CYP2C19 and CYP3A4, and can also inhibit
these isoforms (particularly CYP2D6).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

Do drugs react the same way with each isoform of the CYP

A

Some drugs are metabolised by one CYP isoform but act as competitive inhibitors of other
isoforms. (Quinidine, for example, is a competitive inhibitor of CYP2D6 but not a substrate.)

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

How does ketoconazole react with CYP3A4

A

forms a complex with the Fe3+ form of haem in CYP3A4

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

How can drugs show mechanism based inhibition of CYPs

A

The product of
oxidation binds covalently to the CYP, irreversibly blocking the enzyme. This is also called
‘suicide inhibition’.

115
Q

Why can you not have grapefruit juice on certain drugs

Which drugs does this include

A

A component of grapefruit juice4
(and perhaps some other citrus juices) potently inhibits CYP3A4.

Since CYP3A4 is responsible
for metabolising almost 50% of drugs, this is major problem.

Affected drugs include many dihydropyridine Ca2+ channel blockers (e.g. nifedipine), statins (e.g. simvastatin), antibiotics (e.g. erythromycin) and immunosuppressants (e.g. cyclosporine), amongst others.

116
Q

Which CYP does grapefruit juice affect and where in the body does it happen

A

CYP3A4

The major site of action appears to be CYP3A4 in the intestinal wall rather than the liver.

117
Q

How can xenobiotics affect the impact of CYPs in the long term

A

can affect expression via nuclear receptors

118
Q

Are the nuclear receptors which control CYP expression promiscuous?

A

yes
The flexible ligand binding site of these nuclear receptors allows them to be activated by structurally diverse molecules (usually small and lipophilic)

119
Q

Describe the pathway for activation of CYP genes via nuclear receptors

A

pregnane X receptor (PXR) and constitutive androstane receptor (CAR) heterodimerise
with the retinoid X receptor (RXR), allowing them to bind to xenobiotic response elements (XREs) in upstream promotor regions

CYP3A4 is upregulated

120
Q

Name 2 drugs that can activate PXR

Name a herbal xenobiotic that also does so

A

phenobarbital
rifamycin

St John’s Wort

121
Q

What is St John’s Wort used to treat

A

mild/ moderate depression

122
Q

Which CYP is especially unregulated by XRE

Why is this important for pharmacologists to know

A

CYP3A4

CYP3A4 metabolises 50% of all clinical drugs

123
Q

What is FMO

What does it do

A

Flavin-containing monooxygenases (FMO) in the liver

catalyse oxidation reactions, using FAD rather than haem

124
Q

What is the most abundant FMO in the liver

What can it metabolise (3)

A

FM03

FMO3 can metabolise
structurally diverse drugs, including amphetamines, the anti-psychotic clozapine, and the
histamine H2 receptor antagonist ranitidine

125
Q

What kind of drugs are clozapine and ranitidine? What is the common denominator

A

clozapine: anti-psychotic
ranitidine: H2 histamine receptor antagonist

both metabolised by FMO3

126
Q

What causes fish odour syndrome

A

mutations in FMO3 so TMA is not metabolised and builds up to be released into the sweat and urine causing a fishy smelll

127
Q

What can be released from CYPs

what happens to them

A

highly reactive epoxides

epoxide hydrolases detoxify them

128
Q

What is carbamazepine

A

a prodrug activated by CYPs used to treat epilepsy

129
Q

How is carbamazepine activated and deactivated

A

activated by CYPs to generate a

reactive (and active) epoxide. This is hydrolysed by microsomal EH, which also inactivates the drug.

130
Q

What is often used in conjunction to carbamazepine

why

A

valproic acid (an anticonvulsant)

inhibits microsomal EH, increasing the
concentration of the active metabolite of carbamazepine and delaying its elimination.

131
Q

What is the importance of alcohol dehydrogenase

A

oxides ethanol to ethanal (a toxic acetaldehyde)

this must then be converted to acetate by acetaldehyde dehydrogenase

132
Q

What does disulfiram do in alcohol metabolism

A

inhibits acetaldehyde dehydrogenase

133
Q

Where are esterases found (2)

Name a drug these are important for

A

in the intestinal wall and liver

Aspirin is
hydrolysed to salicylic acid by esterases

134
Q

What is one of the most common Phase 2 reactions

A

Glucuronidation (addition of glucouronic acid)

135
Q

What is glucuronidation in phase 2 catalysed to

why is this reaction important

A

by UDP-glucuronosyltransferases (UGTs)

Many drugs are excreted as glucuonides in urine or bile

136
Q

Which functional groups can glucuronidation occur with

therefore…

A

hydroxyls, carboxyls, sulfuryl, carbonyl, and amide groups, so can occur with many structurally diverse drugs

137
Q

What is the pKa of the carboxylic acid in glucuronic acid

what does this mean

A

pKa of 3-3.5.

It is mostly ionised at physiological pH in cells, in the blood, and in urine.

