Pharmacology Flashcards

1
Q

What are the 3 principal efferent outputs from the CNS?

A
Autonomic NS (PNS and SNS)
Somatic NS
Neuroendocrine system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What are the principal target organs of the ANS?

A

Exocrine glands
Smooth muscle
Cardiac muscle

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

What are the principal target organs of the somatic NS?

A

Skeletal muscle

Including the diaphragm and respiratory muscle

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

What are the principal roles of the neuroendocrine system?

A
Growth 
Metabolism
Reproduction
Development
Salt and water balance
Host defence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are the principal roles of the ANS?

A

Metabolism
Host defence

PNS= Rest and digest
SNS= Fight or flight
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the sympathetic and parasympathetic effects on pupillary constriction?

A

Pupillary constriction= response to light

Sympathetic= need dilated (get info in)
Parasympathetic= constrict in bright room
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What cranial nerve is involved in parasympathetic control of pupillary constriction?

A

Oculomotor

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

What cranial nerve is involved in mediating cephalic and gastric phases of gastric secretion?

A

Vagus (PNS)

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

How does the PNS control the gastric secretion?

A

PNS drives gut including stomach

Vagus mediates:

  • Cephalic secretions
  • Gastric motility and secretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What branch of ANS is dominant at rest?

A

Parasympathetic

Resting HR is about 70bpm
BUT should be 100-110bpm if it was based on the midpoint of the intrinsic rates of vagal (50) and sympathetic (200)

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

What receptors influence the ANS control of the heart?

A

Barorecptors

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

How do baroreceptors affect heart rate speed?

A
Parasympathetic= slows heart
Sympathetic= speeds up heart
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Do the PNS and SNS always innervate different tissues?

A

No

PNS and SNS often innervate the same tissues and do different things

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

How does the SNS act on blood vessels?

A

2 different ways dependent on receptors

Dilation in skeletal muscles

Constriction in skin. mucous membranes and splanchic areas

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

What do all preganglionic fibres of the ANS release?

A

Acetylcholine

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

What are the neurotransmitters released by the postganglionic fibres in the PNS and SNS?

A
PNS= cholinergic (ACh) 
SNS= adrenergic (NA)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What are the lengths of the pre and postganglionic fibres in the PNS and SNS?

A
PNS= long pregang, short postgang
SNS= short pregang, long postgang
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Where do parasympathetic nerve fibres extend from on the spine?

A

Cranial/sacral

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

Where do sympathetic nerve fibres extend from on the spine?

A

Thoracic/lumbar

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

Outline the 3 possible ways neurotransmitters are released in the SNS

A

Short preganglionic fibre-> ACh
Long postganglionic fibre-> NA (acts on effector organ)

Short preganglionic fibre-> ACh (on adrenal medulla but acts like ganglion)
Long postganglionic fibre-> A and NA (via bloodstream, acts on effector organ)

Short preganglionic fibre-> ACh
Long postganglionic fibre-> ACh (acts on effector organ e.g. sweat gland)

DIAGRAM IN LECTURE

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

What is the difference in divergence between the PNS and SNS?

A

PNS= discrete/localised (little
divergence, 1:1 pre vs post)

SNS= coordinated
response (very divergent, up to 1:20 pre vs post)

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

What is the name of the NS of the gut?

A

Enteric

Works with SNS and PNS

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

What neurotransmitters are involved in the somatic nervous system?

A

ACh

Released by 1 long motor neurone from spinal cord to effector organ

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

What is the aim of the somatic nervous system?

A

Skeletal muscle contraction

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

What cholinoceptors and adrenoceptors are located in the ANS?

A

Muscarinic and nicotinic

These are membrane-bound receptors

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

What do muscarinic receptors do?

A

Mediate an affect (found at effector organ)

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

What do nicotinic receptors do?

A

Mediate connection between pre and post ganglionic fibre (found at start of post gang fibre)

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

What stimulates nicotinic and muscarinic receptors?

A
Nicotinic= nicotine/acetylcholine
Muscarinic= muscarine/acetylcholine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Where are nicotinic and muscarinic receptors found?

A

Nicotinic= at all autonomic ganglia
Muscarinic=At all effector organs innervated by
post ganglionic parasympathetic fibres

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

What signalling systems are employed by nicotinic and muscarinic receptors?

A

Nicotinic= type 1- ionotropic (speed important)

Muscarinic= type 2- G protein coupled (slower)
-> generation of 2nd messenger-> activation of cell signalling

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

If you blockade nicotinic AChRs in a person at rest, what would the effect be on the bowel?

A

Constipation

Rest and digest so PNS response
Blocking nicotinic-> blocking PNS (PNS should make gut work well)

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

If you blockade nicotinic AChRs in a person, what would the effect be on heart rate…

a) at rest
b) during exercise

A

a) Blockade at rest: PNS will be dominant at rest (low HR) but blocking means take out this dominant effect so increased HR
b) Blockade during exercise: SNS will be domininant in exercise (increased HR) but blocking means reduced HR

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

What are the subtypes of muscarinic cholinoceptors?

A

M1-M5

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

Where are the subtypes of muscarinic cholinoceptors found?

A

M1= Neural

M2= Cardiac

M3= Exocrine, smooth muscle

M4= Periphery: prejunctional nerve endings (inhibitory)

M5= Striatal dopamine release

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

What do M1, M2 and M3 do?

A

M1= Neural= forebrain – learning and memory (salivary glands, stomach, CNS)

M2= Cardiac= brain – inhibitory autoreceptors (heart)

M3= Exocrine and smooth muscle= hypothalamus – food intake (salivary glands, bronchial/visceral SM, sweat glands, eye)

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

What signalling systems are employed by M1, M2 and M3?

A
M1= Gq  STIM (IP3 DAG)
M2= Gi INHIB (cAMP)
M3= Gq STIM (IP3 DAG)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Where are adrenoceptors found?

A

All effector organs innervated by post ganglionic fibres

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

Which allows for more selective drugs to be used; adrenoceptors or muscarinic cholinoceptors?

A

Andrenoceptors
There are multiple subtypes
Very selective drugs available

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

What stimulates adrenoceptors?

A

NA/A

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

What signalling system is employed by adrenoceptors?

A

Type 2- GPCR

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

What are the subtypes of adrenoceptors?

A
alpha 1
alpha 2
beta 1
beta 2
(beta 3)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What neurotransmitters do nicotinic Rs, muscarinic Rs and adrenoceptors respond to?

A
Nicotinic= ACh
Muscarinic= ACh
Adrenoceptors= NA/A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What happens to heart rate and sweat production during exercise if muscarinic receptors are blocked?

A

Increased HR

Reduced sweat production

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

Summarise biosynthesis, release and metabolism of acetylcholine

A

SEE DIAGRAM

Acetyl CoA + Choline -> (choline acetyl transferase, CAT)-> ACh + CoA

ACh in vesicle-> released into synapse (because of AP Ca release)

ACh binds to R on effector cell

ACh-> (acetylcholinesterase)-> choline + acetate

Choline + acetate reabsorbed into presynaptic neurone

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

Summarise biosynthesis, release and metabolism of noradrenaline

A

SEE DIAGRAM

Tyrosine -> (tyrosine hydroxylase) -> DOPA-> (DOPA decarboxylase)-> Dopamine

Dopamine -> (dopamine B hydroxylase)-> NA

NA released into synapse (because of AP Ca release)

NA binds to adrenoceptor on effector cell

  • Uptake 1= presyn neuron -> NA-> (monamine oxidase A) -> metabolites
  • Uptake 2= postsyn (degradation by COMT)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What causes a reduction in synaptic NA concentrations?

A

Tyrosine hydroxylase

DOPA decarboxylase

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

What causes an increase in synaptic NA concentrations?

A

Uptake 1 transport protein (most increase)
Monoamine oxidase
Cathecol-O-methyl transferase

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

Define: pharmacokinetics

A

The study of how drugs are handled within the body

Including their absorption, distribution, metabolism and excretion (ADME)

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

Define: pharmacodynamics

A

The interactions of drugs with cells and their mechanism of action on the body

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

Define: drug

A

A chemical that affects physiological function in a specific way

OR

A chemical that interacts with a biological system to produce a physiological effect

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

Define: drug target sites

A

Protein complexes key to drug mechanisms of action

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

What are the 4 target sites of drugs?

A

Cell receptors
Ion channels
Transport systems
Enzymes

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

What do the 4 target sites of drugs have in common?

A

They are all proteins

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

What are drug receptors activated by?

A

(Endogenous) NT or hormone

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

What is the general structure of drug receptors?

A

Proteins with cell membranes (usually)

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

What are the 4 subtypes of drug receptors and how are they coupled?

A

Ionotropic (ligand-gated)= DIRECT
E.g. nAChR and GABAR

Metabotropic= G PROTEIN
E.g. mAChR and B1-adrenoceptors

Kinase-linked= DIRECT OR INDIRECT
E.g. Insulin receptors

IC steroid type= VIA DNA
E.g. Steroid/thyroid receptors

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

Location and effectors of ionotropic drug receptors

A
LOCATION= Membrane
EFFECTOR= Channel
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Location and effectors of metabotropic drug receptors

A
LOCATION= Membrane
EFFECTOR= Enzymes or channel
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Location and effectors of kinase-linked drug receptors

A
LOCATION= Membrane
EFFECTOR= Enzyme
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Location and effectors of IC steroid type drug receptors

A
LOCATION= Intracellular 
EFFECTOR= Gene transcription
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What affect is hijacking drug receptors useful pharmacologically?

A

Stimulate or stop a response

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

What kind of drug receptors are nAChRs and GABAAR?

A

Ionotropic (msec)

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

What kind of drug receptors are mAChRs and B1 adrenoceptors?

A

Metabotropic (sec)

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

What kind of drug receptors are insulin receptors?

A

Kinase-linked (mins)

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

What kind of drug receptors are steroid/thyroid receptors?

A

IC steroid type (hours)

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

How are ion channels target sites for drugs?

A

Selective pores

Allow transfer of ions down electrochem gradients

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

What are the 2 types of ion channels (drug target sites)?

A

Voltage-sensitive e.g. VSCC

Receptor-linked e.g nAChR

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

What drugs act on ion channels? (2 e.g.s)

A

Local anaesthetics

Calcium channel blockers (-dipine)

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

How do transport systems act as drug target sites?

A

Transport against conc gradients

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

What drugs act on transport systems? (2 e.g.s)

A

Tricyclic anti-depressants (TCAs)

Cardiac glycosides

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

What are examples of transport systems used as drug target sites?

A

Na+/K+-ATPase (Na out, K in)

NA uptake 1

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

How can drugs interact with enzymes (3 ways with examples)?

A

Enzyme inhibitors e.g. anticholinestarases (neostigmine)

False substrates-> ‘false’ productes e.g. methyldopa

Prodrugs e.g. chloral hydrate-> trichloroethanol

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

What are the unwanted effects of paracetamol?

A

Overload metabolism-> toxic metabolites-> irreversible damage to liver and kidney

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

How can drugs act non-specifically (i.e. not on target sites)? (2 e.g.s)

A

Physiochemical properties e.g. antacids and osmotic purgatives

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

Define: affinity

A

Strength (avidity) of drug binding to R

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

Define: efficacy (intrinsic activity)

A

Ability of the drug to induce a response in the R post-binding

I.e. through conformational change in the R

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

Define: potency

A

Powerfulness of a drug

Depends on affinity and efficacy

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

Define: full agonist

A

Agonist which has the ability to induce a max response in tissue post-binding

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

Define: partial agonist

A

Agonist which can only produce a partial response in tissue

AND

In conjunction with a full agonist may act with antagonism activity

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

Define: selectivity

A

The preference of a drug for a receptor

NOT THE SAME AS SPECIFICITY)

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

Define: structure-activity relationship

A

The activity of the drug is closely related to the structure of the drug

Small changes in the structure may produce large effects on its action

Like LOCK AND KEY theory

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

What does the receptor reserve refer to?

A

The fact that in many tissues, not all Rs need to be occupied in order to achieve the maximal tissue response

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

What does the receptor reserve cause in physiological tissue?

A

Increased sensitivity and speed of response

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

GRAPH: In a log dose response curve, what is the relationship between full agonist and partial agonists?

A

Partial= lower maximum

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

GRAPH: In a log dose response curve, what is the relationship between full agonist and full agonist with lower affinity?

A

Need higher concentration for full agonist with lower affinity (so shift curve to R)

Same maximums

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

Do antagonists have affinity or efficacy?

A

Affinity but no efficacy

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

What are the 2 types of receptor antagonist (i.e. receptor blockade)?

A

Competitive

Irreversible

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

How does competitive antagonism work? What is the affect on DR curve?

A

Same site as agonist
Surmountable
Shifts DR curve to right

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

2 examples of competitive antagonists

A

Atropine

Propranolol

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

How does irreversible antagonism work?

A

Binds tightly OR at different site

Insurmountable

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

1 example of an irreversible antagonist

A

Hexamethonium

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

Which of the following statements is most accurate?

A: A partial agonist will always have a higher efficacy than a full agonist
B: Agonists have higher affinities than antagonists
C: Full agonists that are selective for a given receptor will have the same efficacy
D: Antagonists possess better efficacy than their respective agonists
E: Competitive antagonists will preferentially occupy the relevant receptor in the presence of agonist

A

C: Full agonists that are selective for a given receptor will have the same efficacy

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

A drug acting as an inhibitor at a particular drug target site prevents the removal of neurotransmitter from the synapse. Which type of drug target is this drug acting on?

A: Receptor
B: Voltage-sensitive ion channel
C: Receptor-linked ion channel
D: Transport protein
E: Non-proteinaceous target
A

D: Transport protein

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

What are the 4 types of drug antagonism?

A

Receptor blockade
Physiological antagonism
Chemical antagonism
Pharmacokinetic antagonism

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

What is physiological antagonism? (1 example)

A

Different receptors have opposite effects in the same tissue

E.g. NA and histamine on blood pressure

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

What is chemical antagonism? (1 example)

A

Interaction in solution

E.g. Dimercaprol-> heavy metal complexes (chelating agent)

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

What is pharmacokinetic antagonism? (1 example)

A

Antagonist leads to decreased conc of active drug at site of action

Reduced absorption-> increased metabolism and increased excretion

E.g. barbiturates

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

What is drug tolerance?

A

Gradual decrease in responsiveness to drug with repeated administration (days/weeks)

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

What are the 5 main factors underlying drug tolerance?

A
Pharmacokinetic factors
Loss of receptors
Change in receptors
Exhaustion of mediators
Physiological adaption
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

How do pharmacokinetic factors lead to drug tolerance? (2 examples)

A

Increased rate of metabolism

Barbiturates
Alcohol

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

What is an example of exhaustion of mediator stores leading to drug tolerance?

A

Amphetamine

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

How does loss of receptors lead to drug tolerance?

A

By membrane endocytosis
Receptor down-regulation

Beta-adrenoceptors

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

How do change in receptors lead to drug tolerance?

A

Receptor desensitization-> conformational change

E.g. nAChR at NMJ

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

How do physiological adaption lead to drug tolerance?

A

Homeostatic responses

Tolerance to drug side effects

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

What determines the distinction between the 4 receptor families?

A

Type 1-4 (ionotropic, metabotropic, kinase-linked, IC steroid)

Based on molecular structure and signal transduction systems

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

A 4 year old girl is bitten by a Tiger snake whilst holidaying in Australia. An anti-venom (harvested antibodies) is administered. Which form of antagonism is utilised by the anti-venom?

A: Competitive receptor blockade
B: Physiological antagonism
C: Chemical antagonism
D: Pharmacokinetic antagonism
E: Irreversible receptor blockade
A

C: Chemical antagonism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q
Tolerance to the euphoric effects of drugs of abuse (e.g. heroin & cocaine) can occur after repeated use. Which form of tolerance would not involve any change in the cells that mediate the euphoric effects?
A: Receptor desensitisation
B: Receptor down-regulation
C: Exhaustion of mediator stores
D: Receptor up-regulation
E: Increased metabolic degradation
A

E: Increased metabolic degradation

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

How does a drug achieve its effect?

A

ADME

Administration

Absorption
Distribution (to site of action)
Metabolism
Excretion

Removal

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

What are the most common forms of administration for drugs?

A

Ingestion
Inhalations
Intravenous
Injections (dermal, intramuscular, subcutaneous, intraperitoneal)

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

What are the limitations of drug administration?

A

Ingestion (easy)
Inhalations (very easy but lots exhaled particularly if volatile drugs)
Intravenous (good straight into blood and lymph)
Injections (dermal, intramuscular, subcutaneous, intraperitoneal)= require skill

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

Primary site of drug metabolism

A

Liver

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

What is the difference between systemic and local drugs?

A

Systemic affect entire organism

Local restricted to one area of organism e.g. topical

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

Why is IV the most effective way to get drugs into systemic circulation?

A

Rapid absorption

Directly into blood and lymph

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

How do drug molecules move around the body (2 ways)?

A

Bulk flow transfer i.e. bolus in bloodstream

Diffusional transfer i.e. molecule by moluecule over short distances

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Why are the characteristics of drugs important to allow their transport within the body?

A

Have to cross aqueous and lipid environment

Compartments= aqueous e.g. blood, lymph, ECF, ICF

Barriers= lipid e.g. cell membranes

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

How do drugs cross barriers to allow for absorption?

A

MOST IMPORTANT
Diffusion through lipid
Carrier molecules

ALSO
Diffusing through aqueous pores in the lipid
Pinocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

How are non-polar substances absorbed?

A

Freely dissolve in non-polar solvents

Penetrate lipid membranes freely

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What does the ratio of ionised and non-ionised drugs depend on?

A

Most are weak acids or weak bases
Drugs exist in ionised (polar) and non-ionised (non-polar forms)

Ratio depends on pH

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

What are the difference types of aspirin used for and why?

A

Soluble= relief for headache (rapid), effectively absorbed in stomach because of stomach’s pH

Enteric coated= arthritis, slower releasing, slowly absorbed in intestine

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

How does aspirin act in the stomach?

A

Aspirin is a weak acid so in stomach most aspirin is uncharged
Can diffuse across membranes into cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How does aspirin act in the intestine?

A

Aspirin release is slow because aspirin mainly ionised (charged-> slower) in the neutral ph (7.4) of the intestine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Define: ion trapping

A

Build up of a high conc of a chemical across a cell membrane due to the pKa value of the chemical and difference of pH across the cell membrane

Ion is trapped so slow release in dynamic equilibrium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What factors influence drug distribution?

A

Regional blood flow
EC binding (plasma-protein binding)
Capillary permeability
Localisation in tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

How does regional blood flow affect drug distribution?

A

High metabolically active tissues have denser capillary networks
Blood perfusion changes rapidly in stress

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

How does extracellular (plasma-protein) binding affect drug distribution?

A

If drug is heavily bound to plasma-protein then will persist in bloodstream for a long time
May need to give more drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

How does capillary permeability affect drug distribution?

A

Ionised drugs can go through pore
Unionised can diffuse through cell wall unless bound

Capillary can be continuous, fenestrated or discontinuous

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What are the 2 main routes of drug excretion?

A

Kidney (most drug elimination)
Liver (some are concentrated in the bile, usually large molecular weight conjugates)

NB. entero-hepatic circulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

How is the kidney involved in drug excretion?

A

Glomerulus= drug-protein complexes not filtered

Proximal tubule= active secretion of acids and bases

Proximal and distal tubules= lipid soluble drugs reabsorbed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

Why is iv sodium bicarbonate given with aspirin and what will this do?

A
Na bicarb increases urine pH
Increased urine pH ionizes the aspirin
Ionized aspirin= less lipid soluble 
-> less reabsorbed from the tubule
-> increased rate of excretion 

Increase pH of urine in clinic to remove drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

How is the liver involved in drug excretion?

A

Bile doesn’t have a size limit that it can deal with (unlike kidney)

Large molecular weight drugs excreted through bile because kidney can’t handle it

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

How can excretion happen outside the kidney and liver?

A

Lungs, skin, GI secretions, saliva, sweat, milk, genital secretions

Insignificant amounts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

Why can entero-hepatic cycling be problematic for drug design?

A

Drug/metabolite excreted into gut (via bile) then reabsorbed, take in to liver, excreted again

Leads to drug persistence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What pharmacokinetic factors are important in predicting the time course of drug action?

A

Bioavailability (linked to absorption)
Apparent volume of distribution (linked to distribution)
Biological half life (linked to metabolism)
Clearance (linked to excretion)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

Bioavailability

A

Linked to absorption

Proportion of the administered drug that is available within the body to exert its pharmacological effect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

Apparent volume of distribution

A

Linked to distribution

The volume in which a drug appears to be distributed- an indicator of the pattern of distribution (how much has been distributed and to where)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

Biological half-life

A

Linked to metabolism/excretion

Time taken for the concentration of drug (in blood/plasma) to fall to half its original value

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

Clearance

A

Linked to excretion

Blood (plasma) clearance is the volume of blood (plasma) cleared of a drug (i.e. from which the drug is completely removed) in a unit time

(Related to volume of distribution and rate at which drug is eliminated. If clearance involves several processes, then total clearance is the sum of these processes)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Which of the following drugs would be least likely to penetrate lipid membranes?

Ionised drug
Non-ionised drug
Protein bound drug (still correct but not best)
Lipophilic drug
Hydrophilic drug
A

Ionised drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What is first-order kinetics?

A

SEE GRAPH

First-order kinetics describes the rate of elimination of a drug where the amount of a drug decreases at a rate that is proportional to the concentration of the drug remaining in the body

Dependent on the conc of drug at any given time

Applies to most clinical drugs

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

What is zero-order kinetics?

A

Half life doesn’t really apply to zero order kinetics

There is a constant amount of drug eliminated per unit time

Implies that there is a saturable metabolic process (applies to very few drugs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What is the difference between first and zero order kinetics?

A

First order= most common= rate of elimination is proportional to the plasma drug conc

Zero order= constant amount of drug is eliminated per unit time

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

What is an important use of ethanol phenytoin and why?

A

Police use to check drunk drivers

Rate of elimination is constant

Can use to calculate how much alcohol someone has drunk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Is kinetics (1st order and 0 order) drawn on a log or linear graph? What would happen if it were the other?

A

Drug-conc axis is linear

If it were log…
1st order would be straight
0 order would be curved

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What chemical property does a xenobiotic usually have?

A

Lipophilic foreign compound

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

What does metabolism do to allow lipophilic xenobiotics to be excreted?

A

Metabolism converts lipophilic chemicals to polar derivatives

By reducing or eliminating pharmacological/toxicological activity

Polar derivatives can be readily excreted

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

What is ‘first pass’ metabolism?

A

First time the drug goes through system before entering circulation (normally first metabolism is in the liver but could be skin, gut, kidneys, brain)

‘Clearing the drug’

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

When is ‘first pass’ metabolism not a problem?

A

When drugs don’t need to get into circulation

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

What are the 3 types/stages of metabolic change that drugs undergo?

A

Phase I
Phase II
Excretion

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

Phase I of metabolic change of drugs

A

Oxidation (creates new functional groups)
Reduction (creates new functional groups)
Hydrolysis (unmasks new functional groups)

  • functional group serves as point of attachment for phase II reactions

Often generate a biologically inactive product
Have little effect on drug polarity
Sometimes produce toxic metabolites

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

Phase II of metabolic change of drugs

A
Glucuronidation
Acetylation
Amino acid conjugation
Sulphation
Methylation
Glutathione conjugation

Are conjugation reactions which utilise -OH, -NH2, -SH and -COOH.

Involve a high energy intermediate e.g. UDPGA or PAPS

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

What do Phase I reactions usually do in drug metabolism?

A

Often inactive chemicals but can activate (e.g. prodrug)

After phase I, there is little change in polarity of the drug

Phase I prepares a drug for Phase II metabolism by introducing a functional group (handle) such as –OH, -NH2, -SH or –COOH.

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

Where are cytochrome P450?

A

Family of enzymes embedded in SER

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

How many cytochrome P450 enzymes are there?

A

57

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What do all cytochrome p450 enzymes have in common?

A

All have a haem
Can all metabolise drugs
Predominantly found in liver

NB. Multiple isozymess

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

Why does smoking affect metabolism of some drugs?

A

Smoking-> induces enzymes (p450)-> metabolism of drug changes

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

What effects do drugs have the CYP450 system?

A

Inhibit or induce the system

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

What does cytochrome P450 often mediate?

A

Oxidation

SEE DIAGRAMS OF OXIDATION BY CYP

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

True or false: Metabolism of prodrugs activates their pharmacological activity

A

TRUE

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

True or false: Xenobiotic metabolism only occurs in the liver

A

FALSE

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

True or false: Hydrolysis is a Phase 1 reaction

A

TRUE

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

True or false: Cytochrome P450 uses NADH+ as cofactor

A

TRUE but prefers NAPH

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

True or false: Cytochrome P450 contains Cu2+ at its active site

A

FALSE

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

What effect do Phase I reactions have on drug polarity?

A

Little effect

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

What can be produced by Phase I reactions?

A

Toxic metabolites

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q
What enzymes are used for the following processes:
Glucuronidation
Acetylation
Amino acid conjugation
Sulphation
Methylation
Glutathione conjugation
A

Glucuronidation= glucuronyl transferase

Acetylation= acetyl transferase

Amino acid conjugation= acyl transferase

Sulphation= sulphotransferase

Methylation= methyl transferase

Glutathione conjugation= glutathione-S-transferase

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

What are 3 main features of Phase II reactions?

A

Conjugate is almost always pharmacologically inactive

Less lipid soluble

Easier to excrete

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

How does glucuronidation in Phase II occur? (incl. formula)

A

ROH + very reactive UDPGA -> RO-D-glucuronide

Generate high energy phosphate compound
Glucuronic acid part transferred to an electron rich atom (N, O or S)
This is very important
UDP-glucuronate often excreted in bile

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

How does acetylation in Phase II occur? (incl. formula)

A

RNH2 + high energy CH3COSCoA -> RNHCOCH3 + CoASH

Acetyl CoA acts as donor compound

Donates acetyl group

Acetyl group transferred to an electron rich atom (N, O or S)

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

How does methylation in Phase II occur? (incl. formula)

A

RZH + S-adenosyl -> RZ-CH3 + S-adenosylhomocysteine

S-adenosyl methionine acts as donor compound

Methyl group transferred to an electronrich atom (N, O or S)

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

How does sulphation in Phase II occur? (incl. formula)

A

ROH + PAPS -> ROSO3- + PAP

Energy rich donor required

Paracetamol is very lipophilic so need to make it possible to excrete in urine

Sulfotransferases catalyse transfer of sulphate to substrates

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

How does conjugation with glutathione in Phase II occur? (incl. formula)

A

R-X + GSH -> R-SG + XH

X can be any leaving group

Glutathione is a protective factor, used for the removal of potentially toxic compounds
(Glycine + glutamine + cysteine tripeptide)

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

True or false: Metabolism of lipophilic chemicals facilitates their excretion

A

TRUE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

True or false: Metabolism of drugs prior to entering the systemic circulation is known as “first pass metabolism.”

A

TRUE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

True or false: Phase 2 metabolism includes reduction and acetylation

A

FALSE

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

True or false: Phase 2 metabolism generally increases the polarity of drugs

A

TRUE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

True or false: Conjugation of drugs with glutathione is the most common Phase 2 route of metabolism

A

FALSE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What phase of drug metabolism uses a high energy intermediate e.g. UDPGA or PAPS?

A

Phase II

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What phase of drug metabolism are conjugation reactions which use -OH, -NH2, -SH and -COOH?

A

Phase II

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What phase of drug metabolism prepare a drug by introducing a functional group (handle) such as –OH, -NH2, -SH or –COOH?

A

Phase I

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

Why is drug metabolism important?

A

Biological half-life of the chemical is decreased
Duration of exposure is reduced
Accumulation of the compound in the body is avoided
Potency/duration of the biological activity of the chemical can be altered
Pharmacology/ toxicology of the drug can be governed by its metabolism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What are muscarinic effects?

A

Correspond to those of parasympathetic stimulation
Those replicated by muscarine
Can be abolished by low doses of antagonist atropine

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

What are nicotinic effects?

A

After atropine bloackage of muscarinic actions larger doses of acetylcholine can induce effects similar to those caused by nicotine
Nicotinic responses are smaller (need higher doses of ACh)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What do nicotinic receptors look like?

A
Ligand gated ion channels
5 subunits (α β γ δ ε)
Subunit combo depends on properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

Subunits are found in:
Muscle type nicotinic Rs
Ganglion (neuronal) type nicotinic Rs

A

Muscle type nicotinic Rs= 2α β δ ε

Ganglion (neuronal) type nicotinic Rs= 2α 3β

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What are the muscarinic cholinergic target systems? (8)

A
Eye
Salivary glands 
Lung
Sweat glands 
Heart
Gut 
Bladder
Vasculature
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What are the muscarinic effects on the eye?

A

Contraction of the ciliary muscle (lens bulges) -> accommodation for near vision

Contraction of the sphincter pupillae (circular muscle of the iris)= constricts pupil (miosis) and improves drainage of intraocular fluid

Lacrimation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What causes Glaucoma?

A

Poor drainage of aqueous humour-> increased intraocular pressure

NORMALLY
Aqueous humour drains into anterior and posterior chamber to bathe lens (nutrients and oxygen) and to cornea

Drained through canal of Schlemm into vascular system

GLAUCOMA
Usually occurs when the fluid in the eye cannot drain properly-> increased pressure inside the eye and puts pressure on the optic nerve

The drainage angle is narrowed but production of aqueous humour doesn’t change

Muscarinic drugs affect aqueous humour in eye

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What are the muscarinic effects on the heart?

A

M2 AChR in atria and nodes (inhibitory action)
Decreased cAMP

1) Decreased Ca2+ entry-> decreased cardiac output
2) Increased K+ efflux-> decreased heart rate

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

What are the muscarinic effects on the vasculature?

A

Most blood vessels do not have parasympathetic innervation

ACh acts on vascular endothelial cells to stimulate NO release (via M3 AChR, stimulatory)

NO induces vascular smooth muscle relaxation-> decreased TPR

Important clinically

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

What are the muscarinic effects on the CV system? (4)

A

Decreased heart rate (bradycardia)

Decreased cardiac output (due to decreased atrial contraction)

Vasodilatation (stimulation of NO production)

Sharp drop in blood pressure (due to all the above factors)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What are the muscarinic effects on the non-vascular smooth muscle (lung, gut, bladder)?

