Pharmo Flashcards

1
Q

What are some examples of problems with the patient and population that can result in poor prescribing?

A

Rapid patient turnover
Increased complexity of medical care
Increasingly older patients- more co-morbidities, on more medication, higher risk of side effects
Too many medical students needing to be trained with to enough staff

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

What are some examples of problems with the doctor that can result in poor prescribing?

A

No room for error
Expected to be perfect from day 1
Varying medical school experience- level of teaching or examination
On call medicine- results in sleep deprived, accident prone doctors
On call doctors- don’t know the patient, have to do routine boring jobs
Shift work- lack of continuity of care, working alone more often
Locum- doctors don’t have sufficient training, rules vary differently at different hospitals

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

What are some examples of problems with the pharmaceutical industry that can result in poor prescribing?

A

Vast numbers of new drugs
Clinical evidence for new drugs is usually with selected, healthier patients and/or young volunteers
Some side effects only come to light after the drug is on the market
Blind adherence to guidelines can lead to prescriptions where contraindications or serious interactions exist

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

What two models account for prescription errors?

A

Reasons model of error causation

Swiss cheese model of accident causation

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

What is Reasons model of error causation?

A

Latent conditions –> Error producing conditions –> Active failures –> Accident

(DEFENCES)

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

What is the Swiss cheese model of accident causation?

A

Cheese with holes in act as successive layers of defences, barriers and safeguards to hazards
Holes in the cheese represent active failures and latent conditions
When holes align and hazards persist you get losses

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

What is a latent condition?

A

Problem with the organisational processes and management decisions

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

What is an error producing condition?

A

Problems with the environment, team, individual and task factors that affect performance

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

What is an active failure?

A

Error- slips, lapses, mistakes

Violation

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

What are the defences?

A

Inherent within the system and the individual
Designed to protect against hazards and mitigate consequences of failure
Defences can be inadequate as a result of latent conditions

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

What is an error?

A

Failure of a planned sequence of actions to achieve a desired goal because an adequate plan was incorrectly executed (skill based slip or memory based lapse) or an inadequate plan was executed (rule based or knowledge based mistake)

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

What is a violation?

A

When rules of correct behaviour are consciously ignored

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

What is the basic checklist to reduce error? (15)

A
  1. Consider patient
  2. Correct chart for patient?
  3. Diagnosis and therapeutic aim?
  4. Right drug?
  5. Will illness affect drug distribution/elimination?
  6. Alternative drug?
  7. Patient on non-prescription medication?
  8. Appropriate route of administration?
  9. Correct dosage?
  10. Correct frequency and timing of drug?
  11. Duration of treatment?
  12. Most serious side effects?
  13. TDM required?
  14. How much info/ explanation does the patient need?
  15. Any special prescribing requirements for drug?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What things should we be clear on when deciding the right drug for a patient?

A

Avoid therapeutic duplication
Serious interactions that could lead to failure of treatment
Allergies
Drug spelt correctly
No abbreviation of drug names
Form of the drug- is it correct for the patient?

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

What is the role of the pharmacist in prescription administration?

A

Legal responsibility of pharmacist is to dispense according to prescription- if they suspect an error they must refer it back to the prescriber

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

What is formulary?

A

Previously was a list of formulae for compound medicines

Now is a List of recommended first line drugs for common medical conditions

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

What is the BNF?

A

More comprehensive listing all drugs currently licensed in the UK - widespread use in NHS

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

What three things must a drug show to be listed in the formulary?

A

Efficacy- how effective it is compared to similar drugs/ placebo
Safety- major and minor side effects
Cost- ONLY if efficacy and safety are equally equivalent

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

What is pharmacokinetics?

A

Study of the movement of a drug into and out of the body (what the body does to the drug)

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

What is pharmacodynamics?

A

Study of the drug effect and mechanism of action (what the drug does to the body)

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

What is pharmacogenetics?

A

Effect of genetic variability on the pharmacokinetics/ dynamics of a drug on an individual

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

What are the four broad stages of pharmacokinetics?

A

Absorption
Distribution
Metabolism
Elimination

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

Expand on absorption

A

In what form and route the drug is taken into the body and how this affects its action/effectiveness
How much of the drug is lost on entry into the body

BIOAVAILABILITY

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

What is bioavailability?

A

Fraction of a dose that finds its way into a body compartment (usually the circulation)
100% for IV bolus

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

How is bioavailability calculated?

A

For routes other than IV bolus (F= 100%)
Compare amount reaching the body compartment with IV bioavailability
F = AUC oral / AUC IV

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

What is bioavailability affected by?

A

Absorption- drug formulation, age, food (lipid sol > water sol), vomiting and malabsorption

First pass metabolism - gut lumens, gut wall, liver

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

What is first pass metabolism?

A

Any metabolism occurring before drug enters the systemic circulation
IN gut lumen (gastric acid, proteolytic enzymes, grapefruit juice, insulin), gut wall (P glycoprotein efflux pumps drugs out of intestinal enterocytes and back into the lumen - ciclorporin) and liver (propranolol)

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

Expand on distribution

A

A drugs ability to dissolve in the body

2 key factors- protein binding in systemic circulation and volume of distribution

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

What does protein binding refer to?

A

Drugs bind to albumin (acidic drugs), globulins (hormones), lipoproteins (basic drugs), acid glycoproteins (basic drugs)
Most drugs must be unbound/ free to have an effect

Changes in protein binding causes changes in drug distribution
- only important if: high protein binding, low Vd, narrow therapeutic ratio

Drugs bound or unbound to proteins are still ‘available’!!

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

What is meant by protein binding drug interactions?

A

Protein binding drug interactions - other drugs try to bind and displace drugs bound to proteins

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

What is protein binding affected by?

A

Hypoalbuminaemia
Pregnancy
Renal failure
Displacement by other drugs

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

What is the volume of distribution?

A

Drugs that are not bound to plasma proteins are available for distribution to tissues of the body - tissue fluid, or extensively bound to body tissues
Measure of how widely a drug is distributed in body tissues
Hypothetical but useful with dosing regimens
E.g. 100mg gentamicin dose –> peak plasma conc 5mg/ L has a Vd of 20L

Vd = dose (mg) / peak plasma conc (mg/L) [DRUG]t0
Half life is proportional to Vd

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

Apart from protein binding and Vd, what else is distribution affected by?

A
Specific receptor sites in tissues
Regional blood flow
Lipid solubility
Active transport 
Disease states
Drug interactions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Expand on drug metabolism

A

Phase 1 - make drug more reactive- oxidation, reduction, hydrolysis
Phase 2 - make drug more soluble- conjugation with glucuronic acid, glutathione, sulphuric acid

Either pharmacologically inactive –> active (PRODRUG)
Or pharmacologically active (codeine) –> another active (morphine)

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

What are some examples of CYP450 enzymes involved in phase 1 of drug metabolism?

A

3A
2D6
2C9
2C19

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

Where are CYP450 enzymes made?

A

In the liver (gut/lung)

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

What drugs do 3A enzymes metabolise?

A
Calcium channel blockers
Benzodiazepines 
HIV protease inhibitors
Most statins 
Cyclosporins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What induces 3A?

A

St. John’s wort
Phenytoin
Rifampicin

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

What inhibits 3A?