138
Q

Describe phenytoin metabolism (phases 1 and 2)

A

m involves CYP-dependent hydroxylation (phase 1) followed by
glucuronic acid conjugation (phase 2)

therapeutic doses of phenytoin are close to saturation of this pathway

139
Q

What can phenytoin act as an inhibitor of (2 things involved in drug metabolism)

A

as an inhibitor of CYP2C9 and of glucuronidation.

140
Q

Give 3 uses of codeine and morphine

give 4 side effects

A

pain relief
cough suppression
antidiarrheal

depression,
constipation,
sedation
addiction.

141
Q

Through which receptor does the analgesic effect of codeine and morphine arise

A

µ-opioid receptor

142
Q

True or false

codeine is a prodrug

A

true -

it is a weak µ-opioid agonist, and ineffective at plasma
concentrations usually achieved. It is considered to be a prodrug

143
Q

How does the affinity of codeine to the µ-opioid receptor compare to morphine compare

A

Morphine has a much greater affinity (200-300x).

144
Q

What happens to codeine in the liver

A

Most is directly glucuronidated to codeine-6-glucuronide, and some
converted to norcodeine by CYP3A4
5-15% of the codeine is coverted to morphine

145
Q

How does the affinity of the products of codeine metabolism compare to codeine itself

A

Codeine-6-glucuronide and norcodeine have a similar
affinity for the µ-opioid receptor to codeine
Morphine has a much greater affinity (200-300x).

Morphine and norcodeine are also
glucuronidated. Morphine-6-glucuronide is a high affinity µ-opioid agonist (similar to
morphine) and may be responsible for the some of the actions and side effects of codeine
and morphine. Morphine-3-glucuronide is ineffective at the µ-opioid receptor, but may also
contribute to side effects and toxicity.

146
Q

Why might heroin be considered a prodrug?

the handout does NOT call it this, it’s just for the purpose of this flashcard

A

Heroin (diacetylmorphine) is also converted to morphine for its action

147
Q

What allows heroin to cross the BBB

A

ts acetyl groups

increase its lipophilicity, which means that it can readily cross

148
Q

Where is heroin converted to morphine

A

in the brain (after crossing the BBB)

149
Q

Name a CYP2D6 inhibitor

A

fluoxetine

150
Q

What effect does fluoxetine have on codeine

A

it is a CYP2D6 inhibitor so reduces morphine formation from it
prevents the analgesic effect

151
Q

How do individuals’ abilities to metabolise codeine (5 points)

A

CYP2D6 expression levels vary in humans.

Normal (‘extensive’) metabolisers have one or two functional copies of the CYP2D6 gene.

Ultra-rapid metabolisers have >2 functional copies, and are at high risk of morphine toxicity at normal doses of codeine.

Codeine should also be avoided in breast-feeding mothers who are ultra-rapid metabolisers.

Poor metabolisers lack functional CYP2D6, and do not obtain adequate
pain relief from codeine.

152
Q

What is the effect of CYP3A4 inhibitors on codeine metabolism

A

Codeine metabolism via CYP3A4 is a minor pathway in most people, so CYP3A4 inhibitors have little effect.

However, strong CYP3A4 induction may reduce formation of morphine.

153
Q

What are the 3 routes of paracetamol metabolism following a therapeutic dose

A

major routes:
glucuronidation and sulfonation (both phase 2)

A minor fraction is oxidised to NAPQI, a
reactive metabolite, which is detoxified by conjugation to glutathione (GSH).

154
Q

What happens to the different metabolism pathways in a supratherapeutic dose of paracetamol

A

sulfonation pathway is saturated.

At higher toxic doses gluruonidation is also saturated

thus a higher proportion is converted to NAPQI, leading to depletion of GSH and hepatic cell injury or cell death

155
Q

Which molecule do you want to replace in paracetamol overdose

How can you do this

A

GSH

by exogenous NAC

156
Q

Why might phenytoin lead to paracetamol hepatic injury

what else might increase risk of this

A

phenytoin and pentobarbital are glucuronidation inhibitors

Induction of CYP2E1 by chronic alcohol consumption may also increase hepatotoxicity risk.

157
Q

What is the principle site for excreting water soluble drugs

A

kidneys
(Drugs can be passively filtered from plasma and secreted into the renal tubules. They can also be reabsorbed from the tubules. )

158
Q

What is normal plasma flow

what is normal GFR

A

625ml/min

125ml/min

159
Q

What 2 features must a drug have to be freely filtered by the kidneys/ readily cleared by glomerular filtration

A

polar

not heavily bound to plasma proteins

160
Q

f𝑖𝑙𝑡𝑟𝑎𝑡𝑖𝑜𝑛 𝑟𝑎𝑡𝑒 𝑜𝑓 𝑑𝑟𝑢𝑔 =

A

𝐺𝐹𝑅 × 𝐶𝑓𝑟𝑒e

161
Q

Why does glomerular filtration not immediately change plasma [drug]

A

water and drug filtered in proportion

162
Q

How does the number of binding sites for drugs, and bound drugs themselves change from afferent to efferent arterioles in the kidney

