Pharmacology Flashcards

1
Q

define pharmacokinetics

A

what the body does to a drug (metabolism, absorption etc)

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

define pharmacodynamics

A

what a drug does to the body (i.e. drug-receptor interactions)

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

define a drug

A

a chemical substance of known structure which when administered to a living organism, has a biological effect

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

define a medicine

A

one or more drugs combined and administered with the intentions of achieving a therapeutic effect. not all drugs are medicines and not all medicines contain drugs. medicines have properties which treat disease or restore/correct physiological function by exerting a pharmacological, immunological or metabolic action or making a medical diagnosis

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

define therapeutics

A

the use of drugs to diagnose, prevent and treat illness

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

define formulations

A

how the active drug is combined with other substances to produce a medicine

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

define excipients

A

substances formulated alongside the drug such as bulking agents, preservatives or coatings

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

what are the three different names of a drug?

A

the chemical name (which describes the chemical structure)
the generic name (the class of drug to which a molecule belongs)
the proprietary/trade name (named by the manufacturer)

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

what would the ideal drug do?

A
  • have the desired pharmacological action
  • have acceptable or no side effects
  • reach its target in the right concentration at the right time
  • remain in sufficient concentration at the target for a sufficient time
  • be rapidly and completely removed from the body after action
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

define ligand

A

a molecule that binds to a receptor. it can be exogenous (from outside), endogenous (from inside). it could be a drug, hormone or neurotransmitter and could act on one or many different receptors. they can also be synthetic or natural.

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

define receptor

A

a molecular target for a drug

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

define agonist

A

a molecule which ‘activates’ a receptor

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

define antagonist

A

a molecule which blocks or reduces agonist mediated responses (either by blocking the receptor or by reducing the effects of an agonist)

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

define target

A

a molecule, usually a protein, that is accessed by a drug to produce a therapeutic effect

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

define affinity

A

how well a ligand binds to a receptor, this can be strong or weak. very strong affinity may lead to permanent binding

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

how do drugs bind to receptors?

A

through steric (physical shape) interaction whereby the ligand fits into the receptor like a lock and key

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

what factors effect drug-receptor binding?

A

steric properties = the physical shape of the ligand and the receptor must be complementary
physiochemical properties = local charges across the receptor binding site and local charges on the corresponding site of the ligand must be opposing to have the strongest affinity.
target tissue - this doesn’t actually effect binding but the same drug acting on different tissues will produce different effects

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

how can pharmacogenetics effect responses to therapeutic drugs?

A

genes code for the production of proteins
genetic variation therefore leads to a variation in the proteins produced
proteins are the primary binding site for therapeutic drugs
so variations in proteins may change how drugs bind and the effect they have on the body.

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

what are the common targets for drug action?

A

most are proteins, common target proteins include
receptors
ion channels
carrier proteins
enzymes

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

what are the two different types of antagonist?

A

allosteric antagonists = these bind somewhere other than the ligand binding site but in doing so change the shape or charge of the ligand binding site meaning that the intended ligand can no longer bind to the receptor
orthostatic antagonists = these bind to and block the original ligand binding site, meaning that the intended ligand cannot bind.

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

what are G protein linked membrane receptors?

A

these are membrane receptors which cause a intracellular messenger cascade when a ligand binds to them, producing various effects including:
- altered cellular excitability
- modulation of other ion channels
- down regulation if G protein linked receptors
- long lasting impact on regulation of genes within the cell

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

how do drugs act on ion channels?

A

ion channels allow the movement of charged particles across a membrane, maintaining resting membrane potential. they are physical pores within the membrane but are selective as to what ions they allow through. drugs can either be:
blockers = drug sits within the ion channel, physically blocking it
modulators = drug binds to somewhere on the ion channel, changing its characteristics.

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

how do drugs act on carrier proteins?

A

carrier proteins facilitate transport of larger molecules across a membrane. disruption to carrier proteins results in the changing of membrane potential. drugs can be:
inhibitors = stop transport in carrier proteins
false substrates = act as the intended substrate to enter or change the action of carrier proteins.

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

how do drugs act on enzymes?

A

drugs interact with enzymes in many different ways. some common mechanisms include:
enzyme inhibitors = normal reaction is inhibited or blocked by the drug as the drugs are substrate analogues (same physical shape as the intended substrate) so they fit into the enzymes binding site
false substrates = drug binds to the enzyme and is broken down into abnormal metabolites
pro-drugs = an active drug is produced after enzymatic breakdown of the initial drug.

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

is any drug completely specific to one receptor?

A

no! side effects from drugs arise because although drugs may have specific target molecules, they are likely to bind to other receptors as well, causing a whole load of other effects. this is more likely when the drug is in the body in high concentrations.

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

what four processes determine a drugs time and duration of onset in the body?