A

Macrolides
Cimetidine
Grapefruit juice

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

What drugs do 2D6 enzymes metabolise?

A

Codeine
B blockers
Tricyclics

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

What inhibits 2D6?

A

Fluoxetine
Paroxetine
Haloperidol

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

What drugs do 2C9 enzymes metabolise?

A

Most NSAIDs

Phenytoin

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

What induces 2C9?

A

Ethanol

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

What inhibits 2C9?

A

Fluconazole

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

What drugs do 2C19 enzymes metabolise?

A

Diazepam
Phenytoin
Omeprazole

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

What induces 2C19?

A

Rifampicin

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

What inhibits 2C19?

A

Omeprazole

Isoniazid

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

Expand on Elimination

A

Mostly by kidney (also by lungs, breast milk, sweat, tears, bile)
Elimination by the kidney is dependent on: glomerular filtration (affected by unbound drugs like gentamicin), passive tubular reabsorption (affected by urine flow rate and pH- aspirin), active tubular secretion (penicillin)
1st and 0 order kinetics
Clearance

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

What is 1st order kinetics?

A

Rate of elimination of drug is proportional to drug level
Constant fraction of drug eliminated in unit time
Half life can be defined and so multiple dosing is acceptable

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

What is 0 order kinetics?

A

Rate of elimination is a constant
Most drugs show this at high doses because receptors/ enzymes are saturated
Zero order drugs are likely to show toxicity at high doses (fixed rate/unit time) and so small dose change may cause large increase in dose and toxicity
No half life is calculable so will require TDM

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

What are the three broad categories of causes of poor prescribing?

A

Problems with the patient or population
Problems with the doctor
Problems with the pharmaceutical industry

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

What is clearance?

A

ability of the body to excrete drug (=GFR) / rate of elimination
Measure of hepatic, renal and other secondary forms of elimination such as sweating or biliary elimination
Volume of plasma that is completely cleared of the drug per unit time
Low GFR –> low clearance
Half life is inversely proportional to clearance (so lower the clearance or GFR, the higher the half life)

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

What is TDM?

A

Therapeutic drug monitoring

Important when- zero order kinetics, long half life, narrow therapeutic window, increased risk of drug-drug interaction

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

What is multiple dosing?

A

Repeated (first order) drug administration
Reach a steady state (Cpss) in 3-5 half lives (irrespective of dose/ frequency)
Require 4-5 half lived to completely eliminate most of a drug

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

What body systems can affect clearance and how?

A

HRH
HEART- circulation/ cardiovascular factors affecting blood flow to main organs of elimination
RENAL- factors affecting renal elimination (kidney failure, poor perfusion)
HEPATIC- factors affecting hepatic eliminations (enzyme inducers and inhibitors)

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

What is drug half life?

A

Amount of time over which the concentration of a drug in plasma decreases to one half the concentration value that it had when it was first measured

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

How is half life related to clearance and volume of distribution?

A

Half life is directly proportional to volume of distribution
Half life is inversely proportional to clearance

Half life = (0.693 x volume of distribution) / clearance

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

What is the therapeutic window?

A

Comparison of the concentration of drug with a therapeutic effect (minimal required concentration) to the concentration of drug with a toxic effect (maximal required concentration)

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

How does multiple dosing affect the therapeutic window?

A

Repeated drug administration means that a new steady state can be achieved so that levels stay within the therapeutic window

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

What is steady state equal to?

A

Steady state (Cpss) = dose rate / clearance

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

What is loading dose?

A

When a clinician wants to achieve a steady state quickly within the therapeutic window they can give a loading dose
Once the steady state is reached, it is maintained by the steady state equation: steady state = dose rate / clearance

Loading dose = volume of distribution x steady state (Cpss)

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

In what ways do drugs exert their effects on the body?

A
Interactions with enzymes
Transport systems
Secondary messengers
Hormones
Ion channels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What theory is pharmacodynamics based on?

A

Pharmacodynamics is based around the concept of the receptor theory, whereby the drugs act on receptors to target the molecules.

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

What are the key concepts of the receptor theory?

A
Agonists (partial)
Antagonists (competitive and non competitive)
Specificity
Selectivity
Affinity
Efficacy
Potency
Therapeutic index and window
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What is an agonist?

A

An agonist will bind to the receptor and stabilise it (whilst bound) into its active state.

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

What is an antagonist?

A

An antagonist will bind to the receptor and stabilise it (whilst bound) into its inactive state.

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

What is a partial agonist?

A

A partial agonist will bind to the receptor yet will not fit quite well as well and does not bring about the maximal response

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

What is a competitive antagonist?

A

Competitive antagonists bind to the site in which the natural ligand binds to and can completely remove all response from the receptor

Competitive antagonism occurs where the antagonist can be “out-competed” for the receptor site by simply increasing the agonist concentration, shown on curves by a shift to the right.

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

What is a non competitive antagonist?

A

Non-competitive antagonists bind to another site on the receptor (i.e. where the ligand does not bind) and will partially reduce the overall response of the receptor

Non-competitive antagonism can be either irreversible binding to receptor site or reversible or irreversible binding to a separate site, which is represented by the EC50 staying the same yet a reduction in overall response

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

What is specificity?

A

Specificity relates to the complementary drug and receptors (i.e. good specificity is only working on one receptor)

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

What is selectivity?

A

Selectivity relates to clinical effect of the drug and can be measured with specific therapeutic indices (i.e. good selectivity has minimal side effects)

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

What is affinity?

A

Affinity defines the ability of a drug to bind to a specific receptor type. The terms used to define this are Kd for agonists and Ki for antagonists. These terms indicate the concentration at which half the receptors are occupied at

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

What is efficacy?

A

Efficacy defines the maximal effect of a drug when bound to a receptor. Agonists have 100% efficacy, partial agonists will have reduced affinity, efficacy, or both, and antagonists have affinity yet no efficacy.

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

What is potency?

A

Potency defines the overall response seen by the receptor once the ligand has bound. It is measured by the EC50 which is the concentration where 50% of the maximal response is obtained. EC50 is rarely equal to Kd values. Antagonist potency can also be measured, which can be defined as the concentration that reduces maximal activation of a receptor by 50%.

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

What is the therapeutic index?

A

The therapeutic index is the relationship between concentrations causing adverse effects and concentrations causing desirable effects; it is calculated as LD50 / ED50 and a large therapeutic index is preferred.

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

What is the therapeutic window?

A

The therapeutic window is the range of drug concentrations where they exert a clinically useful effect but without exerting toxic effects; it is the range between the lowest dose that has a positive effect and the highest dose before the negative effects outweigh the positive effects.

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

What are some examples of drugs with a narrow therapeutic window?

A
  • Warfarin
  • Aminophylline
  • Digoxin
  • Aminoglycosides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Besides from pharmacokinetics and pharmacodynamics, what else can affect a drug or the body?

A

Drug interactions with other drugs
Drug interactions with food
Drug interactions with diseases

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

What 4 stages of pharmacokinetics are affected by drug-drug interactions?

A

ADME

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

How is absorption affected by drug-drug interactions?

A

Changes in gut motility can occur to affect absorption; for example, opiates and atrophine slow the gut down to increase Cmax and increase Tmax, whereas metoclopramide speeds the gut up to increase Cmax and decrease Tmax.
Other drugs will interfere with absorption, such as calcium salts bind to tetracyclines and reduce their absorption or cholestyramine binds to Warfarin and digoxin to reduce their absorption.