A

Smooth muscle that does have parasympathetic innervation responds in the opposite way to vascular muscle (IT CONTRACTS)

Lung= bronchoconstriction

Gut= increased peristalsis (motility)

Bladder= increased bladder emptying (increased urination in response to bladder filling)

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

What are the muscarinic effects on exocrine glands?

A

Salivation
Increased bronchial secretions
Increased GI secretions (including HCl production)
Increased sweating (SNS-mediated)

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

What are the overall muscarinic effects? (7)

A
Decreased HR
Decreased BP
Increased sweating
Difficulty breathing
Bladder contraction
GI pain
Increased salivation and tears
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What are the typical, DIRECTLY acting cholinomimetic agonists at muscarinic receptors?

A
Choline esters (bethanechol= M3 selective)
Alkaloids (pilocarpine= non-selective)

Both have chem structure similar to acetylcholine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

How does pilocarpine work? What is it used for? Side effects?

A

Non-selective muscarinic agonist (half life 3-4h, lipid soluble)

Local treatment for glaucoma

SEs= blurred vision, sweating, GI disturbance/pain, hypotension, respiratory distress

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

How does bethanechol work? What is it used for? Side effects?

*NB cevimeline= newer version

A
M3 AChR selective agonist (half life 3-4h) 
Resistant to degradation (slower than ACh)
Orally administered (limited access to brain)

Used to assist bladder emptying and enhance gastric motility

SEs= sweating, impaired vision, nausea, bradycardia, hypotension, respiratory difficulty

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

What are the typical, INDIRECTLY acting cholinomimetic agonists at muscarinic receptors?

A

Increase effect of normal parasympathetic nerve stimulation

Reversible anticholinesterases: physostigmine, neostigmine, donepezil (‘Aricept’)

Irreversible anticholinesterases: ecothiopate, dyflos, sarin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

What do cholinesterase enzymes do?

A

Metabolise acetylcholine to choline and acetate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

How many types of cholinesterases are there and what are they?

A

Two types which differ in distribution, substrate specificity and function:

Acetylcholinesterase (true or specific cholinesterase)

Butyrylcholinesterase (pseudocholinesterase)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

Where do clinically relevant cholinomimetics act?

A

Muscarinic Rs

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

Where is acetylcholinesterase found and how does it work?

A

Found in all cholinergic synapses (peripheral and central)

Very rapid action (hydrolysis; >10 000 reactions per sec)

Highly selective for acetylcholine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

Where is butyrylcholinesterase found and how does it work?

A

Found in plasma and most tissues but not cholinergic synapses

Broad substrate specificity - hydrolyses other esters e.g. suxamethonium

Is principal reason for low plasma acetylcholine

Shows genetic variation

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

What effects do cholinesterase inhibitors have?
Low dose
Moderate dose
High dose

A

Low dose
= enhanced muscarinic activity

Moderate dose
= further enhancement of muscarinic activity
= increased transmission at all autonomic ganglia (nAChRs)

High dose
= depolarising block at autonomic ganglia and NMJ

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

How do reversible anticholinesterase drugs (e.g. physostigmine and neostigmine) work?

A

Compete with ACh for active site on cholinesterase enzyme

Donate a carbamyl group to the enzyme, blocking the active site and preventing acetylcholine from binding

Carbamyl group removed by slow hydrolysis (mins rather than msecs)

Increase duration of ACh activity in the synapse

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

What are physostigmine and neostigmine?

A

Reversive anticholinesterase drugs

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

Where does physostigmine work and what is it used for?

A

Primarily acts at the postganglionic parasympathetic synapse (half life= 30mins)

Used in the treatment of glaucoma, aiding intraocular fluid drainage

Also used to treat atropine poisoning, particularly in children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

How do irreversible anticholinesterase drugs (e.g. organosphosphate compounds- ecothiopate, dyflos, parathion and sarin) work?

A

Rapidly react with the enzyme active site, leaving a large blocking group

This is stable and resistant to hydrolysis - recovery may require the production of new enzymes (days/weeks)

Only ecothiopate in clinical use, but the others are commonly used as insecticides (and nerve gas)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What kind of drugs are ecothiopate and sarin?

A

Irreversible anticholinesterase drugs

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

Where do ecothiopate work and what is it used for? Side effects?

A

Potent inhibitor of acetylcholinesterase

Slow reactivation of the enzyme by hydrolysis takes several days

Used as eye drops in treatment of glaucoma, acting to increase intraocular fluid drainage with a prolonged duration of action

Systemic SEs= sweating,
blurred vision, GI pain, bradycardia, hypotension, respiratory difficulty

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

How do anti-cholinesterase drugs affect the CNS?
Low doses
High doses

A

Non-polar anticholinesterases (e.g. physostigmine; nerve agents) can cross BBB

Low doses
= excitation with possibility of convulsions

High doses
= unconsciousness, respiratory depression, death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

Why are donepezil and tacrine used to treat Alzheimer’s disease?

A

ACh is important in learning and memory

Potentiate central cholinergic transmission-> relieves AD symptoms BUT does not affect degeneration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

What happens in organophosphate poisoning and how is it treated?

A

Organophosphate poisoning e.g. from insecticides or sarin
-> severe toxicity (increases muscarinic activity, CNS excitation, depolarising NM block)

Treatment= IV atropine, articificial respiration, IV pralidoxime

  • pralidoxime works at pre and post ganglionic AChRs
  • atropine works at effector organ
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

What are the 2 main classes of cholinomimetics?

A

Direct (agonists)

Indirect (inhibitors of cholinesterase enzymes)

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

What can high doses of cholinomimetics do?

A

High doses of cholinomimetics activate the parasympathetic NS

BUT ALSO
Can activate all autonomic ganglia and ultimately cause depolarising blockade of nAChRs

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

Anticholinesterase drugs have the ability to increase activity at which synapses within the autonomic nervous system?

A: All autonomic synapses
B: Pre- and post-ganglionic parasympathetic synapses
C: Pre- and post-ganglionic sympathetic synapses
D: Post-ganglionic parasympathetic synapses only
E: Pre-ganglionic sympathetic synapses only

A

B: Pre- and post-ganglionic parasympathetic synapses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

Anticholinesterase drugs can be used to treat which of the following conditions?

A: Asthma
B: Glaucoma
C: Hypotension
D: Motion Sickness
E: Peptic Ulcer Disease
A

B: Glaucoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

Do agonists and antagonists possess affinity and efficacy?

A

Agonists and antagonists possess affinity

Agonists possess efficacy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q
Which of the following drugs has efficacy for the muscarinic acetylcholine receptor?
Acetylcholine
Atropine
Acetyl-cholinesterase
Adrenaline
Acetate
A

Acetylcholine

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

What is ganglion blocking drugs another name for?

A

Nicotinic receptor antagonists

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

How do ganglion blocking drugs/ nicotinic receptor antagonists?

A

Prevent ion from getting through by binding to R and blocking it but also get into ion channel and block it themselves

Nicotinic so not at effector organ (refer to diagram)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

Give 2 examples of nicotinic receptor antagonists?

A

Hexamethonium

Trimetaphan

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

When are nicotinic receptor antagonists most effective?

A

When channel is open

More ACh present, the more active the channel so the more effective the antagonist

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

Why is the nicotinic receptor antagonist block described as ‘incomplete’?

A

Not a total blockade just slows the process down

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

Do nicotinic receptor antagonists have affinity?

A

Technically but affinity is irrelevant if blockading

Physical blockade doesn’t require receptor binding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

What are the parasympathetic and/or sympathetic effects on the eye?

A
SYMP= dilatation (pupil)
PARA= constriction (pupil), contraction (ciliary muscle)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What are the parasympathetic and/or sympathetic effects on the trachea and broncheoles?

A
SYMP= dilates (Ad)
PARA= constriction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

What are the parasympathetic and/or sympathetic effects on the liver?

A

SYMP= glycogenolysis, gluconeogenesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

What are the parasympathetic and/or sympathetic effects on the adipose tissue?

A

SYMP= lipolysis

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

What are the parasympathetic and/or sympathetic effects on the kidney?

A

SYMP= increased renin secretion

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

What are the parasympathetic and/or sympathetic effects on the ureters and bladder?

A
SYMP= relaxes detrusor, constriction of trigone and sphincter
PARA= contraction of detrusor, relaxation of trigone and sphincter
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

What are the parasympathetic and/or sympathetic effects on the salivary glands?

A
SYMP= thick, viscous secretion
PARA= copious, watery secretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

What are the parasympathetic and/or sympathetic effects on the skin?

A

SYMP (CHOLINERGIC)= increased sweating

PARA= piloerection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What are the parasympathetic and/or sympathetic effects on the heart?

A

SYMP=increased HR and contractility

PARA= decreased HR and contractility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

What are the parasympathetic and/or sympathetic effects on the GI system?

A
SYMP= decreased motility and tone, sphincter contraction
PARA= increased motility and tone, increased secretions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

What are the parasympathetic and/or sympathetic effects on the blood vessels?

A

PARA (skeletal muscle)= dilatation

PARA (skin, mucous membranes and splanchnic are)= constriction

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

What does the effect of a blocking nicotinic R depend on?

A

Which arm of the NS is active

E.g. in eye, trachea, ureters/bladder, heart, GI

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q
Which 2 of the following effects would be observed at rest after treatment  with a ganglion blocking drug?
Increased heart rate
Pupil constriction
Bronchodilation
Detrusor contraction
Increased gut motility
A

Increased HR and bronchodilation

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

Why do ganglion blocking drugs cause hypotension?

A

Increased renin secretion

Constriction of blood vessels (skin, mucous membranes, splanchnic areas)

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

What are the effects of ganglion blocking drugs (nicotinic R antagonists) on smooth muscle?

A

Pupil dilation
Decreased GI tone
Bladder dysfunction (relaxes detrusor, constriction of trigone and sphincter
Bronchodilation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

What are the effects of ganglion blocking drugs (nicotinic R antagonists) on exocrine tissue?

A

Decreased exocrine secretions

E.g. from skin, salivary glands, GI system

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

How are hexamethonium and trimetaphan used clinically?

A

Hexamethonium= 1st anti-hypertensive, predominantly a nicotinic R blockage

Trimetaphan= hypotension during surgery, predominantly a R antagonist (very short acting)

Both can do both

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

Why are so few nicotinic receptor antagonists used clinically?

A

Many side effects

Only hexamethonium and trimetaphan used clinically

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

What is a-bungarotoxin? How does it work?

A

From common krait snake venom
Very potent
Irreversible R antagonist-> permanent nicotinic blockade
Useful to snakes because prey can’t move and can’t breathe (diaphragm paralysed)
Affect somatic nervous system

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

Which is more clinically useful, nicotinic or muscarinic receptor antagonists?

A

Muscarinic receptor antagonists

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

What are two examples of muscarinic receptor antagonists?

A

Atropine

Hyoscine

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

What is atropine derived from?

A

Atropa belladonna

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

What is hyoscine derived from?

A

Hyoscyamus niger

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

What physiological responses could be influenced by muscarinic receptor antagonists?

A

ALL PARASYM (+SWEAT= cholinergic sympathetic)

Eye= contriction (pupil), contraction (ciliary muscle)
Trachea and broncheoles= constriction
Ureters and bladder= contraction of detrusor, relaxation of trigone and sphincter
Salivary glands= copious, watery secretion
Skin= increased sweating*
Heart= decreased rate and contractility
GI= increased motility, tone and secretions

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

Which antagonist is more specific; muscarinic or nicotinic?

A

Muscarinic

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

What are the clinical uses of atropine?

A

Muscarinic receptor antagonist

CNS effects

Normal dose- little effect
Toxic dose- mild restlessness-> agitation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

What are the clinical uses of hyoscine?

A

Muscarinic receptor antagonist

CNS effects

Normal dose- sedation, amnesia
Toxic dose- CNS depression or paradoxical CNS excitation (associated with pain)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

Why does hyoscine have more CNS effects than atropine?

A
Atropine= not very M1 selective (M1R seems to do a lot in brain)
Hyoscine= greater permeation into CNS, influence at therapeutic dose, more lipid soluble and M1 selective
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

What are the clinical uses of muscarinic receptor antagonists?

A

Ophthalmic= examination of retina with tropicamide

Anaesthetic premedication

Neurological= motion sickness, hyoscine patch,

Parkinson’s= cholinergic/dopaminergic balance in basal ganglia

Respiratory= asthma/obstructive airways disease

GI= IBS

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

Muscarinic receptor antagonists in ophthalmic exam of retina

A

Tropicamide

Pupil dilates (parasympathetic effect blocked)
Examine back of retine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

Muscarinic receptor antagonists in anaesthetic premedication

A

Antagonist interferes with parasympathetic effects including…
Trachea and bronchioles (blocks constriction)
Salivary glands (blocks copious, water secretion)
Heart (blocks heart rate decrease and contractility decrease)
Also-> sedation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

Muscarinic receptor antagonists in neurological motion sickness

A

Hyoscine patch

Cholinergic sensory mismatch still occurs but won’t be sick

Controls eye movement to maintain vision whilst in motion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

Muscarinic receptor antagonists in Parkinson’s disease

A

Normally cholinergic and dopaminergic systems work together

Cholinergic system is generally inhibitory to dopaminergic system

In PD, lose DAergic neurons so less D1 R activation-> fine control of movement is impeded

At same time cholinergic (parasymp) system is trying to dampen DAergic response

Need to block M4R (cholinergic muscarinic R4) to stop the cholinergic inhibition of DA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

Muscarinic receptor antagonists in asthma/obstructive airway disease

A

Ipratropium bromide (polar so can’t leave lungs easily, given as aerosol)

Antagonist blocks constriction of trachea and bronchioles -> lungs dilate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

Muscarinic receptor antagonists in GI system (IBS)

A

M3 selective antagonist

IBS= overactive bowel so want to suppress that
Block increased motility, tone and secretions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

What are the unwanted effects of muscarinic receptor antagonists?

A

Hot as hell= sweating, thermoregulation

Dry as a bone= secretions

Blind as a bat= cyclopegia

Mad as a hatter= CNS disturbance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q
Which of the following drugs would you
administer to treat an atropine overdose?
Bethanechol 
Ecothiopate
Hyoscine
Physostigmine
Pralidoxime
A

Bethanechol

NB.
Ecothiopate= irreversible (toxic, nasty drug but would cure atropine overdose)
Hyoscine
Physostigmine= reversible
Pralidoxime= can reverse anticholinesterase poisoning

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

What happens following poisoning with anti-cholinesterase (e.g. physostigmine)?

A

Acetylcholinesterase blocked
So ACh doesn’t get broken down to choline and acetate
ACh outcompetes atropine
More ACh binding than atropine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

What is botulinum toxin an example of?

A

A parasympatholytic

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

How does botulinum toxin work?

A

Drug that prevents ACh being exocytosed by interfering with the SNARE complex

Vesicles can’t dock with the membrane

ACh can’t be released

Very toxic

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

What are the subtypes of nicotinic cholinoceptors?

A

a1, a2, B1, B2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

Where are nicotinic cholinoceptors found?

A

On the effector organs of sympathetic pathways

Directly with NA and indirectly via adrenal medulla with A and NAin bloodtream

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
267
Q

How do directly acting sympathomimetics?

A

Mimic the actions of NA/A by binding to and stimulating adrenoceptors (GPCRs)

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

What are the main uses of directly acting sympathomimetics?

A

Principally for their actions in the CVS, eyes and lungs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q
What happens at each type of nicotinic cholinoceptor?
a1
a2
b1
B2
A
a1= PLC, IP3, DAG
a2= decreased cAMP
B1= increased cAMP
B2= increased cAMP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q
Nicotinic cholinoceptor subtypes at....
Eye
Trachea
Liver
Adipose
Kidney
Urinary bladder
Ureter
Male genitalia
Female genitalia
Lacrimal glands
Salivary glands
Skin
Heart
GI
Blood vessels (skeletal and skin/mucous)

Use + symbols to show the relative importance of adrenergic activity

A

SEE DIAGRAM

Eye= a1 ++
Trachea= B2 +
Liver= a1, B2 ++
Adipose= a1, B1 ++
Kidney= B1 ++
Urinary bladder= B2 + (relaxes detrusor), a1 ++ (constricts trigone and sphincter)
Ureter= a1 +
Male genitalia= a1 +++
Female genitalia= a/B
Lacrimal glands= a1+
Salivary glands= a/B
Skin= a1 +++
Heart= B1+++
GI= a/B
Blood vessels= B2 ++ (skeletal), a1 +++ (skin/mucous)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

What are the adrenergic effects on female and male genitalia?

A

Male- stimulates ejaculation (a1 +++)

Female- relaxation of uterus (B2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

What activates adrenoceptors?

A

NA and A

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
273
Q

How does selectivity for NA and A differ by subtype of adrenoceptor?

A

Selectivity for NA:
α1 = α2 > β1 = β2

Selectivity for A:
β1 = β2 > α1 = α2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
274
Q

Describe the NA metabolism feedback system

A

Tyrosine (with tyrosine hydroxylase)->DOPA

DOPA (with dopa-decarboxylase)-> DA

DA (in vesicle with DBH)-> NA

NA-> diffusion into blood, postsynaptic R, extraneuronal uptake, metabolised, presynaptic uptake R

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
275
Q

List 5 directly acting SNS agonists and state what subtype of adrenoceptor they act on

A
Adrenaline (non-selective)
Phenylephrine (α1)
Clonidine (α2)
Dobutamine (β1)
Salbutamol (β2)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
276
Q

Why is adrenaline used in treatment of anaphylaxis?

A

β2 – broncho dilation
β1 – tachycardia
α1 – vasoconstriction

Suppression of mediator release

(A= airways
B= breathing
C= circulation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

How can SNS agonists be used to treat pulmonary obstructive conditions?

A
In asthma (emergencies)
Acute bronchospasm associated with chronic bronchitis or emphysema

B2-> bronchdilation

Suppression of mediator release

Selective B2 agonists preferable

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
278
Q

How can adrenaline be used to treat glaucoma?

A

a1-mediated vasoconstriction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
279
Q

What can adrenaline be used for clinically?

A

Glaucoma
Cardiogenic shock
Spinal anaesthesis
Local anaesthesia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
280
Q

How can adrenaline treat cardiogenic shock?

A

Cardiogenic shock= sudden inability of heart to pump sufficient oxygen-rich blood

Severe heart attack/MI
Cardiac arrest
β1 – positive inotropic actions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
281
Q

How can adrenaline be used during spinal anaesthesia and local anaesthesia?

A

Spinal anaesthesia
Maintenance of blood pressure

Local anaesthesia
Prolongs duration of action
α1 – vasoconstrictor properties

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
282
Q

What are the unwanted actions of adrenaline?

A

Secretions= reduced and thickened mucous

CVS effects= tachycardia, palpitations, arrhythmias, cold extremities, hypertension, overdose -> cerebral haemorrhage, pulmonary oedema

Skeletal muscle= tremor

GIT and CNS= minimal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
283
Q

What is phenylephrine? What is it resistant to?

A

Drug related chemically to adrenaline
Resistant to COMT but not MAO

Selective α1»α2»>β1/β2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
284
Q

What is phenylephrine selective for?

A

α1»α2»>β1/β2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
285
Q

What are the clinical uses of phenylephrine?

A

Vasoconstriction
Mydriatic
Nasal decongestant

E.g. sudafed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

What is clonidine selective for?

A

α2»α1»>β1/2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
287
Q

What are the clinical uses of clonidine?

A

Treatment of hypertension and migraine (now superseded)

Reduces sympathetic tone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
288
Q

How does clonidine reduce sympathetic flow?

A

α2 adrenoceptor mediated
presynaptic inhibition of NA release

Central action in brainstem within

Baroreceptor pathway to reduce

Sympathetic outflow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
289
Q

What is isoprenaline selective for?

A

ß1=ß2»»α1/2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
290
Q

How do isoprenaline and adrenaline differ?

A

Chemically similar but isoprenaline is less susceptible to uptake 1 and MAO breakdown

Plasma half life 2 hours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
291
Q

What are the clinical uses of isoprenaline?

A

Cardiogenic shock
Acute heart failure
Myocardial infarction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
292
Q

Why does reflex tachycardia result form isoprenaline treatment?

A

β2-stimulation in vascular smooth muscle and skeletal muscle results in fall in venous blood pressure which triggers a reflex tachycardia via the stimulation of baroreceptors

Dobutamine better as doesn’t cause reflex tachycardia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
293
Q

What is dobutamine selective for?

A

β1»β2»>α1/2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
294
Q

What is dobutamine used for clinically?

A

Cardiogenic shock

Lacks isoprenaline’s reflex tachycardia

Administration by i.v. infusion. Plasma half life 2 minutes (rapidly metabolised by COMT)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
295
Q

What is salbutamol (ventolin) selective for?

A

β2»β1»>α1/2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
296
Q

What is salbutamol relatively resistant to?

A

MAO and COMT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
297
Q

What are the clinical uses of salbutamol?

A

Treatment of asthma

Treatment of threatened premature labour

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
298
Q

How does salbutamol treat asthma?

A

β2-relaxation of bronchial smooth muscle

Inhibition of release of brochoconstrictor substances from mast cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
299
Q

How does salbutamol treat threatened premature labour?

A

β2-relaxation of uterine smooth muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
300
Q

What are the side effects of salbutamol?

A

Reflex tachycardia
Tremor
Blood sugar dysregulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
301
Q

What are cocaine and ‘cheese’ examples of?

A

Indirectly acting SNS agonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
302
Q

Where do indirectly acting sympathomimetics act?

A

Indirectly acting sympathomimetics are drugs that act at the adrenergic nerve terminal as opposed to the adrenoceptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
303
Q

What does cocaine do?

A

Uptake 1 blocker

Affects DA, NA, A

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
304
Q

What are the effects of cocaine on the CNS and CVS in low and high doses?

A

CNS
Low= euphoria, excitement, increased motor activity
High= activation of CTZ, CNS depression, respiratory failure, convulsions and death

CVS
Low= tachycardia, vasoconstriction, raised blood pressure
High= ventricular fibrillation and cardiac arrest

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
305
Q

What is the ‘cheese reaction’?

A

Tyramine

A dietary AA (in cheese, red wine and soy sauce) acts a ‘false’ neurotransmitter

Not problematic when normal mechanisms for degradation of monoamines are in operation but problem with MAO treatment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
306
Q

How does the cheese reaction lead to increased NA release from tyramine?

A
  1. Some weak agonistic activity by cheese false NT at post synaptic adrenoceptors
  2. Competes with catecholamines for Uptake 1, i.e. it is taken up into adrenergic nerve terminals
  3. Displaces NA from intracellular storage vesicles into cytosol
  4. NA and tyramine compete for sites on MAO
  5. Cytoplasmic NA leaks through the neuronal membrane to act at postsynaptic adrenoceptors

INCREASED NA RELEASE overall but issue is if you’re taking MAO inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
307
Q

What is caused by the cheese reaction in someone taking MAO inhibitors?

A

Hypertensive crisis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

Adrenoceptor functions: a1

A

Vasoconstriction

Relaxation of GIT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
309
Q

Adrenoceptor functions: a2

A

Inhibition of transmitter release
Contraction of vascular smooth muscle
CNS actions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

Adrenoceptor functions: b1

A

Increased cardiac rate and force
Relaxation of GIT
Renin release from kidney

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
311
Q

Adrenoceptor functions: b2

A

Bronchodilation
Vasodilation
Relaxation of visceral smooth muscle
Hepatic glycogenolysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
312
Q

Adrenoceptor functions: b3

A

Lipolysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q
What adrenoceptor antagonists act on the following?:
a1 and b1
a1 and a2
a1
b1 and b2
b1
A
a1 and b1= labetalol (really non-selective but predominantly these two)
a1 and a2= phentolamine
a1= prazosin
b1 and b2= propranolol
b1= atenolol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
314
Q

What does propranolol act on?

A

Adrenoceptor antagonist

B1 and B2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
315
Q

What are SNS antagonists and false transmitters useful for clinically?

A

Hypertension
Cardiac Arrhythmias
Angina
Glaucoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
316
Q

What is hypertension?

A

Increased BP associated with increased risk of other disease
Sign rather than a disease

Sustained diastolic arterial pressure >90mmHg (140/90mmHg or higher)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
317
Q

What are the main elements that contribute to hypertension?

A

Blood volume
Cardiac output
Peripheral vascular tone

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
318
Q

What are the tissue targets for antihypertensives?

A

Sympathetic nerves that release NA (vasoconstrictor)

Kidney
Heart
Arterioles (determine peripheral resistance)
CNS (determines BP pressure set point)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
319
Q

What are the 3 main categories of beta adrenoceptor antagonists?

A

Cardioselective
Nonselective
Drugs with additional a1 antagonist activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
320
Q

How do b-adrenoceptor antagonists act?

A

Competitive antagonism of B1 adrenoceptors (B2 antagonism’s importance is unclear)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
321
Q

What do b-adrenoceptor antagonists do to the CNS, heart and kidney?

A

CNS
Reduce sympathetic tone

HEART
B1- reduce HR and CO (disappears in chronic treatment)

KIDNEY
B1- reduce renin-production-> reduced peripheral resistance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
322
Q

What are the pre-synaptic effects of b-adrenoceptor antagonists?

A

Antagonist blocks facilitatory effects of presynaptic b-adrenoceptors on NA release

Contributes to antihypertensive effect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
323
Q

What are the main unwanted effects of b-adrenoceptor antagonists?

A
Bronchoconstriction 
Cardiac failure 
Hypoglycaemia
Fatigue
Cold extremities
Bad dreams
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
324
Q

Why is bronchoconstriction an unwanted effect of b-adrenoceptor antagonists?

A

Problematic with asthmatics and patients with obstructive lung disease e.g. bronchitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

Why is cardiac failure an unwanted effect of b-adrenoceptor antagonists?

A

Heart disease patients may need some sympathetic drive to the heart to maintain adequate CO

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
326
Q

Why is hypoglycaemia an unwanted effect of b-adrenoceptor antagonists? How can this be reduced?

A

B-antagonists mask the symptoms of hypoglycemia (sweating, palpitations, tremor)

Use of non-selective b-antagonists are more dangerous in such patients since they will also block the b2- receptors driven breakdown of glycogen

B1- selective agents may have advantages since glucose release from the liver is controlled by b2- receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
327
Q

Why is fatigue caused by b-adrenoceptor antagonists?

A

Due to reduced CO and reduced muscle perfusion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
328
Q

Why are cold extremities caused by b-adrenoceptor antagonists?

A

Loss of b-receptor mediated vasodilation in cutaneous vessels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
329
Q

What is propranolol? What does it do?

A

Non-selective beta adrenoceptor antagonist

At rest, very little change in HR, CO or BP but reduces the effect of exercise or stress on these variables

Produces adverse effects (non-selective)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

What is atenolol? What does it do?

A

Cardio-selective (historical name)
B1-selective (selectivity is concentration dependent)

Antagonises NA effects on heart

Affects any tissue with b1 receptors e.g. kidney

BUT most side effects are caused by B2 Rs
(Still not safe for asthmatic patients despite less effect on airways)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
331
Q

What is labetalol? What does it do?

A

B1 and a1 antagonists (4:1 b1 to a1)

Lowers BP by reducing peripheral resistance
a1- in peripheral vascular muscles
b1- in kidney-> decreases renin production

Induces change in HR or CO (effect wanes with chronic use)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
332
Q

What do a-adrenoceptor antagonists cause?

A

Non-selective

Mediate peripheral resistance (-> fall in arterial pressure)
Postural hypotension
CO/HR increases *reflex response to fall in arterial pressure)
Blood flow through cutaneous and splanchnic vascular beds increased
Very small effects on vascular smooth muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

What is phentolamine? What does it do?

A

Non-selective a-antagonist

Causes vasodilatation and fall in BP (blockade of a1 receptors)

Concomitant blockade of a2-Rs tends to increase NA release-> enhances reflex tachycardia

Increased GIT motility-> diarrhoea

NOT USED CLINICALLY NOW

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

What is prazosin? What does it do?

A

Highly selective a1-antagonist

Vasodilatation and fall in arterial pressure= DRAMATIC HYPOTENSIVE EFFECT

Dilation of capacitance vessles-> fall in venous pressure-> CO output decreases

Modest decrease in LDL and increase in HDL cholesterol

Less tachycardia than non-selective antagonists (don’t increase NA release via a2)
Some postural hypotension

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
335
Q

What is methyldopa? What does it do?

A

False transmitter

Antihypertensive agent taken up by noradrenergic neurons

Decarboxylated and hydroxylated to form false transmitter, α-methyl-noradrenaline
Not de-aminated by MAO so accumulates in larger quantities than NA and displaces NA from synaptic vesicles

Some CNS effects, stimulates vasopressor centre in the brainstem to inhibit sympathetic outflow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
336
Q

How does methyldopa work? How does the false transmitter differ from NA?

A

False transmitter released in same way as NA

BUT less active than NA on a1-Rs (so less effective in causing vasoconstriction)
AND more active on presyn a2 Rs (auto-inhibitory fb mechanism stronger-> reduces transmitter release below normal levels)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
337
Q

What are the beneficial results of methyldopa?

A

Antihypertensive
Renal and CNS blood flow well maintained
Suitable for hypertensive pregnant women (no adverse effects on foetus despite crossing blood-placenta barrier)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
338
Q

What are the adverse effects of methyldopa?

A

Dry mouth
Sedation
Orthostatic hypotension
Male sexual dysfunction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
339
Q

What is an arrhythmia?

A

Abnormal/irregular heart beats

Main cause= myocardial ischaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
340
Q

What affect does symapthetic activity have on arrhythmias and AV conductance?

A

Increased sympathetic drive to the heart via b1 precipitates or aggravates arrhythmias (especially common after myocardial infarction)

Sympathetic activity also affects AV conductance and the AV refractory period is increased by b-adrenoceptor antagonists (interferes with AV conduction in atrial tachycardias)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
341
Q

What class II antiarrhythmics are used and why?

A

Propranolol (non-selective b-antagonist drug)
Effects many attributed to b1 antagonism
Reduce mortality of patients with myocardial infarction
Particularly successful in arrhythmias that occur during exercise or mental stress

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
342
Q

What is angina?

A

Pain that occurs when O2 supply to myocardium is insufficient for its needs

Pain= chest, arm, neck

Brought on by exertion or excitement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
343
Q

What are the 3 types of angina?

