Pharmacology Flashcards

(220 cards)

1
Q

define pharmacokinetics

A

what the body does to the drug (process of absorption and distribution)

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

what happens when a drug is administered?

A

absorbed into the blood stream to systemic circulation where it is distributed to tissues

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

define pharmacodynamics

A

the effects of the drug on the body

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

what happens when the drug reaches it’s site of effect?

A

there is pharmacological effect leading to the clinical response

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

what two effects are seen in the clinical response to a drug?

A

toxicity

therapeutic effect

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

what does toxicity describe in relation to clinical response to a drug?

A

the side effects

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

when may an animal be given a drug even when there are contraindications?

A

if the quality of life is sufficiently affected then a cost benefit analysis will indicate that it is worthwhile giving the drug

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

what are the 2 key ways in which drugs work?

A

non-cellular mechanisms

cellular mechanisms

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

what are the 4 main non-cellular ways in which drugs work?

A

physical effects
chemical reactions
physicochemical mechanisms
modification of body fluid composition

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

why do fluids work through non-cellular mechanisms instead of cellular mechanisms?

A

because the effects that they have on cells isn’t direct, it is instead mediated by osmosis that is caused by changes if fluid composition

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

give an example of a drug with physical effects

A

eye lubricant

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

give an example of a drug which works to modify body fluid composition

A

hypertonic saline

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

what are the 4 main cellular mechanisms in which drugs work?

A

physicochemical/biophysical mechanisms
cell membrane structure and function modification
enzyme inhibition
receptor mediated effects

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

give an example of drugs which affect cell membrane structure and function

A

insulin

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

give an example of drugs that work through enzyme inhibition

A

NSAIDs

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

give an example of drugs that work through receptro mediated effects

A

opioids

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

what are the targets for drug action?

A
receptors
enzymes
transporters
ion channels
nucleic acid
miscellaneous
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

how may receptors aid drug action?

A

transduce signal from drug to produce conformational change and influence flow of ions

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

how can enzymes be affected by drug action?

A

may be activated or inhibited

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

how are ion channels affected by drug action?

A

open or close

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

how are transporters influenced by drug action?