A

absorption = process by which drug reaches systemic circulation
distribution = process by which drug reaches target area(s)
metabolism = the breaking down of a drug in the body, usually in the liver
excretion = removal of the drug, or its metabolites, from the body

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

what factors affect absorption of a drug?

A
  • route of administration
  • permeation (getting into the tissues of the body - must be in solution to do this)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

what are the pros and cons of enternal (via gut) administration of drugs?

A

pros:
- low infection risk
- self-administration
cons:
- stomach is a harsh environment
- lots of the drug is lost to first pass metabolism

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

what are the pros and cons of injections?

A

pros:
- rapid bioavailability
- avoids first pass metabolism
cons:
- infection risks
- targeting risks

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

what are the different types of injection?

A

intravascular = directly into bloodstream
intramuscular = into skeletal muscle
subcutaneous
dermal
depot injection = for slow release formulation

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

what are the pros and cons of topical administration of drugs?

A

pros:
- local effects
- low systemic effects
- limited first pass metabolism risks
- low infection risk
cons
- long period of administration
- must be lipid soluble, small molecule and use a carrier molecule in order to permeate skin (this carrier molecule is usually an irritant to speed up absorption which isn’t great)

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

what is first pass metabolism?

A

metabolism of a drug that occurs in the intestine or liver before it reaches systemic circulation. it significantly reduces the bioavailability of the drug.

33
Q

define bioavailability

A

the proportion of a drug that reaches systemic circulation and is free to bind to its target. it is affected by route of administration and formulation of the drug

34
Q

what factors affect the distribution of a drug in the body?

A

protein binding
blood flow
membrane permeation/tissue solubility

35
Q

how does protein binding affect drug distribution?

A

drugs in the bloodstream are either free/unbound or they are bound to plasma proteins. this is a dynamic equilibrium and drugs can bind and unbind to plasma proteins. only unbound drugs can pass through capillary walls and bind to targets on cells etc. so only unbound drugs can produce a pharmacological effect. plasma protein binding decreases with age so drugs may have a greater effect in the elderly as there is more unbound drug in their bloodstream.

36
Q

how does blood flow affect drug distribution?

A

drugs are primarily distributed around the body in the circulatory system. areas with greater perfusion rates (more blood flow) will have greater access to the drug.

37
Q

how does membrane permeability affect drug distribution?

A

how permeable a tissue or cell is will affect how much of the drug can enter it. ficks law = three factors effect diffusion rate: area, distance and concentration gradient.
maximal diffusion will occur when area is high, distance is low and concentration gradient is high.

38
Q

what is the effect of metabolism on a drug?

A

metabolic action usually inactivates a drug. metabolic rate can determine the duration of drug action in this case, faster metabolism of drug = shorter duration of drug action.
however in some cases, metabolism activates a drug (pro-drugs) where the drug is not active until it is metabolised.

39
Q

what are the two phases of metabolism?

A

phase I = often produces toxic metabolites
phase II = converts toxins to soluble metabolites for excretion
(phase II can precede phase I or occur without phase I)

40
Q

explain the process of metabolism

A
  1. phase I metabolism occurs: this involves changing the chemical structure of a drug through processes like oxidation, hydroxylation, dealkynation or deamination. phase I may change the polarisation of a substance, increase it’s water solubility, reduce it’s pharmacological activity or activate pro-drugs. cytochrome p450 enzymes found in the liver commonly catalyse this reaction.
  2. phase II metabolism occurs: this involves the addition of endogenous substances via covalent bonding. this conjugation makes the substance water soluble and inactive.
41
Q

explain the process of renal excretion

A

unbound drugs in systemic circulation are filtered in the kidney via glomerular filtration. in the nephron their PH becomes the same as urine and they pass through to the bladder
(it’s a lot more complex than this but IDK how much we need to know)

42
Q

explain the process of biliary excretion

A

drug is removed from the body through bile and then forms faeces and is excreted from the body. the risks of this include the fact that gut bacteria can convert the drug back into its original active form, or the drug can be reabsorbed through the intestinal wall. either way this is bad as the drug returns to systemic circulation and can prolong drug action.

43
Q

what is drug half-life?

A

the length of time it takes for half of the original bioavailability to remain in the blood plasma. there is a limited number of half lives (4-5) before there is a non-therapeutic dose of the drug left in the plasma.

44
Q

define analgesic

A

a drug that relieves or reduces pain (or the pain experience)

45
Q

What is an NSAID?

A

a non-steroidal anti-inflammatory drug
widely used to reduce pain, reduce inflammatory and reduce high temperatures.

46
Q

what is the main mechanism of NSAID action?

A

the arachidonic acid cascade:
- phospholipase A2 produces arachidonic acid when tissue is damaged.
- COX enzymes convert arachidonic acid into intermediary molecules
- these intermediary molecules are then converted into prostanoids including prostoglandins which activate free nerve endings.
NSAIDs block the action of COX enzymes, meaning no prostoglandin is produced.