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

How is distribution affected by drug-drug interactions? (Class 1 and 2 drugs)

A
Class I (object) drugs are administered at a dose where the number of molecules of that drug is much lower than the number of binding sites available for that drug whereas class II (precipitant) drugs are used at much higher numbers than the number of binding sites available so displace the class I drugs.
Administering a precipitant drug can cause free object drug concentrations to rise to toxic levels, pushing levels out of the therapeutic window. This can however be met with an increased clearance to achieve a similar steady state.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

How is metabolism affected by drug-drug interactions?

A

Drugs can affect the metabolism of themselves or other drugs by two mechanisms of either induction or inhibition (described in previous lecture).

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

How is excretion affected by drug-drug interactions?

A

The primary mechanism affecting drug excretion includes changes in protein binding, inhibition of tubular secretion, and changes to urine flow / pH. Decreased protein binding increases the amount of free unbound drug which accelerates its removal, and inhibition of tubular secretion will result in increased plasma levels of the drug (NSAIDs can act to reduce tubular secretions).

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

How will drug-drug interactions affect the pharmacodynamics of a drug and the therapeutic outcome of the drug action?

A

Interactions will either enhance or reduce therapeutic outcomes through actions on the receptor, and these drug interactions can occur via different receptors or different tissues. This can have several categories.
For example, opiate analgesics and naloxone act antagonistically whereas digoxin toxicity is enhanced by hypokalaemia caused by loop diuretics (e.g. Furosemide). Even using aspirin with Warfarin will cause levels of unbound Warfarin to increase.

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

What are the 5 main classes of drugs that contribute to drug- drug interactions?

A
  • Anticonvulsants
  • Anticoagulants
  • Antidepressants
  • Antibiotics
  • Antiarrhythmics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are some examples of drug-disease interactions? (HRH)

A

The effects of hepatic, renal, or cardiac deficit on both PK and PD are common loci for drug-disease interaction. These drug-disease interactions often lead to exacerbation of systemic toxicity due to their close interdependence.
Renal Disease- Falling GFR will cause a reduced clearance of renally excreted drugs, e.g. digoxin or aminoglycoside antibiotics. Further reduction in GFR can be seen in patients on NSAIDs or ACEis, causing AKI and are considered to be nephrotoxic.
Hepatic Disease- Hepatic disease would cause reduced clearance of hepatically metabolised drugs by a reduced CYP 450 activity. This results in much longer half-lives of drugs resulting in toxicity. Good examples include opiates in cirrhosis, with small doses even resulting in accumulation causing coma.
Cardiac Disease- a falling cardiac output can cause reduced organ perfusion, resulting in both reduced hepatic blood flow and renal blood flow, reducing drug clearance.

Many drugs are bound to albumin, so any hypoalbuminaemia caused by liver failure, malnutrition, nephrotic syndrome, plus many more, higher free drug levels will be seen. This would affect PD and PK significantly.

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

What are some examples of drug- food interactions?

A
  • Cranberry juice can be used therapeutically in UTI treatment, yet also acts to inhibit CYP2C9 so reduce clearance of Warfarin to cause raised INR and increased risk of haemorrhage.
  • Grapefruit juice inhibits several CYP450 isoenzymes, so can reduce clearance of many drugs (e.g. simvastatin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What is an adverse drug reaction?

A

In pharmacology, any unexpected or dangerous reaction to a drug. An unwanted effect caused by the administration of a drug. The onset of the adverse reaction may be sudden or develop over time.

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

What are two types of adverse drug reactions?

A

On target ADRs
OR
Off target ADRs

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

What are on target ADRs?

A

‘On target’ ADRs which are due to exaggerated therapeutic effect of the drug, most likely due to increased dosing or due to factors affecting the PK or PD. A simple example would be use of a certain drug to treat hypertension which could result in dizziness or syncope. Many ‘on target’ ADRs include effects on the same receptor type but found on other tissues.

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

What are off target ADRs?

A

‘Off target’ ADRs involves interaction of other receptor subtypes secondarily to the one intended for the therapeutic effect, which can also occur with metabolites that can subsequently act as a toxin. Inappropriate immune responses also constitute off target ADRs.

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

What increases the risk of ADRs?

A

Use of polypharmacy increases the risk of ADRs, due to effects of drug-drug interactions.

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

What are the 4 sites of actions of drugs?

A

Receptors
Ion channels
Enzymes
Transporters

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

How does a receptor function as site of action of a drug?

A

Receptors are the sensing elements in the system of chemical communication that coordinates the function of all the different cells in the body. Drugs can act by being agonists, partial agonists, or antagonists for known endogenous mediators.

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

How does a ion channel function as a an action site of a drug?

A

Some ion channels incorporate a receptor and open only when the receptor is occupied by an agonist. Thus drugs can act on ion channels indirectly (via G-protein coupled receptors) or directly (drug binds directly to ion channel) to alter its function.

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

How does an enzyme function as an action site for a drug?

A

Drugs can target certain enzymes to act as a competitive or non-competitive inhibitor of the enzyme, which can be reversible and irreversible. They can also act as false substrates, whereby drug molecules undergo chemical transformation to form an abnormal product that subverts the normal metabolic pathway. It should be mentioned that drugs may require enzymatic degradation to convert them from an inactive form (the prodrug) to their active form.

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

How do transporters function as an action site for drugs?

A

Transport of some ions and molecules requires carrier proteins, allowing for facilitated diffusion. Drugs can act to block these transporters, preventing these substances entering the cells.

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

What 4 stages of pharmacokinetics are affected by drug-drug interactions?

A

ADME

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

How is absorption affected by drug-drug interactions?

A

Changes in gut motility can occur to affect absorption; for example, opiates and atrophine slow the gut down to increase Cmax and increase Tmax, whereas metoclopramide speeds the gut up to increase Cmax and decrease Tmax.
Other drugs will interfere with absorption, such as calcium salts bind to tetracyclines and reduce their absorption or cholestyramine binds to Warfarin and digoxin to reduce their absorption.

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

How is distribution affected by drug-drug interactions? (Class 1 and 2 drugs)

A
Class I (object) drugs are administered at a dose where the number of molecules of that drug is much lower than the number of binding sites available for that drug whereas class II (precipitant) drugs are used at much higher numbers than the number of binding sites available so displace the class I drugs.
Administering a precipitant drug can cause free object drug concentrations to rise to toxic levels, pushing levels out of the therapeutic window. This can however be met with an increased clearance to achieve a similar steady state.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

How is metabolism affected by drug-drug interactions?

A

Drugs can affect the metabolism of themselves or other drugs by two mechanisms of either induction or inhibition (described in previous lecture).

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

How is excretion affected by drug-drug interactions?

A

The primary mechanism affecting drug excretion includes changes in protein binding, inhibition of tubular secretion, and changes to urine flow / pH. Decreased protein binding increases the amount of free unbound drug which accelerates its removal, and inhibition of tubular secretion will result in increased plasma levels of the drug (NSAIDs can act to reduce tubular secretions).

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

How will drug-drug interactions affect the pharmacodynamics of a drug and the therapeutic outcome of the drug action?