A

Stable
Unstable
Variable

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
344
Q

What is stable angina?

A

Pain on exertion
Increased demand on heart
Due to fixed narrowing of the coronary vessels e.g. atheroma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
345
Q

What is unstable angina?

A

Pain with less and less exertion, culminating with pain at rest
Platelet-fibrin thrombus associated with a ruptured atheromatous plaque but without complete occlusion of the vessel
Risk of infarction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
346
Q

What is variable angina?

A

Occurs at rest
Caused by coronary artery spasm
Associated with atheromatous disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
347
Q

How do b-adrenoceptor antagonists reduce myocardial oxygen demand in angina?

A

Decrease HR
Decrease systolic BP
Decrease cardiac contractile activity
Reduce the oxygen demand whilst maintaining the same degree of effort

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
348
Q

At low doses what effects do b1-selective agents have on HR and contractile activity?

A

At low doses, b1-selective agents, metoprolol, reduce heart rate and myocardial contractile activity without affecting bronchial smooth muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
349
Q

What are the adverse effects of b1-selective antagonists in angina treatment?

A
Fatigue
Insomnia
Dizziness
Sexual dysfunction
Bronchospasm
Bradycardia
Heart block
Hypotension
Decreased myocardial contractility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
350
Q

What patients should avoid b1-selective antagonists as angina treatment?

A

Patients with bradycardia (heat beat of less than 55 beats/min)
Patients with bronchospasm
Patients with hypotension (systolic pressure less than 90mmHg)
Patients with AV block
Patients with severe congestive heart failure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
351
Q

Outline the production and route of aqueous humour?

A

Produced by blood vessels in ciliary body via the actions of carbonic anhydrase
Flows into posterior chamber, through pupil to anterior chamber
Drains into trabecular network and into veins and canal of Schlemm

Production indirectly related to blood pressure and blood flow in ciliary body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
352
Q

What adrenoceptor antagonists can be used to treat glaucoma?

A

Beta antagonists
Non-selective (block B1 and B2)

Carteolol hydrochloride, levobunolol hydrochloride, timolol maleate

NB. Selective b1 antagonists betaxolol hydrochloride also shown to be effective

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
353
Q

How do b-adrenoceptor antagonists treat glaucoma?

A

Reduce rate or aqueous humor formation by blocking receptors on ciliary body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
354
Q

What can b-adrenoceptor antagonists be used to treat?

A
Glaucoma
Angina
Arrhythmias
Hypertension
Heart failure
Anxiety 
Migraine prophylaxis
Benign essential tremor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
355
Q

Why are beta blockers useful in anxiety?

A

Control somatic symptoms associated with sympathetic overreactivity e.g. palpitations and tremor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
356
Q

What type of receptor is found at neuromuscular junctions?

A

nAChR (different from ganglionic nAChR)

Needs 2 molecules of ACh to stimulate it
Bonds to alpha Rs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
357
Q

List NM blocking drugs and their type

A
COMPETITIVE ANTAGONISTS (non-depolarising)
Tubocurarine
Galamine
Pancuronium
Alcuronium
Atracurium
Vecuronium

AGONISTS (depolarising)
Suxamethonium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
358
Q

What are the main sites/processes of NM blocking drug action?

A

Central processes
Conduction of nerve AP in motor neurone
ACh release
Depolarisation of motor end-plate AP initiation
Propagation of AP along muscle fibre and muscle contraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
359
Q

True or false; tubocurarine and atracurium act postsynaptically?

A

True

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
360
Q

True or false; suxamethonium acts presynaptically?

A

False (postsynaptically)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
361
Q

What NM drug acts on the central processes? How does it work?

A

Spasmolytics (diazepam, baclofen)

Act in SC
Reduce AP generation-> reduced muscle tension

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
362
Q

What NM drug acts on the conduction of nerve AP in motor neurone? How does it work?

A

Local anaesthetics

Reduce AP propagation-> weakness of skeletal muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
363
Q

What NM drug acts on the ACh? How does it work?

A

Hemicholinium
Ca2+ entry blockers
Neurotoxins

Ultimately lead to depletion of ACh
Reduces exocytotic release of Ca
Inhibits release of ACh

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
364
Q

What NM drug acts on the depolarisation of motor end-plate AP initiation? How does it work?

A

Tubocurarine
Suxamethonium

Affect nAChRs on post syn membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
365
Q

What NM drug acts on the propagation of AP along muscle fibre and muscle contraction? How does it work?

A

Spasmolytics (dantrolene)

Affects muscle fibres rather than CNS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
366
Q

NM drugs that act post-synaptically have a number of common features despite being non-depolarising antagonists or polarising agonists. These include…

A

They don’t affect consciousness
They don’t affect pain sensation
Always assist respiration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
367
Q

Suxamethonium:

MOA and pharmacokinetics

A

Depolarising NM blocker (agonist)

MODE OF ACTION
Extends EP depolarisation-> depolarisation block (phase 1)
Fasciculations-> flaccid paralysis

PHARMACOKINETICS
IV admin
Paralysis 5 min (short)
Metabolised by pseudo-cholinesterase in liver and plasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
368
Q

What is suxamethonium used for?

A

Endotracheal intubation

Muscle relaxant for ECT (electroconvulsive therapy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
369
Q

What are the unwanted effects of suxamethonium?

A

Post-operative muscle pains
Bradycardia (direct muscarinic action on heart)
Hyperkalaemia (soft tissue injury or burns-> ventricular arrhythymias or cardiac arrest)
Increased intra-ocular pressure (by action of extraocular skeletal muscles- avoid for eye injuries and glaucoma)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
370
Q

Tubocurarine: MOA

A

Naturally occurring quaternary ammonium compound (alkaloid) but now synthetic drugs available

Competitive nAChR antagonist
70-80% block necessary

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
371
Q

What are the effects of tubocurarine?

A

Flaccid paralysis

Extrinsic eye muscles (double vision)-> small muscles of face, limbs, pharynx-> respiratory muscle-> RECOVERY

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
372
Q

Why is it useful to relax skeletal muscles during surgical operations?

A

Less anaesthetic

Permit artificial ventilation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
373
Q

How can the actions of non-depolarising blockers be reversed?

A

Anticholinesterases

Neostigmine (and atropine)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
374
Q

What are the pharmacokinetics of tubocurarine?

A
IV admin (highly charged)
Doesn't cross BBB or placenta
Paralysis lasts 40-60min
Not metabolised
70% excreted in urine 30% in bile (careful if renal or hepatic function impaired)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
375
Q

What are the unwanted effects of tubocurarine?

A

Hypotension (ganglion blockade-> decreased TPR, histamine released from mast cells)

Tachycardia (may-> arrhythmias)- reflex and blockade of vagal ganglia

Bronchospasm (histamine release)

Excessive secretions (bronchial and salivary)

Apnoea (always assist respiration)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
376
Q

The clinical use of neuromuscular blocking drugs will most likely involve interference with which of following physiological processes?

A: Kidney function
B: Consciousness
C: Body temperature regulation
D: Pain sensation
E: Respiration
A

E. Respiration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
377
Q

Which of the following effects would be observed with a non-depolarising neuromuscular block?

A: Initial muscle fasciculations
B: Irreversible nAChR blockade
C: The block would be enhanced by anti-cholinesterase drugs
D: A flaccid paralysis
E: Increased arterial pressure
A

D. Flaccid paralysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
378
Q

What is phase 4 of the cardiac AP? (Bit under threshold)

A

Spontaneous depolarization (pacemaker potential) that triggers the AP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
379
Q

What is If?

A

Hyperpolarization-activated cycle nucleotide-gated (HCN) channels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
380
Q

What is Ica (T or L)?

A

Transient T or L type Ca channel

L= long lasting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
381
Q

What is Ik?

A

Potassium K channels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
382
Q

What mechanisms regulate heart rate?

A

If
Ica
Ik

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
383
Q

What is the effect of the sympathetic system on cAMP, If and ICa?

A

Increase cAMP
Increase If
Increase Ica

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
384
Q

What is the effect of the parasympathetic system on cAMP and Ik?

A

Decrease cAMP

Increase Ik

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
385
Q

Outline cardiac Ca release following an AP

A
AP excitation (via If) from the SAN induces membrane depolarization 
-> Ca channel gates open-> small release of Ca into cytoplasm (T)

Small Ca current-> release of Ca from SR (Ca-induced Ca release) through RyR2 (L)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
386
Q

What percentage of the free Ca in a cardiac twitch is through L type channels?

A

20-25%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
387
Q

What percentage of Ca necessary for cardiac contraction is released through the RyRs?

A

75-80%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
388
Q

What effect does blood amount have on the heart?

A

Determines contractility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
389
Q

What is the primary determinant of myocardial oxygen demand?

A

Myocyte contraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
390
Q

How does increased HR lead to an increased force of contraction?

A

Increased HR-> more contractions-> increased afterload or contractility (greater force of contraction)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
391
Q

How does increased preload affect contraction?

A

Small increase in force of contraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
392
Q

What is the myocardial oxygen supply?

A

Increased coronary blood flow

Increased arterial O2 content

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
393
Q

How does work affect the myocardial oxygen demand?

A

Increased heart rate, preload, afterload and contractility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
394
Q

List examples of drugs that reduce HR and how

A

B blockers= decrease If and Ica
Ca antagonists= decrease Ica
Ivabradine= decrease If

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
395
Q

How do drugs reduce heart rate (overall)?

A

Prolong rate at which depolarisation occurs

E.g. Ca antagonists reduce ability of tissue to depol

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
396
Q

List examples of drugs that decrease contractility

A

B blockers = decrease contracility

Ca antagonists= decrease ICa

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
397
Q

What receptor to Ca antagonists act on?

A

DHPR (dihydropiridine receptors)

-> decrease Ca entry into cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
398
Q

What are the classes of calcium antagonists?

A

Rate slowing (cardiac and smooth muscle actions)

Non-rate slowing (smooth muscle actions- more potent)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
399
Q

List examples of rate slowing calcium antagonists

A

Phenylalkylamines (e.g. Verapamil)

Benzothiazepines (e.g. Diltiazem)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
400
Q

List examples of non-rate slowing calcium antagonists

A

Dihydropyridines (e.g. amlodipine)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
401
Q

What heart condition can be caused by profound vasodilation (by Ca antagonists)?

A

Reflex tachycardia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
402
Q

How can drugs increase myocardial oxygen supply?

A

Hyperpolarised coronary vessel (due to organic nitrates and K channel openers)

  • > impaired ability to contract
  • > coronary blood flow
  • > improved delivery to the heart
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
403
Q

What effect do organic nitrates and K channel openers have on coronary blood flow?

A

Increase coronary blood flow (increases preload and afterload)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
404
Q

What effect does increasing coronary blood flow have on preload and afterload?

A

Increases both

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
405
Q

What effect does vasodilation have on afterload?

A

Decreases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
406
Q

What effect does venodilation have on preload?

A

Decreases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
407
Q

K channel openers affect which of the following?

Coronary blood flow
Arterial O2 content
Heart rate
Preload 
Afterload
Contractility
A

Coronary blood flow
Preload
Afterload

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
408
Q

Nitrates affect which of the following?

Coronary blood flow
Arterial O2 content
Heart rate
Preload 
Afterload
Contractility
A

Coronary blood flow
Preload
Afterload

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
409
Q

Ivabradine affects which of the following?

Coronary blood flow
Arterial O2 content
Heart rate
Preload 
Afterload
Contractility
A

Heart rate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
410
Q

Beta blockers affect which of the following?

Coronary blood flow
Arterial O2 content
Heart rate
Preload 
Afterload
Contractility
A

Heart rate

Contractility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
411
Q

CCBs affect which of the following?

Coronary blood flow
Arterial O2 content
Heart rate
Preload 
Afterload
Contractility
A

Heart rate

Contractility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
412
Q

What is angina?

A

Myocardial ischaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
413
Q

What can be used to treat angina?

A

Beta blocker or calcium antagonist as background anti-angina treatment

Ivabradine is a newer treatment

Nitrate as symptomatic treatment (short acting)

Other agents e.g. K channel opener if intolerant to other drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
414
Q

What are the side effects of beta blockers?

A

Worsening of cardiac failure (C.O. reduction)

Bradycardia (heart block)(due to less conduction through AV node)

Bronchoconstriction (blockade of β2 in airways)

Hypoglycaemia (in diabetics on insulin)(decreased glycogenolysis/ gluconeogenesis)

Cold extremities and worsening of peripheral arterial disease (blockade of β2 in skeletal muscle vessels)

MAYBE....
Fatigue
Impotence
Depression
CNS effects (lipophilic agents) e.g. nightmares
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
415
Q

What causes the side effects of beta blockers?

A

Unwanted effects can be due to actions on B1 (and sometimes B2 Rs due to partial selectivity)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
416
Q

What are the side effects of Verapamil (Ca channel blocker)?

A

Bradycardia and AV block (Ca channel block)

Constipation (gut Ca channels in 25% patients)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
417
Q

What are the side effects of Dihydropyridines?

A

10-20% patients

Ankle oedema (vasodilation means more pressure on capillary vessels)

Headache/flushing (vasodilation)

Palpitations (vasodilation/reflex adrenergic activation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
418
Q

What are the side effects of K channel openers and nitrates?

A

Ankle oedema (vasodilation means more pressure on capillary vessels)

Headache/flushing (vasodilation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
419
Q

What are the aims of treatment for arrhthymias/dysrhythmias?

A

Reduce sudden death
Prevent stroke
Alleviate symptoms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
420
Q

How can you classify arrhythmias?

A

By site of origin

Supraventricular arrhythmias (e.g. amiodarone, verapamil)

Ventricular arrhythmias (e.g. flecainide, lidocaine)

Complex (supraventricular + ventricular arrhythmias) (e.g. disopyramide)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
421
Q

What is the Vaughan-Williams classification of anti-arrhythmic drugs?

A

Class I-IV (divided by mechanism of action)
Little clinical significance

1= Sodium channel blockade 
2= Beta adrenergic blockade 
3= Prolongation of repolarisation (membrane stabilisation)
4= Calcium channel blockade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
422
Q

What is adenosine used for?

A

Anti-arrhythmic

Used IV to terminate supraventricular tachyarrhythmias (SVT)

Short-lived actions (20-30s) so safer than verapamil

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
423
Q

What is verapamil used for? How does it work?

A

Anti-arrhythmic

Reduction of ventricular responsiveness to atrial arrhythmias

Depresses SA automaticity and subsequent AVN conduction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
424
Q

What is amiodarone used for? How does it work?

A

Anti-arrhythmic

Superventricular and ventricular tachyarrhythmias

Complex action probably involving multiple ion channel blocks (prolongs AP-> prolongs hyperpolarisation-> reduces likelihood of reentry rhythms)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
425
Q

What are the adverse effects of amiodarone?

A

Accumulates in body (half life 10-100 days)
Photosensitive skin rashes
Hypo- or hyper-thyroidism
Pulmonary fibrosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
426
Q

How does digoxin work?

A

Anti-arrhythmic

Cardiac glycosides

Binds to K binding site

  • > interferes with Na and K exchange (inhibits Na-K-ATPase)
  • > less Na outside the cell so less Na/Ca exchange
  • > more Ca remains inside cardiac muscle
  • > more Ca stored in SR and then released
  • > increased contractility (POSITIVE INOTROPIC EFFECT)

-> central vagal stimulation causes increased refractory period and reduced rate of conduction through the AV node (STIMULATE VAGAL TONE TO SLOW HR DOWN)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
427
Q

What is digoxin used for?

A

Atrial fibrillation and flutter

Via vagal stimulation reduces the conduction of electrical impulses within the AV node (fewer impulses reach the ventricles and ventricular rate falls)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
428
Q

What are the adverse effects of digoxin?

A

Dyshrythmias (AV conduction block, ectopic pacemaker activity)

Hypokalamia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
429
Q

Why does hypokalamia lower the threshold for digoxin toxicity?

A

Digoxin binds to k binding site (directly competing with K) so if you have very low K then less competition for this target (so increased effects of digoxin)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
430
Q

How do endothelial cells impact smooth muscle?

A

Endothelial cells produce a raft of substances that can impact smooth muscle tone

Variscosities alongside nerve release NT (NA) when nerve is activated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
431
Q

What do the drugs affecting vascular resistance usually target?

A

Arterioles due to their contribution to bp

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
432
Q

What determines vascular tone of an arteriole?

A

Arteriolar smooth muscle normally displays a state of partial constriction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
433
Q

What is the formula for BP?

A

BP= CO x TPR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
434
Q

What is hypertension defined as?

A

Consistently above 140/90mmHg

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
435
Q

What does hypertension increase the risk of?

A

Strokes
Heart failure
MI
Chronic kidney disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
436
Q

What drugs impact vascular tone?

A

ACE inhibitor (ACEi)
Angiotensin receptor blocker (ARB)
Calcium channel blocker (CCB)
Thiazide-like diuretic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
437
Q

What is the order drugs are given for hypertension?

A

STEP 1
ACEi or ARB for <55y olds
CCB or thiazide-type diuretic for >55y olds and all Afro-Caribbeans

STEP 2
CCB or thiazide-like diuretic AND ACEi or ARB
(For Afro-Caribbeans prefer ARBs to ACEis)

STEP 3
ACEi/ARB with CCB and thiazide diuretic

STEP 4 (resistant hypertension)
Low-dose spironolactone
B blocker or a blocker
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
438
Q

How does the RAAS system change in hypertension?

A

Decreased Na reabsorption
Decreased renal perfusion pressure
Increased sympathetic NS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
439
Q

Outline the RAA system

A

Angiotensinogen is converted to angiotensin I (by renin from kidney)

AT1 converted to AT2 (by angiotensin converting enzyme ACE)

AT2

  • > SNS activation/thirst
  • > vasoconstriction
  • > salt and water retention
  • > aldosterone secretion

Bradykinin converted to inactive metabolites by ACE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
440
Q

Where does ACEi act?

A

On ACE

i.e. stops AT1-> AT2 and bradykinin->inactive metabolites

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
441
Q

Where does ARB act?

A

Prevents AT2’s actions

i. e. stops;
- > SNS activation/thirst
- > vasoconstriction
- > salt and water retention
- > aldosterone secretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
442
Q

How does ACEi work?

A

Inhibits the somatic form of ACE

Prevents conversion of AT1 to AT2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
443
Q

What are ACEi used to treat?

A
Hypertension
Heart failure
Post-myocardial infarction
Diabetic nephropathy
Progressive renal insufficiency
Patients at high risk of cardiovascular disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
444
Q

Example of ACEi?

A

Enalapril

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
445
Q

How do ACEi treat hypertension?

A

TPR x CO= BP
(Increased TPR-> increased BP
Increased venous return-> increased cardiac contractility (Starling’s law) and CO)

So preventing vasoconstriction and salt and water retention-> lowered venous return and lowered TPR-> lowered BP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
446
Q

How do ACEi treat heart failure?

A

In heart failure

  • Increased vasoconstriction-> increased afterload and cardiac work
  • Increased venous return-> fluid retention and congestion (-> oedema)

Prevent this with ACEi which -> decreased vasoconstriction and salt/water retention

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
447
Q

Overall, how do ACEis treat heart failure and hypertension?

A

Decreased vasoconstriction (decreases TPR) and decrease salt/water retention (decreases CO)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
448
Q

Example of ARB?

A

Iosartan

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
449
Q

How do ARBs work?

A

Antagonists of type 1 (AT1) Rs for Ang II-> prevents renal and vascular actions of Ang II

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
450
Q

How can ARBs be used to treat heart failure and hypertension?

A

Prevent vasoconstriction, salt/water retention and aldosterone secretion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
451
Q

What are the side effects of ACEi?

A

Generally well tolerated (less than ARB)

Cough (linked to bradykinin)
Hypotension
Hyperkalaemia (can have K supplements or K sparing diuretics)
Fetal injury
Renal failure in patients with renal artery stenosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
452
Q

What are the side effects of ARB?

A

Generally well tolerated (more than ACEi)

Urticaria/angioedema
Hypotension
Hyperkalaemia (can have K supplements or K sparing diuretics)
Fetal injury
Renal failure in patients with renal artery stenosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
453
Q

Why can ACEi lead to hyperkalaemia?

A

Prevent aldosterone from being produced

Leads to build up of K in blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
454
Q

How does smooth muscle contract?

A

Membrane depol -> VGCCs open
Ca enters and binds to calmodulin (CaM)
Ca-CaM binds to and activates myosin light chain kinase (MLCK)
MLCK mediated phosphorylation-> smooth muscle contraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
455
Q

How do CCBs work?

A

Block calcium channels

Dihydropyridines (DHPs) e.g. amlodipine= more selective for blood vessels, doesn’t cause negative inotropy

Non-DHPS (rate-limiting) e.g. varapamil= large inotropic effect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
456
Q

Why are DHPs (a type of CCB) used to treat hypertension?

A

Dihydropyridines inhibit Ca entry into vascular smooth muscle cells

Decreased TPR-> decreased BP

NB. Vasodilation can -> reflex tachycardia and increased inotropy thus increased myocardial O2 demand

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
457
Q

Why can alpha blockers be used as anti-hypertensives?

A

Antagonise a1 adrenoceptor antagonists

Low side effects

Reduce BP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
458
Q

Give examples of alpha blockers used in hypertension?

A

Phentolamine

Prazosin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
459
Q

Outline the ‘reward’ pathways in the brain activated by drugs of abuse

A

Mesolimbic DA system

Drugs hijack the body’s natural reward pathway (which is set of DA producing neurones from ventral tegmental area to nucleus accumbens)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
460
Q

Why are drugs abused?

A

DA release causes feelings of reward

Generally drugs are more powerful than natural process

(Rewarding stimulus on VTA-> NAcc DA release)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
461
Q

What are the main routes of administration in drugs of abuse?

A
Snort= intra-nasal 
Eat= oral solid or liquid
Smoke= inhalational
Inject= intra-venous
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
462
Q

How do the different routes of administration in drugs of abuse alter absorption?

A

Snort= mucous membranes of nasal sinuses (slow absorption)

Eat= GI tract (v. slow absorption)

Smoke= small airways and alveoli (rapid absorption- slightly faster than injecting)
-NB. Alveoli= very little barrier to drug

Inject= veins (rapid absorption)

In all systems- drug has to get into venous system then back to heart and then ejected to brain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
463
Q

Outline classifications of drugs of abuse

A

Narcotics/painkillers (e.g. opiates)

Depressants (downers e.g. alcohol, benzodiazepines, barbiturates)

Stimulates (uppers e.g. cocaine, amphetamine, caffeine, nicotine, metamphetamine)

Miscellaneous (e.g. cannabis, ecstasy)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
464
Q

How does the route of administration affect onset of euphoria?

A

In ascending order for onset of euphoria

Oral < intranasal < intravenous < inhalational

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
465
Q

What are the types of cannabis/marijuana?

A

Hashish/resin= trichomes (glandular hairs)

Hash oil- solvent extraction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
466
Q

How has marijuana dosing changed over time?

A

Concentration of THC has been increasing because of farming etc.

Now more potent forms of cannabis

Massive increase in dose now (skunkweed/netherweed) than was administered in past (reefer)

Dose related= very important, more powerful effect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
467
Q

What is the difference in the pharmacokinetics of cannabis orally and by inhalation?

A

Oral= 5-15%

  • Delayed onset/slow absorption
  • First pass metabolism

Inhalation= 25-35%

Percentage shows bioavailability (i.e. how much actually gets into bloodstream)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
468
Q

Outline the pharmacokinetics of cannabis

A

Very lipid soluble (slowly accumulate in poorly perfused fatty tissues-> fatty conjugates)

LIVER= Substantial proportion of the deposit in fat seems to consist of fatty acid conjugates of 11-OH-THC (very potent)

GIT= 65%

URINE= 25%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
469
Q

What is the problem with the metabolism of cannabis?

A

Major metabolite from cannabis broken down in liver-> 11-hydroxy-THC

VERY POTENT

Also, GIT 65%= enterohepatic recycling

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
470
Q

How does enterohepatic recycling affect cannabis?

A

Potent metabolite (11-OH-THC) from broken down cannabis-> metabolite largely excreted by bile-> very lipid soluble drug so excreted into intestines and then reabsorbed back into gut-> continuous effects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
471
Q

Why is the plasma cannabinoid concentration and degree of intoxication poorly correlated?

A

Poor correlation because potent metabolite (11-OH-THC) and enterohepatic recycling not taken into account

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
472
Q
How long after smoking a cannabis 
cigarette will the effects persist in the body?
5 hours
12 hours
7 days
30 days
10 years
A

30 days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
473
Q

When can cannabinoids be measured in the blood?

A

Cannabinoids measured in blood up to 30 days later

Peak levels in fat after 5 days

Redistributed back in blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
474
Q

Where are CB1 and CD2 receptors and what are they for?

A

GPCRs in the brain that respond to cannabinoids (and endogenous anandamide)

CB1 receptors in the hippocampus/cerebellum/cerebral cortex/basal ganglia (lots in the brain)

CB2 receptors on immune cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
475
Q

How does cannabis lead to euphoria?

A

Process of disinhibition of DA-producing neurons

Cannabinoid Rs on GABA interneurones

Lots of GABA interneurons to suppress the neuronal system (suppresses reward system until euphoria switched on)

So cannabis switches of the inhibition-> increased firing rate of DA- producing neurons-> more DA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
476
Q

What is the anterior cingulate cortex (ACC) involved in?

A

Performance monitoring with behavioural adjustment in order to avoid losses

i.e. used in changing environments as we constantly need to adapt our behaviour by focusing on goal-relevant info and selecting the most appropriate behaviour

477
Q

How is the anterior cingulate cortex affected in cannabis users?

A

Hypoactivity (so reduced monitoring)

Can impair ACC and can induce Schizophrenia trait in theory

478
Q

What are effects of THC and THC with cannabidiol?

A

THC and cannabidiol= elation, giggling etc.

THC alone= really unpleasant, depressing, paranoid-> psychosis

(Seems to be THC that causes the problem, cannabidiol seems to have protective effects)

479
Q

How does cannabis affect food intake?

A

Positive effect on orexigenic neurones in lateral hypothalamus

  • Indirectly switches off inhibition of GABA to increase MCH which increases neuronal activity
  • Directly switches on orexin neurones-> increased orexin production-> act to increase appetite

(Doesn’t affect leptin signals to the brain)

480
Q

How does cannabis act as an immunosuppressant?

A

Large scale depressant effects
Disrupts B and T cell function
Decrease NK and macrophage activity

Profound immunosuppressant effect

Not an issue if not chronic

481
Q

What parts of the brain are affected by cannabis?

A

Food intake= hypothalamus
Memory loss= limbic regions (amnesic effects, reduced BDNF)
Psychomotor performance= cerebral cortex

Also, psychosis and schizophrenia

482
Q

What are the peripheral effects caused by cannabis?

A

Immunosuppressant
Tachycardia/vasodilation (used in treatment)
Medulla- low CB1 receptor expressor

483
Q

Why is it important that the medulla has low CB1 receptor expression?

A

Can’t overdose

Doesn’t affect cardio/resp control centres in medulla

484
Q

Why do eyes appear pink after smoking cannabis?

A

Conjunctivae

485
Q

When is cannabis used medically?

A

MS/STROKE/PAIN= regulatory (increased regulation of CB receptors)

FERTILITY= pathology (increased regulation of CB receptors)

OBESITY= Endocannabinoids and CB1 receptors are up-regulated in the liver and adipose tissue so in theory should help with obesity (but -> suicide)

486
Q

How is cannabis eliminated?

A

11-hydroxy-THC

65% via gut (enterohepatic cycling)
25% urine

487
Q

What is rimonabant?

A

Used for autoimpairment

Inhibits Gi/o

488
Q

What are dronabinol and sativex used for?

A

Autoprotection

489
Q

What plant does cocaine come from?

A

Erythroxylum coca

Leaves= 0.6-1.8%

490
Q

What are the pharmacokinetics/dosing of cocaine?

A

Paste mushed up= 80% cocaine (organic solvent)

Cocaine HCL= dissolve in acidic solution (IV, oral, intranasal)- NB. can’t be smoked

Crack= precipitate with alkaline alkaline solution (e.g. baking soda) (INHALATION)

Freebase= dissolve in non-polar solvent (e.g. ammonia and ether) (INHALATION)

491
Q

How does speed of euphoria onset change with way cocaine is administered?

A

Smoked/IV, snort, oral

Oral (ionized in GIT so not quickly absorbed)= slowly absorption, prolonged action

492
Q

How is cocaine metabolised?

A

Metabolised very quickly

Broken down-> inactive metabolites (75-90% - ecgonine methyl ester, benzoylecgonine)

Half life 20-90 mins depending on administration

493
Q

How do the cocaine pharmacokinetics contribute to the addictive potential of the drug?

A

Very euphoric very suddenly (quick onset so associated with drug taking-> addiction)
But doesn’t take long

494
Q

How is cocaine used as a local anaesthetic?

A

Therapeutic effect

Blocks predominantly sodium channels-> no nerve conduction locally

495
Q

How does cocaine work?

A

Inhibits reuptake

Blocking the transporter-> increase life of NT in synapse

Not very selective so increases NA/Ad, 5-HT and DA

496
Q

Does cocaine influence dopamine affinity/efficacy for the DA receptor?

A

Ability of NT/chem to bind to receptor (affinity) and activate the receptor (efficacy)

SO doesn’t affect NT binding

497
Q

How does cocaine cause euphoria?

A

Very powerful euphoric affect
Blocks DA re-uptake (binds to DA transporter)
So DA stays in synapse

498
Q

When does cocaine cause mild and sever symptoms?

A

Dose and chronicity affect what effects exist

MILD= positive/reinforcing
SEVERE= negative/stereotypic
499
Q

How can cocaine cause myocardial infarctions?

A

Cocaine stimulates the sympathetic nervous system by inhibiting catecholamine reuptake at sympathetic nerve terminals, stimulating central sympathetic outflow, and increasing the sensitivity of adrenergic nerve endings to NA

Cocaine also acts like a class I antiarrhythmic agent (local anesthetic) by blocking sodium and potassium channels, which depresses cardiovascular parameters

Of these 2 primary, opposing actions, enhanced sympathetic activity predominates at low cocaine doses, whereas the local anesthetic actions are more prominent at higher doses

In addition, cocaine stimulates the release of endothelin-1, a potent vasoconstrictor, from endothelial cells and inhibits nitric oxide production, the principal vasodilator produced by endothelial cells

Cocaine promotes thrombosis by activating platelets, increasing platelet aggregation

500
Q

How is cocaine eliminated?