A

both 20% higher in the efferent arterioles than in the afferent arterioles

163
Q

What factors preserve the

equilibrium between bound and free drug in the kidney (before and after glomerulus)

A

Glomerular filtration does not immediately change the plasma concentration of a drug, since water and drug are filtered in proportion. In addition, the loss of water means that the number of binding sites for drugs, and bound drugs themselves, are 20% higher in the efferent arterioles than in the afferent arterioles

164
Q

How does an increase in plasma protein binding affect clearance of a drug if the drug is ONLY filtered

A

decreases clearance

165
Q

Which transporters secrete drugs from peritubular capillary plasma to the tubule

A

two families of SLC transporter: OATs and OCTs

166
Q

What do OATs in the nephron transport

A
acidic drugs in their negatively charged anionic form (eg, penicillin) 
endogenous acids (eg uric acid.) Glucuronide and sulphide conjugates  (such as conjugates of paracetamol
167
Q

What do OCTs transport

A

organic bases in their protonated cationic form (eg morphine)

168
Q

True or false

drugs can compete at OATs and OCTs

A

true

Probenecid and penicillin are both transported by OATs.

Probenecid prolongs the action of penicillin by reducing its tubular secretion.

169
Q

How does drug secretion into the tubule affect plasma [drug]

what is the effect of secretion on drug bound to plasma protein

A

lowers the free concentration in the plasma, since drug movement is not accompanied by water.

shifts the equilibrium between bound and free drug. More drug is released from the plasma proteins.
If secretion is fast enough the newly released drug is also secreted

170
Q

If secretion is fast enough the drug newly released from plasma protein is also secreted. what does this mean for renal clearance

A

an increase in plasma protein binding has little effect on renal clearance.

171
Q

When can a drug have a clearance greater than GFR

A

Drugs that are rapidly secreted and not reabsorbed have clearance greater than GFR and can approach RPF

172
Q

What is required for drugs to be reabsorbed from the tubule

A

If the drug can cross the renal tubule then it will diffuse down its concentration gradient (out of tubule)

173
Q

Which drug traits mean they are poorly or well reabsorbed from the renal tubule

A

Lipid soluble drugs are poorly excreted by the kidney since they are readily reabsorbed.

In contrast, polar drugs are poorly reabsorbed and are readily excreted

174
Q

How does urine pH affect excretion of drugs

A

alters the dissociation of acidic and basic drugs in the tubular lumen. This can lead to ion trapping

175
Q

How does urine pH compare to plasma pH

how does this affect excretion of drugs

A

urine is usually more acidic

favours excretion of basic drugs in their cation form, and reabsorption of acidic drugs

176
Q

How can you increase the excretion of acidic drugs in urine

what is this used for

A

by increasing urine pH through infusion of NaCO3

following an overdose of eg aspirin, barbiturates

177
Q

How does increasing urine pH affect excretion of basic drugs

A

increases reabsorption, so decreases excretion, of basic drugs

178
Q

Why do the effects of urine pH on drug excretion explain the importance of hepatic metabolism for renal excretion

A

conjugated metabolites are usually more water soluble, so freely filtered.

They can also be secreted via OATs.

Since they are more polar than their parent drug, they are less likely to be reabsorbed from the tubule.

179
Q

How are drugs excreted in faeces

A

hepatocytes secrete drugs and their metabolites from plasma into bile using similar transporters to the renal tubules. Bile is delivered to the small intestine. There, the drug can be excreted in faeces

180
Q

Are all drugs that enter the small intestine in bile excreted in feces?

A

no
once they have entered the small bowel they can be excreted in faeces; unmodified drugs can be reabsorbed; and conjugates can be hydrolysed, potentially regenerating an active drug

181
Q

which drug conjugate is particularly likely to be hydrolysed once it has entered the small bowel in bile

A

Glucuronides

182
Q

What happens to drug conjugates that are hydrolysed after entering the small bowel in bile

A

hydrolysed drug can then be either excreted in faeces or reabsorbed.

If it is reabsorbed then is called enterohepatic circulation. This slows the rate of elimination of a drug from the plasma, prolonging the effect of the drug

183
Q

What must pharmacologists be aware of when studying dosing patterns for animals that perform coprophagy

A

active drugs can enter the feces and so be taken into the animal that ingests the feces

184
Q

Given 2 examples of coprophagy causing problems for pharmacologists

Give a further example of a similar problem that did not arise from coprophagy

A

In one study, foals were treated with erythromycin to treat an infection. Mares practising coprophagy with their foal’s faeces had a high incidence of colitis because of the erythromycin.
In another study, rabbits showed a rebound in plasma concentration of a drug 24 hours after i.v. injection.

untreated horses were housed in a box previously used for naproxen-treated horses. The untreated horses were found to excrete naproxen in their urine.
caused by cross-contamination of bedding and ingestion of contaminated straw

185
Q

What does first order kinetics refer to

A

the rate of elimination is directly proportional
to the plasma concentration.
A chart of rate of elimination versus plasma concentration would give a straight line.