A

carry a molecule across the membrane

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

give an example of receptors which are targeted by drugs

A

mu opioid receptor

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

give an example of enzymes which are targeted by drugs

A

COX 1 and 2

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

give an example of a drug that may be moved by transporters

A

insulin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
give an example of a drug that affects ion channels
local anaesthetics
26
define ligand
a substance that forms a complex with a receptor (biomolecule) to serve a biological purpose (e.g. analgesia in the case of opioids)
27
what is the role of a receptor?
interacts with extracellular physiological signals and converts them to intracellular effects
28
describe the 3 step process involved in a receptor converting extracellular signalling to intracellular effects
receives a signal transduces signal to effector mechanism
29
what are the 2 ways receptors will be targeted by drugs?
agonist or inverse agonist | antagonist
30
what are the main effects of agonists/inverse agonists on receptors?
direct through ion channel opening or closing | transduction mechanisms leading to enzyme activation/inhibition, ion channel modulation and DNA transcription
31
what is an example of a receptor agonist/inverse agonist?
dexmedetomidine
32
what is the effect of antagonists on receptors?
prevents binding of other agonists/antagonists | produces no effect and blocks endogenous mediators
33
give an example of a receptor antagonist
atipam
34
what are the 2 effects of drugs on ion channels?
blockers | modulators
35
what is the effect of blockers on ion channels?
permeation blocked so no entry or exit
36
what is the effect of modulators on ion channels?
increased or decreased opening probability
37
where do modulators and blockers sit within ion channels?
blockers sit within the channel itself | modulators sit in the sub-unit
38
what are the 3 ways drugs can affect enzymes?
inhibition false substrate prodrug
39
how does a false substrate affect the enzyme?
is structurally similar to substrate but not the same so an abnormal metabolite is produced
40
how does prodrug worth with an enzyme?
makes use of the enzyme function to convert prodrug to active drug. An active drug is produced by the reaction
41
how do transporters work to produce drug effects?
normal transport to allow drug to enter cell inhibition of transporter to block transport false substrate leading to accumulation of abnormal compounds
42
what are the 4 main types of receptors?
ligand gated ion channels G-protein coupled receptors kinase linked receptors nuclear receptors
43
what are the fastest acting receptors?
ligand gated (milliseconds)
44
what are the slowest acting receptors?
nuclear receptors (hours)
45
how do ligand gated ion channels work?
lead to hyperpolarisation or depolarisation and therefore cellular effects
46
how do G-protein coupled receptors work?
change in cell excitability leads to activation of second messengers which cause protein phosphorylation. This leads to cellular effects
47
how do kinase linked receptors work?
receptor phosphorylates protein leading to gene transcription, protein synthesis and then cellular effects
48
how do nuclear receptors work?
receptors in the nucleus cause gene transcription this then leads to protein synthesis and cellular effects.
49
give an example of a ligand gated ion channel
nicotinic ACh receptor
50
give an example of a G-protein coupled receptor
muscarinic ACh receptor
51
give an example of a kinase linked receptor
cytokine receptor (linked to inflammation)
52
give an example of nuclear receptors
oestrogen receptors
53
define affinity
how well/avidly a drug binds to it's receptor
54
define intrinsic activity/ efficacy
magnitude of a drugs effect once bound (0-1)
55
what is a full agonist able to do once bound to the receptor?
generate a maximal response
56
describe the affinity and intrinsic activity of full agonists
high affinity and high intrinsic activity
57
give an example of a full agonist drug
morphine is a full agonist at the mu opioid receptor
58
what type of receptor is a mu opioid receptor?
G-protein coupled receptor
59
what is a partial agonist drug?
one that has intrinsic activity of less than 1
60
what does receptor occupancy of a partial agonist drug lead to?
a sub maximal effect even if receptors are fully bound
61
give an example of a partial agonist drug
buprenorphine binds to the mu opioid receptor but does not produce a maximal effect even if the dose is increased
62
what is an inverse agonist?
drug binds and exerts an opposite effect to the endogenous agonist
63
give an example of an inverse agonist
antihistemines
64
define antagonist
exhibits affinity but no intrinsic activity