A

Ecgonine methyl ester, benzoylecgonine

Urine (75-90%)

501
Q

What systems does cocaine affect?

A

Transporter inhibitor= euphoria (CNS effects) and CVS problems

502
Q

What is nicotine?

A

Nicotine is a potent parasympathomimetic alkaloid

Present in particulate matter in cigarettes (smoking= 5% particulates and 95% volatile)

Particulates= tar droplets with alkaloid-> deep in lungs and then absorbed by blood

503
Q

What are the pharmacokinetics/dosing of nicotine?

A

Most replacement mechanisms try to maintain nicotine levels

Nicotine spray – 1mg (20-50%)

Nicotine Gum – 2-4mg (50-70%)

Cigarettes – 9-17mg (20%)

Nicotine Patch – 15-22mg/day (70%)

504
Q

Why is there no buccal absorption of nicotine?

A

Cigarette smoke is acidic i.e. no buccal absorption

(50% does get to lungs but very ionised so not great at getting into lungs and bloodstream except in lower/deeper lung)

Absorption in alveoli independent of pH

505
Q

What is the pKa of nicotine?

A

7.9

506
Q

How is nicotine metabolised?

A

Hepatic CYP2A6-> (70-80%)-> cotinine

Less addictive than cocaine but similar metabolism

Vast majority (75-80%) quickly metabolised in liver-> inert metabolite

Longer half life (t1/2-4 h) than cocaine but still short (so less addictive)

507
Q

What are the pharmacodynamics of nicotine? (What does it bind to?)

A

Bind to nicotinic AchR

Ion-channel linked R
Increases channel opening
Increases ion flux

Mediates autonomic function-> impact parasymp and symp actions

508
Q

How does nicotine cause euphoria?

A

Nicotine binds to nAChR and directly stimulates neurone at level of VTA
Firing rate of neurone increases
More DA produced

509
Q

What are cardiovascular effects of nicotine?

A

Increase sympathetic stimulation (CNS and adrenals)

Increase HR and SV

NA induced vasoconstriction (of coronary arterioles and skin arterioles)

Vasodilation of skeletal muscle arterioles (due to symp system)= increased lipolysis/FFA/VLDL, decreased HDL, increased TXA2, decreased NO

510
Q

What are the metabolic effects of nicotine?

A

Increased metabolic rate

Appetite suppressed by nicotine and increased metabolic rate -> suppresses weight gain

511
Q

What are the neurodegenerative effects of nicotine?

A

Seems to have therapeutic value in development of PD and AD

PD= increased brain CYPs-> neurotoxins
(Chronic cig smokers-> increased cytochrome p450-> increased ability to metabolise neurotoxins-> less neurotoxin degradation of DAergic toxins-> less PD)

AD= decreased B-amyloid toxicity and APP (amyloid precursor protein)
(Chronic cig smokers-> decreased B amyloid toxicity and APP-> decreased AD risk)

512
Q

How long is the onset of euphoria with nicotine?

A

Seconds

513
Q

How does caffeine cause euphoria?

A

Oral and small doses so minor euphoria

Adenosine receptor antagonist

Adenosine activates A1 receptor-> negative effect on DA R function and DA secretion by mesolimbic pathway
So caffeine does opposite= increases DA release

514
Q

Can chocolate be considered a drug of abuse?

A

Is naturally rewarding but doesn’t directly addictive doesn’t affect DA

515
Q

Outline the epidemiology of alcohol?

A

Western Europe= bad for alcohol consumption especially Ireland

516
Q

How do you calculate the absolute amount of alcohol?

A

% ABV x 0.78= g alcohol/100ml

ABV= alcohol by volume

517
Q

How do you calculate units of alcohol?

A
         1000

1 unit= 10ml or 8g of absolute alcohol

518
Q

What is the recommended amount of alcohol per week?

A

Up to 14 units a week
for males and females

Should be spread throughout the week and with ‘drink free’ days

519
Q

What is binge drinking?

A

> 8 units in one sitting

520
Q

What is the legal driving limit?

A

0.08

Blood levels= 0.01% = 10mg/100ml blood

521
Q

How is alcohol administered?

A

Oral alc-> 20% absorbed from stomach and 80% from small intestine

522
Q

How does having a full stomach affect the speed of onset with alcohol?

A

Speed of onset= depends on gastric emptying (directly proportional)

Alc needs to get to small intestine

Stomach doesn’t empty if you’ve eaten a meal (slowly releases to SI)

So drinking on full stomach means alc stays in stomach more

523
Q

Explain the metabolism of alcohol

A

90% metabolised
10% not metabolised= some of it can be breather out (use breathalyser)

85% occurs in liver
(Alcohol->acetaldehyde)

524
Q

How does alcohol get converted to acetaldehyde?

A

75% alcohol dehydrogenase

25% mixed function oxidase (upregulates in chronic alc drinking-> tolerance [reversible])

525
Q

How does alcohol tolerance build up?

A

Mixed function oxidase (which is involved in converting alcohol to acetaldehyde) upregulates in chronic alc drinking-> tolerance

Reversible

526
Q

What is acetaldehyde?

A

Produced in first stage of alcohol metabolism (metabolite of alcohol)

Toxic

527
Q

How much metabolised alcohol is metabolised in the liver?

A

85%

First pass hepatic metabolism

528
Q

What percentage of metabolised alcohol is metabolised in the GIT?

A

15%

Alcohol -> alcohol dehydrogenase-> acetaldehyde

529
Q

Why are blood alcohol levels higher in women?

A

Women are fatter than men, have less body water (50%) and less ADH

Men have more body water (59%) and less adipose tissue

Alcohol is water soluble

530
Q

Why can disulfiram be effective as alcohol aversion therapy?

A

Prevents aldehyde dehydrogenase (which convert acetaldehyde to acetic acid)

Causes build up of acetaldehyde-> lots of negative effects of alcohol-> makes drinking more unpleasant

531
Q

What causes ‘asian flush’?

A

Genetic polymorphism (particularly prevalent in asians/middle easterns)

People can’t build up acetaldehyde

Causes build up of acetaldehyde-> lots of negative effects of alcohol

532
Q

What are the pharmacodynamics of alcohol?

A

Low pharmacological potency (‘alcohol as a key fits into a lot of locks’)

Very high levels to induce effects (poor affinity and efficacy)

533
Q

What are the effects of alcohol on the CNS?

A

Acute effects

Primary= depressant
(CNS agitation may occur)

Disinhibition-> excitation but only at low dose

Degree of CNS excitability depends on environment and personality

534
Q

Why are there acute CNS effects due to alcohol?

A

Increased pre- vs post-synaptic allopregnenolone (steroid)

Allosteric modulation (decreased NMDA Rs)

Neurotransmitters (deceased Ca channels)

535
Q

How can alcohol cause euphoria?

A

Works similarly to opiates and cannabis
Switches off GABA (disinhibition)
Alcohol uses u receptor
Not as powerful as heroin (less selective)

536
Q

What parts of the brain does alcohol affect? What does this cause?

A

Corpus Collosum= Passes info from the left brain (rules, logic)
to the right brain (impulse, feelings) and vice versa

Hypothalamus= Controls appetite, emotions, temperature, and pain sensation

Reticular Activating System= Consciousness

Hippocampus= Memory

Cerebellum= Movement and coordination

Basal Ganglia= Perception of time

537
Q

How does alcohol cause cutaneous vasodilation?

A

Cutaneous vasodilation (acts on arterioles via acetaldehye)

Decreases Ca entry
Increases prostaglandins

Explains facial flush

538
Q

How does alcohol cause the heart to speed up?

A

Speeds heart up
Mostly centrally mediated

Alc impairs the relationship between brain and baroreceptor (NORMALLY= stimulates parasympathetic and inhibits sympathetic from increasing heart rate)

Alc changes this, lose inhibition of sympathetic-> allows high firing from symp nerve-> increased HR

539
Q

How does alcohol affect the cardiovascular system?

A

Cutaneous vasodilation

Tachycardia

540
Q

How does alcohol affect the endocrine system?

A

ACUTE= Diuresis (polyuria)

CHRONIC= increased ACTH secretion and decreased testosterone secretion

541
Q

Why does alcohol cause (diuresis) polyuria?

A

Posterior pituitary effects
Probably acetaldehyde not alcohol
Prevents VP secretion from post pit (depressant effect)
Less water retention-> more urine production

542
Q

What are the chronic CNS effects of alcohol?

A

COMMON
Dementia= cortical atrophy/ volume cerebral white matter

Ataxia= cerebellar cortex degeneration

LEADS ON TO…
WENICKE-KORSAKOFF SYNDROME (due to thiamine deficiency)
= Chronic alcoholics get too much caloric intake from alc
So low thiamine for a length of time-> impaired brain metabolism

NB. Wernicke’s encephalopathy – (affects 3rd ventricle and aqueduct, initialyy reversible)
Korsakoff’s psychosis-(affects dorsomedial thalamus, irreversible hippocampal damge)

543
Q

What are the chronic effects of alcohol on the liver?

A

Lipids build up in liver

  • NAD+ mostly lost
  • Triacylglycerol builds up in liver

Generation of free radicals

Blood and hepatic cytokine changes (e.g. increased IL-6 and TNF-a)

544
Q

Why do lipids build up in the liver?

A

1)
Alcoholics use large amount of NAD+ to metabolise alc

NAD+ normally= to convert glucose to pyruvate and pyruvate to acetyl coa and acetyl co into citric acid cycle and fatty acid cycle

So diverting NAD+ (in alcoholic) means…

  • Pyruvate-> lactate
  • Acetyl coA-> ketones
  • Lipids building up in liver

2)
Triacylglycerol builds up in liver because glycerol and fatty acids in blood can’t go to hepatocyte mitochondria

545
Q

Why does chronic alcohol lead to free radical generation in hepatitis?

A

Generation of free radicals

Predominantly mixed function oxidase enzymes-> generates oxygen free radicals

Becomes big problem chronically (elevated in liver)-> generate inflam stimulus-> hepatitis (reversible if stop drinking)

Prolonged inflam-> cytokine release-> structural changes in liver

CIRRHOSIS NOT REVERSIBLE (if inflam is prolonged long enough)

546
Q

What happens to the liver in cirrhosis?

A

Liver cells can’t regenerate
Not reversible
At some point, liver can’t manage the metabolic demands of the body

Hepatocyte regeneration decreased
Fibroblasts increases (supportive framework)
Active lier tissue decreases

547
Q

What are the beneficial CVS effects of low dose alcohol?

A

Low dose alcohol is positive (probably protective against some cardiac disease)

Decreased mortality from coronary artery disease (men 2-4 units/day)
Increase HDLs
Increase tPA levels
Decreased platelet aggregation

548
Q

What are the beneficial GIT effects of low dose alcohol?

A

Alcohol is absorbed from stomach
Chronic alcoholic= constantly have acetaldehyde in stomach
Damages stomach mucosa and possibly causes cancer

549
Q

What are the chronic effects on the endocrine system of alcohol?

A

Increased ACTH secretion (high cortisol, like Cushing’s effects)

Decreased testosterone secretion (-> feminisation)

550
Q

What are the constituents of blood?

A

45% blood cells (99% erythrocytes)

55% blood plasma

551
Q

What blood constituents are involved in clotting?

A

BLOOD CELLS
Platelets

PLASMA CLOTTING FACTORS
Procoagulants= prothrombin, factors V, VII-XIII
Fibrinogen

Anticoagulants= plasminogen, TFPI, proteins C&S, antithrombin

552
Q

What is TFPI?

A

Tissue factor pathway inhibitor

553
Q

What do procoagulants lead to?

A
Physiologically= Clotting
Pathologically= Thrombophilia
554
Q

What do anticoagulants lead to?

A
Physiologically= Haemophilia
Pathologically= Bleeding
555
Q

What is haemostasis?

A

Essential physiological process- blood coagulation prevents excessive blood loss

556
Q

What is thrombosis?

A

Pathophysiological process where blood coagulates within blood vessel and obstructs blood flow

Red thrombi can embolise

557
Q

What is the difference between red and white thrombi?

A

RED= venous thromboses= high fibrin components (clot becomes life-threatening if it dislodges from the vessel-> embolises)

WHITE= arterial thromboses= high platelet components

558
Q

What is atherosclerosis?

A

Pathophysiological process- thrombus forms within atherosclerotic plaque

Plaque can rupture-> thrombus released into lumen (ischaemia)

Arterial thromboses (white thrombi)

559
Q

What is Virchow’s triad?

A

Rate of blood flow
Consistency of blood
Blood vessel wall integrity

560
Q

How does rate of blood flow contribute to coagulation?

A

Blood flow is slow/stagnating -> no replenishment of anticoagulant factors and balance adjusted in favour of coagulation

561
Q

How does consistency of blood contribute to coagulation?

A

Natural imbalance between procoagulation and anticoagulation factors e.g. Factor V leiden

562
Q

How does blood vessel wall integrity contribute to coagulation?

A

Damaged endothelia-> blood exposed to procoagulation factors

563
Q

How does Virchow’s triad go from physiology to thrombogenic?

A

Physiological:
Anti-coagulants= pro-coagulants

Thrombogenic:
Increased pro-coagulants
Decreased anti-coagulants

564
Q

What is the cell-based theory of coagulation?

A
  1. INITIATION
    Anticoagulants-> small scale production of thrombin
  2. AMPLIFICATION
    Antiplatelets-> large scale thrombin production on the surface of platelets
  3. PROPAGATIONS
    Thrombolytics-> thrombin mediated generation of fibrin strands
565
Q

What causes the coagulation process?

A

Virchow’s triad

Cell-based theory of coagulation

566
Q

List some important anti-coagulants

A

Antithrombin
Vitamin K
Factors II and X

567
Q

What happens in the initiation of thrombosin production?

A

Small-scale thrombin production

  1. TISSUE FACTOR (TF)
    TF bearing cells activate factors X and V forming-> prothrombinase complex
  2. PROTHROMBINASE COMPLEX
    This activates factor II (prothrombin) creating factor IIa (thrombin)
  3. ANTI-THROMBIN (AT-III)
    AT-III inactivates fIIa and fXa
568
Q

What are other names for factors IIa and II?

A
IIa= thrombin
II= prothrombin
569
Q

How can anticoagulants be used to stop the initiation of thrombin production?

A

Inhibit factor IIa
- Dabigatran (oral) - factor IIa inhibitor

Inhibit factor Xa
- Rivaroxaban (oral) - factor Xa inhibitor

Increase activity of AT-III

  • Heparin (IV, SC) - activates AT-III (decrease fIIa & decrease fXa)
  • Low-molecular weight heparins (LMWHs, e.g.Dalteparin) - activate AT-III (decrease fXa)

Reduce levels of other factors

  • Warfarin (oral) - vitamin K antagonist
  • Vitamin K - required for generation of factors II, VII, IX and X
570
Q

What is dabigatran used for?

A

Factor IIa inhibitor

Anticoagulant to reduce thrombin production initiation

571
Q

What is rivaroxaban used for?

A

Factor Xa inhibitor

Anticoagulant to reduce thrombin production initiation

572
Q

What is heparin used for?

A

Activates AT-III (decreases fIIa and fXa)

Anticoagulant to reduce thrombin production initiation

573
Q

What are low molecular weight heparins (e.g. dalteparin) used for?

A

Activates AT-III (decreases fXa)

574
Q

What is warfarin used for?

A

To reduce the levels of other factors
Vitamin K antagonist
(Vit K required for generation of factors II, VII, IX and X)

575
Q

What are the indications for when you use anticoagulants to stop initiation of thrombin production?

A

Main indications for diseases that involve venous thrombosis (red thrombi)

Deep vein thrombosis and pulmonary embolism
Thrombosis during surgery
Atrial fibrillation - prophylaxis of stroke

576
Q

How can anti-platelets be used to prevent amplification of platelets?

A

PLATELET ACTIVATION AND AGGREGATION

Thrombin
- Factor IIa -> activates platelets

Activated platelet

  • Changes shape
  • Becomes ‘sticky’ and attaches other platelets
577
Q

How can you prevent platelet activation?

A

PREVENT PLATELET ACTIVATION/AGGREGATION
Clopidogrel (oral) - ADP (P2Y12) receptor antagonist

INHIBIT PRODUCTION OF TXA2
Aspirin (oral) - irreversible COX-1 Inhibitor
(NB: High doses no more effective BUT more side-effects)

PREVENT PLATELET AGGREGATION
Abciximab (IV, SC)- monoclonal ab
(Limited use AND only by specialists)

578
Q

What is clopidogrel used for?

A

ADP (P2Y12) R antagonist

Inhibits platelet activation/aggregation

579
Q

What is aspirin used for?

A

Inhibit production of TXA2

Aspirin irreversibly Inhibits platelet activation/aggregation

580
Q

What is abciximab used for?

A

Inhibits platelet aggregation by inhibiting GPIIb/IIa receptor

581
Q

What are the indications for when you stop platelet activation?

A

Arterial thrombosis

Acute coronary syndromes - myocardial infarction
Atrial fibrillation - prophylaxis of stroke

582
Q

What happens in the propagation of the thrombolytics?

A

Generation of fibrin strands

  1. Activated platelets
    - Large-scale thrombin production
  2. Thrombin
    - Factor IIa-> binds to fibrinogen and converts to fibrin strands
583
Q

What is the mechanism of thrombolytics?

A

Anticoagulants and anti-platelets - DO NOT remove pre-formed clots

  • Convert plasminogen plasmin
  • Plasmin - protease degrades fibrin
  • Alteplase (IV) - recombinant tissue type plasminogen activator (rt-PA)
584
Q

How are platelets activated?

A

Thrombin - binds to protease-activated receptor (PAR) on platelet surface.
PAR activation rise in intracellular Ca2+
Ca2+ rise exocytosis of adenosine diphosphate (ADP) from dense granules

ADP RECEPTORS
ADP activates P2Y12 receptors platelet activation/ aggregation

CYCLO-OXYGENASE
PAR activation liberates arachidonic acid (AA)
Cyclo-oxygenase (COX) generates thromboxane A2 (TXA2) from AA

GLYCOPROTEIN IIb/IIIa RECEPTOR (GPIIb/IIIa)
TXA2 activation expression of GPIIb/IIIa integrin receptor on platelet surface
GPIIb/IIIa - involved in platelet aggregation

585
Q

What indications do you use thrombolytics for?

A

Arterial and venous thrombosis

  • Stroke= first-line treatment
  • ST-elevated MI
586
Q

What causes DVT? How can it develop into a PE?

A

‘Red’ thrombus-> deep vein of the leg (e.g poplietal vein)

Caused by:

  • Decreased rate of blood flow
  • Damage to endothelium

Thrombus detachment-> pulmonary embolism

587
Q

What is the management strategy for DVT?

A

Restore balance between coagulants and anti-coagulants

Decreased levels of anti-coagulant factors
= Anticoagulants

588
Q

What is an NSTEMI?

A

Non-ST elevated myocardial infarction (MI)

An acute coronary syndrome

589
Q

What causes an NSTEMI? How can it be treated?

A

‘White’ thrombus -> partially occluded coronary artery

Caused by:

  • Damage to endothelium
  • Atheroma formation
  • Platelet aggregation

Need to:

  1. Reduce lipid accumulation
  2. Reduce platelet aggregation
  3. Dissolve thrombus

Management:
a) Prevent further arterial occlusion
Decrease platelet activation/aggregation
= Anti-platelets (1 and 2)

b) Dissolve clot
= Thrombolytics (3)

590
Q

What diseases are associated with thrombosis?

A

ARTERIAL
Brain= stroke
Heart= acute coronary syndromes (-> atrial fibrillation)
Arteries= aortic aneurysm, peripheral arterial disease

VENOUS
Deep vein thrombosis
Pulmonary embolism

591
Q

How do you treat a stroke (associated with arterial thrombosis)?

A

Anti-platelets

Thrombolytics

592
Q

How do you treat an acute coronary syndromes (associated with arterial thrombosis)?

A

Anti-platelets

Thrombolytics

593
Q

How do you treat atrial fibrillation (associated with arterial thrombosis)?

A

Anti-platelets

Anticoagulants

594
Q

How do you treat aortic aneurysms/peripheral arterial disease (associated with arterial thrombosis)?

A

Anti-platelets

595
Q

How do you treat DVT (associated with venous thrombosis)?

A

Anticoagulants

596
Q

How do you treat PEs (associated with venous thrombosis)?

A

Anticoagulants

Thrombolytics

597
Q

Differentiate between the terms haemostasis and thrombosis

A

Haemostasis is a physiological process preventing blood loss whereas thrombosis is a pathophysiological process

598
Q

Explain the process of coagulation and identify the actions of drugs that affect production or activation of clotting factors

A

The cell based theory of coagulation consists of three stages

Anticoagulants inhibit stage 1 and affect the activity/ production of clotting factors

599
Q

Explain the process of platelet activation and identify the action of specific antiplatelet drugs

A

Platelets are activated by thrombin, which causes Ca2+ rise, ADP release and GPIIb/IIIa receptor expression

Anti-platelet drugs fall into 3 main categories:
COX inhibitors
P2Y12 receptor antagonists
GPIIb/IIIa receptor antagonists

600
Q

What are the 3 main categories of anti-platelet drugs?

A

COX inhibitors
P2Y12 receptor antagonists
GPIIb/IIIa receptor antagonists

601
Q

Explain the actions of fibrin and identify the role of thrombolytic drugs

A

Fibrin is produced from fibrinogen and stabilises blood clots
Plasmin can degrade fibrin strands and thrombolytic drugs activate plasmin

602
Q

Understand which of these classes of drugs can be used in specific clinical situations

A

Anticoagulants: deep vein thrombosis, pulmonary embolism, during surgical procedures, atrial fibrillation

Antiplatelets: acute coronary syndromes (STEMI & NSTEMI), atrial fibrillation

Thrombolytics: STEMI, stroke

603
Q

What kind of lipoproteins are the main drivers of atherosclerosis?

A

LDL

604
Q

What are the main differences between LDL and HDL in circulating in solution?

A

LDLs have apoprotein b which allows them to circulate in an aqueous environment

(HDL- apoprotein A1)

605
Q

What is the exogenous pathway of lipid metabolism?

A

Dietary triglycerides and cholesterol-> intestine

Chylomicron out of intestine-> (via LP lipase)-> FFA and chylomicron remnant

FFA-> skeletal muscle and adipose tissue

Chylomicron remnant-> remnant receptor in liver or to form atheroma

606
Q

What is the endogenous pathway of lipid metabolism?

A

Liver is key organ

From liver= Large VLDL, small VLDL, IDL, LDL (also can be formed from the larger version)

LDL receptor on liver

607
Q

What do chylomicrons do?

A

Whilst chylomicrons transport triglyceride from the gut to the liver

608
Q

What is VLDL?

A

VLDL is the analogous particle that transports triglycerides from the liver to the rest of the body

609
Q

What is reverse cholesterol transport?

A

As cholesterol cannot be broken down within the body, it is eliminated intact

Transported via HDL from peripheral tissues to liver

610
Q

Why is HDL protective from atherosclerosis?

A

HDL acts as a vehicle for the transport of cholesterol for elimination (peripheral tissues-> liver)

HDL as a protective factor against the development of atherosclerosis

611
Q

How is inflammation involved in atherosclerosis?

A

Monocyte goes into subendothelial space and converted into foam cells

Foam cells are loaded with cholesterol and cholesterol esters

Inflam process-> atherosclerosis

612
Q

Outline endothelial dysfunction in atherosclerosis?

A

Endothelial dysfunction in atherosclerosis is characterised by a series of early changes

Changes:

  • Greater permeability of the endothelium
  • > Up-regulation of endothelial adhesion molecules
  • > Leukocyte adhesion
  • > Migration of leukocytes into the artery wall

-> Lesion formation

613
Q

What is the fatty streak in atherosclerosis?

A

Fatty steak= earliest recognisable lesion of atherosclerosis

Caused by the aggregation of lipid-rich foam cells, derived from macrophages and T lymphocytes (within the intima)

Common, may increase in size, remain static or ever disappear

614
Q

What later lesions occur after fatty streaks in atherosclerosis? How are they formed?

A

Smooth muscle cells

Steps= smooth muscle migration, T cell activation, foam cell formation and platelet adherence and aggregation

615
Q

How is a complicated atherosclerotic plaque?

A

Plaque= advance stage in atherosclerotic process

Results from death and rupture of the lipid-laden foam cells in the fatty streak

Migration of vascular smooth muscle cells (VSMCs) to the intima and laying down of collagen fibres results in the formation of a protective fibrous cap over the lipid core

The fibrous cap separates the highly thrombogenic lipid-rich core from circulating platelets and other coagulation factors

Lesions expand at the shoulders by continued leukocyte adhesion

616
Q

What characterises stable atherosclerotic plaques?

A

Stable atherosclerotic plaques are characterised by a necrotic lipid core covered by a thick VSM-rich fibrous cap

617
Q

What are the types of atherosclerotic lesions?

A

Coronary artery at lesion-prone location

Type II lesion

Type III (preatheroma)

Type IV (atheroma)

Typve V (fibroatheroma)

Type VI (complicated lesion)

618
Q

What happens to remnant lipoproteins?

A

Remnants= VLDL, chylomicron remnant, IDL)

Contribute to atherosclerosis (not LDL alone)

Remnant lipoproteins

  • > remnants
  • > modified remnants
  • > MCP-1, macrophage, foam cells

Macrophage-> cytokines-> adhesion molecules

619
Q

What are vulnerable atherosclerotic plaques characterised by?

A

Characterised by a thin fibrous caps, a core rich in lipid and macrophages, and less evidence of smooth muscle proliferation

620
Q

What are vulnerable plaques prone to?

A

Rupture and ulceration

Followed by rapid development of thrombi

(NB. Size of plaque doesn’t appear to predict whether a plaque is prone to rupture)

621
Q

Where does plaque rupture usually occur? What is it associated with?

A

Rupture usually occurs at sites of thinning (particularly at shoulder area of plaque) and is associated with regions with relatively few smooth muscle cells but abundant macrophages and T cells

Associated with greater influx and activation of macrophages, accompanied by release of matrix metalloproteinases involved with the breakdown of collagen

622
Q

What modifies LDL cholesterol?

A

Modified by other risk factors

  • Low HDL cholesterol
  • Smoking
  • Hypertension
  • Diabetes
623
Q

What is a 10% increase in LDL cholesterol associated with?

A

An approximate 20% increase in risk for CHD

624
Q

What happens when HDL is low? What causes it to be low?

A

The lower the HDL cholesterol level, the higher the risk for atherosclerosis and CHD

HDL cholesterol tends to be low when triglycerides are high

HDL cholesterol is lowered by smoking, obesity and physical inactivity

625
Q

What are examples of of lipid-lowering therapies?

A
Inhibitors of HMG CoA reductase (statins)
Fibrates
Bile acid sequestrants (resins)
Nicotinic acid and its derivatives
Probucol
626
Q

What are bile acid sequestrants? SEs?

A

Potent cholesterol-lowering agents

NB. compliance can be a problem as patients may object to the taste and texture, and common adverse events are gastrointestinal bloating, nausea and constipation

627
Q

What is nicotinic acid used for? SEs?

A

B-complex vitamin= with lipid-lowering properties

Very effective at increasing HDL cholesterol levels and is indicated for all dyslipidaemias except congenital lipoprotein lipase deficiency

Limited by the incidence of adverse events, which include flushing, skin problems, gastrointestinal distress, liver toxicity, hyperglycaemia and hyperuricaemia

628
Q

What are fibrates? SEs?

A

Effective triglyceride-lowering drugs (effective for patients with type III hyperlipoproteinaemia)

Main mechanism= activation of PPAR (peroxisome proliferator activated) alpha receptors

In some patients they modestly lower LDL cholesterol and raise HDL cholesterol

However, in the majority of patients they are only moderately successful in reducing LDL cholesterol

629
Q

What is probucol?

A

Prescribed for the treatment of high cholesterol levels

Only a modest LDL cholesterol-lowering effect, and there is no evidence that it reduces CHD risk

630
Q

What is the mechanism of action of statins?

A

Affects cholesterol synthesis pathway
Inhibit HMG-CoA reductase

Increase number of LDL receptors on hepatocytes
Leads to reduced elevated LDL (reduced LDL-C particles)

631
Q

List examples of statins and their % change on LDL and HDLs?

A

Simvastatin= LDL -32%, HDL +8%

Fluvastatin= LDL -22%, HDL +2%

Atorvastatin= LDL -38%, HDL +6%

Rosuvastatin= LDL -52%, HDL +14%

Pravastatin= LDL -32%, HDL +3%

632
Q

Who are statins effective for?

A

Patients with establishes CVD (secondary prevention)

Benefits of primary prevention unknown

633
Q

What do PPARs do?

A

Peroxisome proliferator activates receptors

Alpha in liver
Gamma in adipose tissue

Affecting them-> decreased plasma fatty acids and triglycerides

634
Q

What is ezetimibe?

A

Inhibits cholesterol absorption from SI (CETP inhibitor)

Absorbed then activated as glucuronide

Added to simvastatin

635
Q

What is CETP?

A

Cholesterol ester transfer protein

Involved in reverse cholesterol transport

636
Q

What are the problems with cept inhibitors?

A

Adverse effects of torcetrapib “off target”

? due to activation of aldosterone synthesis leading to raised BP

Other “rapibs” do not have same effect

637
Q

What is PCSK9?

A

Secreted inhibitor of the LDLR

Potential candidates for PCSK9 inhibitory therapy= familial hypercholesterolemia

Approved in UK and USA

638
Q

What are the clinical uses of NSAIDs?

A

Analgesic (for mild-to-moderate pain)
E.g. toothache, headache, backache postop pain, dysmenorrhea

Antipyretic (reduction of fever)
E.g. influenza

Anti-inflammatory
E.g. Rheumatoid arthritis, osteoarthritis, musculo-skeletal inflam, soft tissue injuries, gout

639
Q

How do NSAIDs affect prostanoids?

A

Inhibit synthesis of prostanoids

640
Q

How do NSAIDs inhibit prostanoid synthesis?

A

Inhibit cyclo-oxygenases (COXS 1 and 2)

Prevent formation of prostaglanding H2 from arachidonic acid from phospholipid membranes

Means they aren't converted (with specific synthases) into:
Prostacyclin
Prostaglandin L2
Prostaglandin E2
Prostaglandin F2
Prostaglandin D2
Thromboxane A2
641
Q

What are prostanoids?