186
Q

Which drug elimination methods follow Michaelis Menten kinetics

A

Elimination by metabolism and carrier-mediated drug transport (e.g. tubular secretion) which are both dependent on enzymes

187
Q

For many drugs, the therapeutic concentration is much lower than Km, so the rate of
elimination is linear with respect to concentration. Why?

A

𝑟𝑎𝑡𝑒 𝑜𝑓 𝑟𝑒𝑎𝑐𝑡𝑖𝑜𝑛 =
𝑉𝑚𝑎𝑥. 𝐶
————-
𝐾𝑚 + C

if C

188
Q

True or false

renal filtration will always be linear

A

true
Renal filtration is not enzyme dependent and cannot be saturated.
The rate of elimination in this manner with always be linear.

189
Q

Compare a first order and zero order reaction

A

A first order reaction has a rate proportional to concentration of a single reactant.

a zero order reaction is independent of the concentration of the reactant. (When elimination is saturated and has reached the fixed, maximum rate, the elimination is zero order.)

190
Q

What is clearance considered a measure of (2)

A

a measure of the body’s ability to eliminate a drug, normalised to plasma concentration

s sometimes thought of as the flow of arterial blood that would be completely cleared of a drug at that rate of elimination

191
Q

If a rug is cleared through different routes, how do they relate?

A

Clearances through different routes are additive:

𝑡𝑜𝑡𝑎𝑙 𝑐𝑙𝑒𝑎𝑟𝑎𝑛𝑐𝑒 = ℎ𝑒𝑝𝑎𝑡𝑖𝑐 𝑐𝑙𝑒𝑎𝑟𝑎𝑛𝑐𝑒 + 𝑟𝑒𝑛𝑎𝑙 𝑐𝑙𝑒𝑎𝑟𝑎𝑛𝑐𝑒 + ⋯

192
Q

If elimination is first order, clearance will be…

A

constant

193
Q

What does the single compartment model assume

A

the body behaves
like a single, well-mixed container into which a dose of drug, D, is rapidly added by IV injection.

i.v. injection means that the model ignores absorption.

The [drug] in the plasma, C, is in rapid equilibrium with the drug in any extravascular tissues that the drug can access

Since ‘well mixed’, we can ignore distribution

assumes elimination is 1st order kinetics

194
Q

In the single compartment model, concentration of drug in the plasma, C, is in rapid equilibrium with the drug
in any extravascular tissues that the drug can access. Does this mean [drug] everywhere is equal to plasma [drug]

A

no

rather that these concentrations are proportional to the plasma concentration at all times

195
Q

What does the single compartment model assume for elimination

what does this mean

A

assumes that the drug elimination follows first order kinetics.

This means that the rate of elimination is proportional to the plasma concentration.

Elimination may be via metabolism or excretion, or both

196
Q

How does Vd change over time in the single compartment model

A

Since the compartment is well mixed, Vd remains constant throughout

197
Q

How to work out [drug] in plasma at any time in the single compartment model

A

C=X/Vd

X=The amount of drug remaining in the compartment
Vd= volume of distribution

198
Q

How to calculate rate of change of plasma concentration in single compartment model

A

dC
—– =-Ke.C
dt

199
Q

How to workout the rate of increase in amount of drug in the body in single compartment model?

A

dX
—– = -Ke.X
dt

200
Q

How do work out Vd in single compartment model

A

Vd=CL/Ke

201
Q

What is the equation you should use to plot plasma concentration against time (single compartment model)

How is this made into a straight line
What is the important information that can be gathered from this line?

A

C=Co.e^(-Ke.t)
where C0 is the plasma concentration at time zero

ln(𝐶) = ln(𝐶o) − 𝑘𝑒.t

The slope is –ke and the vertical intercept is ln(C0).

202
Q

What is the equation for half life in the single compartment model

A

t0.5= 0.693/Ke

203
Q

How can you find the clearance from a graph of conc vs time (SINGLE COMPARTMENT MODEL)

A

area under the curve

AUC=Co/Ke

CL=D(i.v.)/AUC

204
Q

What is the 2 compartment model used to model

A

describes a situation where the drug does not appear to distribute instantaneously.
Instead, the drug appears to distribute to some parts of the body more rapidly than others

205
Q

How does distribution work in the 2 compartment model

A

drug is injected into a central compartment.
This represents the blood (and therefore plasma), and often represents other well-perfused tissues, such as the liver, kidney, heart, brain and lungs.

The drug is eliminated from this
compartment by metabolism or excretion. The drug can also slowly distribute into a peripheral compartment.

This often represents poorly-perfused tissues, such as skin, fat and skeletal muscle (at rest).

206
Q

What is the time course for drug distribution through the 2 compartments in the 2 compartment model

A

When the drug is injected, it rapidly distributes through the central compartment.

It will be at a high plasma concentration as the volume of distribution is relatively low.
Over time, it will also slowly distribute into the peripheral compartment. This increases the volume of distribution.

