Pharmacodynamics & Pharmacokinetics Flashcards

1
Q

What is the definition of a drug?

Agonist?

Antagonist?

Partial agonist?

How do drugs produce an effect?

On what 4 proteins can drugs have an effect?

A

Chemical agent affecting a biological process

Drug that binds to receptor to produce a biological response

Binds to receptor but does not produce a biological response (basal level)

Binds to receptor & produces small biological response but less than agonist

Interacting with endogenous proteins

  1. Receptors
  2. Transporters
  3. Enzymes
  4. Ion channels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What are the 4 classes of receptors in a cell?

What are the 2 types of ligands that bind to which receptors?

A
  1. Ligand-gated ion channel (ionotropic)
  2. G-protein coupled receptors (metabotropic)
  3. Kinase-linked receptors (dimerise on activation)
  4. Nuclear receptors

Hydrophilic - 1, 2, 3
Hydrophobic - 4 (diffuse across plasma membranes)

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

What are the 2 types of ion selectivity of ionotropic receptors?

What is the structure of the nAChR?

How many times does it span the membrane?

What happens once ACh binds?

What happens to the state of the cell?

So what type of NT is it?

A
  1. Cationic (Ca2+, Na+, K+)
  2. Cl-

made up of 2 alpha subunits, gamma subunit, beta subunit & delta subunit to form a-helices central pore/gate

4 times

2 ACh bind at alpha subunits to change conformation to allow Na+ flow into cell

Depolarises (becomes more positive)

Excitatory

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

What is the structure of the GABA-A ionotropic receptor?

Where are the 3 binding sites?

What happens on its activation?

What happens to the cell?

Hence what kind of response?

What structure do both nAChR & GABA receptors have in common?

A

2 alpha, 2 beta & 1 gamma subunit

2x GABA binding site at a-ß interface
1x benzodiazepine site at a-gamma interface

Changes shape of channel such that chloride ions allowed flow into the neurone

Hyperpolarised

Inhibitory (brain)

Alpha helices spanning the membrane with exterior N-terminal tail with ligand binding domain & C-terminal tail on exterior too

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

What is the structure of the glutamate superfamily of ionotropic receptors? 4 things

What is a type of glutamate receptor?

What is the Q/R-site?

What are the characteristics of the glutamate receptors? 3 things

What kind of response is produced?

A

Polypeptides that span the membrane 3 times, has 2 glutamate binding sites exteriorly, flip/flop region exteriorly and interior Q/R-site (between TM1-2)

AMPA

Selectivity filter to close ions from passing through membrane to regulate the polarisation state of the membrane

  1. Very rapid as ligands are the effectors at post-synaptic membrane
  2. Not useful for signalling downstream as very localised at membrane
  3. 1ms speed

Excitatory & inhibitory

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

How are GPCR characteristics different to ionotropic? 2

Why are GPCRs so important in the pharmaceutical industry?

What is the structure of a GPCR?

How are G-proteins activated?

How is the G-protein reassembled?

What are the 4 classes of G protein & what do they do?

How does PKA regulate gene expression?

A
  1. Slower of 100ms - minutes
  2. Extrasynaptic - can regulate gene expression, create new synapses

Associated most with disease - pain, asthma, obesity, cancer etc so most drugs target these

7-transmembrane domain spanning the membrane - with glycosylation at N-terminal extracellular domain & G-protein coupling domain internally

Ligand binding promotes exchange GDP to GTP on alpha subunit so alpha & beta-gamma dissociate

Alpha subunit has GTPase activity (like a GAP) so hydrolyses GTP into GDP & re-associates with beta-gamma

Gs = AC stimulation
Gi = AC inhibition
Go = Ca2+ & K+ ion channel regulation
Gq = PLC-ß stimulation
G12/13 = activates small GTPases to regulate actin cytoskeleton

Regulates transcription factor CREB which binds to CRE promoter to regulate expression of genes included in long-term memory

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

What ligands bind to tyrosine kinase receptors?

What pathway do they regulate?

What does the pathway do? 2 things

What is the cascade of the pathway from tyrosine kinase ligand binding? 7

A

Growth factors

MAP pathway

Change protein activity (phosphorylation) & gene expression (pro-survival & cell proliferation)

  1. Receptor dimerises & kinase domains phosphorylate tyrosine residues on intracellular domain
  2. SH2 domains of GRB2-Sos binds to pY
  3. Sos of GRB2-Sos acts on Ras-GDP & becomes Ras-GTP
  4. Ras-GTP phosphorylates Raf (MAPKKK) for its activation
  5. Raf kinase phosphorylates MEK kinase
  6. MEK phosphorylates MAPK x 2 to form active homodimer
  7. MAPK phosphorylates Rsk
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What G protein activates PLC-ß?

What is the signalling cascade of PLC-ß in 3 steps?

What is the significance of lithium in this cycle?

What is lithium used to treat?