A

Interactions will either enhance or reduce therapeutic outcomes through actions on the receptor, and these drug interactions can occur via different receptors or different tissues. This can have several categories.
For example, opiate analgesics and naloxone act antagonistically whereas digoxin toxicity is enhanced by hypokalaemia caused by loop diuretics (e.g. Furosemide). Even using aspirin with Warfarin will cause levels of unbound Warfarin to increase.

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

What are the 5 main classes of drugs that contribute to drug- drug interactions?

A
  • Anticonvulsants
  • Anticoagulants
  • Antidepressants
  • Antibiotics
  • Antiarrhythmics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What are some examples of drug-disease interactions? (HRH)

A

The effects of hepatic, renal, or cardiac deficit on both PK and PD are common loci for drug-disease interaction. These drug-disease interactions often lead to exacerbation of systemic toxicity due to their close interdependence.
Renal Disease- Falling GFR will cause a reduced clearance of renally excreted drugs, e.g. digoxin or aminoglycoside antibiotics. Further reduction in GFR can be seen in patients on NSAIDs or ACEis, causing AKI and are considered to be nephrotoxic.
Hepatic Disease- Hepatic disease would cause reduced clearance of hepatically metabolised drugs by a reduced CYP 450 activity. This results in much longer half-lives if drugs resulting in toxicity. Good examples include opiates in cirrhosis, with small doses even resulting in accumulation causing coma.
Cardiac Disease- a falling cardiac output can cause reduced organ perfusion, resulting in both reduced hepatic blood flow and renal blood flow, reducing drug clearance.

Many drugs are bound to albumin, so any hypoalbuminaemia caused by liver failure, malnutrition, nephrotic syndrome, plus many more, higher free drug levels will be seen. This would affect PD and PK significantly.

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

What are some examples of drug- food interactions?

A
  • Cranberry juice can be used therapeutically in UTI treatment, yet also acts to inhibit CYP2C9 so reduce clearance of Warfarin to cause raised INR and increased risk of haemorrhage.
  • Grapefruit juice inhibits several CYP450 isoenzymes, so can reduce clearance of many drugs (e.g. simvastatin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What is an adverse drug reaction?

A

In pharmacology, any unexpected or dangerous reaction to a drug. An unwanted effect caused by the administration of a drug. The onset of the adverse reaction may be sudden or develop over time.

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

What are two types of adverse drug reactions?

A

On target ADRs
OR
Off target ADRs

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

What are on target ADRs?

A

‘On target’ ADRs which are due to exaggerated therapeutic effect of the drug, most likely due to increased dosing or due to factors affecting the PK or PD. A simple example would be use of a certain drug to treat hypertension which could result in dizziness or syncope. Many ‘on target’ ADRs include effects on the same receptor type but found on other tissues.

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

What are off target ADRs?

A

‘Off target’ ADRs involves interaction of other receptor subtypes secondarily to the one intended for the therapeutic effect, which can also occur with metabolites that can subsequently act as a toxin. Inappropriate immune responses also constitute off target ADRs.

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

What increases the risk of ADRs?

A

Use of polypharmacy increases the risk of ADRs, due to effects of drug-drug interactions.

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

What are the 4 sites of actions of drugs?

A

Receptors
Ion channels
Enzymes
Transporters

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

How does a receptor function as site of action of a drug?

A

Receptors are the sensing elements in the system of chemical communication that coordinates the function of all the different cells in the body. Drugs can act by being agonists, partial agonists, or antagonists for known endogenous mediators.

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

How does a ion channel function as a an action site of a drug?

A

Some ion channels incorporate a receptor and open only when the receptor is occupied by an agonist. Thus drugs can act on ion channels indirectly (via G-protein coupled receptors) or directly (drug binds directly to ion channel) to alter its function.

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

How does an enzyme function as an action site for a drug?

A

Drugs can target certain enzymes to act as a competitive or non-competitive inhibitor of the enzyme, which can be reversible and irreversible. They can also act as false substrates, whereby drug molecules undergo chemical transformation to form an abnormal product that subverts the normal metabolic pathway. It should be mentioned that drugs may require enzymatic degradation to convert them from an inactive form (the prodrug) to their active form.

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

How do transporters function as an action site for drugs?

A

Transport of some ions and molecules requires carrier proteins, allowing for facilitated diffusion. Drugs can act to block these transporters, preventing these substances entering the cells.

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

What are the main 2 classes of contraceptive drugs?

A

Progesterone only pills (POP)

Combined oral contraceptive pill (COCP)

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

How does POP work?

A

Low-dose Progestogen (Progesterone analogue).
It causes thickening of cervical mucus to inhibit sperm transport. It inhibits endometrial implantation and causes suppression of gonadotropin secretion

119
Q

What are the instructions upon administration of POP?

A

Oral – daily at the same time each day, starting at day 1 of the menstrual cycle. (If delay >3hours, contraceptive effect may be lost)

120
Q

To whom would you usually prescribe POP?

A

Contraception – more suitable for heavy smokers & HTN/Heart Disease, DM patients or other contraindications of oestrogen therapy

121
Q

When is POP not advised to be prescribed?

A

Pregnancy, arterial disease, liver disease, carcinoma of breast or genital tract

122
Q

What are some adverse effects of taking POP?

A

Menstrual irregularities & poor cycle control, NV, headaches, weight gain, breast tenderness

123
Q

What drug interactions can POP have?

A

Metabolised by CYP450 so affected by inhibitors & inducers. Inducers can lead to contraception failure

124
Q

How does COCP work?

A

It contains both an Oestrogen (usually Ethinylestradiol, 20-50mg) and a Progestogen (a Progesterone analogue). It mimics the luteal phase of the menstrual cycle and suppress gonadotropin release by negative feedback (inhibits follicular maturation and ovulation)

125
Q

What are the instructions upon the administration of COCP?

A

Oral - One a day for 21 days, then break, placebo or iron pill for 7 days

126
Q

To whom would you usually prescribe COCP?

A

Patients wanting Contraception &

Patients with menstrual symptoms- Uterine bleeding

127
Q

When is COCP not advised to be prescribed?

A

Pregnancy, breast feeding, Hx or Heart Disease risk factors, HTN, hyperlipidaemia, any prothrombotic coagulation abnormality, DM, liver disease, carcinoma of breast of genital tract

128
Q

What are some adverse effects of COCP?

A

Venous Thrombo-embolism, HTN, decreased glucose tolerance, mood swings, acne, NV, amenorrhoea of variable duration on pill cessation
Weight gain

129
Q

What drug interactions can COCP have?

A

Metabolised by CYP450 so affected by inhibitors & inducers. Inducers can lead to contraception failure e.g. carbamezepine and phenytoin (anti-epileptics). Broad spectrum antibiotics (e.g. Amoxicillin) :Enterohepatic recirculation of oestrogen increases the efficacy of the COCP. If gut flora is killed by a broad spectrum antibiotic this is reduced and may cause contraception failure

130
Q

What are some other forms of contraception (not the main ones)?

A

Depot progesterone
Morning after pill
Progesterone receptor modulators
Copper IUD

131
Q

How does depot progesterone work?

A

Same as POP

132
Q

How is depot progesterone given to the patient?

A

IM implant of progesterone

133
Q

Who would you give depot progesterone to?

A

Patients wanting long term contraception

134
Q

How does the morning after pill work?