This will affect the plasma concentration and the rate of elimination.

Drug in the peripheral compartment must return to the central compartment to be eliminated

207
Q

How does plotting C against time in 2 compartment model differ from 1 compartment

A

may be difficult to see any difference from the single compartment model.

However, when ln(C) is plotted against time the chart is no longer a straight line. This is indicative of a distribution phase

208
Q

Why is ln(C) plotted against time in 2 compartment model different from single compartment

A

indicative of a distribution phase. The plasma
concentrations soon after injection are higher than would be predicted by a straight line
from the later data. The later, straight-line phase is called the terminal phase

209
Q

How do you know if a parameter refers to the terminal phase of the ln(C) vs time graph in 2 compartment model

A

often given the subscript z

210
Q

When does graph of ln(C) vs t show that elimination is entirely dictated by first order kinetics

A

when distribution is complete (terminal phase)

211
Q

How do you work out the Ke and half life from the terminal phase of ln(C) vs t graph (2 compartment)

How do you find AUC in terminal phase

A

gradient = -Kz

t0.5=0.693/Kz

AUCz=C1/Kz
where C1 is the plasma concentration at the start of the terminal phase

212
Q

How do you find the total AUC for ln(C) vs t in 2 compartment model

A

divide AUC before terminal phase into trapezia and work out area of each:
1/2(h1+h2)b

𝐴𝑈𝐶𝑡𝑜𝑡𝑎𝑙 = 𝐴1 + 𝐴2 + ⋯ + 𝐴n + 𝐴𝑈𝐶z

213
Q

How do you find the volume of distribution of the terminal phase (2 compartment model)

A

Vz=CL/Kz

Vz refers to the terminal phase volume of distribution, i.e, once the drug has distributed between the central and peripheral compartments.

214
Q

How is single oral dose added to 1 or 2 compartment models

what is absorption affected by

A

dose is added to a separate G.I. compartment

Absorption from the G.I. compartment is proportional to the amount of dose remaining, so the rate of absorption decreases with time. The rate of absorption will also be affected by the formulation of the drug.

215
Q

In a single oral dose model, how can you show slower absorption

A

Sustained release preparations can be used for slower absorption and more sustained effect

216
Q

Is all of a drug given orally usually absorbed?

A

no
First pass metabolism is a major cause of loss of drug.
The proportion of drug that is absorbed is the factional bioavailability, F.

217
Q

In the 2 compartment model with a single oral dose, which compartment is the drug absorbed into

A

central

It is possible to model absorption into a central compartment with distribution into a peripheral compartment, but this is often not necessary unless absorption is much more rapid than distribution between central and peripheral compartments
(does not change approach to analysis)

218
Q

|Describe the graph of Plasma concentration against time for a drug taken as a single oral dose (5)

A

C shows dramatic initial increase as drug is absorbed

rate of elimination also increases as plasma concentration increases.

As drug is absorbed, the amount of drug remaining in the G.I. tract is reduced, so rate of absorption decreases.

The maximum concentration occurs when the absorption has decreased and elimination has decreased sufficiently for them to balance.

As absorption continues to decrease the kinetics become dominated by the rate of elimination.

During the terminal phase there is no further absorption and only elimination

219
Q

How is Kz found for a single oral dose of a drug

How is AUC found?

How is CL found?

A

slope of the chart of ln(C) against time = -Kz

AUCz= C1/Kz
total AUC = trapezia + AUCz

AUCtotal

220
Q

In the equation:
Oral Dose x F
——————– = CL
AUC total

what does F represent

How does F affect the dose

how is F determined

A

F= fractional bioavailability

reduces the dose reaching the central compartment

can be determined by comparing the pharmacokinetics of a single i.v. dose and a single oral dose

221
Q

What is the equation to find F

A

AUC oral dose(iv)
————–x————
AUC(iv) oral dose

222
Q

During i.v. infusion at a constant rate, what is the rate at which the drug accumulates in the body?

A

difference between the rate of infusion and the rate of elimination.

at steady state rate of infusion is equal to the rate of elimination

223
Q

During i.v. infusion at a constant rate, the rate at which drug accumulates in the body is the difference between the rate of infusion and the rate of elimination. Give this in equation format

A

dX/dt= rate in - rate out

=Rin - CL.C

224
Q

Give the equation for plasma concentration of drug at steady state

what should be noted

A

Rin
—– =Css
CL

Css is only determined by the rate of infusion and by the clearance. Vd has no effect.
If the Rin is doubled, the Css is also doubled.

225
Q

How do you calculate the time taken to reach a desired conc when you are constantly infusing the drug?

A

𝐶𝑡 = 𝐶𝑠𝑠(1 − 𝑒^−𝑘𝑒.𝑡)

thus

t= (-1/Ke).ln[1-Ct/Css]

226
Q

What is a loading dose

what is a draw back of this

A

To get to Css immediately, we could give a large, bolus i.v. injection in addition to slow infusion.
dose needs to be sufficient to give a plasma concentration of Css when it is distributed throughout the volume of distribution.

might not be the best approach as it could give very high peak concentrations in the brain and heart.