65
give an example of an antagonist
atipam antagonises a-2 adrenoreceptor against dexmedetomidine
66
define potency
the dose of a drug required to produce a response (more potent = less drug required for response)
67
what is therapeutic index a measure of?
maximum non-toxic dose divided by minimum effective does
68
what is the issue with using therapeutic index?
based on data that may not be clinically relevant effective dose can be extremely variable depending on what is being treated doesn't account for idiosyncratic reactions
69
what does ADME stand for?
absorption distribution metabolism excretion
70
what is ADME involved with?
what the body does to the drug
71
why is it important that drugs can cross cell membranes?
to enable them to reach their site of action so they may have an effect
72
what are the 5 main ways that drugs cross cell membranes?
``` aqueous diffusion passive lipid diffusion facilitated diffusion pinocytosis active transport ```
73
what is the law which governs the rate of transfer of drug across biological membranes?
Fick's law of diffusion
74
what are the 5 elements of Fick's law of diffusion?
``` diffusion coefficient surface area of tissue membrane thickness partition coefficient concentration gradient ```
75
what is the partition coefficient?
how drugs distribute themselves in water vs in oil
76
what sort of molecules are drug molecules usually?
small weak acids or weak bases
77
what is the level of ionisation of a drug determined by?
pKa of drug and pH of the surrounding tissue
78
how do non-ionised drugs cross biological membranes?
passive diffusion
79
how do ionised drug molecules cross biological membranes?
selective transport mechanisms, facilitated diffusion or pinocytosis
80
what process in the body may affect drug absorption?
infection/inflammation can change tissue pH and make it more acidic
81
what is drug absorption mainly influenced by?
route of administration and drug formulation | peripheral shutdown if present
82
define bio-availability
the fraction of a dose reaching the systemic circulation after administration IM compared to the same does administered IV
83
what are the 8 key routes of drug administration?
``` IV IM SC oral inhalational epidural transmucosal (oral and rectal) transepithelial (skin, cornea, nasal mucosa) ```
84
where does drug absorption primarily take place after oral administration?
small intestine
85
what types of drug molecules are usually poorly absorbed through oral administration?
low lipid soluble and strong acids and bases
86
what are the 4 main factors which affect GI absorption?
gastrointestinal motility splanchnic blood flow particle size and formulation physicochemical factors
87
how can gastrointestinal motility affect GI absorption of drugs?
fast and slow can mean that not enough of the drug is absorbed
88
how may splanchnic blood flow be reduced and so reduce drug absorption?
dehydration and reduced BP
89
what is first pass metabolism of drugs?
enzymes in gastrointestinal wall and liver that metabolise drugs
90
what is the fastest route of drug administration?
IV
91
what is challenging about IV drugs?
access can be difficult
92
what influences the rate of IV injection?
peak concentration (faster admin = higher peak concentration)
93
define peak concentration of drugs
maximum serum concentration of a drug in a specific area
94
what can be used to maintain plasma concentrations of IV drugs?
constant rate infusions (CRI) or target controlled infusions (TCI)
95
describe the basic path of IV drugs once administered
right heart - lungs - systemic circulation
96
why is IM injection variable?
local blood flow | diffusion through tissue
97
why may IM injections produce an adverse response?
they are often painful but varies from drug to drug
98
what must happen on administration of IM drugs before injection?
aspirate (draw back)
99
how does absorption of SC injection compare to IM?
slower and more unpredictable
100
what influences absorption of SC injections?
injection site, temperature of skin, dehydration and shock
101
what drugs are given through inhalation?
drugs that can be vapourised or aerosolised
102
what is a benefit of epidural, transmucosal or transepithelial administration of drugs?
direct to site of action so doses can be lower
103
define apparent volume of distribution
amount of drug administered / plasma concentration
104
what are the 5 key factors which determine drug distribution?
``` protein binding tissue binding organ blood flow membrane permeability drug solubility ```
105
what are the 2 proteins involved in plasma protein binding?
albumin | alpha-1 acid glycoprotein
106
what does albumin bind to during plasma protein binding of drugs?
weak acids
107
what does alpha-1 acid glycoprotein bind to during plasma protein binding of drugs?
weak bases
108