47
Q

what are the three types of COX enzyme?

A

COX 1 = found mostly in endoplasmic reticulum of most cells/tissues. involved in gastric protection, blood flow and platelet aggregation
COX 2 = found in the nuclear membranes of most cells, fibroblasts, macrophages and endothelial cells. involved in inflammation, pain and fever. key target for pain medications then, although there is some functional overlap between COX 1 and COX 2.
COX 3 = found in the CNS of animals, may or may not be active in humans, very poorly understood.

48
Q

what are some key examples of NSAIDs?
and which COX enzyme are they selective/have greater affinity for?

A
  • aspirin (COX1)
  • ketoprofen (COX1)
  • fenoprofen (equally selective for COX1 and COX2)
  • celecoxib (COX2)
49
Q

what are the four main therapeutic effects of NSAIDs?

A
  • anti-inflammatory
  • analgesic
  • antipyretic
  • platelet aggregation
50
Q

explain the anti-inflammatory effects of NSAIDs

A

this effect comes from the inhibition of COX 2 enzymes and subsequently their derived prostoglandins which themselves are powerful vasodilators but also release other vasodilating chemicals such as histamines or substance P

51
Q

explain the analgesic effects of NSAIDs

A

this effect comes from the inhibition of COX 2 enzymes and their derived prostoglandins which prevents peripheral sensitisation of nociceptive free nerve endings. NSAIDs also inhibit COX 2 in the dorsal horn of the spinal cord which means that central sensitisation of 2nd order neurons does not occur

52
Q

explain the antipyretic effects of NSAIDs

A

pyrogens stimulate PGE2 (prostoglandin E2) in the hypothalamus, which inhibits temperature sensitive neurons, affecting homeostasis and leading to a fever. NSAIDs block COX 2 which is responsible for forming PGE2, so none is created and neurons in the hypothalamus remain temperature sensitive.

53
Q

explain the platelet aggregation effect of NSAIDs

A

platelets produce thromboxane A2 which changes surface proteins on platelets, allowing them to bind together and cause blood clots. COX 1 inhibition reduces thromboxane production, reducing the likelihood of clotting. Aspirin covalently bonds to COX 1 meaning that this bonding is permanent and platelets never recover the ability to aggregate after aspirin use, meaning new platelets need to be developed in order for blood to clot again.

54
Q

what percentage of adverse drug reactions in the UK are to NSAIDs?

A

around 25%

55
Q

what are the areas that commonly see side effects to NSAIDs?

A
  • gastrointestinal
  • respiratory
  • renal
  • liver
56
Q

explain the gastrointestinal side effects of NSAIDs

A

prostoglandins promote the production of alkali mucus which lines the stomach and stops stomach acid from digesting the stomach itself. COX 1 selective NSAIDs like aspirin block the production of prostoglandin, leading to less alkali mucus, which in turn can lead to aspirin induced gastritis or ulceration.

57
Q

explain the respiratory side effects of NSAIDs

A

arachidonic acid also normally converts into leukotrienes which are bronchoconstrictors. when COX enzymes are blocked, they stop converting arachidonic acid into intermediary molecules, which leaves a large amount of unused arachidonic acid left. this means that more arachidonic acid is converted into leukotrienes. this may not be harmful in all people but can induce asthma like symptoms which may be additionally harmful to people with pre-existing respiratory conditions.

58
Q

explain the renal side effects of NSAIDs

A

prostoglandins promote vasodilation and glomerular filtration. when they are blocked, this leads to reduced renal filtration and therefore greater sodium retention in the kidney. this in turn reduces the efficacy of the kidney and ultimately leads to kidney damage

59
Q

explain the hepatic (relating to the liver) side effects of NSAIDs

A

this is a less common side effect of NSAIDs but has the tendency to be severe when it does occur. multiple mechanisms have been proposed:
- retention of bile (cholestasis): bile is held in the liver and builds up, damaging liver cells.
- mitochondrial damage: eventually leads to programmed cell death
- inhibition of prostoglandin E2: this prostoglandin is cytoprotective and protects against the triggering of programmed cell death. when this prostoglandin is blocked, programmed cell death in the liver is triggered.
- reactive metabolites cause autoimmune response: metabolites of NSAIDs are reactive in such a way that they may trigger an immune response, more extreme immune responses cause the immune system to attack liver cells.

60
Q

what is paracetamol?

A

not strictly an NSAID as it is non-opioid. it has analgesic and anti-pyretic effects but these are poorly understood. it is a poor peripheral inhibitor of COX 1 and 2 enzymes but may act centrally on COX 2 or even COX 3. does not affect platelet aggregation or gut mucosa.
usual dose is 1g 3/4 times daily, cannot exceed 4g per day.