A
E.g.s= 
Prostacyclin
Prostaglandin L2
Prostaglandin E2
Prostaglandin F2
Prostaglandin D2
Thromboxane A2

Lipid mediators derived from arachidonic acid

Widely distributed
Not stored pre-formed
Receptor-mediated

642
Q

What are the main isoforms of COX? How does this affect drugs?

A

2 main isoforms

Most NSAIDs reversibly inhibit both isoforms to varying degrees (e.g. ibuprofen)

Some selectively inhibit one (e.g. coxib family like celecoxib reversibly inhibits COX-2)

643
Q

What are the known receptors of prostanoids?

A

10 known

DP1, DP2, EP1, EP2, EP3, EP4, FP, IP1,IP2, TP

All GPCRs but have effects independent of G proteins

644
Q

What does PGE2 normally do?

A

Can activate 4 receptors

cAMP-dependent and independent downstream mechanisms

Gastroprotection
Renal salt and water homeostasis
Bronchodilation
Vasoregulation (dilation and constriction depending on receptor activated)

Unwanted actions

645
Q

What are the unwanted effects of PGE2?

A
Increased pain perception
Thermoregulation
Acute inflammatory response 
Immune responses
Tumorigenesis
Inhibition of apoptosis
646
Q

How do PGE2 analogues lower the pain threshold? How can you prevent this?

A

Stimulation of PG receptors sensitizes the nociceptors which cause pain both acutely and chronically

Co-injection of a COX-2 inhibitor prevents or reduces the duration of prolonged pain

647
Q

How do prostanoids lower the pain threshold?

A

Mechanisms are not entirely clear

Possibly:
EP1 receptors
EP4 receptors (in periphery and spine)
Endocannabinoids (neuromodulators in thalamus, spine and periphery)

Not mutually exclusive

648
Q

Why is PGE2 pyrogenic?

A

PGE2 stimulates hypothalamic neurones initiating a rise in body temperature

PGE2 in CSF-> increase temp

649
Q

What is the role of PGE2 in inflammation?

A

PGE2 binds to EP3 Rs on mast cell-> Gi signalling via PLC (IP3) and PI3K (PIP3)

Histamine released (degranulation)
-> increased vascular permeability 

IL-6 from nucleus-> increased leukocyte recruitment

650
Q

What are the desirable physiological actions of PGE2 and prostanoids?

A

Gastroprotection
Renal salt and water homeostasis
Bronchodilation
Vasoregulation (dilation and constriction depending on receptor activated)

651
Q

What is the role of PGE2 in gastric cytoprotection?

A

PGE2 downregulates HCl secretion

PGE2 stimulus mucus and bicarbonate secretion

652
Q

Why do NSAIDs increase the risk of ulceration?

A

PGE2 normally gastric cytoprotective

NSAIDs inhibit PGE2

NB. Celecoxib- few ulcers than conventional NSAIDs

653
Q

How does PGE2 regulate salt and water homeostasis?

A

COX-1 and COX-2 found in kidney

COX-1= collecting duct
COX-2= macula densa and proximal ascening limb
COX-1 and COX-2=

654
Q

How does PGE2 regulate salt and water homeostasis?

A

COX-1 and COX-2 found in kidney

PGE2 increases renal blood flow

Can cause renal toxicity:
Constriction of afferent renal arteriole
Reduction in renal artery flow
Reduced glomerular filtration rate

655
Q

Where are COX-1 and 2 found in kidney?

A

COX-1= collecting duct
COX-2= macula densa and proximal ascening limb
COX-1 and COX-2= glomerulus

656
Q

Why should NSAIDs not be taken by asthmatics?

A

Causes bronchodilation

COX-inhibition favours production of leukotrienes (bronchoconstrictors)

PGE2 seems to be protective normally

657
Q

How do we know that prostanoids involved in vasoregulation?

A

Serious unwanted CV effects from NSAIDs (prostanoid action removed)

658
Q

What are the unwanted CV effects of NSAIDs?

A

Vasoconstriction
Salt and water retention
Reduced effect of antihypertensives

Hypertension
Myocardial infarction
Stroke

659
Q

Are the CV effects of COX-2 inhibitors or conventional NSAIDs worse?

A

COX-2 inhibitors

660
Q

Why do COX-2 inhibitors have CV effects?

A

Affect vascular enothelial and smooth muscle cells-> enhanced probability of coronary atherothrombosis

Affect cardiomyocytes-> increased risk of heart failure

affect kidney-> increased LT CV risk and increased risk of heart failure

661
Q

True or false; all NSAIDs increase risk of GI bleeds and CVS events?

A

True

662
Q

How does NSAID use vary with analagesis use and anti-inflammatory use?

A

ANALGESIC USE
Usually occasional
Relatively low risk of side effects

ANTI-INFLAM USE
Often sustained
Higher doses
Relatively high risk of side effects

663
Q

How can you limit the GI effects of NSAIDS?

A

COX-2 selective NSAIDs (but cardio=worse)

Topical application

Minimise NSAID use in patients with history of GI ulceration

Treat H pylori if present

Administer with omeprazole or other proton pump inhibitor

Minimise NSAID use in patients with other risk factors and reduce risk factors where possible e.g. alcohol consumption, anticoagulant or glucocorticoid steroid use

664
Q

How does aspirin work?

A

Selective for COX-1
Binds irreversibly

Has anti-inflammatory, analgesis and anti-pyretic actions

reduces platelet aggregation

665
Q

How do prostanoids affect platelet aggregation?

A

Platelets-> thromboxane A2 = promotes platelet aggregation

Endothelial cells-> prostacylcin (PGI2)= inhibits platelet aggregation

666
Q

How does aspirin affect platelet aggregation?

A

COX-1 makes thromboxane A2 so inhibition of this by aspirin-> reduced platelet aggregation
No nucleus: no resynthesis of COX-1

NB. PGI2 (inhibits platelet aggregation) synthesised by COX-1 and 2
Nucelated: replenished COX-1 and COX-2

667
Q

Anti-platelet actions of aspirin are due to….

A

Very high degree of COX-1 inhibition which effectively suppresses TxA2 production by platelets
Covalent binding which permanently inhibits platelet COX-1
Relatively low capacity to inhibit COX-2
Need low dose to allow endothelial resynthesis of COX-2

668
Q

What are the major SEs of aspirin? Why are they more likely than with other NSAIDs?

A

Gastric irritation and ulceration
Bronchospasm in sensitive asthmatics
Prolonged bleeding times
Nephrotoxicity

Side effects are more likely with aspirin than other NSAIDS because it inhibits COX covalently and irreversibly, rather than its selectively for COX-1

669
Q

Why is paracetamol not an NSAID?

A

Doesn’t have anti-inflam effect

Is a good analgesic (mild-to-mod) and anti-pyretic

NB. legislation exists

670
Q

Why can paracetamol cause irreversible liver failure?

A

If glutathione is depleted-> the metabolite oxidises thiol groups of key hepatic enzymes-> cell death

671
Q

What is the antidote for paracetamol poisoning?

A

Add compound with -SH groups (usually IV acetylcysteine, sometimes oral methionine)

Needs to be administered early

672
Q
Aspirin is unique amongst NSAIDS because: 
It has no effect on COX-1
It has no effect on COX-2
It binds covalently to COX enzymes
It binds covalently to TP receptors  
It causes gastric ulceration
A

ANSWER= It binds covalently to COX enzymes

It has no effect on COX-1
(It effectively inhibits COX-1)

It has no effect on COX-2
(It has some effect on COX-2)

It binds covalently to TP receptors
(Untrue)

It causes gastric ulceration
(True but not unique to aspirin)

673
Q
Inhibition of which enzyme will reduce platelet aggregation with fewest side effects? 
COX-1
COX-2
Prostacyclin synthase
Prostaglandin E synthase
Thromboxane A2 synthase
A

ANSWER= Thromboxane A2 synthase (causes platelet aggregation but not much else)

COX-1
(No- rate limiting for multiple prostanoids)
COX-2
(No- rate limiting for multiple prostanoids)

Prostacyclin synthase
(No- prostacyclin reduces platelet aggregation)

Prostaglandin E synthase
(No- multiple complex effects)

674
Q

Assertion: Inhibition of PGI2 synthesis by low dose aspirin decreases the risk of stroke
Because : Decreased PGI2 reduces platelet aggregation

Assertion true, reason true and explains assertion
Assertion true, reason true but does not explain assertion
Assertion true, reason false
Assertion false, reason true
Assertion false, reason false

A

ANSWER= Assertion false, reason false

Synthesis of PGI2 (prostacyclin) is inhibited by low dose aspirin, but it is not this action which decreases the risk of stroke, because PGI2 actually reduces platelet aggregation. It’s the inhibition of thromboxane synthesis

675
Q

Define: nausea

A

Subjective unpleasant sensation in throat and stomach: often precedes vomiting

Often preceded by salivation, sweating and increased HR

676
Q

Define: vomiting

A

Forceful propulsion of stomach contents out of the mouth

Often preceded by salivation, sweating and increased HR

677
Q

Outline the vomiting pathway

A

Deep breath is taken, glottis is closed and larynx is raised to open the upper oesophageal sphincter

Soft palate is elevated to close off posterior nares

Diaphragm is contracted sharply downward-> creates negative pressure in thorax-> facilitates opening of oesophagus and distal oesophageal sphincter

Simultaneously with downward movement of the diaphragm - muscles of adbominal walls are vigorously contracted-> squeezes stomach and elevates intragastric pressure

With pylorus closed and oesophagus relatively open-> route of exit cleared

678
Q

What is the consequence of acute nausea?

A

Interferes with mental and physical activity

679
Q

What is the consequence of chronic nausea?

A

Very debilitating

680
Q

What is the consequence of severe vomiting?

A

Dehydration

Loss of gastric hydrogen and chloride ions may lead to hypochloraemic metabolic alkalosis (raised blood pH)

Contributes to a reduction in renal bicarbonate excretion and an increased in bicarbonate reabsorption accompanied by increased sodium reabsorption in exchange for potassium, leading to the hypokalaemia

681
Q

What components are involved in pathways of nausea/vomiting?

A

CNS= cortex, thalamus, hypothalamus, meninges

Vestibular system= H1 R?, M1 R

GI tract and heart= mechanoR, chemoR, 5-HT R

Chemoreceptor trigger zone (area postrema)= chemoR, D2 R, NK1 R, (5HT3 R)

Vomiting center (nucleus of tractus solitarius)= H1 R, M1 R, NK1 R, (5-HT3 R)

682
Q

Outline the pathway in nausea/vomiting

A

CNS and vestibular system

-> Chemoreceptor trigger zone (area postrema)

CN IX or X (from GI tract and heart to vomiting centre in medulla oblongata)

-> Parasympathetic and motor efferent activity

683
Q

What pathways feed into the vomiting centre?

A

Cortex (via intracerebral projections)

Vestibular system (via projections from vestibular nuclei)

  • Direct to vomiting centre
  • Via chemoreceptor trigger zone (then to vom centre via intracerebral projections)

Peripheral pathways

  • Direct to vomiting venter (via vagus, splanchnic and glosspharyngeal nerves, sympathetic ganglia)
  • Via chemoreceptor trigger zone (via vagus and splanchnic nerves and then to vom centre via intracerebral projections)
684
Q

What is promethazine?

A

A phenothiazine derivative
Mixed receptor antagonist
Used as an anti-emetic

685
Q

What is the mode of action of promethazine?

A

Competitive antagonist at histaminergic (type H1), cholinergic (muscarinic, M) and dopaminergic (type D2) receptors

Order of potency of antagonistic activity: H1> M > D2 receptors

Acts centrally (vestibular nucleus, CTZ, vomiting centre) to block activation of vomiting centre

686
Q

What does promethazine act on?

A

Achm and H1 in vestibular system

D2 in chemoreceptor trigger zone

Achm in vomiting centre

687
Q

How can promethazine be used as an anti-emetic?

A

Used prophylactically for motion sickness (some benefit taken after onset of nausea and vomiting)

Disorders of the labyrinth e.g. Meniere’s disease

Hyperemesis gravidarum

Pre-and post-op (sedative and anti-muscarinic action also useful)

NB. Other uses= relief of allergic symptoms, anaphylactic emergency, night sedation/insomnia

688
Q

What is Meniere’s disease?

A

Inner ear-> balance problems

689
Q

What is hyperemesis gravidarum?

A

Pregnancy complications

Lead to severe sickness

690
Q

What is the pharmacokinetics of promethazine?

A

Administer orally
Onset of action 1-2 hours
Maximum effect circa 4 hours
Duration of action 24 hours

691
Q

What are the unwanted effects of promethazine?

A
Dizziness
Tinnitus
Fatigue
Sedation (‘do not drive or operate machinery')
Excitation in excess
Antimuscarinc side-effects
692
Q

What is metoclopramide (domperidone)?

A

D2 R antagonist

Closely related to phenothiazine group

693
Q

Outline the mode of action of metoclopramide: domperidone

A

Order of antagonistic potency:
D2 > H1 > Muscarinic Rs

Acts centrally, especially at CTZ (D2 receptors)

Prokinetic effects in the GI tract

694
Q

What are the prokinetic effects in the GI tract caused by metoclopramide:domperidone?

A

Increases smooth muscle motility (from oesophagus to small intestine

Accelerated gastric emptying

Accelerates transit of intestinal contents (from duodenum to ileo-coecal valve)

695
Q

What is metoclopramide: domperidone used for?

A

To treat nausea and vomiting associated with:
- Uraemia (severe renal failure)
- Radiation sickness
- GI disorders
- Cancer chemotherapy (high doses) e.g. cisplatin (intractable vomiting)
- Parkinson’s disease treatments which stimulate dopaminergic transmission in CTZ e.g. L-DOPA, DA agonists
(Stays mainly in periphery but little action on trigger zone)

Not effective against motion sickness

696
Q

What are the pharmacokinetic consideration of metoclopramide: domperidone?

A

May be administered orally; rapidly absorbed; extensive first pass metabolism (may also be given i.v.)

Metoclopramide crosses BBB

Crosses placenta

NEED TO TAKE CARE WITH BIOAVAILABILITY OF CO-ADMINISTERED DRUGS

697
Q

How can digoxin and nutrient supply be affected by metoclopramide: domperidone?

A

Affects the bioavailability of co-administered drugs

Absorption and hence effectiveness of digoxin may be reduced (due to the prokinetic effects with the increased transit in the early GIT this can effect other drug absorption)

Nutrient supply may be compromised- especially important in conditions such as diabetes mellitus

698
Q

Outline the unwanted effects of meotclopramide:domperidone?

A

In CNS (Metoclopramide only due to CNS blockade of DA Rs; Domperidone does not cross main BBB)
Drowsiness
Dizziness
Anxiety
Extrapyramidal reactions (children more susceptible than adults- Parkinsonian-like syndrome)

In the endocrine system (stimulates PL release)
Hyperprolactinaemia
Galactorrhoea
Disorders of menstruation

699
Q

What are the pharmacokinetic considerations of metoclopramide: domperidone?

A

When administered orally – rapidly absorbed but effective concentration entering the body is decreased by extensive 1st pass metabolism

May be given intravenously

Crosses the BBB (Metoclopramide)

Crosses the placenta

700
Q

What is the mode of action for hyoscine?

A

Muscarinic receptor antagonist

Order of antagonistic potency: Muscarinic >D2 = H1 receptors

Acts centrally, especially in the vestibular nuclei and CTZ to block activation of vomiting centre

701
Q

How can hyoscine be used as an anti-emetic?

A

Prevention of motion sickness
Has little effects once nausea/ emesis is established
In operative pre-medication

702
Q

Where does hyoscine act on the nausea pathway?

A

Achm in vestibular system
D2 (central) in chemoreceptor trigger zone
Achm in vomiting centre

703
Q

What are the unwanted effects of hyoscine?

A
Typical anti-muscarinic SEs:
Drowsiness
Dry mouth
Cycloplegia
Mydriasis
704
Q

What is cycloplegia?

A

Paralysis of the ciliary muscle of the eye resulting in a loss of accommodation

705
Q

What is mydriasis?

A

Dilation of the pupil of the eye

706
Q

What are the pharmacokinetic considerations of hyoscine?

A

Can be administered orally (peak effect in 1-2 hours), intravenous, transdermally

707
Q

What is the mode of action of ondansetron?

A

Acts to block transmission in visceral afferents and CTZ

708
Q

How is ondansetron used as an anti-emetic?

A

Main use in preventing anticancer drug-induced vomiting, especially cisplatin
Radiotherapy-induced sickness
Post-operative nausea and vomiting

709
Q

What is ondansetron?

A

Serotonin R (5-HT3) antagonists

710
Q

Where does ondansetron act in the nausea pathway?

A

D2 (central) in chemoreceptor trigger zone

5HT3 receptors in GI tract

711
Q

What are the pharmacokinetic considerations of ondansetron?

A

Administer orally
Well absorbed
Excreted in urine

712
Q

What are the unwanted effects of ondansetron?

A

Headache
Sensation of flushing and warmth
Increased large bowel transit time (constipation)

713
Q

Why/when is ondansetron used in combination with corticosteroids?

A

Use alone- efficacy may wear off

Corticosteroids, such as dexamethasone, may be used in combination with 5-HT3 receptor antagonists for high or moderately high emetogenic chemotherapy (without= low)

Improved efficacy of combined therapy may be due to anti-inflammatory properties of corticosteroids

714
Q

How are cannabinoids used as anti-emetics?

A

THC isolated from Marijuana or the synthetic agent Nabilone

Effective at treating emesis from anti-cancer drugs which other antiemetic’s are not very effective against e.g. Cisplatin

Act at a number of cites within CNS via the CB1 Rs (located pre-synaptically and decrease the release of NTs associated with triggering the vomiting process)

Also inhibit prostaglandin synthesis which has been implicated in emesis from anti-cancer drugs

715
Q

What are the major forms of IBD?

A

Ulcerative colitis
Crohn’s disease
(Distinction incomplete in 10% patients- indeterminate colitis)

716
Q

What are the risk factors for IBD?

A

Genetic predisposition (163 loci- particularly Caucasian europeans)

Environmental factors= smoking (especially CD), diet/obesity, gut microbioma

Obesity (CD not UC)

717
Q

How do gut flora and autoimmune diseases relate?

A

Defective interaction between mucosal immune system and gut flora

10x more gut bacteria than host cells

Complex interplay between host and microbes-> disrupted innate immunity and impaired clearance-> prof-inflammatory compensatory responses-> granuloma formation and physical damage

718
Q

Outline the autoimmune background to Crohn’s disease

A

Th1-mediated e.g. IFNγ, TNFα, IL-17, IL-23

Florid T cell expansion
Defective T cell apoptosis

719
Q

Outline the autoimmune background to UC

A

Th2-mediated e.g. IL-5, IL-13

Limited clonal expansion
Normal T cell apoptosis

720
Q

What gut layers are affected in CD?

A

All layers

721
Q

What gut layers are affected in UC?

A

Mucosa/submucosa

722
Q

What regions are affected in CD?

A

Any part of GI

Patchy inflamed areas

723
Q

What regions are affected in UC?

A

Rectum, spreading proximally

Continuous inflamed areas

724
Q

Are abcesses/ fissures/ fistulae more common in CD or UC?

A

Common in CD

Uncommon in UC

725
Q

Can surgery cure CD or UC?

A

Not always CD

Can cure UC

726
Q

What are the clinical features of IBD?

A

Apthous ulcers

Anaemia, uveitis, fevers, sweats, jaundice

Primary sclerosing cholangitis

Abdominal pain

Right iliac fossa mass/pain

Arthritis, arthraligia

Weight loss

Skin rash (pyoderma, erythema nodosum)

Diarrhoea, blood mucus

727
Q

What can be used as IBD therapies?

A

SUPPORTIVE (for acutely sick patient)
Fluids (electrolyte replacement)
Blood transfusion/oral iron
Nutritional support

SYMPTOMATIC: ACTIVE DISEASE
Glucocorticoids e.g. prednisolone
Aminosalicylates e.g. mesalazine
Immunosuppressives e.g. azathiprine

SYMPTOMATIC: PREVENTION OF REMISSION
Glucocorticoids
Aminosalicylates
Immunosuppressives

POTETIALLY CURATIVE
Microbiome manipulation
Biologic therapies

728
Q

Why are aminosalicylates used in UC and CD?

A

Anti-inflammatory

UC
First line in inducing and maintaining remission
Good evidence base

CD
Literature unclear
Ineffective in inducing remission
Less clear cut than utility in UC

729
Q

What are aminosalicylates?

A

Anti-inflammatory

Mesalazine or 5-ASA (amionsalicyclic acid)

Olsalazine (2 linked 5-ASA molecules)

730
Q

How do aminosalicylates work as anti-inflammatory drugs?

A

Inhibition of IL-1, TNF-𝛼, and platelet activating factor (PAF)

Decreased antibody secretion

Non-specific cytokine inhibition

Reduce cell migration (macrophages)

Localised inhibition of immune responses

731
Q

Why do you need to 5-ASA with other drugs?

A

5-ASA derivatives activated by gut flora

Depends where they are absorbed

Olsalazine metabolised by colonic flora and absorpsed by colon

732
Q

What is better topically- 5-ASA or steroids in IBD?

A

Topical 5-ASA beter than topical steroids in inducing UC remission

Combined oral and topical 5-ASA better at inducing remission in UC than oral 5-ASA alone

733
Q

Outline the use of glucocorticoids in IBD

A

UC
Use of glucocorticoids in decline
Can be used topically (enema) or IV if very severe
Aminosalicylates are better

CD
GCs remain drugs of choice for inducing remission
Likely to get SEs if used to maintain remission

734
Q

Give examples of glucocorticoids

A

Prednisolone
Fluticasone
Budesonide

735
Q

Why are glucocorticoids used in IBD?

A

Powerful anti-inflammatory and immunosuppressive drugs

Derived from the hormone cortisol

Activate IC glucocorticoid Rs which can then act as positive or negative transcription factors

When given systemically- chronic GCs-> many unwanted effected

736
Q

What are the strategies to minimise unwanted effects of GCs?

A

Administer topically- fluid or foam enemas or suppositories

Use a low dose in combination with another drug

Use an oral or topically administered drug with high hepatic first pass metabolism e.g. Budesonide so little escapes into the systemic circulation

737
Q

Are GCs or budesonides better at inducing remission in active CD?

A

Standard oral glucocorticoids better than budesonide at inducing remission in active CD

738
Q

Should GCs be used to treat UC or CD?

A

Case by case basis

Avoid in UC

Used in CD - Budesonide preferred if disease is mild

739
Q

What immunosuppressive agents are used to treat IBD?

A

Azathioprine and 6-mercaptopurine

  • No advantage over placebo in active CD
  • Some success in UC

Methotrexate
- Has demonstrable efficacy in some IBD patients

Cyclosporin
- Useful in severe UC only

740
Q

What is azathioprine?

A

Immunosuppressive pro-drug (purine antagonist)

Activated by gut flora to 6-mercaptopurine

Mainly used to maintain remission in CD

May allow reduction in glucocorticoids

Slow onset- 3 to 4 months treatment for clinical benefit

741
Q

What does 6-MP do?

A

Formed by azathioprine (prodrug) breaking down

Interferes with DNA synthesis and cell replication

It impairs:

  • Cell- and antibody-mediated immune responses
  • Lymphocyte proliferation
  • Mononuclear cell infiltration
  • Synthesis of antibodies

It enhances:
- T cell apoptosis

742
Q

What are the unwanted effects of 6-MP?

A

Nearly 10% patients have to stop treatment because of SEs

Pancreatitis
Bone marrow suppression
Hepatotoxicity
Increased risk (4 fold) of lymphoma and skin cancer

743
Q

What is 6-MeMP?

A

Hepatotoxic

Inactive form of of 6-MP (with TPMT)

744
Q

What converts 6-MP to 6-TU (inactive)?

A

XO (xanthine oxidase)

Allopurinol inhibits XO

745
Q

Outline the pathway from azathioprine to its derivatives?

A

Azathioprine-> 6-MP-> 6-TIMP

-> w/ TPMT-> 6-MeMP (inhibition of de novo puring synthesis)
OR
-> w/ IMPD/GMPS/kinases -> 6-TGN (incorporation into DNA)

746
Q

What does 6-TGN do?

A

Beneficial but also causes myelosuppression

747
Q

What is methotrexate used for in IBD?

A

Acts as folate antagonist

Reduces synthesis of thymidine and other purines

Not widely used as monotherapy due to SEs

Demonstrable effect in CD (inducing and maintaining remission)

748
Q

What biologic therapies are used in IBD?

A

Potentially curative

Anti-TNFα e.g. Infliximab
Anti-α-4-integrin e.g. Natalizumab

749
Q

Are probiotics useful in IBD?

A

No evidence for probiotics in CD

Some evidence for probiotics for maintenance of remission in UC (as effective as 5-ASA in inducing and sustaining remission in UC)

750
Q

What is FMT in IBD?

A

Faecal microbiota replacement

Insufficient evidence

751
Q

Why is Rifaximin used as antibiotic treatment in IBD?

A

To manipulate the microbiome

Interferes with bacterial transcription by binding to RNA polymerase

Induces and sustains remission in moderate CD

May be beneficial in UC
- REDUCES INFLAMMATORY MEDIATOR mRNA IN UC

752
Q

What biological therapies are approved for use in IBD?

A

Anti- TNFα antibodies
E.g. Infliximab (iv)
Other antibodies effective but have more SEs

753
Q

Is anti-tumour necrosis factor alpha used to treat IBD?

A

Successfully in treatment of CD (potentially curative)

Some evidence of effectiveness in UC

754
Q

How do anti-TNFα antibodies in IBD work?

A

Anti-TNFα reduces activation of TNF α receptors in the gut

Reduces downstream inflammatory events

Also binds to membrane associated TNFα

Induces cytolysis of cells expressing TNFα

Promotes apoptosis of activated T cells

755
Q

What is the pharmacokinetics of infliximab?

A

Infliximab given IV

Very long half-life (9.5 days)

Benefits can last for 30 weeks after a single infusion

Most patients relapse after 8 – 12 weeks

Therefore repeat infusion every 8 weeks

756
Q

What are the problems of infliximab?

A

Up to 50% responders lose response within 3 years due to production of anti-drug antibodies and increased drug clearance

Attempts being made to optimise dosing regimens

757
Q

What are the adverse effects of infliximab?

A

TUBERCULOSIS
4x to 5x increase in incidence
Also risk of reactivating dormant TB

SEPTICAEMIA
Increased risk

HEART FAILURE
Worsening

DEMYELINATING DISEASE
Increased risk

MALIGNANCY
Increased risk

Can be immunogenic- azothiaprine reduces risk, but raises TB / maligancy risk

758
Q

When should infliximab be used?

A

Early use better than last resort

Combined infliximab and azathioprine therapy may be more effective than antibody alone

759
Q

In IBD, budesonide causes fewer unwanted systemic effects than prednisolone because:

a. It can be administered topically
b. It can be co-administered with another drug
c. It has a higher potency at therapeutic doses
d. It has a lower potency at therapeutic doses
e. It is metabolised and inactivated locally

A

e. It is metabolised and inactivated locally

  • -
    a. It can be administered topically (True but not unique to budesonide)
    b. It can be co-administered with another drug (True but not unique to budesonide)
    c. It has a higher potency at therapeutic doses (Potencies are similar)
    d. It has a lower potency at therapeutic doses (Potencies are similar)
760
Q

The mechanism of action of Azathioprine in IBD:

a. Interferes with purine biosynthesis
b. Is a direct reduction of protein synthesis in the GI tract
c. Is blocked by co-administration with allopurinol
d. Means that it increases side-effects caused by infliximab
e. Needs activation of the drug by metabolism to 5-ASA

A

a. Interferes with purine biosynthesis

  • -
    b. Is a direct reduction of protein synthesis in the GI tract (it will reduce protein synthesis indirectly)
    c. Is blocked by co-administration with allopurinol (untrue- allopurinol inhibits metabolism)
    d. Means that it increases side-effects caused by infliximab (untrue- it reduces SEs of infliximab)
    e. Needs activation of the drug by metabolism to 5-ASA (untrue- it needs activation to 6-mercaptopurine)
761
Q

What kinds of therapy is used for treating gastric and dodenal ulcers?

A

TRIPLE THERAPY
Antibiotics
Inhibitors of gastric acid secretion
Cytoprotective drugs

Antacids

762
Q

What is peptic ulcer disease?

A

Area of damage to the inner lining of the stomach (gastric ulcer) or upper part of duodenum (duodenal ulcer)

Imbalance of factors which protect or damage GI barrier

763
Q

When is pain felt in a gastric ulcer?

A

Pain at mealtimes, when gastric acid is secreted

764
Q

When is pain felt in a duodenal ulcer?

A

Pain relieved by a meal as pyloric sphincter closes- pain 2-3h after a meal

765
Q

What is more common- duodenal or gastric ulcers?

A

Duodenal:gastric
4:1

766
Q

What is important to protect the GI mucosal barrier?

A

The integrity of the gastrointestinal mucosal barrier is important in maintaining a disease free state

767
Q

What is the use of protective factors in the gut?

A

Lubricate ingested food and protect the stomach and duodenum from attack by acid and enzymes

Mucous from gastric mucosa creates GI mucosal barrier

HCO3- ions trapped in mucous generate a pH6-7 at mucosal surface

Locally produced prostaglandins stimulate mucous and bicarbonate production (paracrine action) and inhibit gastric acid secretion

768
Q

What factors that convert food into chyme can damage the mucosal barrier?

A
PARIETAL CELLS
Acid secretion (isotonic solution of HCl, pH<1) from parietal cells of the oxyntic glands in the gastric mucosa

CHIEF CELLS
pepsinogens from the chief cells which can erode the mucous layer

769
Q

What colour do you stain parietal (oxyntic) cells?

A

Pink

770
Q

What factors may cause damage to mucosal GI barrier?

A

Increased acid and/or decreased bicarbonate production

Decreased thickness of mucosal layer

Increase in pepsin type I (breaks down proteins into smaller peptides)

Decreased mucosal blood flow

Infections with Helicobacter pylori

771
Q

What are the risk factors for peptic ulcers?

A

Genetic predisposition
Stress
Diet, alcohol, smoking

772
Q

What are the aims of treatment of peptic ulcers?