what is the effect of plasma protein binding of drugs during distribution?
doesnt elicit a response. It takes up drug and removes it from circulation so preventing it from working on receptors
109
what happens to the unbound/free fraction of the drug that is not plasma protein bound?
can interact with receptors/diffuse across membranes
110
why may plasma protein binding vary from patient to patient?
those with low albumin will have a larger unbound/free fraction
111
what are the 2 types of tissue binding of drugs?
specific and non-specific
112
where are drugs initially distributed to?
organs with high blood flow
113
what is an advantage of drugs being initially distributed to organs with high blood flow?
good for anaesthetics as they go to the brain first
114
what is a disadvantage of drugs being initially distributed to organs with high blood flow?
anaesthetic drugs will negatively affect the heart quickly as this also has good blood flow
115
how does membrane permeability affect drug distribution?
higher membrane permeability = faster effect
116
what may happen to highly lipid soluble drugs?
accumulate in fat and then be released as the fat is metabolised. This can lead to the 'hangover' effect and keep the plasma concentration of a drug higher for longer leading to prolonged recovery
117
what must be assumed about all drugs with regards to pregnant or lactating animals?
drugs will cross placenta and be secreted into milk
118
define drug clearance
the volume of plasma from which a drug is completely removed per unit time
119
define half life of a drug
the time taken for plasma drug concentration to fall to 50% of it's initial value
120
what is the equation used to calculate drug half-life?
K= clearance / volume of distribution
121
what is the termination of drug effects caused by?
primarily biotransformation then excretion
122
where does biotransformation usually occur?
in the liver
123
where does excretion of drugs usually happen?
the kidneys
124
what compounds does the kidney excrete most easily?
polar water soluble compounds
125
why is biotransformation in the liver so crucial?
most drugs are lipophillic and highly plasma protein bound which the kidneys cannot excrete easily. Biotransformation converts them into easily excreted molecules
126
what is the primary organ of metabolism?
the liver
127
what are the 2 stages of hepatic metabolism?
I: conversion of drug to polar metabolite II: conjugation with substrates
128
what reactions are used in phase I of hepatic metabolism?
oxidative, hydrolytic or reductive
129
what is required in phase II of hepatic metabolism?
energy
130
what is the aim of phase II hepatic metabolism?
to make resultant polar compound more readily excreted
131
what reaction does not occur in phase II hepatic metabolism of cats that can cause longer lasting effects of propofol?
glucuronidation
132
what reaction does not occur in phase II hepatic metabolism of dogs?
acetylation
133
what are the 4 effects of hepatic metabolism?
drugs may be activated drugs may have active metabolites drugs may have toxic metabolites enzymes involved in metabolism may be inhibited or induced (long term drug use)
134
what type of excretion of drugs is most common?
renal excretion
135
what are the 3 other types of excretion of drugs from the body?
biliary exhalation GI tract
136
what are the 2 types of renal excretion of drugs?
active | passive
137
where does active renal excretion of drugs occur?
tubular secretion
138
where does passive renal excretion of drugs occur?
glomerular filtration
139
what may need to happen to drug doses in animals with renal compromise?
may need to be altered
140
what happens during enterohepatic circulation of drugs?
drug is metabolised (often through glucuronidation) billiary excretion deglucuronidation by gut microflora reabsorption from gut
141
what can be explained by enterohepatic circulation of drugs?
longer lasting effects of drugs
142
how long does enterohepatic circulation continue for?
until plasma concentration drops
143
how may antimicrobial agents be classified?
according to the type of organism against which they are effective
144
what do bacteriostatic antimicrobials do?
arrest bacterial multiplication so stop proliferation and allow individuals immune system to clear the infection
145
what do bacteriocidal antimicrobials do?
act by primarily killing bacteria so useful for immunocompromised animals
146
what is the outcome of concentration dependent antibiotics related to?
peak antibiotic concentration achieved at the site of infection relative to the MIC
147
what is MIC?
minimum inhibitory concentration (HIGH DOSE)
148
what is the outcome of time dependent antibiotics related to?
concentration of drug being kept above MIC for high proportion of time between each dose (SUSTAINED DOSE)
149
what are the 3 mechanisms of action of antibiotics?