A

Very high oral dose of progesterone (1.5mg) alone or a Progestogen with an oestrogen to prevent implantation

135
Q

What are the instructions for prescribing the morning after pill?

A

Oral up to 72hrs after sex

136
Q

When is the morning after pill useful?

A

Emergency contraception after unprotected sex

137
Q

When would the morning after pill not be advised to be used?

A

COCP contraindications

Need to ask about cycle and when they had sex to determine if woman’s already pregnant- illegal (abortion)

138
Q

How do progesterone receptor inhibitors work?

A

delays or inhibits ovulation- emergency contraception 120 hours after sex

139
Q

How does the copper IUD work?

A

Stops or kills sperm- emergency contraception 120 hours after sex

140
Q

How does hormone replacement therapy work?

A

Oestrogen and Progesterone given to replace lost hormones

141
Q

How is HRT administered?

A

Orally or topically (patch/gel)

142
Q

What is HRT used for?

A

Menopause

Control well being

143
Q

When would you not give HRT?

A

Don’t give oestrogen alone unless patient has had a hysterectomy- would cause endometrial hyperplasia and cancer

144
Q

What are some adverse effects of HRT?

A

Adverse effect on lipid and thrombophilia profile
Risks:
Unopposed oestrogens- endometrial and ovarian cancers
Increase risk of breast cancer, IHD, stroke thromboembolism, uterine bleeding

145
Q

What are some benefits of HRT?

A

Decreased risk of colorectal cancer, increased sexual function, small reduction in bone loss

146
Q

What is combine HRT and why is it useful/unuseful?

A

Combined HRT- patient given same dose of oestrogen and progesterone for 28 days continuously; woman has no withdrawal bleed, woman does have risk of endometrial hyperplasia- so only given when woman has had a hysterectomy

147
Q

What is sequential HRT and why is it useful/unuseful?

A

Sequential HRT- patient given oestrogen only up to day 14-16 and then progesterone added; will stimulate withdrawal bleed; reduced risk of endometrial cancer as endometrial hyperplasia does not occur; can be given to patients who haven’t had a hysterectomy

148
Q

How does anti oestrogen medication work?

A

Opposes oestrogen receptors

149
Q

When would you use anti oestrogen medication?

A

Fertility problems- Chlomiphene

Breast cancer and ovulation induction- Tamoxifen

150
Q

What is chlomiphene used for?

A

Anti oestrogen used for fertility problems

151
Q

How does chlomiphene achieve helping fertility problems?

A

Induces ovulation by inhibiting binding of oestrogen and thus causing an increase in GnRH, LH and FSH

152
Q

What is tamoxifen used for?

A

Breast cancer and ovulation induction

153
Q

How does tamoxifen achieve helping breast cancer and inducing ovulation?

A
  • Reduces oestrogen binding- limiting progress/ recurrence of breast cancer
154
Q

How does anti progesterone work?

A

Antagonises effects of progesterone
Sensitises uterus to prostaglandins
Partial agonist for progesterone receptors and inhibits action of progesterone

155
Q

When is anti progesterone used?

A

Medical termination of pregnancy and inducing labour (not in UK)

156
Q

How does anti androgen work?

A

Weak progesterogenic effect by competing with dihydrotestosterone

157
Q

What is anti androgen used for?

A

Hisuitism- Cyproterone

158
Q

How does selective oestrogen receptor mediators work?

A

Act of oestrogen receptors

Have different actions depending on where the receptor is found

159
Q

When are SORMs used?

A

Osteoporosis in post menopausal women- Raloxifene

160
Q

What are the adverse effects of using SORMs?

A

No proliferative effects on endometrium, oestrogenic effects on bone, lipid metabolism, blood coagulation, reduced risk of invasive breast cancer, increased hot flushes

161
Q

What are the 5 main drug groups for treating hyperlipidaemia?

A
Statins
Fibrin acid derivatives
Cholesterol absorption inhibitors
Bile acid sequestrants 
Niacin
162
Q

What are some examples of statins?

A
Simvastatin
(short HL 1-4hrs)
Atorvastatin
(HL = 20 hours)
Rostuvastatin
(HL = 20hours)
Pravastatin
163
Q

Which statin has the shortest half life? What is it?

A

Simvastatin 1-4 hours

164
Q

How do statins work?

A

HMG-CoA Reductase inhibitor. Prevents cholesterol synthesis in the liver. Lower liver cholesterol concentration stimulates the production of LDL receptors, increasing LDL removal from the plasma

165
Q

What is the route of administration of statins?

A

Oral

166
Q

When are statins useful?

A

Hyperlipidaemia which has not responded to changes in diet & exercise
Secondary prevention in patients with serum cholesterol >5.5mmol/L

167
Q

When should statins not be used?

A

Pregnancy, breast feeding, liver disease

168
Q

What are some adverse effects of statin usage?

A

Serious ADRs tend to be limited even at the highest doses of statin given.
Myalgia, myopathy & rhabdomyolysis, increased transaminase levels, GI disturbances, arthralgia, headache

169
Q

What are some drug interactions of statin?

A

CYP450 inducers & inhibitors. Inhibitors increase risk of myopathies as drug spends more time in the plasma & can interact with muscle

170
Q

What are some examples of fibrin acid derivatives?

A

Bezafibrate
Ciprofibrate
Gemfibrozil

171
Q

How do fibrin acid derivatives work?

A

Agonist at Peroxisome Proliferator-Activated Receptor- α. This causes LDL decrease, HDL increase, TAG decrease

172
Q

How are fibric acid derivatives administered?

A

Oral

173
Q

When are fibric acid derivatives used?

A

Hyperlipidaemia which doesn’t respond to dietary control

174
Q

When should fibric acid derivatives not be used?

A

Pregnancy, breast feeding, gall bladder disease, severe renal or hepatic impairment, Hypoalbuminaemia

175
Q

What are some adverse effects of fibric acid derivatives?

A

GI disturbances, dermatitis, pruritis, rash, impotence, headaches, dizziness, blurred vision

176
Q

What are some drug interactions of fibric acid derivatives?

A

Increased chance of myalgia & myopathies when taken with statins

177
Q

What’s an example of a cholesterol absorption inhibitor? (CAI)

A

Ezetimibe

178
Q

How do CAIs work?

A

Blocks NPC1L1 in the intestinal brush border to inhibit cholesterol absorption. Increases hepatic LDL receptors

179
Q

What is the route of administration of CAIs?

A

Oral

180
Q

When are CAIs used?

A

Hyperlipidaemia resistant to dietary control in statin intolerant patient

181
Q

When should CAIs not be used?

A

Breast feeding

182
Q

What are some adverse effects of CAIs?

A

GI disturbances, headache

183
Q

What are some examples of bile acid sequestrants?

A

Colestipol

Colestyramine

184
Q

How do bile acid sequestrants work?

A

They bind to bile acids in the intestine to prevent reabsorption and conversion to cholesterol in to bile acids in the liver.

185
Q

What’s the administration of bile acid sequestrants?

A

Oral

186
Q

When are bile acid sequestrants used?

A

Elevated cholesterol resulting from a high LDL concentration

187
Q

When should bile acid sequestrants not be used?

A

Biliary obstruction

188
Q

What are some adverse effects of bile acid sequestrants?