A

Eliminate cause of mucosal damage

Promote ulcer healing

773
Q

Why are antibiotics good drugs for treating peptic ulcers?

A

Eradicate H. pylori

774
Q

Why are inhibitors of gastric acid secretion good drugs for treating peptic ulcers?

A

Prevent gastric acid production

775
Q

Why are cytoprotective drugs good drugs for treating peptic ulcers?

A

Promote healing

776
Q

Why are antacids good drugs for treating peptic ulcers?

A

Neutralise gastric acid

777
Q

What is H. pylori?

A

Gram-negative bacterium

Inflammation in the stomach (gastritis) and ulceration of the stomach or duodenum (peptic ulcer disease) is the result of an infection of the stomach caused by the bacterium

778
Q

How can you see if H. pylori is present in the stomach?

A

Patients swallow urea labelled with an uncommon isotope

In the subsequent 10–30 minutes, the detection of isotope-labelled carbon dioxide in exhaled breath indicates that the urea was split

Urea split if urease (the enzyme that H. pylori uses to metabolize urea) is present in the stomach which means H. pylori bacteria are present

779
Q

Why is antibiotic therapy useful in peptic ulcers?

A

50-80% worldwide are chronically infected

10-20% go on to develop peptic ulcer disease or neoplasia

Most people with duodenal ulcers and gastric ulcers are infected

780
Q

What do we know about methods of transmission of peptic ulcers?

A

Transmitted by saliva

In poor countries= overcrowding, contact with animals and contaminated faeces-> poor hygiene associated with greater transmission (SOCIOECONOMIC CONDITIONS)

781
Q

What is included in “triple therapy” in peptic ulcer treatment?

A
  1. Antibiotics (not just single)
  2. Drugs which reduce gastric acid secretion
  3. Drugs which promote healing
782
Q

What are examples of gastric acid secretion inhibitors?

A

Proton pump inhibitors

Histamine type 2 (H2) receptor antagonists

Anti-muscarinics

783
Q

What is SIH?

A

Somatostatin inhibiting hormone

Inhibits release of gastrin from G cells and Histamine from H cells

784
Q

What stimuli act on the parietal cells?

A

ACh from the vagal nerve

Gastrin -> releases histamine from the H cells which act on the parietal cells

Prostaglandins E2 and I2 are local hormones (inhibit acid production and promotes a good blood supply)

785
Q

How does gastrin act on parietal cells?

A

Stimulatory hormone produced in the antrum in response to food and vagal PNS-> then releases histamine from the H cells which act on the parietal cells

Gastrin also indirectly increases pepsinogen secretion, stimulates blood flow and increases gastric motility

786
Q

What is the role of secretin in peptic ulcers?

A

Regulates water homeostasis and the pH of the duodenum

787
Q

What is omeprazole?

A

Proton pump inhibitor

Inhibits the basal and stimulated gastric acid secretion from the parietal cells by 90%

788
Q

How does omeprazole work?

A

Irreversible inhibitors of the H+/K+ ATPase

Inactive at neutral pH

As it is a weak base it accumulates in the cannaliculi of parietal cells

789
Q

Why is omeprazole’s action prolonged?

A

As it is a weak base it accumulates in the cannaliculi of parietal cells

This concentrates its action there and prolongs its duration of action (2-3 days) and minimizes its effect on ion pumps elsewhere in the body

790
Q

Define: oesophagitis

A

Inflammation of the lining of the oesophagus

Most cases are due to reflux of stomach acid which irritates the inside lining of the oesophagus

791
Q

What are the uses of proton pump inhibitors?

A

Peptic ulcers which are resistant to H2 antagonists

Component of triple therapy

Gasteoesophageal reflux disease (GORD), oesophagitis

Prophylaxis of peptic ulcers in the intensive care setting, and among high-risk patients prescribed aspirin, NSAIDs, as antiplatelets and anticoagulants

792
Q

What are the pharmacokinetics of proton pump inhibitors?

A

Orally active

Administered as an enteric coated slow-release formulation

793
Q

What are the unwanted effects of proton pump inhibitors?

A

Rare (short term use)

Long-term and/or high-dose administration associated with several potential SEs e.g. enteric infections (C. diff), community acquired pneumonia, and hip fracture

794
Q

What drugs act on the histamine type 2 (H2) receptor antagonist?

A

Cimetidine, ranitidine

795
Q

What are cimetidine/ ranitidine?

A

Inhibits gastric acid secretion from the parietal cells by 60%

Less effective at healing ulcers than PPIs

796
Q

How do cimetidine/ranitidine work?

A

Competitive antagonism of H2 histamine receptors

797
Q

What are the pharmacokinetics of cimetidine/ ranitidine?

A

Orally administered, well absorbed

Ranitidine is longer acting than cimetidine

798
Q

What are the unwanted effects of cimetidine/ ranitidine?

A

Rare (dizziness, headache)

Fewer side effects with Ranitidine (Zantac, available OTC)

Relapses likely after withdrawal of treatment, >90% recurrence within 1 year after initial healing

799
Q

Are antimuscarinics useful anti-ulcer drugs?

A

Little use alone

More effective combination therapies

800
Q

Give examples of cyto-protective drugs

A

Drugs that enhance mucosal protection mechanisms and/ or build a physical barrier over the ulcer

Sucralfate
Bismuth chelate
Misoprostol

801
Q

What is sucralfate?

A

Cyto-protective drug

Polymer containing aluminium hydroxide and sucrose octa-sulphate

802
Q

How does sucralfate act?

A

Acquires a strong -vecharge in an acid environment

Binds to positively charged groups in large molecules (proteins, glycoproteins) resulting in gel-like complexes

-> coat and protect the ulcer, limit H+ diffusion and pepsin degradation of mucus

Increase prostaglandins, mucous and HCO3- secretion and reduces the number of H. pylori

803
Q

What are the unwanted effects of sucralfate?

A

Most of the orally administered Sucralfate remains in the GIT

May cause constipation or reduced absorption of some other drugs e.g. antibiotics and digoxin

804
Q

What is bismuth chelate (pepto-bismol)?

A

Acts like sucralfate

Used in triple therapy (where resistance to drugs has been shown)

Has anti-inflammatory actions in the stomach

Weak anti-biotic and antacid properties

805
Q

What is misoprostol?

A

Stable prostaglandin analogue

Orally active

Co-prescribed with chronically used oral NSAIDs

806
Q

How does misoprostol act?

A

Mimics the action of locally produced PG to maintain the gastroduodenal mucosal barrier

807
Q

Why is misoprostol used with NSAIDs?

A

Misoprostol= stable prostaglandin analogue

NSAIDs block the COX enzyme required for PG synthesis from arachidonic acid

Therefore, there is a reduction in the natural factors that inhibit gastric acid secretion and stimulate mucus and HCO3- production

808
Q

What are the unwanted effects of misoprostol?

A

Diarrhoea, abdominal cramps, uterine contractions (not to given during pregnancy)

809
Q

What are antacids?

A

Mainly salts of Na+, Al3+ and Mg2+

May be effective in reducing duodenal ulcer recurrence rates

810
Q

What speed of effects do the following have?
Sodium bicarbonate
Aluminium hydroxide
Magnesium trisilicate

A

Sodium bicarbonate has rapid effects

Aluminium hydroxide and magnesium trisilicate have slower actions

811
Q

How do antacids work?

A

Neutralise acid, raises gastric pH, reduces pepsin activity

812
Q

How are antacids administered?

A

Taken orally- primarily used for non-ulcer dyspepsia (OTC)

813
Q

What are the problems associated with triple combination therapy?

A

Compliance
Resistance to antibiotics (may be superseded by vaccination)
Adverse response to alcohol (metronidazole interferes with alcohol metabolism)

814
Q

Give examples of triple combination

A

1
Metronidazole/amoxycillin
Clarithromycin
PPI

2
H2 R antagonist
Clarithromycin
Bismuth

815
Q

What is GORD/GERD?

A

Stomach and duodenal contents reflux into the oesophagus (oesophagitis)

OCCASIONAL
Occasional and uncomplicated GERD
Heart burn, may treat by self medication with antacids and H2 antagonists (OTC)

CHRONIC
Chronically may progress to pre-malignant mucosal cells and potentially oesophageal adenocarcinoma

816
Q

How can GERD be treated?

A

PPIs (drug of choice) or H2 antagonists (less effective)

Combine with drugs that increase gastric motility and emptying of the stomach e.g. Dopamine D2 receptor antagonist (metoclopramide)

817
Q

What is an adverse drug event?

A

Preventable or unpredicted medication event- with harm to patient

818
Q

Why is methotrexate important in adverse drug events?

A

High doses for treatment of cancers
Low doses= rheumatoid arthritis
Adverse events happen when given too much or too frequently

819
Q

What percentage of ADRs are preventable?

A

30-60%

Substantial morbidity and mortality

820
Q

How are ADRs classified?

A

Onset
Severity
Type

821
Q

What are the possible onsets for ADRs?

A
Acute= within 1 hour, e.g. anaphylaxis
Sub-acute= 1 to 24 hours
Latent= >2 days
822
Q

What are the possible severities of ADRs?

A
Mild= requires no change in therapy
Moderate= requires change in therapy, additional treatment, hospitalisation 
Severe= disabling or life-threatening
823
Q

What can severe ADR cause?

A
Results in death
Life-threatening
Requires or prolongs hospitalisation
Causes disability
Causes congenital anomalies
Requires intervention to prevent permanent injury
824
Q

What is Type A ADR?

A

70% of adverse reactions

Extension of pharmacologic effect
Usually predictable and dose dependent
E.g. atenolol and heart block, anticholinergics and dry mouth, NSAIDs and peptic ulcer

825
Q

What is the ADR profile of paracetamol?

A

Up to threshold= very safe drug

Then toxicity increases very rapidly and mainly effects live

826
Q

What is the ADR profile of digoxin?

A

Toxicity at any dose, not a threshold

827
Q

What is Type B ADR?

A

Idiosyncratic or immunologic reactions

Includes allergy and “pseudoallergy”

Rare (even very rare) and unpredictable

E.g., chloramphenicol and aplastic anemia, ACE inhibitors and angioedema

828
Q

What is Type C ADR?

A

Associated with long-term use

Involves dose accumulation

E.g., methotrexate and liver fibrosis, antimalarials and ocular toxicity

829
Q

What is Type D ADR?

A

Delayed effects (sometimes dose independent)

Carcinogenicity (e.g. immunosuppressants)

Teratogenicity (e.g. thalidomide)

830
Q

What is Type E ADR?

A

Withdrawal reactions= Opiates, benzodiazepines, corticosteroids

Rebound reactions= Clonidine, beta-blockers, corticosteroids

“Adaptive” reactions= Neuroleptics (major tranquillisers)

831
Q

Why is clonidine withdrawal so dangerous?

A

Rebound effect

Potent anti hypertensive
Makes you drowsy and tired
Miss dosses and bp suddenly increases (can lead to strokes or death)
Missing one dose now manageable

Before 170/10
During 145/90
After 220/130

832
Q

What is the ABCDE classification of adverse drug reactions?

A
Augmented pharmacological effect
Bizarre
Chronic
Delayed
End-of-treatment
833
Q

What are the kinds of allergy classes?

A

Type I-IV

834
Q

What is Type I of allergic reactions?

A

Immediate, anaphylactic (IgE)

E.g. anaphylaxis with penicillins

835
Q

What is Type II of allergic reactions?

A
Cytotoxic antibody (IgG, IgM)
E.g. methyldopa and hemolytic anemia
836
Q

What is Type III of allergic reactions?

A

Serum sickness (IgG, IgM)
antigen-antibody complex
E.g. procainamide-induced lupus

837
Q

What is Type IV of allergic reactions?

A
Delayed hypersensitivity (T cell)
E.g. contact dermatitis (more common)
838
Q

What are pseudoallergies?

A

Similar presentation to a true allergy

Due to different causes

May be due to alterations in the metabolism of histamine

Can be the cause of some forms of food intolerance

839
Q

How are Aspirin/NSAIDs and bronchospasm an example of pseudoallergies?

A

Happens mostly to asthmatics
Blocking COOX pathways which usually makes prostaglandins
AA converted to make leukotrienes= pro inflam-> bronchospasm

840
Q

How is ACE inhibition and cough/angioedema an example of pseudoallergies?

A

ACE inhibitors stop breakdown of inflammatory peptides e.g. bradykinin particularly in lung

Accumulated can -> cough by acting on sensory nerves of lung

841
Q

What are the common causes of ADRs?

A
Antibiotics
Antineoplastics
Anticoagulants
Cardiovascular drugs
Hypoglycemics
Antihypertensives
NSAID/Analgesics
CNS drugs
842
Q

Why are ADRs increasing in frequency?

A

Increasing polypharmacy

So more people having interactions

843
Q

How are ADRs detected?

A

SUBJECTIVE REPORT
Patient complaint

OBJECTIVE REPORT
Direct observation of event
Abnormal findings
Physical examination
Laboratory test
Diagnostic procedure
844
Q

Why are rare events not usually detected before drug is marketed?

A

Need a lot of patients to see adverse reactions

845
Q

What is the yellow card scheme?

A

Voluntary
Used by doctors, dentists, nurses, coroners and pharmacists

For established drugs= only report serious adverse reaction
For ‘black triangle’ drugs (newly licensed)= report any suspected adverse reactions

846
Q

Why is it hard to calculate drug-drug interaction incidence?

A

True incidence difficult to determine

Data for drug-related hospital admissions do not separate out drug interactions (focus on ADRs)

Lack of availability of comprehensive databases

Difficulty in assessing OTC and herbal drug therapy use

Difficulty in determining contribution of drug interaction in complicated patients

847
Q

What are the pharmacodynamic drug interactions?

A

Additive, synergistic, or antagonistic effects from co-administration of two or more drugs

848
Q

Give an example of synergistic drug interactions

A

Antibiotics

849
Q

Give an example of overlapping toxicities drug interactions

A

Overlapping toxicities - ethanol and benzodiazepines

850
Q

Give an example of antagonistic drug interactions

A

Anticholinergic medications (amitriptyline and acetylcholinesterase inhibitors)

851
Q

What are pharmacokinetic drug interactions?

A

Alteration in absorption
Protein binding effects
Changes in drug metabolism
Alteration in elimination

852
Q

What is chelation?

A

Irreversible binding of drugs in the GI tract
So can’t be absorbed

E.g. Tetracyclines, quinolone antibiotics - ferrous sulfate (Fe+2), antacids (Al+3, Ca+2, Mg+2), dairy products (Ca+2)

853
Q

How do protein binding interactions lead to competition in drug reactions?

A

Competition between drugs for protein or tissue binding sites

Increase in free (unbound) concentration may lead to enhanced pharmacological effect

Metabolism interactions

854
Q

What happens in phase 1 and 2 in drug metabolism and elimination?

A

Phase 1= original drug altered but then cleared (-> liver/kidney or to phase 2)

Phase 2= altered once and then again before being excreted by kidney (-> kidney)

Also could be directly excreted unchanged by kidney

855
Q

What happens to the drug before the kidney?

A

Converted from lipid to water soluble so easier to get rid of in the kidney

856
Q

What happens in phase I drug metabolism?

A

Oxidation
Reduction
Hydrolysis

857
Q

What happens in phase II drug metabolism?

A

Conjugation

Glucuronidation
Sulphation
Acetylation

858
Q

What happens to drug metabolism with co-administration of other drugs?

A

Inhibited or enhanced

CYP 450 system= most extensively studied

CYP3A4, CYP2D6, CYP1A2, CYP2B6, CYP2C9, CYP2C19 and others

859
Q

What are CYP 450 substrates metabolised by?

A
Single isozyme (predominantly)
Multiple isozymes
860
Q

What drugs have CYP 450 metabolised by a single isozyme?

A

Few examples of clinically used drugs

Examples of drugs used primarily in research on drug interactions

861
Q

What drugs have CYP 450 metabolised by a multiple isozymes?

A

Most drugs metabolized by more than one isozyme

E.g. Imipramine: CYP2D6, CYP1A2, CYP3A4, CYP2C19

862
Q

What happens if you co-administer a CYP 450 substrate with CYP 450 inhibitor?

A

If co-administered with CYP450 inhibitor, some isozymes may “pick up slack” for inhibited isozyme

I.e. block one subtype of enzyme others will kick in and start metabolising the drug

863
Q

List examples of CYP 450 inhibitors

A

Cimetidine
Erythromycin and related antibiotics
Ketoconazole etc
Ciprofloxacin and related antibiotics

Ritonavir and other HIV drugs
Fluoxetine and other SSRIs
Grapefruit juice

864
Q

List examples of CYP 450 inducers

A
Rifampicin
Carbamazepine
(Phenobarbitone)
(Phenytoin)
St John’s wort (hypericin)

Many inducers in plants and vegetables which may have affect on drug metabolism (not known in much detail)

865
Q

What is the difference in time between inhibition and induction? Why?

A
Inhibition= very rapid (hours)
Induction= hours/days becuase need time for transcription and translation
866
Q

Where do drug elimination interactions usually happen?

A

Almost always in renal tubule

867
Q

What kind of drug elimination interaction happens between probenecid and pencillin?

A

Good

868
Q

What kind of drug elimination interaction happens between lithium and thiazides?

A

Bad

869
Q

Outline the drug elimination interaction between lithium and thiazides

A

Thiazide reduces clearance so toxic accumulation in blood

Increasing secretion of sodium salt but lithium is retained

870
Q

Why is levodopa & carbidopa a deliberate interaction?

A

Allows lower doses to be used because not broken down in periphery

871
Q

Why is ACE inhibitors & thiazides a deliberate interaction?

A

Enhance each others antihypertensive effects

872
Q

Why is penicillins &gentamicin a deliberate interaction?

A

Severe staphylococci infections prevented

873
Q

Why is salbutamol & ipratropium a deliberate interaction?

A

Treatment of asthma and COPD

874
Q

What are opiates?

A

An alkaloid derived from the poppy

Natural product

Papaver somniferum

875
Q

Are opioids synthetic?

A

Can be natural, synthetic or semi-synthetic

876
Q

What part of morphine contributes to analgesia? Why? How can it be made into an antagonist?

A

Tertiary form of nitrogen permits receptor anchoring (binds drug to R)

Can be made into antagonist if you extend side chain to 3+ carbons (determines if you can activate R)

(Quaternary nitrogen-> decreased analgesia because can’t get into CNS)

877
Q

How is morphine altered to become codeine?

A

Add methyl to hydroxyl group at position 3

Required for binding

Codeine is a prodrug

Much less potent than morphine

878
Q

How is morphine altered to become heroin? Why is this important?

A

Acetylated (oxidised) hydroxyl groups 3 and 6

Diacetyl morphine-> increases lipid solubility (penetrates tissues much better)
Converted to morphine in the tissue

879
Q

What are the similarities between the structure of methadone and morphine?

A

Tertiary nitrogen remains
Phenyl group
Quaternary carbon

Look different but have same key features

880
Q

What are the similarities between the structure of fentanyl and morphine?

A

Tertiary nitrogen remains
Phenyl group
NO quaternary carbon (tertiary)

Make it more powerful

881
Q

Why is the oral route of morphine not very efficient?

A

Orally (but not well absorbed from stomach because remains mostly ionised= weak base)

Relatively well absorbed from SI

Heavily metabolised in liver (lots of first pass)

Only about 20% gets into blood

882
Q

What is the pKa of most opioids?

A

> 8

Weak bases

883
Q

How do you ensure high bioavailability of morphine?

A

Inject
IV

Still distributes to brain relatively slowly because not great lipid solubility

884
Q

Which opioids are more/less lipid soluble?

A

MOST-> LEAST
Methadone/fentanyl
Heroin
Morphine

NB. More lipid soluble- more potent

885
Q

What is morphine metabolised to? Why is this important?

A

Various metabolites
Including Morphine-6- glucuronide (10% - active metabolite)

Length of action prolonged by active metabolites

886
Q

Why is fentanyl used as an analgesic and methadone used to help wean people off heroin?

A

Fentanyl is metabolised very quickly (fast onset, quickly cleared)

Methadone is slowly metabolised (prolonged effects, long acting)

887
Q

How much codeine is metabolised to morphine?

A

5-10%

888
Q

How is codeine metabolised?

A

CYP2D6 metabolism is slow but converts codeine to morphine i.e. codeine is a prodrug for morphine

889
Q

What deactivates codeine?

A

CYP3A4

890
Q

Why do some people not respond well to codeine?

A

2D6 polymorphism

891
Q

What metabolises morphine?

A

Morphine is the major exception- metabolised by uridine 5 diphosphate glucoronosyltransferase

892
Q

How do opioids work?

A

Act via specific opioid receptors (mu, delta, kappa)

893
Q

What are examples of endogenous opioid peptides? What do they affect?

A
Endorphins= pain/mood/CVS
Enkephalins= pain/mood/CVS
Dynorphins/neoendorphins= appetite
894
Q

What receptors do endorphins act on? Where are these?

A

Opioid
Mu or delta-> mu

Thalamus, amygdala, n.acc, PAG

895
Q

What receptors do enkephalins act on? Where are these?

A

Opioid
Delta

N.acc, caerebral cortex, amygdala

896
Q

What receptors do dynorphins act on? Where are these?

A

Opioid
Kappa

Appetite

897
Q

What is the cellular mechanism of action of opioids?

A

Depressant

  • > hyperpolarisation (increases K+ efflux )
  • > diminishes inward Ca current
  • > decrease adenylate cyclate activity

All work to decrease activity of cell

898
Q

Why might opioids be taken?

A

Analgesia

Euphoria

Depression of cough centre (anti-tussive)

Depression of respiration (medulla)

Stimulation of chemoreceptor trigger zone (nausea/ vomiting)

Pupillary constriction

GI effects

899
Q

How do opioids have an affect of analgesia?

A

Decrease pain perception
Increase pain tolerance

Maybe central pain perception

900
Q

Where can opioids act on the pain pathway to prevent pain perception?

A

Pain perceived in PNS
Pain relayed to DH
Spinothalamic tracts relay up to brain

Thalamus= mu (sorting of signals)
Cortex= mu, delta (adds emotional response)
901
Q

Outline the pain tolerance pathway

A

Integrating centre (PAG)= mu, kappa

  • Receives + from thalamus
  • Receives +/- from cortex

Info relayed to NRM= delta
- Also info from NRPG (independent of thalamus, essentially reflex)= mu, delta

NRM inhibits DH (decreases sensation of pain)

Locus coeruleus also has negative effect (SNS suppresses sensation of pain- fight/flight)

902
Q

Why is pain tolerance needed?

A

Recognise pain initially (to avoid damage)
But need to depress feelings of pain= tolerance
Too much pain is harmful

903
Q

What information can the DH process?

A

Some is automatic= not processed, just direct inhibitory effect

But structures within SC can be involved in processing info= substantia gelatinosa
Further processes descending info

904
Q

Where can opioids act?

A

NRPG
PAG
Periphery at site of tissue injury

905
Q

Why do opioids cause euphoria?

A

Opiates act at mu receptors
Depress firing rate of GABA acting on VTA
Causes more DA release by NAcc-> euphoria

906
Q

What leads to coughing? Why are opioids used as anti-tussives?

A

Stimulation of mechano-or chemoreceptors (throat, resp passages or stretch Rs in lungs)= respond to irritants

Afferent impulses to cough centre (medulla)
OPIOIDS ACT ON- Ach/NK C-fibres relay to Vagus (stop info to cough centre)

Efferent impulses via parasympathetic and motor nerves to diaphragm, intercostal muscles and lung
OPIOIDS ACT ON - 5HT1A receptors (negative feedback receptors) (stop cough centre)

Increased contraction of diaphragmatic, abdominal and intercostal (ribs) muscles-> noisy expiration (cough)
OPIOIDS HAVE PERIPHERAL EFFECTS on mu-opioid receptors in airway vagal sensory neurons
AND OPIOIDS INHIBIT both eNANC nerve activity and cholinergic contraction of smooth muscles

907
Q

Wy do opioids cause respiratory depression?

A

Most dangerous side effect

Interfere with central chemoreceptors (which respond to PaCO2)
- Disconnect resp control centre from amount of CO2 in blood

Act directly in medullary control centre

  • Pre-Botzinger complex (part that deals with breathing rhythm)
  • Opioids suppress this area
908
Q

Why do opioids cause nausea/vomiting?

A

Activate mu receptors in chemoreceptor trigger zone-> medullary vomiting center-> vomiting reflex

Probably due to interference with GABA (switch off GABA-> feel nauseous)

909
Q

Why do opioids cause miosis?

A

Pinpoint pupils (e.g. in heroin deaths/overdose)

Mu opioid receptors in Edinger-Westphal nucleus

Opioids activate parasympathetic nerve independent of what optic nerve is doing

Probably relates to GABA (switched off- parasymp nerve starts firing like crazy)

910
Q

Why do opioids cause gastrointestinal disturbance?

A

Opioids have significant inhibitory effects on gut-> constipating

ENTERIC NS (controls gut function)
Several opioid receptor types can be demonstrated on myenteric neurons

Both kappa and mu receptor agonists regulate cholinergic transmission in the myenteric plexus

911
Q

Why do opioids cause urticaria?

A

Not well understood

In some people, opioids (with combination of the N-methyl group and the 6-hydroxyl group) cause histamine release

Affects mast cells near skin

PKA activates-> histamine release

(NB. not allergy but looks like it)

912
Q

What determines opioid tolerance? How is it developed?

A

Not PK, this is issue tolerance

Opioids taken chronically will upregulate levels of arrestin in tissue

Arrestin promotes R internalisation (normal process to recycle Rs BUT upregulating-> too many Rs internalised)

Develop tolerance to opioids

913
Q

What is opioid withdrawal associated with?

A
Psychological craving
Physical withdrawal (resembling flu)

Depressant effects in opioid users-> cells upregulate adenylate cyclase to compensate

When withdraw drugs-> left with cells with too much adenylate cyclase (slowly returns to normal)

914
Q

What happens in an opioid overdose?

A

Coma

Respiratory depression

Pin-point pupils

Hypotension

915
Q

How can you treat an opioid overdose?

A

Naloxone (opioid antagonist) i.v.

916
Q

What is the target organ of diuretics?

A

Kidney

917
Q

In the PCT cell, outline ionic composition/channels?

A

Free movement of H2O and Na across apical membrane from lumen into cell

SODIUM
Need to constantly remove sodium from cell to maintain conc gradient
- Na/K exchange (Na out, K in)

WATER
Sodium removal also helps passage of water (back towards blood)
- And helped by oncotic pressure

PARACELLULAR= big gap junctions
Allow movement of H2O, Na, CL, HCO3-

CARBONIC ANHYDRASE
Enzyme to allow movement of bicarb ions from apical to basal sides of cell
- CA on outside of apical surface (ensures bicarb and H ions are converted to CO2 and H2O which can move into cell more easily by diffusion)
- Cytoplasmic CA converts it back to H and HCO3 (bicarb)
- Bicarb then cotransported with sodium back towards blood

AMINO ACIDS
Na/H transporter (Na in, H out)
Coupled with glucose/AA transport at apical membrane into cell

Exporting exogenous proteins also happens

918
Q

How do drugs leave the kidney?

A

Transported across PCT cell

Lost in urine

919
Q

How much of the filtered load is reabsorbed in the PCT?

A

Of the filtered load, up to 70% is reabsorbed in PCT

920
Q

In the DESCENDING LIMB outline ionic composition/channels?

A

Water moves down osmotic gradient

Aquaporins present

921
Q

In the ASCENDING LIMB outline ionic composition/channels?

A

Very little through paracellular route
Pretty much impermeable to water

Move NaCl from one side to other

  • Na/2Cl/K transporter moves ions across cell into blood
  • K also moves in opposite direction into lumen
922
Q

Outline the countercurrent effect

A

PUMP NA FROM ACSCENDING TUBULE
Descending limb-
permeable to water

Ascending limb- impermeable to water

Na leaves the ascending limb and enters medullary interstitium

Fluid in ascending limb decreases in osmolarity

ADJUST OSMOLARITY OF DESCENDING TUBULE
More concentrated
medullary interstitium draws water from the permeable descending limb

Fluid in descending limb
increases in osmolarity

MORE FLUID FLOWS INTO TUBULE
More fluid enters and forces fluid from descending to
ascending limb
- This fluid
has increased in osmolarity due to increased Na+ conc
in the medulla

SECOND ROUND OF NA PUMPING
Ascending limb –
impermeable to water

Na+ leaves the ascending
limb and enters medullary
Interstitium

Fluid in ascending limb
decreases in osmolarity

923
Q

Why is the countercurrent multiplier effect in the LOH important?

A

Drives the reabsorption of water (particularly in PCT)

Why loop diuretics are so powerful

924
Q

In the DCT outline ionic composition/channels?

A

EARLY DCT
Predominant effect driven by NaCl cotransport protein
- Reabsorbed together and transferred across into the blood

LATE DCT
Aldosterone= steroid= most important in driving Na reuptake
- Binds to mineralocorticoid R-> moves to nucleus-> increases production of Na channels and Na/K/ATPase

AQP2 via VP determine water reabsorption

925
Q

In the COLLECTING DUCT outline ionic composition/channels?

A

Aldosterone= steroid= most important in driving Na reuptake
- Binds to mineralocorticoid R-> moves to nucleus-> increases production of Na channels and Na/K/ATPase

AQP2 via VP determine water reabsorption

926
Q

How do diuretics work?

A

Inhibit the reabsorption of Na+ and Cl-
i.e. increase excretion

(1 kind= increases the osmolarity of tubular fluid
i.e. decrease the osmotic gradient across the epithelia)

927
Q

What are the 5 main diuretic classes (with eg)?

A

Osmotic diuretics e.g. mannitol

Carbonic anhydrase inhibitors e.g. acetazolamide

Loop diuretics e.g. frusemide (furosemide)

Thiazides e.g. bendrofluazide (bendroflumethiazide)

Potassium sparing diuretics e.g. amiloride, spironolactone.

928
Q

Which diuretic classes are used clinically?

A

Loop diuretics
Thiazides
K sparing diuretics

929
Q

How do osmotic diuretics e.g. mannitol work?

A

Pharmacologically inert= don’t affect kidney tubule cells
Just change osmolarity of filtrate

Filtered by glomerulus but not reabsorbed

No ACTION OF NA REABSORPTION

ACTION OF H2O REABSORPTION

  • Increase osmolarity of tubular fluid
  • Decrease H20 reabsorption where nephron is freely permeable to water i.e. PCT, desc loop and CT
930
Q

Where do carbonic anhydrase inhibitors work?