227
Q

What is the equation for loading dose

A

𝐿𝐷 (𝑖𝑛𝑓𝑢𝑠𝑖𝑜𝑛) = 𝐶𝑠𝑠 × 𝑉d

228
Q

What happens to a drug after it has stopped being infused constantly?

How does it behave

A

drug will be eliminated from the body by metabolism and/or excretion.

behaves in the same way as exponential decay after i.v. injection in the single compartment model, as distribution has already occurred during the infusion.

229
Q

Relate AUC, Css, and Ke in an equation

A

Css
—— = AUC
Ke

230
Q

How can you determine steady state volume distribution from CL

A

CL
—– = Vss
Ke

231
Q

How can you achieve Sustained, effective plasma concentrations

A

constant iv infusion

multiple dose regimen, either of i.v. bolus injections, or oral tablets.

232
Q

How does multiple dose regime reach effective sustained plasma conc of drug

A

dose is repeated with a dose interval, τ
If the second dose is given before all of the first dose has been eliminated then the drug will accumulate, achieving a higher peak concentration with the second dose and so on.

Assuming that drug elimination follows first order kinetics, average rate of elimination will increase as drug plasma concentration increases. Css is reached when the amount of drug eliminated in each dose interval is equal to the amount of the dose.

From this point, the C will vary between the Cmax ss and minimum concentration (Cmin, ss; immediately prior to the next dose).

233
Q

How does changing dose frequency and amount in multiple dose regime change the steady state

A

Half the dose, twice as often (same dose rate) will produce the same Cav, ss, but with less
variation about above and below this value.

234
Q

What is Cav ss

A

average concentration at steady state (Cav, ss) that is achieved by multiple dosing

v similar to Css from constant infusion

235
Q

True or false

At Css, the entire dose must be eliminated just before the next doser

A

true

average rate of absorption (Rav, in) must equal the average rate of elimination

236
Q

average rate of absorption (Rav, in) must equal the average rate of elimination. Express this as 2 equations

A

Rav,in=

tau

or

CL.Cav,ss

237
Q

Give a formula for Cav,ss in terms of AUC and tau

A

AUCss
———– = Cav,ss
tau

238
Q

Assuming first order kinetics, how does AUCss compare to AUC for an equivalent single iv dose? Why?

A

AUCss is the same as AUC for an equivalent single i.v. dose

because AUC = Dose/CL in both circumstances. If the elimination is still first order, the clearance will be the same

239
Q

Give an equation for Cmax,ss involving dose iv, Vd and an exponential

Cmaxss=…

A

dose iv 1
———-x —————
Vd 1-e^(-Ke.τ)

240
Q

How do you calculate loading dose to reach Css for multiple doses

A

𝐿𝐷 (𝑚𝑢𝑙𝑡𝑖𝑝𝑙𝑒 𝑑𝑜𝑠𝑒𝑠) = 𝐶𝑚𝑎𝑥,𝑠𝑠. 𝑉𝑠s

241
Q

What is the problem with using Vss in calculations

A

s if the drug undergoes extensive redistribution between compartments
The initial Vd will be smaller than Vss, making the initial plasma concentration much higher than predicted and risk of toxicity.

242
Q

How does achieving Css with multiple oral doses differ from multiple iv doses

A

absorption after an oral dose will be slower than direct i.v. injection.

Without knowing the rate constant for absorption, we cannot predict the Cmax ss and Cminss

However, if absorption is fairly rapid, we might use the i.v. as an approximation

243
Q

Describe behavior of elimination in zero order kinetics

A

elimination rate is constant and is not dependent on plasma concentration.

244
Q

How does plasma concentration of drug differ between first order and zero order kinetics

A

in zero order, If we infuse at a constant rate, plasma concentration will continue to increase.

Compare with first order kinetics, where constant infusion leads to a steady state plasma concentration, Css.

If we increase the rate of infusion then Css does not increase
proportionally, as in first order kinetics, but increases exponentially.

245
Q

Why must you be careful when increasing doses of phenytoin

when can it be dangerous at normal doses

A

Therapeutic concentrations of phenytoin are close to saturation for hydroxylation by CYP2C9. A small increase in dose rate can lead to a much greater plasma concentration and toxicity.

Polymorphisms in CYP2C9 that reduce its reaction rate, and inhibitors of CYP2C9
(e.g. valproic acid), can make this more likely at ‘normal’ dose rates

246
Q

Can zero order kinetics become first order kinetics

A

In zero-order kinetics, the rate of elimination is constant. A constant amount of drug is eliminated per unit time. The concentration declines at a constant rate until the concentration is sufficiently low for first order kinetics to be re-established

247
Q

Give an example of first order kinetics becoming zero order kinetics

A

ethanol

Metabolism of ethanol saturates at fairly low alcohol intake.
Above this, elimination follows zero order kinetics.

Chronic alcohol consumption can increase the rate of ethanol metabolism through induction of CYP2E1.