61
Q

how is paracetamol metabolised in the body?

A

phase I metabolism produces NAPQI which is a toxic metabolite and so damages cells if left in the body in high concentrations. Phase II metabolism uses glutathione to convert NAPQI into a non-toxic glutathione conjugate which can then be excreted from the body.

62
Q

why is the maximum dose of paracetamol 4g daily?

A

because there is only enough glutathione in the body to convert 4g worth of NAPQI. any more than 4g of paracetamol and there will be left over, toxic, NAPQI in the body.

63
Q

what should you consider when suggesting what type of analgesic/NSAID someone should use

A
  • pre-existing medical conditions (if someone already has asthma do not suggest a COX 1 selective enzyme, similarly if they have stomach issues, COX 1 selective won’t help this)
  • risk of side effects (COX 2 selective NSAIDs have higher risk of cardiac side effects so not great in older people probs)
  • why they need the drug (if someone needs blood thinners, COX 1 selective probs helpful as it stops clotting)
64
Q

what is an opioid?

A

a compound resembling opium in its physiological effects
- they bind to specific opioid receptors
- they mimic the action of endogenous peptide neurotransmitters such as endorphins, enkephalins or dymorphins

65
Q

what are some examples of natural opioids?

A

morphine and codeine

66
Q

what are some examples of semi-synthetic opioids?

A

buprenorphine, hydrocodone, oxycodone, oxymorphone etc.

67
Q

what are some examples of synthetic opioids?

A

fentanyl, methadone, tramadol etc.

68
Q

what is the general mechanism of opioid action on opioid receptors?

A

opioid receptors in the PNS are Mu or delta fibres. many opioid receptors are G protein coupled receptors where binding of opioids to the receptor causes an intracellular cascade which results in changes in activity of voltage gated ion channels and overall nerve excitability. opioid receptors decrease activity of Ca2+ channels and increase activity in K+ channels. this decreases neuronal excitability and sensitivity of synapses. this happens to both 1st and 2nd order neurons.

69
Q

how do opioids affect peripheral sensitisation?

A

Mu receptors are located on free nerve endings, activation of these receptors on free nerve endings reduces 1st order nociceptive sensitivity and guards against peripheral sensitisation

70
Q

what are the widespread effects of opioids in the CNS?

A

spinal - inhibition of nociception
supraspinal effects vary in the brain stem and limbic system. includes modulation of descending control of nociceptive receptors.

71
Q

what are mild opioids?

A

these are opioids that include morphine analogues (such as codeine) and synthetic derivatives (such as tramadol) which work by weakly activating opioid receptors. the stronger the opioid the greater efficacy they have at the receptor.

72
Q

why is codeine a special opioid?

A

codeine is a pro-drug, it is metabolised in the liver by cytochrome P450 2D6. some of it is converted into morphine which is a much stronger opioid. people with fast metabolism for codeine (varies depending on genetics) will therefore have a higher dose of morphine in the liver which may lead to side effects such as respiration depression or GI motility issues.

73
Q

what are strong opioids?

A

these are opioids including morphine analogues (such as diamorphine which is literally heroine but can be prescribed as a pain killer) and synthetic derivatives such as fentanyl (far more potent than morphine but has a shorter half-life). these are strong agonists of opioid receptors.

74
Q

what are the potential short term side effects of opioids?

A
  • constipation (downregulation of nerve cells in the gut)
  • depression of cough reflex (downregulation of nerve cells in the brain stem)
  • respiratory depression (depression of neurons in the brain stem which are responsible for inspiration. overdoses literally stop you from being able to breathe in)
  • nausea/vomiting (either due to constipation or depression of sensory systems)
  • tolerance effects (adaptation of 2nd messenger cascade can lead to downregulation in sensitivity of nerves so more opioid is needed for same effect)
  • physical dependence
  • euphoria
75
Q

what are the potential long term side effects of opioid use?

A
  • immune suppression
  • decreased sex hormone production
  • opiate induced hyperalgesia (extended use of opiates causes glial cell activity to switch, causing them to produce more pro-inflammatory chemicals rather than less and leads to greater perception of pain)
76
Q

how can antidepressants be used as an analgesic?

A

neurons in the descending control pathway release serotonin and norepinephrine. antidepressants block the reuptake of these neurotransmitters and leave more in the synapses in the spine. therefore more is available to close the pain gate.

77
Q

how can anticonvulsants be used as an analgesic?

A

gabapentin and pregabalin bind to alpha-2-delta subunits of voltage gated Ca2+ channels, which results in reduced Ca2+ inflow, reducing excitability in the spinothalamic tract.

78
Q

how can local anaesthetics be used as an analgesic?

A

local anaesthetics such as lidocaine block voltage gated Na+ channels from the inside of the cell (it partitions into the cytoplasm of cells). meaning that no action potentials can be generated.