A

GI disturbances, very few systemic side effects as they are not absorbed

189
Q

How does niacin work?

A

Inhibits lipoprotein-a synthesis

190
Q

What are some adverse effects of niacin?

A

Skin flushing & itching, dry skin, skin rashes

191
Q

What is diabetes?

A

Vascular disease leading to small and large vessel damage in which there’s premature death from cardiovascular disease

192
Q

What is the cause of type I DM?

A

Insulin deficiency

193
Q

What is the cause of type II DM?

A

Insulin resistance (decreased insulin sensitivity) and eventually insulin deficiency

194
Q

What are the stages of management of type II diabetes?

A

Diet and lifestyle changes- no pharmacological intervention
Drugs- beta cell stimulators and insulin sensitisers (GLP-1)
Insulin

195
Q

What diet and lifestyle advice can be given to a diabetic?

A

Lose weight by limiting fat intake whilst increasing proportionate calories of complex carbohydrates - helps keep HbA1c levels stable
Reduction in alcohol
Stop smoking
Increasing exercise
Some clinicians give anti obesity drugs to obese patients before diabetic medication

196
Q

What are the two categories of beta cell stimulator drugs?

A

Sulphonylureas

Meglitidines

197
Q

What are some examples of sulphonylureas and their half lives?

A

Tolbutamide (t1/2= 4 hrs, acts upto 6-12 hrs)

Glibencamide (t1/2= 10 hrs, acts upto 18-24 hrs)

Glipizide (t1/2= 7 hrs, acts upto 16-24 hrs)

198
Q

How do sulphonylureas work?

A

They antagonise B cell K+/ATP channel activity; decrease in K+ current causes depolarisation; Ca2+ influx and insulin release

199
Q

How do sulphonylureas affect HbA1c?

A

Reduces HbA1c between 1-2%

200
Q

What is the route and frequency of sulphonylureas?

A

Oral

1/day

201
Q

What are some indications of sulphonylureas?

A

Diabetes mellitus, in patients with residual β-cell activity

202
Q

What are some contraindications of use of sulphonylureas?

A

Breastfeeding women, elderly, renal and hepatic insufficiency
Obese people as it can cause weight gain

203
Q

What are some adverse drug reactions of sulphonylureas?

A

Hypoglycaemia (esp in elderly, with missed meals or excess alcohol)
GI disturbances
Weight gain (not helpful in obese patients)
Highly protein bound

204
Q

What are some examples of Meglitidines? And their half lives?

A

Repaglinide
Nateglinide
(t1/2 = 1-3 hrs)

205
Q

How do Meglitidines work?

A

They antagonise B cell K+/ATP channel activity decrease in K+ current; causes depolarisation; Ca2+ influx and insulin release

206
Q

How do Meglitidines differ from sulphonylureas?

A

Meglitidines work faster than sulphonylureas
Meglitidines have a relatively lower risk of hypoglycaemia than sulphonylureas
Meglitidines not associated with weight gain (useful for OBESE patients)

207
Q

How do Meglitidines affect HbA1c?

A

Reduces HbA1c by 1%

208
Q

What is the route and frequency of Meglitidines?

A

Oral

1/day

209
Q

What are some indications of Meglitidines use?

A

Uncontrolled non-insulin dependent diabetes

210
Q

What are the two categories of insulin sensitiser drugs?

A

Biguanides

Thiazolineinediones

211
Q

What’s an example of a Biguanide? And it’s half life?

A

Metformin (t1/2 = 2/3 hrs)

212
Q

How do Biguanides work?

A

Increases insulin receptor sensitivity (skeletal and adipose)
Inhibits hepatic gluconeogenesis
Reduces hyperglycaemia but doesn’t cause hypoglycaemia

213
Q

How do Biguanides affect HbA1c?

A

Reduces HbA1c by upto 2%

214
Q

What is the route and frequency of biguanides?

A

Oral

2,3 / day

215
Q

What is an indication of Biguanides use?

A

T2DM – endogenous insulin presence required

First line of treatment always/ for overweight patients (NICE)

216
Q

What are some contraindications of Biguanides use?

A

Compromised HR (CKD

217
Q

What are some adverse drug reactions of Biguanides?

A
GI disturbances (thus give slow dose titrations)- wind, loose stools etc.
Lactic acidosis
Vitamin B12 deficiency
218
Q

What are some advantages of Biguanides use?

A

Does not induce hypoglycaemia when reducing hyperglycaemia
Also reduces LDLs and VLDLs
Weight neutral- used for overweight patients
Reduced risk of cancer (bowel etc.)

219
Q

What are some examples of Thiazolineinediones?

A

Rosiglitazone
Piogliatazone

(t1/2 = 7 hrs but metabolites have prolonged t1/2 upto 150 hrs)

220
Q

How do Thiazolineinediones work?

A

Reduction in gluconeogenesis and increased glucose uptake (peak effects after 1-2 months)
PPAR-gamma agonist (receptor found on adipose tissue) – stimulates uptake of fatty acids, making cells more dependent on glucose remaining in blood- uptake of glucose (provided endogenous insulin). By reducing circulating fatty acid concentrations and lipid availability in liver and muscle, the drug improves patient’s sensitivity to insulin.

221
Q

How do Thiazolineinediones affect HbA1c?

A

Reduce HbA1c by 1-1.5%

222
Q

What is the route and frequency of Thiazolineinediones?

A

Oral

1/day

223
Q

What’s an indication of Thiazolineinediones use?

A

Uncontrolled non-insulin dependent diabetes

224
Q

What are some contraindications of Thiazolineinediones use?

A

Compromised HRH function (especially heart failure, can cause oedema)

225
Q

What are some adverse drug reactions of Thiazolineinediones?

A
Highly protein bound
Oedema
Increases in LDL
GI disturbance
Weight gain
Fractures in post menopausal women – inhibits osteoblast function
Bladder cancer
226
Q

What are some advantages of Thiazolineinediones use?

A

Don’t induce hypoglycaemia

Increases in HDL

227
Q

What other therapeutic drugs can be used for the management of diabetes?

A

Glucagon like peptide 1 analogues
Dipeptidyl peptidase 4 inhibitors
Alpha gluosidase inhibitors
Sodium glucose Cotransporter 2 inhibitors

228
Q

What is an example of a GLP1 analogue?

A

Exenatide

229
Q

What is the action of GLP1 analogues?

A

GLP normally secreted from L cells in small intestine upon the ingestion of food
Pancreas- Beta cell- enhances glucose dependent insulin secretion; Alpha cell- suppresses post prandial glucagon secretion
Liver- reduces hepatic glucose output
Stomach- slows rate of gastric emptying
Brain- promotes satiety and reduces appetite
Muscle- increased uptake of glucose

230
Q

What is the route and frequency of GLP1 analogues?

A

SC injection – 1,2 / day

231
Q

What are the indications of GLP1 use?

A

Used after Drugs before Insulin, only if:
HbA1c has reduced by 1% in last 6 months AND
Weight has decreased by 3 % in last 6 months

232
Q

What are some adverse drug reactions of GLP1 analogues?

A

Nausea and other GI symptoms

233
Q

What are some advantages of GLP1 analogues use?

A

Promotes Weight loss by promoting satiety and reducing appetite

234
Q

What is an example of a DPP4 inhibitor?

A

Sitagliptin

235
Q

How do DPP4 inhibitors work?