248
Q

What does general anaesthesia involve (4)

A

loss of consciousness
loss of reflexes,
muscle relaxation,
inability to feel pain (analgesia).

249
Q

Give 6 attempts at producing analgesia used through the ages

what were the earliest inhalation anaesthetics (2)

A
poppy extracts (opioids), 
henbane,
 acupuncture, 
carotid compression, 
boiled mandrake 
coca leaves

ether and nitrous oxide

250
Q

What did Queen Victoria use to reduce labour pains

A

chloroform

251
Q

What does the Meyer-Overton correlation suggest about the MOA of anaesthetics

what properties of GAs do not fit (3)

A

general anaesthetics act through a common and non-specific mechanism by accumulating in lipid bilayers and altering membrane function

maximum molecule size cut-off, discovery of some lipid soluble molecules that did not cause anaesthesia, and stereo-specificity of some anaesthetics such as isoflurane.

252
Q

What did Franks discover about the potency of anaesthetics

A

demonstrated that anaesthetic potency also correlated with their ability to
inhibit a lipid-free protein (luciferase)

253
Q

What conclusion have the observations of Franks, Meyer and Overton lead to about the binding os GAs

A

general anaesthetics bind to a hydrophobic pocket in one or more target proteins

254
Q

What is the Meyer-Overton correlation

A

Meyer (1899) and Overton (1901) independently demonstrated a strong correlation between anaesthetic potency and its solubility in olive oil (oil:water partition coefficient in experiments on tadpoles, or oil:gas partition coefficient for delivery by alveolar ventilation)

255
Q

Which receptors/ channels are GAs thought to act on

A

3 main ones:
GABA A
K2P family
NMDA receptors

additional protein targets:
glycine receptors
HCN channels
Nav channels

256
Q

Name 3 things that act on the Cl- current through GABA A receptors

A

many general anaesthetics
ethanol
barbiturates

257
Q

What does activation of GABA A receptors result in

A

hyperpolarises the post-synaptic membrane and reduces neuronal activity

258
Q

Describe the structure of GABA A receptors

How does this relate to the action of different anaesthetics

A

pentomeric with a variety of different combinations, usually with 2α, 2β and γ isoforms.

i.v. anaesthetics propofol and etomidate act on the β subunits, at related but distinct sites, whereas volatile anaesthetics appear to act on α and β subunits

259
Q

How are mutations in GABA A receptors related to GA potency

A

Point mutations in GABAA α and β subunits have been shown to reduce anaesthetic potency.
Notably, mutations in the α subunit reduce or abolish the effect of many volatile anaesthetics with no effect on propofol or etomidate, whereas mutations in the β subunit affect both volatile and i.v. anaesthetics (e.g. the β3 mutation N265M).

However, not all anaesthetics are affected by mutations in GABAA subunit.

260
Q

Which ion channel does isoflurane activate

A

Some volatile anaesthetics, such as isoflurane and nitrous oxide, activate members of the Two pore domain K+ channel (K2P) family, leading to hyperpolarisation and reduced neuronal activity

261
Q

Give a mutation study that involves halothane

A

mutation in TASK3 (M159A) abolished the effect of halothane and isoflurane, suggesting that this amino acid may be involved in binding of these anaesthetics.

262
Q

What normally activates NMDA receptors

Which GAs affect them? \What is a possible MOA here?

A

glutamate

Some general anaesthetics, particularly nitrous oxide and xenon, may also inhibit the excitatory NMDA receptors
competition with glycine, an essential co-agonist for NMDA receptor activation.

263
Q

Which area of the brain is responsible for all the clinical effects of GAs

A

There may be no one area of the brain that is target site for anaesthetics, responsible for all the clinical effects, although the thalamus seems to be an important site

264
Q

Why must anaesthesia

be carefully monitored and controlled

A

Increasing concentrations progressively affect many brain functions.
Higher concentrations can lead to respiratory failure and death

265
Q

Name 3 GAs that are administered iv

what are they good for

A

Thiopental (thiopentone),
propofol
etomidate

effects have rapid onset so used for rapid induction

266
Q

Where does thiopental act

A

it’s lipophilic so can cross BBB to induce anaesthesia in 10s of seconds

267
Q

Describe the pharmacokinetics of thiopental

A

binds to plasma proteins. It is metabolised in the liver. This metabolism is close to saturation, so can show zero order kinetics of metabolism if maintained by infusion or repeated injection.
However, consciousness is rapidly recovered after a single i.v. injection. This is because the initial fall in plasma concentration is caused by redistribution, not metabolism

268
Q

Describe the distribution of thiopental

A

Following injection, the plasma concentration is immediately high.
Thiopental rapidly equilibrates into high blood flow tissues, such as the brain, heart, kidney and liver.

The initial Vd is relatively low. Over the course of minutes, thiopental equilibrates into lower blood flow tissues, such as the skin and muscle. This change in drug distribution increases the Vd, decreasing the plasma concentration.