A

Gq

  1. Converts PIP2 into IP3 & DAG
  2. IP3 activates Ca2+ channels in ER causing release from ER into cytoplasm - Ca2+ involved in membrane hyperpolarisation (-ve), muscle contraction, enzyme activation
  3. DAG activates PKC which phosphorylates GSK-3

Inositol-1-phosphatase is inhibited by Li (breakdown from IP -> inositol) therefore increasing intracellular IP3/DAG signalling

Bipolar disorder - treats mania

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

What enters through nuclear receptors?

What are the effects?

How long does it take to have an effect?

What happens when the drug is stopped being taken?

A

Drugs to alter gene expression

New proteins are synthesised/not synthesised

Steroids take ~3 weeks

Takes time to reverse to basal state & to degrade protein

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

What is the definition of a ligand?

Agonist?

Antagonist?

Affinity?

Potency?

Efficacy?

Inverse agonist?

What is the difference between a full agonist and partial agonist?

Reversible competitive antagonist?

Irreversible antagonist?

A

Specific molecule that binds to a receptor

Drug that binds to receptor & produces pharmacological effect

Drug that binds to receptor & produces no direct cellular effect (no pharmacological response - blocks agonist action)

How tightly a drug binds to a receptor

Concentration of a drug that gives 50% response - ED50

Concentration of a drug that gives 100% maximal response

Reduces basal level response - full R vs R*

Full agonist = equilibrium shift to R* (active) completely - no R binding
Partial agonist = binds to both R and R* but more R* (active)

Drug binds reversibly to R & effects can be overcome by increasing agonist concentration

Antagonist dissociates slowly/not at all - no change in antagonist occupancy of receptors when [agonist] increased

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

What are the 6 processes of drugs in the body?

What can administration route affect? 3 things

How does route of administration affect drug absorption? 4 things

A
  1. Administration (in)
  2. Absorption
  3. Distribution
  4. Biotransformation
  5. Inactivation/elimination
  6. Excretion (out)

kk

  1. How much drug enters body
  2. How much drug enters site of action
  3. Magnitude of drug effect

kk

  1. Absorbing surface
  2. Number of membranes to cross
  3. Amount of drug destroyed e.g by metabolism
  4. Amount of depot binding
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are the pros of oral administration? 5

What are the cons of oral administration? 3

What are the pros of intravenous administration? 3

What are the cons of intravenous administration? 3

What is the pro of intramuscular administration?

What are the cons of intramuscular administration? 2

What are the pros of subcutaneous inhalation? 4

What are the cons of subcutaneous inhalation? 2

A

Safe, done at home, no needles, economical, don’t need sterile environment

Slow & variable absorption from gastrointestinal tract, subject to first-pass metabolism (stomach pH), less predictable blood levels

Rapid absorption, accurate blood concentration, easy to control dose

Overdose danger, can’t be reversed readily, requires sterile & medical techniques

Slow & even absorption

Irritation at site injection, needs sterile technique

Slow prolonged absorption, large absorption surface (lungs), rapid onset, no injection equipment

Irritation nasal passage, dangerous/small particles can be inhaled & harm lungs

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

What is topical administration?

What are the pros of topical administration? 2

What are the cons of topical administration? 1

What is transdermal administration?

What are the pros of transdermal administration? 1

What are the cons of transdermal administration? 2

What is epidural administration?

What are the pros of epidural administration? 2

What are the cons of transdermal administration? 3

What is intranasal administration?

What are the pros of intranasal administration? 5

What are the cons of intranasal administration? 2

A

Applying drug to a surface e.g skin (cream)

Localised action & effect, easy self-administration

Can be absorbed into general circulation

Place patch onto upper epidermis/skin

Controlled & prolonged absorption

Local irritation, only lipid soluble drugs (phospholipids)

Injection into epidural space around spinal cord

Bypasses blood-brain barrier, rapid CNS effect

Irreversible, needs anaesthetist, possible nerve damage

Administered by way of nasal structures

Easy to use, local & systemic effects, rapid, no first-pass metabolism, bypasses blood-brain barrier

Not all drugs can be atomised, irritation nasal mucosa

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

How does diffusion & transportation across membranes affect absorption?

How does lipid solubility affect absorption?

How is lipid solubility measured? When it is higher what does this mean?

How does ionisation affect absorption?

What is the pH in the gastrointestinal tract?

What does the lower pKa value mean?

A

Concentration from high to low - potentially no diffusion gradient

Membrane made up of phospholipids - drugs of high lipid solubility can passively diffuse through membrane so absorption increased

Partition coefficient - higher permeability

Drugs that ionise in water (e.g NaCl into Na+ Cl-) depending on pH and pKa - such that if they are highly charged will not pass through the membrane & hence poorly absorbed from gastrointestinal tract

5-6 - where ionisation happens

Higher the Ka so stronger brønsted acid & hence greater ability to donate protons - so ionised at lower pH . pKa = pH where 50% drug is ionised

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

What happens when the pH is kept the same in the environment?

When it is changed?