A

Act proximally

931
Q

Where do loop diuretics work?

A

LOH

932
Q

Where do thiazides work?

A

DCT

933
Q

Where do K sparing diuretics work?

A

Late DCT and collecting duct

934
Q

How do carbonic anhydrase inhibitors e.g. acetazolamide work?

A

ACTION OF NA REASBORPTION
Inhibit Na and HCO3 reabsorption in PCT

Act on carbonic anhydrase (apical and cytoplasmic) -> more bicarb in filtrate (not transferred across cell)
-> affects some of the Na reuptake (Na retention in lumen increases-> more Na and therefore water lost in urine)

ACTION ON H2O REABSORPTION
Increased tubular fluid osmolarity and decreased osmolarity of medullary interstitium-> decreased H2O reabsorption in CT

OTHER
Increased bicarb delivery to DCT-> K loss
Ca and Mg affected-> loss of K recycling

NB. Earlier you act in kidney-> give kidney more time to adapt (better to use other diuretics)

935
Q

How do loop diuretics e.g. frusemide work?

A

Act on ascending limb (very powerful because affect countercurrent multiplier)
Via affect Na/2Cl/K cotransport protein
Sodium chloride mostly reasborbed
Potassium recycling drives the positive lumen potential
BUT with loop diuretics-> reduce K movement by interfering with positive lumen potential-> less transfer of electrolyte

ACTION ON NA REABSORPTION
Inhibit Na and Cl reabsorption in ascending limb (30%)

ACTION OF H2O REABSORPTION
Increase tubular fluid osmolarity and decrease osmolarity of medullary interstitium-> decreased H2O reabsorption in CT

OTHER
Increased Na delivery to DCT-> K loss (due to increased Na/K exchange)= like thiazides
Ca and Mg affected-> loss of K recycling

936
Q

How do thiazides e.g. bendroflumethiazide work?

A

Less powerful, act on DCT
Prevent NaCl reuptake-> more water being lost out of kidney

ACTION ON NA REABSORPTION
Inhibit Na and Cl reabsorption in early DCT=5-10%

ACTION ON H2O REABSORPTION
Increased tubular fluid osmolarity-> decreased H2O reabsorption in the collecting duct (promote fluid loss)

OTHER
Increased Na delivery to DCT-> K loss (due to increased Na/K exchange)= like loop diuretics
Lose Mg (like loop diuretics)
Increased Ca reabsorption (maybe, not understood)

937
Q

What effect do diuretics have on renin secretion long term?

A

If chronically taking diuretics-> reduced sodium load passing through DCT in blood-> detected by macula densa cells-> stimulates renin production which counters the effects of diuretic (promotes Na and water reabsorption)

Particularly loop diuretics and thiazides

938
Q

Which diuretic would have the most powerful effect on renin secretion?

A

Loop diuretics (and thiazides)

Loop diuretic sensitive transporter especially affected

939
Q

How do potassium sparing diuretics e.g. spironolactone and amiloride work?

A

Act on collecting duct and late DCT (no longer need to worry about Na/K exchange)

ACTION ON NA REABSORPTION
Inhibit Na reabsorption (and concomitant K secretion) in early distal tubule (5%)

ACTION ON H2O REABSORPTION
Increase tubular fluid osmolarity-> decreased H2O reabsorption in collecting duct

OTHER
Decreased reabsorption of Na to distal tubule increases H retention (decreased Na exchange)

Mechanism depends on class

940
Q

What are the classes of K sparing drugs?

A

Aldosterone receptor antagonists (mineralocorticoid receptor affected) e.g. spironolactone

Inhibitors of aldosterone-sensitive Na channels e.g. amiloride

941
Q

What are the common side effects of diuretics?

A
Hypovolemia
Metabolic alkalosis
Hyperuricemia
Metabolic acidosis
Hyperkalemia
Hypokalemia
Hyponatremia
942
Q

What side effects do loop diuretics cause?

A

30% loss-> Hypovolemia and hyponatremia

Cl loss-> Metabolic alkalosis

? -> Hyperuricemia

Na/K exchange-> Hypokalemia

943
Q

What side effects do thiazides cause?

A

10% loss-> Hypovolemia and hyponatremia

Cl loss-> Metabolic alkalosis

? -> Hyperuricemia

Na/K exchange-> Hypokalemia

944
Q

What side effects do carbonic anhydrase inhibitors cause?

A

HCO3- loss-> Metabolic acidosis

945
Q

What side effects do potassium sparing diuretics cause?

A

Less Na/K exchange-> Hyperkalemia

946
Q

Why do you get hyperuricemia with thiazides and loop diuretics?

A

High conc of diuretic competes with uric acid for transporter (organic anion basal transporter)
Uric acid levels in blood start to build up

947
Q

What diuretics are used to treat hypertension?

A

Thiazide
= First line in most countries
= Good for salt sensitive hypertension

(Other drugs also used for hypertension)

948
Q

How are thiazides used to treat hypertension?

A
Initial response (4-6 weeks)= loss in blood volume
After 4-6 weeks= plasma volume restored, reduced TPR

TPR reduced by activated eNOS (endothelium), Ca channel antagonism and opening of K Ca channel (smooth muscle)

949
Q

How do diuretics treat heart failure and oedema?

A

Heart failure activates SNS which is bad

Loop diuretics used-
30% Na load (better with K sparing diuretics)

Acute reduction in congestion (decreased ventricular filling pressures and improvment

Prompt diuretic effects (furosemide within 30 mins, peaks at 1.5 hours)

Resistance happens quickly (but great acutely)

950
Q

Outline what happens at a GABAergic synapse

A

GABA synthesised from glutamate (converted by GAD)

GABA taken up into vesicles ready for arrival of AP down axon (mostly short axon interneurones)

Release mechanism= diffuses across cleft and reacts with recepto

Stimulates post-synaptic GABA A Rs (linked to chloride channel)

GABA stimulation means chloride enters-> potential drops down to -90mV (less likely to be activated)

GABA reuptake by selective carriers

Can be repackaged and used again or broken down by metabolic enzymes e.g. SSA

951
Q

How does GABA go to succinic semialdehyde?

A

GABA transaminase (GABA-T)

Mitochondrial enzyme

952
Q

How does succinic semialdehyde go to succinic acid?

A

Succinic semialdehyde dehydrogenase (SSDH)

Mitochondrial enzyme

953
Q

What is sodium valproate used for?

A

EPILIM anti-convulsants used for epileptics

Inhibits GABA T and SSDH

954
Q

What is vigabatrin used for?

A

SABRIL
Suicide inhibitor
Binds covalently to GABA-T

955
Q

What happens when you inhibit GABA metabolism?

A

Large increase in brain GABA

956
Q

What is the GABA-A receptor complex? How is binding enhanced?

A

Binding of GABA

Linkage of GABA R and BDZ
R proteins by GABA modulin-> opening of chloride channel (chloride ions enter postsynaptic cell)

Presence of benzodiazepine (BDZ) receptor enhances GABA action to increase affinity of GABA and GABA R

Barbiturates bind to a different place and enhance GABA binding (not reciprocal)

957
Q

What is bicuculline?

A

Competes with GABA (competitive antagonist)

958
Q

What is flumazenil?

A

Competive benzodiazepine receptor antagonist

959
Q

What are the actions of BZs and BARBs on the GABAA R complex? What are the differences?

A

No activity alone (allosteric action)

Different binding sites and different mechanisms

BZs increase frequency of openings
BARBs increase duration of openings

BARBs less selective than BZs
Decreased excitatory transmission and has other membrane effects

960
Q

What are the clinical uses of BZs and BARBs?

A
Anaesthetics (BARBs only, thipentone)
Anticonvulsants (diazepam, clonazepam, phenobarbital)
Anti-spastics (diazepam)
Anxiolytics
Sedatives and hypnotics
961
Q

What is an anxiolytic?

A

Drug to remove anxiety without impairing mental or physical activity
Minor tranquilisers

962
Q

What is a sedative?

A

Reduce mental and physical activity without producing loss of consciousness

963
Q

What is a hypnotic?

A

Drug that induces sleep

964
Q

What should anxiolytics, sedatives and hypnotics do?

A
Have wide margin of safety
Not depression respiration
Produce natural sleep (hypnotics)
Not interact with other drugs
Not produce 'hangovers'
Not proudce dependence
965
Q

Give an example of a sedative and hypnotic barbiturate

A

Amobarbital
Severe intractable insomnia
Half life= 20-25h

966
Q

What are the unwanted effects of barbiturates?

A

Low safety margins= depress respiration, overdosing lethal

Alter natural sleep (reduce REM)-> hangovers and irritability

Enzyme inducers

Potentiate effect of other CNS depressants (e.g. alcohol)

Tolerance

Dependence-> withdrawal syndrome (insomnia, anxiety, tremor, convulsions, death)

967
Q

Give examples of barbiturates

A

Phenobarbitone
Pentobarbitone
Thiopentone

968
Q

What kind of drug is chloral hydrate (to tricholerthanol)?

A

Hypnotic

969
Q

What do benzodiazepines do?

A

All act at GABA A receptors
All similar
Pharmacokinetics determine use

970
Q

Outline the pharmacokinetics of benzodiazepines

A

Well absorbed P.O.
Peak plasma conc after 1h
IV used for status epilepticus

Bind plasma proteins strongly
Highly lipid soluble (so wide distribution)

Usually extensive metabolism (liver)

Excreted in urine, glucoronide conjugates

Action varies greatly

Short or long acting (slow metabolism and or active metabolites)

971
Q

How are benzodiazepines metabolised?

A

LONG ACTING
Diazepam (32h)-> nordiazepam (60h)-> oxazepam (8h)-> glucoronide

Diazepam (32h)-> temazepam (8h)-> oxazepam (h)-> glucoronide

Chlordiazepxodie (12h)-> nnordiazepam (60h)-> oxazepam (8h)-> glucoronide

Nitrazepam (28h)-> glucuronide

SHORT ACTING
Temazepam (8h)-> oxazepam (h)-> glucoronide

Lorazepam (12h)-> glucuronide

SEE DIAGRAM!

972
Q

What is diazepam?

A

Valium

Long-acting anxiolytic

973
Q

What can oxazepam cause?

A

Hepatic impairment

974
Q

Give examples of long-acting anxiolytics

A

Diazepam (valium)
Chlordiazepoxide (librium)
Nitrazepam (NB. has daytime anxiolytic effects)

975
Q

Give examples of short-acting sedatives and hypnotics

A

Temazepam

Oxazepam

976
Q

What are the advantages of benzodiazepines?

A

Wide margin of safety (overdose just causes prolonged sleep= rousable), flumazenil

Mild effect on REM sleep

Don’t induce liver enzymes

977
Q

What are the unwanted effects of benzodiazepines?

A

Sedation, confusion, amnesia, ataxia (impaired manual skills)

Potentiate other CNS depressants (alcohol, barbs)

Tolerance (less than barbs, ‘tissue’ only)

Dependence (withdrawal syndrome, similar to barbs but less intense)

Withdraw slowly

Free plasma concentration increases e.g. by aspirin, heparin

978
Q

What is zopiclone? How does it work?

A
Sedative 
Short acting (half life 5h)
Acts at BZ receptors (cyclopyrrolone)
Similar efficacy to BZs
Minimal hangover effects but dependency still a problem
979
Q

Give an example of antidepressant anxiolytics

A

SSRIs

Effective, delayed response, popular

980
Q

Give examples of antiepileptic anxiolytics

A

Valproate

Tiagabine

981
Q

Give examples of antipsychotic anxiolytics

A

Olanzapine
Quetiapine
Marked side-effects so not often used

982
Q

What is propanolol?

A

Anxiolytic

Improves physical symptoms (tachycardia, B2 and tremor, B2)

983
Q

What is buspirone?

A

5HT1A agonist (anxiolytic)

Fewer side effects (less sedation)
Slow onset of action (days and weeks)

984
Q

Benzodiazepines are used to treat ‘panic attacks’ and other anxiety states. By what mechanism do they produce their anti-anxiety effects?

A: Inhibition of GABA breakdown
B: Activation of 5HT1A receptors
C: Enhancement of the action of GABA at GABA-A receptors
D: Inhibition of GABA reuptake
E: Enhancement of the action of GABA at GABA-B receptors

A

C: Enhancement of the action of GABA at GABA-A receptors

985
Q

Which of the following drugs is commonly used in the treatment of insomnia?

A: Thiopental
B: Phenytoin
C: Baclofen
D: Sodium valproate
E: Temazepam
A

E: Temazepam

986
Q

What is the nigrostriatal pathway?

A

Cell bodies originate in the substantia nigra zona compacta and project to the striatum

Control of movement

987
Q

What is the mesolimbic pathway?

A

Cell bodies originate in the vetnral tegmental in the ventral tegmental area and project to the nucleus accumbens, frontal cortex, limbic cortex and olfactory tubercule

Involved in emotion

988
Q

What is the tuberoinfundibular system?

A

Short neurones running from the arcuate nucleus of the hypothalamus to the medial eminence and pituitary gland

Regulate hormone secretion

989
Q

What dopamine receptors are in the D1 family?

A

D1 and D5

990
Q

What dopamine receptors are in the D2 family?

A

D2, D3 and D4

991
Q

How many case of PD are familial?

A

8% of all cases

992
Q

What is PD believed to be caused by?

A

Combination of environmental, oxidative stress, altered protein metabolism and risk genes

993
Q

What are the main signs of PD?

A

Rest tremor (shaking of limb when relaxed)
Rigidity (stiffness, limbs feel heavy and weak)
Bradykinesia (slowness of movement)
Postural abnormality (forward tilt of trunk)

994
Q

What is the main problem with current PD drugs?

A

Treat the clinical symptoms don’t slow the degenerative process

995
Q

What are the presenting symptoms of PD?

A

Pill-rolling rest tremor
Difficulty with fine movements e.g. micrographia
Poverty of blinking
Impassive face
Monotomy of speech and loss of volume of voice
Disorders of posture- flexion of the neck and trunk
Lack of arm swing
Loss of balance- lack of righting reflex, retropulsion
Short steps, shuffling gait

Unilateral onset
Spreads to both sides
Generally worsen

996
Q

What are the non-motor symptoms of PD?

A
Depression
Sleep disturbances
Pain
Taste and smell disturbances
Cognitive decline and Demention
Autonomic
- Constipation
- Postural hypotension
Urinary frequency and urgency
Impotence
Increased sweating
997
Q

What areas of the brain are affected in PD?

A

Substantia nigra

Lewy bodies

Also, locus coruleus, dorsal vagus nucleus, nucleus basalis of Mynert

998
Q

What are the stages of PD and what is affected in these stages?

A

STAGES 1-2
Dorsal motor nucleus of vagus
Raphe nucleus
Locus coeruleus

STAGE 3
Substantia nigra pars compacta

STAGE 4
Amygdala
Nucleus of Meynert
Hippocampus

STAGE 5-6
Cingulate cortex
Temporal cortex
Frontal cortex
Parietal cortex
Occipital cortex

NB. Stages 1-3 presymptomatic

999
Q

What biochemical change happens in PD?

A

Marked reduction in caudate nucleus and putamen dopamine content

Symptoms appear when 80-85% of DAergic neurons are lost and 70% of striatal dopamine

Compensatory mechanisms mean symptoms take time to appear

1000
Q

What is the purpose of L-DOPA?

A

DOPA is precursor to dopamine (converted by dopa decarboxylase in brain)
Dopamine can’t cross BBB so give L-dopa

1001
Q

Why is L-DOPA given with another drug?

A

Needs to be given with peripheral DOPA decarboxylase inhibitor

Because otherwise L-DOPA metabolised to DA in periphery (SEs include nausea and vomiting)

1002
Q

What are the major preparations of L-DOPA

A

Sinamet (Carbidopa + L-DOPA)

Madopar (Benserazide + L-DOPA)

1003
Q

What does L-DOPA treat?

A

Hypokinesia, rigidity and tremor
Start with low dose and increase until max benefit without SEs
Effectiveness declines over time

1004
Q

What are the SEs of L-DOPA?

A

ACUTE
Nausea
Hypotension
Psychological effects= confusion, disorientation and nightmares

CHRONIC
Dyskinesias
“On-Off” effects

1005
Q

What can be used to treat nausea caused by L-DOPA?

A

Doperidone (peripheral acting antagonist)

1006
Q

What are dyskinesias that appear as chronic SEs of L-DOPA?

A

Abnormal movements of limbs and face
Can occur within 2 years of treatment
Disappear if reduce dose but clinical symptoms reappear

1007
Q

What are the chronic “on-off” effects of L-DOPA

A

Rapid fluctuations in clinical state

Off periods may last from minutes to hours

1008
Q

What do dopamine agonists do?

A

Act on D2 receptors

1009
Q

Give examples of dopamine agonists

A

Bromocriptine
Pergolide
Ropinerol

1010
Q

What are the benefits of using dopamine agonists to treat PD?

A

Smoother and more sustained response
Actions independent of dopaminergic neurons
Incidence of dyskinesias is less
Can be used in conjunction with L-DOPA

1011
Q

What are the adverse effects of dopamine agonists?

A

Common= confusion, dizziness, nausea and vomiting, hallucinations
Rare= constipation, headache, dyskinesias
Problems with heart valves
Addictive behaviours

1012
Q

What is Deprenyl (selegiline)?

A
MAO inhibitor (selective for MAO-B)
Predominates in dopaminergic areas of CNS 
Doesn’t have peripheral SEs
1013
Q

What is Deprenyl used to treat?

A

PD
Cab be given alone in early stages
Given in combo with L-DOPA (so can reduce L-DOPA dose by 30-50%)

1014
Q

What are the SEs of Deprenyl?

A

Rare

Hypotension, nausea and vomiting, confusion and agitation

1015
Q

What is resagiline?

A

MAO inhibitor
Neuroprotective properties by inhibiting apoptosis
Promotes anti-apoptosis genes
May be useful in PD- clinical trials

1016
Q

What are COMT inhibitors?

A

Catechol-O-methyl transferase inhibitors

E.g. tolocapone (CNS and peripheral) and Entacapone (peripheral)

1017
Q

What does tolocapone do?

A

COMT inhibitor
CNS= Prevents breakdown of dopamine in the brain
PNS= Stops 3-OMD formation so increases penetration of L-DOPA across the BBB (reduce L-DOPA dosage)

1018
Q

What does entacapone do?

A

Peripheral COMT inhibitor

PNS= Stops 3-OMD formation so increases penetration of L-DOPA across the BBB (reduce L-DOPA dosage)

1019
Q

What is 3-OMD (in COMT inhibition)?

A

COMT in the periphery converts L-DOPA to 3-OMD
3-OMD and L-DOPA compete to be transported into the brain
More 3-OMD means less L-DOPA enters brain
So inhibiting this (with COMT inhibitors) is beneficial to increase L-DOPA reaching the brain

1020
Q

What are the SEs of COMT inhibitors?

A

Cardiovascular complications

1021
Q

What percentage of the general population are affected by Schizophrenia?

A

1%

1022
Q

What groups of symptoms is Schizophrenia comprised of?

A

Positive symptoms
Negative symptoms
Cognitive deficits

1023
Q

What are the positive symptoms in schizophrenia?

A

Hallucinations
Delusions
Disorganized thoughts

1024
Q

What are the negative symptoms in schizophrenia?

A

Reduced speech (alogia)
Lack of emotional and facial expressive (affective flattening)
Diminished ability to begin and sustain activities (avolition)
Decreased ability to find pleasure in everyday (anhedonia)
Social withdrawal (asociality)

1025
Q

What are the cognitive deficits in schizophrenia?

A

Memory
Attention
Planning
Decision making

1026
Q

What happens once someone has been diagnosed with schizophrenia?

A

10-20% resolved illness, never returns (with or without treatment)
30-35% illness recurs repeatedly with full recovery after each episode
30-35% illness recurs repeatedly with incomplete recovery and a persistent defective state develops (more pronounced with each successive relapse)
10-20% illness pursues a downhill course from start

1027
Q

What is the role of dopamine in schizophrenia?

A

Excessive dopamine transmission in the mesolimbic and striatal region leading to positive symptoms (mediated through D2 receptors)
Dopamine deficit in pre-frontal region (mediated by D1 receptors) leads to negative symptoms

1028
Q

What is the evidence that dopamine is involved in schizophrenia?

A

Dopamine agonists e.g bromocriptine, and amphetamine can induce various psychotic reactions
Typical anti-schizophrenic drugs are dopamine receptor antagonists and there is a strong correlation between antipsychotic potency and activity in blocking D2 receptors

1029
Q

What is the glutamate theory of schizophrenia?

A

NMDA receptor antagonists e.g. phencyclodine, ketamine etc. produce psychotic symptoms
Reduced glutamate concentrations and glutamate receptor densities in post-mortem schizophrenic brain
In mice with reduced NMDA receptor expression- mice show stereotypical behaviours like schizophrenia and respond to antipsychotic therapy
Glutamate and dopamine exert effects on GABAergic striatal neurons (then project to the thalamus and constitute a sensory ‘gate’)
Too little glutamate (excitatory) or too much dopamine (inhibitory), disables the gate, allowing uninhibited sensory input to reach the cortex

1030
Q

How do glutamate and dopamine relate to the ‘sensory gate’ in schizophrenia?

A

Glutamate (excitatory) and dopamine (inhibitory) exert effects respectively on GABAergic striatal neurons (then project to the thalamus and constitute a sensory ‘gate’)
Too little glutamate or too much dopamine disables the gate, allowing uninhibited sensory input to reach the cortex

1031
Q

What is the genetic basis of Schizophrenia?

A

Strong but incomplete hereditary tendency
Risk genes but no single gene responsible
Weak associations of polymorphisms
Neuregulin 1 seems most robust
8 other susceptibility genes (all associated with glutamate or dopamine transmission)

1032
Q

What is the mechanism of antipsychotics? What do they do?

A

Neuroleptic drugs are antagonists at dopamine ‘D2 like’ receptors

Block other receptors too e.g. 5HT

Treat positive symptoms not negative ones
Delayed effects
Initial increase in DA synthesis declines over time

1033
Q

What kinds of side effects can antipsychotics cause?

A
Exprapyramidal
Sedation
Weight gain
Hyperglycaemia
Anticholinergic
Orthostatic hypotension
1034
Q

What are atypical antipsychotics?

A

Antipsychotics that are second generation compounds producing less extrapyramidal side effects

1035
Q

What can neuroleptics be used to?

A

Anti-psychotic

Anti-emetic

1036
Q

Outline the anti-emetic effect of neuroleptics

A

Blocking dopamine receptors in the chemoreceptor trigger zone

E.g. Phenothiazine= effective at controlling vomiting and nausea induced by drugs (e.g. chemotherapy) , renal failure

Many neuroleptics also have blocking action at histamine receptors so are effective at controlling motion sickness

1037
Q

What are the extrapyramidal SEs of antipsychotics?

A

Acute dystonia

Tardive dyskinesias

1038
Q

What are are SEs of antipsychotics?

A

Extrapyramidal (acute dystonia and tardive dyskinesias)

Endocrine effects (prolactin secretion)

Blocking alpha adrenoceptors= orthostatic hypotension

Blocking 5HT Rs= weight gain

Blocking cholinergic muscarinic receptors= anti-muscarinic side effects e.g blurring of vision, increased intra-ocular pressure, dry mouth, constipation, urinary retention

1039
Q

How do antipsychotics lead to breast swelling?

A

DA inhibits PL secretion via D2 receptors
Lowering D2 Rs means increased serum prolactin concentrations
Breast swelling in men and women and sometimes lactation in women

1040
Q

What is acute dystonia? How does it relate to antipsychotics?

A

Blockade of DA Rs in nigrostriatal system can induce PD-like side effects

Involuntary movements e.g. muscle spasm, protruding tongue, fixed upward gaze, neck spasm etc.
Often accompanied by Parkinson’s features
Occur in the first few weeks, often declining with ongoing therapy. Reversible on drug withdrawal or anti-cholinergics

1041
Q

What is tardive dyskinesia? How does it relate to antipsychotics?

A

Blockade of DA Rs in nigrostriatal system can induce PD-like side effects

Involuntary movements, often involving the face and tongue, but also limb and trunk
Occur in about 20% of patients after several months or years of therapy (hence ‘tardive’)
More associated with typical antipsychotics
Made worse by drug withdrawal or anti-cholinergics
May be related to proliferation in pre-synaptic DA D2 receptors or glutamate excitotoxic striatal neurodegeneration

1042
Q

What does general anaesthesia do?

A

Loss of consciousness (at low concentration)
Suppression of reflex responses (at high concentration)

Loss of consciousness
Suppression of reflex responses
Relief of pain (analgesia)
Muscle relaxation
Amnesia
Relief of pain (analgesia)
Muscle relaxation
Amnesia
1043
Q

List types of general anesthetics

A
GASEOUS/INHALATION
Nitrous oxide
Diethyl ether 
Halothane
Enflurance

INTRAVENOUS
Propofol
Etomidate

Structurally dissimilar

1044
Q

Outline the mechanism of action of general anaesthetics

A

Either reduced neuronal excitability or altered synaptic function

Different for IV and inhalation

1045
Q

What did Meyer and Overton show with anaesthetic? What were the problems with this?

A

More lipid soluble= better anaesthetic

Anaesthetic potency increases in direct proportion with oil:water partition coefficient

Problems:
At relevant anaesthetic concentrations, change in bilayer was minute
2. How would this change impact membrane proteins?

1046
Q

What are the targets of inhaled GAs?

A

ALTERED SYNAPTC FUNCTION
1) GABA Rs (halogenated compounds-> enhance depressive)
Via interaction with glycine receptors (homologous to GABA A Rs) and GABA A
a1 subunit-> suppression of reflex responses

2) NMDA-type glutamate Rs (NO and ketamine-> diminish excitatory)
Blocked (probably by competing with co-agonist glycine)

3) Neuronal nicotinic ACh receptor
Decreased activity with more halogenated compound

REDUCED NEURONAL EXCITATION
4) TREK (background leak) K channel
Enhanced-> increased hyperpolarisation of neurones for longer (harder to excite)

1047
Q

What are the targets of IV GAs?

A

Via GABA A receptors (enhancement-> depressant effects)

GABA RS
B3 subunit common on GABA A-> suppression of reflex responses
a3 subunit common on GABA A-> amnesia

1048
Q

How does neuroanatomy affect the loss of consciousness function of GAs?

A

Depress excitability of
thalamocortical neurons

Influences reticular
activating neurons
(RAS carries all the cortical varieties of consciousness)

  • ACh released from cholinergic nerve terminals projecting from RAS to the thalamus and cortex
  • Anaesthetics can directly hyperpolarize thalamocortical neurons by activating TREK channels or potentiating GABA A receptors
1049
Q

How does neuroanatomy affect the suppression of reflex responses by GAs?

A

Depression of reflex pathways in spinal cord

Possible because of high density of GABA Rs in the DH of the SC

1050
Q

How does neuroanatomy affect the amnesia of GAs?

A

A5 subunit of the GABAAR is rare but lots in hippocampus

GAs decrease synaptic transmission in hippocampus and amygdala

1051
Q

What is the difference in administration between inhalation and intravenous anaesthetic inhalation?

A

IV
Inject drug into blood
Then can freely access target tissue (brain)
How long it lasts is dependent on its metabolism by the liver

INHALED
Less well they dissolve in blood= quicker the onset of action
Drug diffuses from airway-> alveoli-> across into blood

Depends of blood:gas coefficient

1052
Q

What happens when an inhaled a drug has a high blood:gas coefficient?

A

High blood:gas coefficient (less lipid soluble) so higher percentage disappears into blood, slower process into brain (slower induction time)
Takes longer until equilibrium with the brain partial pressure of the gas

1053
Q

What happens when an inhaled a drug has a low blood:gas coefficient?

A

Low blood:gas coefficient (more lipid soluble) so lower percentage disappears into blood, faster process into brain (faster induction time)
Quicker to equilibrium with the brain partial pressure of the gas

1054
Q

What is the blood:gas coefficient?

A

The blood/gas partition coefficient describes how the gas will partition itself between the two phases (blood and brain) after equilibrium has been reached

1055
Q

True or false; a poorly soluble GA will have a slow onset of action?

A

False

Will be rapid

1056
Q

Inhalation vs intravenous GA?

A

IV (often first for induction)
Fast induction
Less coughing and excitatory phenomena
NB. Not in control, reliant on liver metabolism

INHALATION (often good to maintain depth and controlling)
Rapidly eliminated
Rapid control of depth of anaesthesia (brain and lungs very similar levels so quick to reverse)
NB. Airway irritation can lead to the cough reflex being initiated

1057
Q

What GAs are commonly used in GAs?

A

To lose consciousness and suppress reflexes:
Induction= propofol (IV)
Maintenance= enflurane (inhalation)

1058
Q

Alongside GAs, what drugs would you use in surgery?

A

Relief of pain (analgesia)= opioid (e.g. IV fentanyl)
Muscle relaxation = neuromuscular blocking drugs (e.g. suxamethonium
Amnesia= benzodiazepines (e.g. IV midazolam)

1059
Q

What is a local anaesthetic?

A

A drug which reversibly blocks neuronal conduction when applied locally

1060
Q

List examples of local anaesthetics

A
Procaine
*Cocaine
Tetraqcaine (amethocaine)
Cinchocaine (dibucaine)
*Lidocaine (lignocaine)
Prilocaine
Bupivacaine
Benzocaine (doesn't have amine side chain)
1061
Q

What are the structural components of local anaesthetics?

A

Aromatic region (important for mechanism of action and metabolism)
Ester or amide bond
Basic amine side-chain

NB.
Ester= procaine, cocaine, tetraqcaine, cinchocaine
Amide= lidocaine, prilocaine, bupivacaine

1062
Q

How do local anaesthetics interact with sodium channels?

A

HYDROPHILLIC PATHWAY
LA (a weak base) is injected

Non-ionised form needed as it passes through connective tissue sheath and passes inside axon (e.g. nociceptive neuron)

Equilibrium established between ionised and non-ionised form

Cationic (ionised) form is needed as it has the LA activity by binding to inside of VSSC-> stereochemically blocks channel (reduces generation and propagation of APs)

Use-dependent because binding site is inside ion channel (needs to be open)

HYDROPHOBIC PATHWAY
Lipid soluble LAs e.g. benzocaine
Passes towards inside of axon (can drop into channel and ionize and block channel even if it isn’t being used)

1063
Q

What are the effects of LAs?