High blood flow tissues rapidly equilibrate with the decreasing plasma concentration. The consequence is that brain concentration falls, and consciousness is regained.

Equilibration into fat is even slower because it has very low blood flow. However, because thiopental is so lipophilic, it readily accumulates in fat.

269
Q

Why can thiopental give an anaesthetic hangover

A

slowly, thiopental is metabolised by the liver. As it is metabolised, thiopental slowly equilibrates back from the low blood flow tissues and the fat. This leads to a very slow decrease in plasma concentration. The plasma concentration is sub-anaesthetic but can be high enough to give side effects

270
Q

why does thiopental require >1 injection or constant infusion

what must you be aware of

A

consciousness is rapidly recovered

With each additional injection, the rapid fall in C by redistribution ends at a higher level and may be above the minimum conc for anaesthesia. Recovery is then dependent on metabolism and slow accumulation in fat.

The time to waking can become hours rather than minutes.

271
Q

What has replaced thiopental

why (3)

A

Propofol

has more rapid redistribution than
thiopental, and has more rapid metabolism. may also have an anti-emetic effect.

272
Q

How does etomidate compare to Propofol

A

Etomidate has a wider therapeutic window for anaesthesia over cardiovascular depression, but a higher rate of vomiting and nausea during recovery has been reported compared to propofol.

273
Q

How are volatile GAs administered

A

inhalation

274
Q

What is a key determinant of rate of induction of volatile GAs

A

They must cross the alveoli epithelium into the blood then cross into the brain.

The blood:gas partition co-efficient is key determinant of rate of induction because it affects how quickly the alveoli air comes into equilibrium with the inspired air.

275
Q

What does the blood: gas partition coefficient describe

A

the ratio of the concentration of the anaesthetic in blood to alveolar gas when the partial pressures are in equilibrium.

276
Q

How can you increase the blood: gas partition coefficient

A

by increasing solubility by increasing the binding to the lipids and proteins in blood that GAs can bind to (eg albumin, triglycerides, serum cholesterol and RBC membranes)

277
Q

Describe how the partial pressure of anaesthetic gas changes as it is inhaled and absorbed

A

The partial pressure of anaesthetic gas in inspired air (Pinsp) is high. During inhalation it is diluted by the residual air in the alveoli, leading to a lower partial pressure in the alveoli (Palv).

Anaesthetic gas diffuses into the blood in pulmonary capillaries, increasing the Pblood, bringing it rapidly close to equilibrium with Palv.

Blood circulates around the body, perfusing the tissues (particularly high blood flow tissues, including the brain. Anaesthetic gas diffuses from capillaries into tissues, increasing the partial pressure there (e.g. PCNS).

278
Q

What does induction of volatile anaesthetics require

A

equilibration between P(insp) and P(CNS)

this involves many equilibria
Pinsp↔Palv, Palv ↔Pblood, and Pblood ↔PCNS.

diffusion between compartments is rapid

279
Q

what does the rate of approach to equilibrium between Pinsp and Palv depend on

use 2 contrasting scenarios to demonstrate

A

blood:gas partition coefficient.

If the blood:gas partition coefficient is high, more gas in the alveoli crosses into the blood to approach equilibrium. The alveoli partial pressure stays relatively low and it takes longer for equilibrium between Pinsp and Palv to be achieved. Induction is relatively slow.

In contrast, if the blood:gas partition coefficient is low, less gas crosses to achieve equilibrium, so Palv rapidly increases. Equilibrium between inspired air and alveoli is rapidly achieved, and induction is rapid.

280
Q

How does cardiac output affect induction rate of volatile GAs

How does alveolar ventilation rate affect it?

A

slows induction rate, since more gas is removed during each breath.

Low alveolar ventilation rate also slows induction rate, since less anaesthetic is delivered to the alveoli in a given time

281
Q

Do volatile GAs accumulate in fat?

A

yes
This is slow since fat blood flow is very low. Complete redistribution equilibrium can take a long time to achieve. The extent of accumulation depends on the length of the procedure.

282
Q

What affects the rate of recovery from volatile GAs

A

The rate at which Palv equilibrates with Pisnp (now equal to 0 when the pump is turned off)

blood:gas partition coefficient

283
Q

How are volatile GAs removed from the body

A

delivered to the lungs by blood flow and is removed from the alveoli on each breath

Anaesthetic in the body (blood and tissues) is acting as a reservoir of gas to keep Palv > Pinsp

284
Q

How does e blood:gas partition coefficient affect recovery from volatile GAs

A

If the blood:gas partition coefficient is low (poorly soluble), most of the anaesthetic rapidly crosses into the alveoli and is removed (i.e. at
equilibrium between Pblood and Palv, the concentration of gas in the blood is low). The amount of anaesthetic in the body falls quickly and recovery of consciousness is rapid.

If blood:gas partition coefficient is high, Pblood and Palv rapidly equilibrate, but at this equilibrium the concentration of anaesthetic gas in the blood is high. This is a greater reservoir to maintain Palv > Pinsp for longer. Therefore, recovery is slow.