Therefore what is absorption going to be like when the pH is similar to pKa?

If aspirin has a pKa of 3.5, where is it most likely going to be absorbed and why?

What would happen to aspirin at a higher pH?

A

Less ionisation, greater lipid solubility so greater absorption

More ionisation, less lipid solubility, less absorption

Less ionisation, more lipid solubility & more absorption

More readily absorbed in the stomach as pH ~ 2 - high [H+] means protonated aspirin (non ionised as COOH group) so can cross hydrophobic lipid membranes into bloodstream

Readily donates protons, so COOH -> COO- and is ionised

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

For a drug in the blood to enter the brain, it must: 3 things

What is the blood-brain barrier for?

Why can some drugs not pass the blood-brain barrier?

What are the 5 different pharmacokinetic factors determining drug action?

A
  1. Be un-ionised at blood pH (no charge)
  2. High partition coefficient (lipid soluble)
  3. Or use existing transport mechanisms - e.g drug that mimics a neurotransmitter through those transporter mechanisms like LDOPA (looking like dopamine)

Protecting neurones from systemically circulating cytotoxic agents

Not lipid soluble & ionised at blood pH (7.4)

  1. Route of administration
  2. Absorption/distribution
  3. Binding - receptors & plasma proteins (depot binding) or stored temporarily in bones/fat
  4. Inactivation/biotransformation
  5. Excretion - liver metabolites with urine/faeces
16
Q

How does depot binding of a drug affect its action?

How does depot binding of a drug affect its interactions?

What is an example of this?

How does depot binding of a drug affect its termination?

What affects depot availability?

A

Drug binds to depot sites/silent receptors & reaches unbound <=> bound receptor equilibrium - when drug leaves bloodstream it is released slowly from depot sites so has a slower effect/action (lower blood conc) on patients with larger capacity depot sites

Some drugs bind to depot sites - meaning that if they are already occupied by a different drug - taking a second will show more effects as they are less likely to bind to the depot sites as they are already occupied & blood conc of the 2nd drug will be higher

Valine and imipramine

Thiopental is very lipid soluble & moves across blood-brain barrier quickly - but blood levels fall quickly as a result (blood conc) due to depot binding so moves back from brain into blood to maintain equilibrium - but within 30 minutes 90% initial levels lost to depot sites

Age, sex, body size

17
Q

What is drug biotransformation?

What type of metabolism is it?

What factors affect biotransformation? 6

What is the result of metabolism?

What can active metabolites do?

What is the definition of the half-life?

What is first-pass metabolism?

What is the impact of this?

A

Drug metabolism in liver - breakdown, detoxification & removal

Oxidation to become water soluble

Age, metabolism, disease, pregnancy, genetic polymorphisms (enzymes), environment (psychological)

Metabolites - formed by enzyme interacts altering drug composition - have unique properties different effects from original drug which can be helpful, harmful or neutral

Last longer than original drug & influence rate at which other drugs metabolised - impact drug interactions & tolerance

Time taken for one half of an administered drug to be lost through biological processes

Orally administered drugs go directly from gastrointestinal tract to the liver before entering the bloodstream where cytochrome P450 transform it (oxidation) into water-soluble metabolites for excretion

Influences plasma level of drugs & therapeutic dosage

18
Q

eDescribe first-order kinetics of drug metabolism

Describe zero-order kinetics of drug metabolism

How does grapefruit juice affect first-pass metabolism?

What 5 factors affect drug elimination?

A

Rate of metabolism is proportional to plasma concentration of the drug (exponential) - e.g 50% removed each time frame

Rate metabolism constant regardless of the amount of drug taken - e,g alcohol/ethanol & some anti-depressants

Inhibits CYP450 3A4 & hence biotransformation & causes drug accumulation

  1. Renal function - acidic drug in alkaline urine makes drug ionised & so more easily soluble in water for excretion - hence acidic urine & acidic drug can facilitate re-absorption into blood (poisoning)
  2. Hepatic function - liver disease (hepatitis) & cytochrome P450 functioning
  3. Metabolites can interfere with renal excretion (change pH)
  4. Drug effects of same drug - e.g diuretics helps remove water from blood & release sodium into urine
  5. Polypharmacy - effects of additional drugs - neutralising stomach acids with antacids & increase pH urine
19
Q

What does excretion do & prevent?

What is the main site of elimination & 4 others?

How does the liver biotransform drugs?

How can you acidify urine to allow excretion of amphetamine in an overdose?

A

Terminates effect of drug & prevents accumulation of it and its metabolites

  1. Renal/kidneys
  2. Liver/hepatic
  3. GI tract
  4. Lungs
  5. Exocrine glands e.g sweat, saliva, milk

Into ionised/water-soluble so inhibits re-absorption as not lipid-soluble (through membranes) so trapped in kidney tubules & excreted with urine

Administrating intravenous ammonium chloride - therefore amphetamine (alkaline) is ionised in acidic tubular urine & more likely to be excreted as water-soluble & shortens duration overdose