interference with cell wall leading to cell rupture due to osmotic pressures interference with structure of the plasma membrane interference with microbial DNA or its replication or repair
150
what are the 6 principles of antimicrobial therapy?
``` make a diagnosis remove barriers to cure decide whether chemotherapy is necessary select the best drug administer the drug in optimum dose and frequency and by the optimum route test for cure ```
151
how should be the best drug be selected?
specificity pharmacokinetic factors the patient
152
what are pharmacokinetic factors?
right dose at the right time
153
what could be involved in removing barriers to cure?
holistic approach e.g. debriding wound
154
what affects the breakdown of local anaesthetics?
whether there are ester and amide linkages
155
where are side effects of local anaesthetic seen?
CVS | CNS
156
when are the 5 main occasions for local anaesthetics to be used?
part of balanced anaesthesia (e.g. epidural under GA) Anaesthesia in a standing/conscious patient Post-operative pain relief Desensitisation for procedures (e.g. EMLA) Lameness investigation in horses
157
what is the difference between epidural and spinal anaesthesia?
epidural - injected into the epidural space via a catheter | spinal - injected directly into CSF via a much smaller needle
158
give an example of when local anaesthesia may be used
infraorbital block for dental surgery
159
give an example of when topical local anaesthesia may be used
desensitisation of feline larynx prior to endotracheal intubation
160
what is potency a measure of in pharmacology?
drug activity expressed in terms of the amount required to produce an effect of given intensity
161
what increases as local anaesthetic potency increases?
toxicity
162
what 4 factors affect the duration of local anaesthetic action?
ease of penetration into nerve cell and amount of drug reaching the sodium channel strength of binding to the sodium channel speed of removal metabolism of the local anaesthetic
163
what is ease of penetration into nerve cell and amount of drug reaching the sodium channel of local anaesthetics due to?
lipid solubility
164
what is speed of removal of local anaesthetic dependent on?
tissue perfusion
165
what may be added to local anaesthetics to reduce speed of removal?
vasoconstrictors (e.g. adrenaline)
166
how are ester links in local anaesthetics hydrolysed?
plasma esterases
167
does CSF contain esterases?
no - esters cannot be broken down there and must diffuse out into other tissues
168
what are amide linkages broken down by?
cytochrome P450 enzymes
169
what may prolong or limit metabolism of amide local anaesthetics?
hepatic disease
170
what can increase the breakdown of amide local anaesthetics?
drugs such as barbiturates induce enzymes which will increase breakdown
171
what can decrease the breakdown of amide local anaesthetics?
drugs that inhibit the P450 enzymes e.g. midazolam
172
what are the 3 main formulations of Lidocaine?
sterile solutions for parenteral use aerosol sprays for nasal/airway use topical patches
173
how water soluble are local anaesthetics?
poor
174
how is the low water solubility of local anaesthetics over come?
making a salt solution
175
what is the effect of making a salt solution to increase water solubility of local anaesthetics?
lowers pH of solution if a hydrochloride salt which can cause stinging on injection
176
what is baricity?
weight of one substance compared with the weight of an equal volume of another substance
177
what is the baricity comparator for spinal anaesthesia?
CSF
178
why is baricity essential in spinal local anaesthetics?
because we don't want anaesthetics to spread higher into epidural space, as it would effect respiration
179
how are solutions made heavier to address issues baricity in local anaesthetics?
glucose is added
180
what are the key effects of vasoconstrictors in local anaesthetics?
reduces risk of toxicity reduces bleeding at the injection site reduces speed of systemic absorption and so prolongs duration of action
181
what happens when local anaesthetics are absorbed systemically?
they are available for systemic effects
182
what must happen to a local anaesthetic drug for it to be systemically active?
must be unbound and unionized
183
what types of local anaesthetics have a longer duration of action and lower risk of toxicity?
those which bind more easily to plasma proteins
184
what is the effect of lower pH on local anaesthetic binding?
reduces affinity for protein binding and increases risk of toxicity
185
what are the 2 levels of toxicity of local anaesthetics?
neurotoxicity | cardiovascular toxicity
186
what must be considered when injecting local anaesthetic?
dose injection site care with smaller patients
187
what effect does site of injection of local anaesthetic have?