A

Inhibits DPP enzyme that breaks down GLP-1, therefore increasing GLP and decreasing blood glucose levels

236
Q

What’s an example of an alpha glucosidase inhibitor?

A

Acarbose

237
Q

What’s the action of alpha Glucosidase inhibitor?

A

Inhibits SGLUT2 in PCT and therefore encourages glucose secretion via kidneys

238
Q

What are some adverse drug reactions of alpha glucosidase inhibitors?

A

Flatulence and diarrhoea

High doses associated with elevation of ALT

239
Q

What are some adverse drug reactions of sodium glucose cotransporters 2 inhibitors?

A

Thrush / UTIs

Polyuria and Thirst

240
Q

What are some different formulations of insulin? And their onset, PETA and duration of action?

A

Ultra Rapid : Onset = 0.2 – 0.5 ; Peak = 1 ; Duration = 3 – 4

Short Acting : Onset = 0.5 – 1 ; Peak = 2 – 5 ; Duration = 6 - 12

Intermediate : Onset = 1.5 – 3 ; Peak = 4 – 10 ; Duration = 16 – 24

Long Acting Onset = 4 – 6 ; Peak = 8 – 30 ; Duration = 18 – 36

241
Q

Generally describe the first and second line treatment of Type II DM?

A

FIRST LINE TREATMENT
Always give Biguanide (metformin), UNLESS
- Patient is intolerant to Biguanide (metformin)
- Patient is not overweight
–> In which case give Sulphonylurea (Tolbutamide)

SECOND LINE TREATMENT

  • Over time if HbA1c rises >7%- add a Sulphonylurea (Tolbutamide)
  • Over time if HbA1c rises >7.5%- add a Thiazolineinediones (Pioglitazone), or start insulin therapy

If on this regime, HbA1c levels rise higher than 7.5%, doses will be titrated upwards to regain adequate glycaemic control

242
Q

How is acute treatment of diabetes mellitus monitored?

A

In patients with advanced diabetes, monitoring several times a day is required to determine dosing level with insulin
Regime in non-insulin therapies is frequently determined by dietary habit
With careful monitoring and control of their diet patients can stay within normal glucose ranges

243
Q

How is chronic treatment of diabetes mellitus monitored?

A

Glucose in the blood will react with the terminal valine of the Hb molecule to produce glycosylated Hb (HbA1c)
% HbA1c is a good indicator of how effective blood glucose control has been
As RBCs spend ~3 months in circulation the %HbA1c is related to average blood glucose concentration over previous 2-3 months
Poorly controlled diabetics have a HbA1c >10%
In combination therapy, new medications are added at HbA1c values of 7 or 7.5%

244
Q

What are the two main combination therapies of insulin and when are they used?

A

2 x intermediate dosages of insulin
OR
1 x long acting dosage of insulin + 2 x short acting dosages of insulin (with meals)

245
Q

What are some adverse drug reactions of insulin?

A

Local reactions
Hypoglycaemia (coma) – overdose
Rarely- immune resistance

246
Q

What are three forms of anti obesity drugs?

A

Orlistat
Sibutramine
Rimonabant

247
Q

What is an alternative to anti obesity medication?

A

Bariatric surgery

248
Q

How does orlistat work?

A

Gastric and pancreatic lipase inhibitor

Reduces the conversion of up to 30% dietary fat to fatty acids and glycerol

249
Q

What are some adverse drug reactions of orlistat?

A

Broad GI disturbances

Soft fatty stools, flatus, faecal discharge/ incontinence

250
Q

How does sibutramine work?

A

NA and serotonin reuptake inhibitor
Appetite suppression
Increased thermogenesis

251
Q

What are some adverse drug reactions of sibutramine?

A

Increased heart rate and blood pressure

252
Q

How does Rimonabant work?

A

Endocannabinoid antagonist

253
Q

What are some adverse drug reactions of Rimonabant?

A

Depression- currently withdrawn in UK by NICE

254
Q

What is the virus life cycle?

A
  1. Influenza virus binds to cell via Hemagglutinin onto sialic acid sugars on the surface of epithelial cells.
  2. Entry of virus into cell via endocytosis
  3. ATP driven proton entry into the endosome, allowing fusion of the viral membrane with the internal endosomal membrane
  4. Entry of protons into the virus itself via the viral M2 Ion Channel. Low pH inside the virus results in breakdown in the viral coat of the nucleocapsid core. This releases viral RNA into the host cytoplasm
  5. Virus replicates using host cell machinery
  6. Viral protein assembly
  7. New virus buds off the cell membrane, but many remain attached by re-attaching to the sialic acid on the cell surface
  8. Viral Neuramidase enzyme breaks this bond, allowing viral release
255
Q

What are the three classes of Influenza virus?

A
o Influenza A
• Most dangerous
• Multiple host species which are able to infect humans- e.g. bird flu and swine flu
• Exhibits antigenic drift and shift
• Drift = different each year
• Shift = leads to an epidemic
• New vaccine development is necessary to treat the pre-absorbed virion when it is vulnerable to the pre-vaccinated immune system
o Influenza B
• No animal reservoir
• Lower mortality than influenza A
o Influenza C
• Common cold like
256
Q

What are the two types of drugs used to treat influenza?

A

M2 ion channel blockers- developed from tricyclics amines

Neuraminidase inhibitors

257
Q

What are some examples of M2 ion channel blockers? Which is preferred and why?

A

o Amantadine

o Rimantadine- preferred as amantadine has a 5-10% higher risk of ADRs

258
Q

How do M2 channel blockers work?

A

Inhibits the un-coating of a virus, therefore preventing it from being able to infiltrate into the cell. This occurs by the action of:
• Inhibiting H+ influx into the virus cell itself by blocking the M2 Ion channel., therefore preventing the change in pH which normally stimulates the viral un-coating
• So viral RNA not released into host cell

259
Q

What is the route of administration of M2 channel blockers?

A

Oral

260
Q

What are some indications of M2 channel blockers?

A

Prophylaxis and treatment of acute Influenza A in groups at risk.

261
Q

What are some contraindications of M2 channel blockers use?

A

Ineffective against group B
Rapid emergence of M2 mutations in H5N1 viruses
Resistance can develop quickly as only a single point mutation is needed in order to change the shape. This causes the binding site to move away from the channel, so that when the drug binds it will no longer block the channel
E.g. amantadine in chicken feed leading to resistance

262
Q

What are some ADRs of M2 channel blockers?

A

Amantadine has more marked ADR risk than Rimantadine of ~5-10%, therefore Rimantadine is usually preferred
Dizziness
Hypotension
GI disturbance
Confusion, insomnia and hallucination can be problematic in the elderly (CNS)
Is nephrotoxic in high doses

263
Q

What are some examples of neuraminidase inhibitors?

A

o Zanamivir

o Oseltamivir

264
Q

How do neuraminidase inhibitors work?

A

Inhibits neuraminidase enzyme which cleaves the virus from receptors on the membrane, once the virus has been produced. It causes aggregation of the virus at the cell surface, therefore preventing the virus from spreading throughout the body and therefore to other people also.
Sialic acid analogues, with very high binding affinities (Ki ~ 0.1nM) and potency ( ED50 ≈ 30nM) for Neuramidase.
The receptor is not involved with antigenic shift or drift

265
Q

What is the route of administration of neuraminidase inhibitors ?