A

Prevent generation and conduction of APs

Do NOT influence resting membrane potential

May also influence channel gating and surface tension

Selectively block small diameter fibres and non-myelinated fibres

1064
Q

What is the pKa of LAs?

A

8-9

Weak bases

1065
Q

How can LAs be administered?

A

SURFACE

INFILTRATION ANAESTHESIA

IV REGIONAL ANAESTHESIA

NERVE BLOCK ANAESTHESIA

1066
Q

Outline surface LA administration

A

Mucosal surface (mouth, bronchial tree)
Spray (or powder)
High concentrations-> systemic toxicity

1067
Q

Outline infiltration anaesthesia

A

Directly into tissues-> sensory nerve terminals
Minor surgery
Adrenaline co-injection (NOT extremities)

1068
Q

Outline IV regional anaesthesia

A

IV distal to pressure cuff
Limb surgery
Systemic toxicity of premature cuff release

1069
Q

Outline nerve block anaesthesia

A

Close to nerve trunks e.g. dental nerves
Widely used- low doses, slow onset
Vasoconstrictor co-injection

1070
Q

Outline spinal anaesthesia

A

Sub-arachnoid space-> spinal roots
Abdominal, pelvic, lower limb surgery
Reduced BP-> prolonged headache
Glucose (increased specific gravity)

1071
Q

Outline epidural anaesthesia

A

Fatty tissue of epidural space-> spinal roots
Abdominal, pelvic, lower limb surgery and painless childbirth
Slower onset-> higher doses
More restricted action (less effect on BP)

1072
Q
Lidocaine
Property=
Absorption (mucous membranes)= 
Plasma protein binding= 
Metabolism=
Plasma half life=
A
Property= amide
Absorption (mucous membranes)= good
Plasma protein binding= 70%
Metabolism= hepatic N-dealkylation
Plasma half life=2h
1073
Q
Cocaine
Property=
Absorption (mucous membranes)= 
Plasma protein binding= 
Metabolism=
Plasma half life=
A
Property= good
Absorption (mucous membranes)= good
Plasma protein binding= 90%
Metabolism= liver and plasma, non-specific esterases
Plasma half life= 1h
1074
Q

What are the unwanted effects of lidocaine?

A

CNS (paradoxical)
Stimulation
Restlessness, confusion
Tremor

CVS (Na channel blockage)
Myocardial depression
Vasodilatation
Decreased BP

1075
Q

What are the unwanted effects of cocaine?

A

CNS (sympathetic actions)
Euphoria
Excitation

CVS (sympathetic actions)
Increased CO
Vasoconstriction
Increased BP

1076
Q

Lidocaine:

A: Inhibits reuptake of 5-hydroxytryptamine
B: Blocks voltage-gated K+ channels
C: Is a competitive muscarinic cholinoceptor antagonist
D: Is a weak base
E: Is a general anaesthetic

A

D: Is a weak base

1077
Q

Which ONE of the following statements about local anaesthetics is INCORRECT? They:

A: Cause blockade of voltage-sensitive sodium channels
B: Block rapidly firing neurones more readily than more slowly firing neurones
C: Enhance action potential propagation
D: Are largely ionised at physiological pH
E: Have their durations of action increased if injected with adrenaline

A

C: Enhance action potential propagation

1078
Q

What kind of disorder is depression?

A

Affective disorder (a type of psychosis)

1079
Q

What are the symptoms of depression?

A
EMOTIONAL( psychological)
Misery, apathy, pessimism
Low self-esteem
Loss of motivation
Anhedonia

BIOLOGICAL (somatic)
Slowing of thought AND action
Loss of libido
Loss of appetite, sleep disturbance

1080
Q

What is unipolar depression or depressive disorder?

A

Mood swings in same direction

Relatively late onset

Reactive depression

  • stressful life events
  • non-familial

Endogenous depression

  • unrelated to external stresses
  • familial pattern

Drug treatment same for endogenous and reactive

1081
Q

What is bipolar depression or manic depression?

A

Oscillating depression and mania

Less common, early adult onset

Strong hereditary tendency

Drug treatment (Lithium)

1082
Q

What is ECT?

A

Electroconvulsive therapy

Electroconvulsive therapy (ECT) is a procedure, done under general anaesthesia, in which small electric currents are passed through the brain, intentionally triggering a brief seizure

Seems to cause changes in brain chemistry that can quickly reverse symptoms of certain mental illnesses

1083
Q

What is the monoamine theory of depression?

A
Depression= functional deficit of central MA transmission
Mania= functional excess

Main biochemical theory of depression

Down regulation of a2, B NA and 5HT receptors

HPA axis (increased CRH levels)
Hippocampal neurodegeneration
1084
Q

What is the pharmacological evidence supporting the monoamine hypothesis of depression?

A

Tricyclic antidepressants-> block NA and 5HT reuptake-> improves mood

MAO inhibitors-> increase stores of NA and 5HT-> improves mood

Reserpine-> inhibits NA and 5HT storage-> lowers mood

a-methyltyrosin-> inhibits NA synthesis-> lowers mood (calms manic patients)

Methyldopa-> inhibits NA synthesis-> lowers mood

ECT-> increases CNS responses to NA and 5HT> improves mood

1085
Q

What is the mode of action of TCAs?

A

E.g. amitriptyline

Neuronal monoamine re-uptake inhibitors

  • Enhance NA and 5HT in the brain
  • Less effective on DA transport

Also other receptor actions

  • a2
  • mAchRs
  • histamine
  • 5-HT

Delayed down-regulation of B-adrenoceptors and 5HT Rs

1086
Q

What are the pharmacokinetics of TCAs?

A

Rapid oral absorption

Highly PPB (90 - 95%)

Hepatic metabolism (active metabolites -> renal excretion of glucuronide conjugates)

Plasma half life= 10-20 hrs

1087
Q

What are the unwanted effects of TCAs?

A

AT THERAPEUTIC DOSES (given o.d. once reaches baseline)
Atropine-like effects (amitriptyline)
Postural hypotension (vasomotor centre)
Sedation (H1 antagonism)

ACUTE TOXICITY
CNS= excitement, delirium, seizures -> coma, respiratory depression

CVS= cardiac dysrhythmias, ventricular, fibrillation and sudden death

NB. Risk of attempted suicide

1088
Q

What are the main drugs TCA interact with?

A

PPB= increased TCA effects (aspirin, phenytoin)

Hepatic microsomal enzymes= increased TCA effects (neuroleptics; oral contraceptives)

Potentiation of CNS depressants (alcohol)

Antihypertensive drugs (monitor closely)

1089
Q

What is the mode of action of MAOIs?

A

E.g. phenelzine

MAO-A: NA and 5HT
MAO-B: DA
Most are non-selective MAOIs

Irreversible inhibition so long duration of action

Rapid effects= increased cytoplasmic NA and 5HT

Delayed effects= clinical response, downregulation of B-adrenoceptors and 5HT2 receptors

Also inhibit other enzymes

1090
Q

Outline the pharmacokinetics of MAOIs

A

Rapid oral absorption

Short plasma half life (few hrs) but longer duration of action

Metabolised in liver-> excreted in urine

1091
Q

What are the unwanted effects of MAOIs?

A

Atropine-like effects (< TCAs)

Postural hypotension (common)

Sedation (seizures in overdose)

Weight gain (possibly excessive)

Hepatotoxicity (hydrazines- rare)

1092
Q

What are the drug interactions of MAOIs?

A

Very serious problem

‘Cheese reaction’= tyramine-containing foods and MAOI -> hypertensive crisis (throbbing headache, increased BP, intracranial haemorrhage

MAOIs and TCAs -> hypertensive episodes (avoid)

MAOIs and pethidine -> hyperpyrexia, restlessness, coma and hypotension

Moclobemide: reversible MAO-A inhibitor (RIMA) can reduce drug interactions but also decreases duration of action

1093
Q

Why is moclobemide useful?

A

Rtyramine-containing foods and MAOI -> hypertensive crisis (throbbing headache, increased BP, intracranial haemorrhage

1094
Q

What is the mode of action for SSRIs?

A

E.g. fluoxetine

Selective 5-HT re-uptake inhibition

Less troublesome side-effects, safer in overdose

But less effective vs severe depression

1095
Q

Outline the pharmacokinetics of SSRIs

A

Oral administration

Plasma half life= 18-24 hrs

Delayed onset of action (2-4 weeks)

Fluoxetine competes with TCAs for hepatic enzymes (avoid co-administration)

1096
Q

What are the unwanted effects of SSRIs?

A

Nausea, diarrhoea, insomnia and loss of libido

Interact with MAOIs (avoid co-administration)

Increased suicidality (< 18 years old)

Fewer SEs than TCAs and MAOIs

1097
Q

What is the most commonly prescribed antidepressant drug?

A

Fluoxetine (‘Prozac’)= an SSRI

1098
Q

What is venlafaxine?

A

Dose-dependent reuptake inhibitor
5HT > NA > DA
2nd Line treatment for severe depression

1099
Q

What is mertazapine?

A

a2 receptor antagonist
Increases NA and 5HT release
Other R interactions (sedative)
Useful in SSRI-intolerant patients

1100
Q

What are the main drug types used to treat depression?

A

TCAs
MAOIs
SSRIs

1101
Q

Tricyclic antidepressant drugs (TCAs) work largely by:

A: Antagonism at 5HT receptors
B: Inhibiting central DA reuptake
C: Blocking VSCCs
D: Inhibition of central NA &amp; 5HT reuptake 
E: Enhancement of the action of GABA
A

D: Inhibition of central NA & 5HT reuptake

1102
Q

The ‘cheese reaction’ is most likely to be caused by:

A: Tricyclic antidepressants (TCAs)
B: Selective serotonin reuptake inhibitors (SSRIs)
C: Monoamine oxidase inhibitors (MAOIs)
D: Reversible MAO-A inhibitors (RIMAs)
E: α2-Adrenoceptor antagonists
A

C: Monoamine oxidase inhibitors (MAOIs)

1103
Q

Outline the epidemiology of Alzheimer’s disease

A

Main risk factor= age
Huge economic cost in the UK but low research investment
AD and dementia are leading cause of death in UK
Genetic basis= APP, PSEN, ApoE (hereditary= 8%)

1104
Q

What are the symptoms of AD?

A

Memory loss= especially recently acquired information

Disorientation and confusion= forgetting where they are

Language problems= stopping in the middle of a conversation

Personality changes= becoming confused, fearful, anxious

Poor judgement= such as when dealing with money

1105
Q

What is the normal physiological process of amyloid?

A

Amyloid precursor protein (APP) cleaved by alpha-secretase
sAPPa released but C83 fragment remains
C83 is digested by y-secretase
Products removed

1106
Q

What is the pathophysiological process of amyloid (amyloid hypothesis)?

A

APP cleaved by beta secretase
sAPPb released but C99 fragment remains
C99 digested by y-secretase releasing B-amyloid protein
AB forms toxic aggregates

1107
Q

What is the normal physiological tau process?

A

Soluble protein present in axons

Important for assembly and stability of microtubules (important for structure and function of neuronal cells)

1108
Q

What is the pathophysiological tau process (tau hypothesis) in AD?

A

Hyperphosphorylated tau is insoluble-> self-aggregates to form neurofibrillary tangles
These are neurotoxic and result in microtubule instability

1109
Q

What is the inflammation hypothesis of AD?

A

Involves microglia
Phenotype changes of specialised CNS immune cells (similar to macrophages)

Increased release of inflammatory mediators and cytotoxic proteins
Increased phagocytosis
Decreased levels of neuroptective proteins

Increased inflammatory load on NS

1110
Q

What are the main hypotheses for AD?

A
Amyloid hypothesis (cleavage by beta secretase)-> AB plaques
Tau hypothesis (hyperphosphorylated)-> neurofibrillary tangles and neuronal instability
Inflammation hypothesis (activity of microglial cells)-> inflammatory load on NS increased
1111
Q

What drug classes can be used to treat AD?

A

Anticholinesterases= donepezil, rivastigmine, galantamine

NMDA receptor blockers= memantine

Treat symptoms not pathophysiology

1112
Q

What anticholinesterases are used to treat AD?

A

DONEPEZIL
Reversible cholinesterase inhibitor
Long plasma half-life

RIVASTIGMINE
Pseudo-reversible AChE and BChE inhibitor
8 hour half-life
Reformulated as transdermal patch

GALANTAMINE
Reversible cholinesterase inhibitor
7-8 hour half-life
7 nAChR agonist

1113
Q

What NMDA receptor blocker is used to treat AD?

A

MEMANTINE
Use-dependent non-competitive NMDA receptor blocker with low channel affinity
Only licensed for moderate-severe AD
Long plasma half-life

1114
Q

What are the major AD treatment failures that have occurred?

A

y-secretase inhibitors
- Tarenflurbil (binds to APP molecule) and semagacestat (small y-secretase inhibitor) failed clinical trials

B-amyloid immunotherapy

  • active= vaccines (in development)
  • passive= antibodies e.g. solanezumab and bapineuzumab (humanised monoclonal antibodies)

Tau aggregation
- In clinical trials now= methylene blue (currently used for methaemoglobinaemia)

1115
Q

What are the main differences in the membranes of gram positive and negative bacteria and mycolic bacteria?

A

GP=prominent peptidoglycan cell wall
GN= outer membrane with lipolysaccharides
MB= outer mycolic acid layer

1116
Q

How are prokaryotic proteins synthesised?

A

NUCLEIC ACID SYNTHESIS
Dihydropterate (DHOp) produced from paraminobenzoate (PABA)
Converted into dihydrofolate (DHF)

Tetrahydrofolate (THF) produced from DHF by DHF reductase
THF is important in DNA synthesis

DNA REPLICATION
DNA gyrase (topoisomerase-> releases tension)

PROTEIN SYNTHESIS
Ribosomes produce protein from RNA templates
Differ from eukaryotic ribosomes

1117
Q

How do eukaryotics and prokaryotic ribosomes differ?

A
P= 30s and 50s
E= 40s and 60s
1118
Q

How can antibiotics inhibit protein synthesis?

A

NUCLEIC ACID SYNTHESIS
Sulphonamides inhibit DHOp synthase
Trimethoprim inhibits DHF reductase

DNA REPLICATION
Fluoroquinolones (e.g. Ciprofloxacin) inhibit DNA gyrase and topoisomerase IV

The rifamycins (e.g. Rifampicin) inhibits bacterial RNA polymerase

PROTEIN SYNTHESIS
Ribosomes can be inhibited (e.g. macrolides like erythromycin, aminoglycosides, chloramphenicol and tetracyclines)

1119
Q

How are bacterial walls synthesised?

A

PEPTIDOGLYCAN (PtG) SYNTHESIS
A pentapeptide is created on N-acetyl muramic acid (NAM)
N-acetyl glucosamine (NAG) associates with NAM forming PtG

PtG TRANSPORTATION
PtG is transported across the membrane by bactoprenol

PtG INCORPORATION
PtG is incorporated into the cell wall when transpeptidase enzyme cross-links PtG pentapeptides

1120
Q

How do bacterial wall inhibitors work?

A

PtG SYNTHESIS
Glycopeptides (e.g. Vancomycin) bind to the pentapeptide preventing PtG synthesis

PtG TRANSPORTATION
Bacitracin inhibits bactoprenol regeneration preventing PtG transportation

PtG INCORPORATION
B-lactams bind covalently to transpeptidase inhibiting PtG incorporation into cell wall

CELL WALL STABILITY
Lipopeptide - (e.g. daptomycin) disrupt Gram +ve cell walls
Polymyxins - binds to LPS and disrupts Gram -ve cell membranes

1121
Q

Give examples of beta lactam antibiotics

A

Carbapenems
Cephalosporins
Penicillins

1122
Q
A patient with a chest infection is prescribed a beta-lactam antibiotic. Which one of the following drug classes is a beta-lactam antibiotic?
Carbapenems
Glycopeptides
Lipopeptides
Macrolides
Sulphonamides
A

Carbapenems

1123
Q
Considering the bacterial structure and the drug mechanism of action, which class of drug is less likely to be effective in the treatment of E Coli infections?
Glycopeptides
Macrolides
Rifamycins
Quinolones
Sulphonamides
A

Glycopeptides

Because e. coli is GN

1124
Q

What are the main causes of antibiotic resistance?

A

Unnecessary prescription
(50% of antibiotic prescriptions not required)

Livestock farming
(30% of UK antibiotic use in livestock farming)

Lack of regulation
(OTC availability in Russia, China, India)

Lack of development
(No new antibiotic drug classes in years)

1125
Q

What are the types of antibiotic resistance?

A

Additional target
Different DHF reductase enzyme produced

Hyperproduction
Over-production of DHF reductase

Drug Permeation
Decreased drug influx, increased efflux systems

Enzyme alteration
Mutations in DNA gyrase enyme

Destruction enzymes
Production of beta-lactamase

1126
Q

How does ‘additional target’ antibiotic resistance work? Give an example

A

Bacteria produce another target that is unaffected by the drug

E.g. E. Coli produce different DHF reductase enzyme making them resistant to trimethoprim

1127
Q

How does ‘hyperproduction’ antibiotic resistance work? Give an example

A

Bacteria significantly increase levels of DHF reductase

E.g. E. Coli produce additional DHF reductase enzymes making trimethoprim less effective

1128
Q

How does ‘drug permeation’ antibiotic resistance work? Give an example

A

Alteration to the enzyme targeted by the drug
Enzyme still effective but drug now ineffective

E.g. S. Aureus= mutations in the ParC region of topoisomerase IV confers resistance to quinolones

1129
Q

How does ‘enzyme alteration’ antibiotic resistance work? Give an example

A

Reductions in aquaporins and increased efflux systems

E.g. Primarily of importance in GN bacteria

1130
Q

How does ‘destruction enzyme’ antibiotic resistance work? Give an example

A

B-lactamases hydrolyse C-N bond of the B-lactam ring

E.g. Penicillins G and V (GP)
Flucloxacillin and Temocillin -> lactamase resistant
Amoxicillin->broad spectrum
GN activity
Co-administered with Clavulanic acid
1131
Q

Why is amoxicillin sometimes given with clavulanic acid?

A

Stops it being beta lactam resistant

1132
Q

How can fungal infections be classified?

A

Can be classified in terms of tissue/organs

Superficial= outermost layers of skin
Dermatophyte= skin, hair or nails
Subcutaneous= innermost skin layers
Systemic= primarily respiratory trac
1133
Q

What are the most common categories of anti-fungal drugs in the UK?

A
Azoles= fluconazole
Polyenes= amphotericin
1134
Q

How do azoles work?

A

Anti-fungalsls (inhibits membrane sterol synthesis)

Inhibit cytochrome P450-dependent enzymes involved in membrane sterol synthesis
Fluconazole (oral) -> candidiasis and systemic infections

1135
Q

How do polyenes work?

A

Anti-fungals (form membrane channe

Interact with cell membrane sterols forming membrane channels
Amphotericin (IV) -> systemic infections

1136
Q

Following surgery for mitral valve prolapse a patient is placed on anticoagulant and antiplatelet therapy using warfarin and aspirin. Describe the mechanisms of actions of these two drugs (2 marks)

A

Warfarin- vitamin K antagonist

Aspirin- CO inhibitor

1137
Q

Regular blood monitoring was subsequently initiated with the aim to maintain the international normalised ratio (INR) between 3-3.5. Explain the potential complications of an INR below this range AND above this range (2 marks)

A

Below the range increases risk of thrombosis

Above the range increases risk of bleeding

1138
Q

Unfortunately, the patient develops infective endocarditis and is prescribed a penicillin, an aminoglycoside and a rifamycin antibiotic. Describe how these three drugs work (2 marks)

A

Pencilin- binds covalently to transpeptidase inhibiting PtG incorporation into cell wall
Rifamycin- inhibits bacterial RNA polymerase
Aminoglycoside- protein synthesis inhibitor

1139
Q

What are the main targets for antibiotics?

A
IC TARGETS
Nucleic acids 
DNA gyrase 
RNA polymerase 
Bacterial ribosomes 

CELL MEMBRANE TARGETS
Peptidoglycan (PtG) synthesis PtG incorporation
Membrane stability

1140
Q

Differentiate between the drugs used to treat fungal infections

A
Azoles= inhibit ergosterol production 
Polyenes= bind to ergosterol and create pores
1141
Q

Describe the structure of a virus

A

Envelope proteins
Lipid envelope
Capsid (protein shell surrounding the genetic material of the virus)
Genetic material (RNA and DNA)

1142
Q

Outline what causes viral hepatitis?

A

Liver hepatocytes

Hep B involves reverse transcriptase as a drug target
Hep C involves RNA polymerase as a drug target

Only chronic infection requires treatment

1143
Q

How is chronic hep B treated?

A

Tenofovir nucleotide analogue, given sometimes with Peginterferon alfa

1144
Q

How is chronic hep C treated?

A

Ribavirin and Peginterferon alfa
Ribavirin nucleoside analogue prevents viral RNA synthesis

Boceprevir protease inhibitor
Most effective against Hep C genotype 1

1145
Q

What do hep C treatments depend on?

A

Aim to cure the virus

Depends on:
HCV genotype (genetic structure of the virus)
Viral load
Past treatment experience
Degree of liver damage
Ability to tolerate the prescribed treatment
Need for liver transplant

1146
Q

Outline the HIV life cycle

A

ATTACHMENT AND ENTRY
Viral membrane proteins interact with leukocyte membrane receptors
- HIV Glycoprotein (GP)120 attaches to CD4 receptor
- GP120 also binds to either CCR5 or CXCR4
GP41 penetrates host cell membrane
Viral capsid endocytosis

REPLICATION AND INTEGRATION
Within cytoplasm- reverse transciptase enzyme converts viral RNA into DNA
DNA transported into nucleus and integrated into host DNA

ASSEMBLY AND RELEASE
Host cell’s ‘machinery’ utilised to produce viral RNA and essential proteins
Virus is assembled within cell -> mature virion is released

1147
Q

How do HIV entry inhibitors interfere with attachment and entry in the HIV life cycle?

A

ENFUVIRTIDE
Binds to HIV GP41 transmembrane glycoprotein
Subcutaneous- people don’t like it

MARAVIROC
Blocks CCR5 chemokine receptor

1148
Q

How do HIV replication inhibitors interfere with replication in the HIV life cycle?

A

Nucleoside RT inhibitors
Activated by 3 step phosphorylation process
E.g. Zidovudine

Nucleotide RT inhibitors
Fewer phosphorylation steps required
E.g. Tenofovir

Non-nucleoside RT inhibitors
No phosphorylation required
Not incorporated into viral DNA
E.g. Efavirenz

1149
Q

How do HIV integration inhibitors interfere with integration in the HIV life cycle?

A

Viral integrase inserts viral DNA into host DNA

Inhibited by integrase inhibitors e.g. raltegravir

1150
Q

How do HIV protease inhibitors interfere with assembly and release in the HIV life cycle?

A

HIV protease cleaves Gag precursor protein (which encodes all viral structural proteins)

Protease inhibitors (PI)
Saquinavir (always given with ritonavir)
1151
Q

Why is ritonavir given with protease inhibitors e.g. saquinavir?

A

Low dose Ritonavir reduces PI metabolism ( co-administered as ‘booster’)

1152
Q

Outline the herpes simplex virus

A

VIROLOGY
Double-stranded DNA
Surrounded by tegument and enclosed in a lipid bilayer

TROPISM
HSV-1 -> cold-sores
HSV-2 -> genital herpes

1153
Q

How is herpes simplex virus treated?

A

Acylcovir

A nucleoside analogue

1154
Q

Outline the influenza virus

A

VIROLOGY
Multipartite single stranded RNA virus
Envelope protein neuraminidase -> release

TROPISM
Nose, throat and bronchi

1155
Q

How is influenza treated?

A

Doesn’t fully work

Neuraminidase inhibitor = oseltamivir

1156
Q

Distinguish between different types of virus and describe how they use the host cell to replicate

A

HIV= retrovirus, leukocytes
Hepatitis= DNA and RNA viruses, hepatocytes
Herpes Simplex= DNA virus
Influenza= RNA virus

1157
Q

Summarise the mechanisms of action of antiretroviral drugs

A

Entry inhibitors= Enfuvirtide and maraviroc

RT inhibitors= nucleoside analogues (Zidovudine) and non-nucleoside analogues (Efavirenz)

Integrase inhibitors= raltegravir

Protease inhibitors= saquinavir

1158
Q

Describe the actions of other (non-antiretroviral) antiviral drugs

A

Nucleotide analogues= ribavirin, acyclovir

Protease inhibitors= boceprevir

Neuraminidase inhibitors= oseltamivir

1159
Q

Outline general seizures

A

Begins simultaneously in both hemispheres of brain
Genetic disorder?

Types of general seizure include:
Tonic-clonic seizures
Absence seizures
Myoclonic seizures

1160
Q

What are the types of general seizure?

A

Tonic-clonic seizures
Absence seizures
Myoclonic seizures

1161
Q

Outline partial or focal seizures

A

Begins within a particular area of brain and may spread out
May be the result of an injury or insult to the brain
More common in adults

Types of seizure include:
Simple or complex partial
Temporal lobe epilepsy

1162
Q

What are the types of partial seizures?

A

Simple or complex partial

Temporal lobe epilepsy

1163
Q

How can you measure brain activity?

A

Electroencephalography (EEG)
Magnetoencephalography (MEG)
Functional magnetic resonance imaging (fMRI)

1164
Q

What are the brain firing patterns?

A

High to low Hz

Gamma: awareness- hyperactive 
Beta: awareness- thinking
Alpha: awareness- relaxed
Theta: drowsiness, meditation
Delta: deep-sleep
1165
Q

What happens to brain firing patterns in seizures?

A

Irregular and asynchronous firing patterns due to neuronal over-activity

1166
Q

What happens in a glutamatergic synapse?

A

VGSC opens -> membrane depolarisation

VGKC opens -> membrane repolarisation

Ca2+ influx through VGCCs ->  vesicle exocytosis
Synaptic vesicle associated (SV2A) protein allows vesicle attachment to presynaptic membrane

Glutamate activates excitatory post-synaptic receptors (e.g. NMDA, AMPA and kainate receptors)

1167
Q

What is carbamazepine?

A

Voltage-gated Na channel blocker

Used for partial seizures and tonic-clonic seizures
Stabilises inactive state of channel

1168
Q

Outline the phamacokinetics of carbamazepine

A

Induces the expression of hepatic enzymes
16-30 hour half-life
Dangerous in individuals with HLA-B*1502 allele

1169
Q

What is phenytoin?

A

Voltage-gated Na channel blockers

Indicated for most forms of epilepsy (not absence)
Class 1b channel blocker

1170
Q

Outline the pharmacokinetics of phenytoin

A

Fast onset (10mins) and long half-life (10-20 hours

1171
Q

What is retigabine?

A

VGKG enhancer

Potassium channel opener specific for KV7 alpha subunit
Only licensed for adjunctive treatment 
Fast onset (30mins); 10h half-life
1172
Q

What is ethosuximide?

A

VGCC blocker

T-type Ca2+ channel antagonist
Mainly used for absence seizures
Long half-life (50 hours)

1173
Q

What is gabapentin?

A

VGCC blocker

Thought to inhibit a2d subunit
Indicated for partial seizures

1174
Q

What is levetiracetam?

A

Anti-convulsant

Affects glutamate exocytosis
Binds to synaptic vesicle associated protein (SV2A)  preventing glutamate release
Monotherapy for focal seizures
Fast-onset (1 hour), half-life (10 hours)

1175
Q

What is topiramate?

A

Anti-convulsant

Inhibits GluK5 subunit of kainate receptor
Also affects VGSCs and GABA receptors
Indicated for most types of epilepsy
Fast-onset (1 hour), long half-life (20 hours)

1176
Q

What is perampanel?

A

Anti-convulsant

Selective inhibitor of AMPA receptor
Only licensed for as an adjunct for partial seizures
Fast-onset (1 hour), long half-life (24 hours)

1177
Q

Give an overview of drugs that interfere with glutamatergic synapse?

A

VGSC antagonist: e.g phenytoin, carbamazepine

VGKC enhancer: retigabine

VGCC antagonist: ethosuximide (T-type antagonist); gabapentin (2 blocker)

SV2A inhibitor: Llvetiracetam

Glutamate receptor antagonist: perampanel (GluA), topiramate (GluK)

1178
Q

What happens in a GABAergic synapse?

A

GABA can be released tonically and also following neuronal stimulation

GABA activates inhibitory post-synaptic GABAA receptors

GABAA receptors are chloride (Cl-) channels -> membrane hyperpolarisation

GABA is taken up by GAT

GABA is metabolised by GABA transaminase (GABA-T)

1179
Q

Outline clonazepam

A

Anticonvulsant
GABAAR drug
Benzodiazepine (BZD) indicated for ALL forms of epilepsy
Fast-onset (2h), long half-life (30h)

1180
Q

Outline phenobarbital

A

Anticonvulsant
GABAAR drug
Indicated for most forms of epilepsy except absence seizures
Acts as a sedative in adults and may cause behavioural disturbances in children
Interacts with numerous drugs
Very fast-onset (20mins), long half-life (60h)

1181
Q

Outline tiagabine

A

Anticonvulsant
Selective inhibitor of GAT-1 (GABA transporter)
Adjunctive treatment for partial seizures
Fast onset (45min), short half-life (6h)

1182
Q

Outline sodium valproate

A

Anticonvulsant
Indicated for ALL forms of epilepsy
Inhibits GABA transaminase
Fast onset (1h), half-life (12h)

1183
Q

Outline vigabatrin

A

Anticonvulsant
Irreversibly inhibits GABA transaminase enzyme
Monotherapy for infantile spasm or as an adjunct for partial seizures

1184
Q

What drugs are used for tonic-clonic seizures?

A

Valproate

1185
Q

What drugs are used for absence seizures?

A

Valproate

1186
Q

What drugs are used for myoclonic seizures?

A

Topiramite
Ethosuximide
Valproate

1187
Q

What drugs are used for partial or focal seizure seizures?

A

Carbamazepine

1188
Q

How can you treat epilepsy?

A

Glutamate inhibition= VGSCs, VGKCs, VGCCs, SVA2 and GluRs

GABA enhancement= GABARs, GABA transporter and GABA transaminase