determines the rate of uptake into the systemic circulation
188
at what concentrations of local anaesthetic is CNS toxicity often seen?
lower than CVS
189
what are the symptoms of generalised CNS depression that are proportional to the unbound drug?
minor behavioral changes muscle twitching and tremors tonic-clonic convulsions CNS depression/respiratory depression and death
190
how is CNS toxicity treated?
symptomatic treatment Benzodiazapines to control seizures O2 supplementation intubation and control of ventilation if respiratory seen/needed
191
what are the 2 main symptoms of CVS toxicity due to local anaesthetic?
hypotension | dysrhythmias
192
how does CVS toxicity from local anaesthetic lead to hypotension?
depression of myocardial activity direct relaxation of vascular smooth muscle loss of vasomotor sympathetic tone
193
how does CVS toxicity due to local anaesthetic cause dysrhythmias?
commonly seen with bupivacaine lipophilicity means that there is rapid entry to open sodium channels during systole drug remains bound to sodium channel during diastole presents as re-entrant arrythmias
194
how is CVS toxicity from local anaesthetic treated?
symptomatic treatment manage bradycardias with an anticholinergic fluid therapy with inotropic support if needed intralipid IV could be used to mop up local anaesthetic
195
how can local anaesthetic toxicity be prevented?
don't exceed 'safe' maximum dose if a greater volume is needed dilute the local anaesthetic with 0.9% NaCl use appropriately sized syringes to draw up dose (accuracy) use appropriately sized needles to minimise tissue trauma aspirate before injection to confirm you are not in a blood vessel
196
what volatile agents are licensed for use in dogs and cats?
isoflurane and sevoflurane
197
what was suggested by CEPSAF to do with gas induction of anaesthesia?
gas induction was associated with increased risk of anaesthetic related death
198
describe the characteristics of the ideal inhalational agent
``` non-irritant to mucous membranes minimal effects on CVS and respiratory function rapid uptake and elimination non-toxic non-flammable and chemically stable easily vaporised good analgesia and muscle relaxation ```
199
what does MAC stand for?
minimum alveolar concentration
200
what is MAC?
concentration of VA required to prevent response to supramaximal noxious stimuli
201
what can MAC be used to compare?
potency of agents
202
what is the difficulty of using MAC as a reliable measure of potency?
measured in a lab and there are factors which affect it
203
what factors can MAC be reduced by?
``` other drugs (e.g. premed) hypothermia pregnancy extremes of age (geriatric and neonatal) hypothyroidism ```
204
what factors can MAC be increased by?
hyperthermia neonates hyperthyroidism administration of catecholamines and sympathomimetics
205
define vapour
gaseous state of a substance that at ambient temperature and pressure is a liquid
206
define gas
exists in gaseous form at room temperature at sea level
207
define partial pressure
pressure that individual gas exerts in a mixture of gases
208
define partition coefficient
ration of the concentration of a compound in two solvents at equilibrium
209
how does concentration of VA in inspired air affect uptake of anaesthetic gases?
drugs move down their concentration gradient, higher concentration of inspired air VA will lead to faster uptake
210
what is the mechanism of action of anaesthetic agents?
unclear - site of action is the brain
211
how does alveolar ventilation affect uptake of anaesthetic gases?
good alveolar ventilation leads to enhanced uptake of agent
212
how does alveolar ventilation affect recovery?
increased alveolar ventilation improves recovery as anaesthetic gas is blown off
213
is panting effective in alveolar ventilation?
no - just moves animals dead space air so exchange is not occurring
214
how does blood gas solubility affect distribution of anaesthetic gases?
lower blood gas solubility leads to faster onset and recovery from anaesthesia
215
how does cardiac output affect distribution of anaesthetic gases?
lower cardiac output leads to faster onset
216
how does blood tissue solubility affect distribution of anaesthetic gases?
impacts how well distributed agent is (increased solubility = better distribution)
217
how can concentrations of anaesthetic gases be reduced so their side effects are reduced?
provision of effective analgesia so that less VA is required during surgery to keep patient comfortable
218
what are the main side effects of inhalant anaesthetics?
dose dependent respiratory and cardiovascular depression
219
what is the role of balanced anaesthesia?
reduction of side effects while benefiting from desired effects of drugs
220
why is monitoring of anaesthetic so essential?
supportive therapy of any issues can be provided quickly (e.g. fluids or ventilation)