A

Zanamivir- inhaled

Oseltamivir- Oral (80% Bioavailability)

266
Q

What are some indications of neuraminidase inhibitor use?

A

Treatment of Influenza A or B virus within 48 hours after onset of symptoms when influenza is endemic in the community

267
Q

What are some contraindications of neuraminidase inhibitor use?

A

Breast feeding
Zanamivir has low bioavailability therefore is given as a dry powder inhalant. It is not used for prophylaxis.
Oseltamivir is a pro-drug and by contrast is well absorbed, with 80% bioavailability. This enables it to be given orally for both treatment and prophylaxis.
Gives rise to:
35-38% reduction in severity

268
Q

What are some ADRs of neuraminidase inhibitors

A
Headache
Nose bleed
Respiratory depression (rarely)
Bronchospasm
GI disturbances
269
Q

How have clinical trials affected the use of oseltamivir?

A

Phase III Clinical Trials have directly informed therapeutic strategy.
Trial outcomes are discussed with respect to:
• Severity of symptom related to dose
o Reduction in symptom severity in Placebo: 75mg:150mg treatment groups showed no difference in outcome between the two active groups with decrease in symptoms score of about 40% vs. Placebo.
• Timing of initiation of treatment and illness duration
o The earlier treatment is started after symptom onset the shorter the duration of symptoms. The time window for significant reduction goes up to 48 hours. Little benefit accrues past this time.
• Mortality
o It appears that Oseltamivir could offer ≈ 70% reduction in risk of mortality in one Canadian study. Importantly, this was achieved even when dosing as delayed as long as 64 hours after symptom onset.
• Prophylaxis
o Treatment for six weeks with 75 mg significantly reduced incidence of flu in both healthy adults and frail elderly subjects.

270
Q

How does resistance to oseltamivir arise?

A
  • The Neuramidase Enzyme also appears to be evolutionary conservative, which is a great advantage for any antimicrobial therapeutic although reports of resistance in various HA and NA surface glycoprotein antigens subtypes have been reported.
  • In the small number of those treated for H5N1 bird flu resistance appears to be high. For other types highly variable values have been reported for H1N1. However the same degree of resistance to zanamivir is not apparent.
271
Q

What is the aim of antibacterial treatment?

A

Treatment aims to selectively target the invading bacteria with minimal effect upon the host
o Achieved by exploiting the differences that exist between the structure and physiology of the prokaryotic bacterial cells and the host eukaryotic cells

272
Q

What are some antibacterial targets?

A

Peptidoglycan cell wall- Peptidoglycan cell wall only present in prokaryotic cell
Nucleic acids- Bacterial genome is a single, circular strand of DNA unenclosed by a nuclear envelope, in contrast to eukaryotic chromosomal arrangement within the nucleus
Protein synthesis- Bacterial ribosome (50s+30s subunits) is different to the mammalian ribosome (60s+40s subunits)
Cytoplasmic membrane- Bacterial plasma membrane does not contain any sterols, unlike mammalian

273
Q

What antibacterials target the peptidoglycan cell wall?

A

Penicillin
Cephalosporin
Glycopeptides

274
Q

What antibacterials target the nucleic acid?

A

Antifolates
Quinolones
Rifampicin

275
Q

What antibacterials target protein synthesis?

A
Aminoglycosides
Tetraclyclines
Macrolides
Chloramphenicol
Fusidic acid
276
Q

What antibacterials target the cytoplasmic membrane?

A

Polymixins

277
Q

What are the two main usages of antibacterials?

A

Prophylaxis- given to people who are at increased risk of infection
• Peri-operative- prevention of surgical site infections
• Short term- Meningitis contact
• Long term- Asplenia (encapsulated bacteria), immunodeficiency
Significant bacterial infection- given go people with a cultured, proven infection
• Empirical treatment

278
Q

What is the difference between bacteriostatic and bacteriocidal?

A

Bacteriostatic (inhibit bacterial growth, but do not kill them) OR
Bactericidal (kill the bacteria)

279
Q

What does the ideal antibacterial give?

A

o Clean killing of infecting bacteria
• Minimal impact on non-target commensal organisms
• No resistance in any surviving pathogens
o No adverse effects on patient

280
Q

What factors help determine a likely infectious agent?

A
o Anatomical Site
o Duration of illness
o Past medical history
o Occupational history
o Travel history
o Time of year
o Age
o Personal background
281
Q

What factors determine which antibiotic is likely to be effective against a bacteria?

A
o Community or healthcare onset?
o Severity of infection
o Baseline rate of resistance
o Immune status of patient
• Immunocompromised patients will need IV Antibiotics immediately
282
Q

What are the four main things that need to be considered for antibacterial choice?

A

o Efficacy- maximal
o Cost- minimal
o Administration Route- oral, IV, SC etc.
o Safety- patient age, toxicity, drug interactions, allergies, pregnancy/breast feeding, organ function

283
Q

What are some antibacterial ADRs?

A
  • Pharmacological- toxicities, drug interactions
  • Allergic reactions
  • Impact on normal flora- Clostridium Difficile infection
284
Q

What is therapeutic drug monitoring?

A
  • To ensure- adequate and non-toxic dose
  • Used for drugs with Zero order kinetics (where half life cannot be determined)
  • Used with: aminoglycosides (inc. gentamicin), vancomycin
285
Q

What are two genetic causes of antibacterial resistance?

A

Chromosomal gene mutation

Horizontal gene transfer

286
Q

Describe chromosomal gene mutation wrt antibiotic resistance

A

o Chromosomal gene mutates in one bacteria in a population, conferring a resistance to antibiotic
o Antibiotic kills all other bacteria, acting as a selection pressure, giving resistant bacteria an advantage
o Population of antibiotic resistant bacteria daughter cells

287
Q

Describe horizontal gene transfer wrt antibiotic resistance

A

Transformation
o Bacteria with antibiotic resistance gene releases DNA
o Uptake of DNA by recipient cell, conferring antibiotic resistance
Transduction
o Phage infected, antibiotic resistant Bacterial donor cell
o Phage passes the DNA conferring resistance to recipient cell
Conjugation
o Connection is made between antibiotic resistant donor cell, and recipient cell
o Plasmid containing resistance gene is replicated and passes from donor cell to recipient cell
o Plasmid may even become incorporated into recipient cell DNA

288
Q

What are 4 different mechanisms of antibiotic resistance?

A

Antibiotic Inactivation
o Production of enzyme that inactivate the drug
• E.g. β-lactamase which inactivates Penicillins
Alteration of Drug Binding Site
o Modified binding sites so drugs no longer have affinity for them
• E.g. Bacterial ribosome alteration, meaning Aminoglycosides and Erythromycin cannot bind
Alteration of Metabolic Pathways
o Development of altered metabolic pathways
• E.g. bacteria can become resistant to Trimethoprim due to acquired changes in their Dihydrofolate Reductase enzyme, which gives it very little affinity for the drug
Reduced Intracellular Antibiotic Concentration
o Active Efflux Mechanisms
• E.g. Active transport mechanisms used (e.g. p-glycoprotein) to pump a drug out of the bacterial cell because it accumulates to an effective level
o Decreased permeability
• E.g. Some bacteria become resistant to Tetracycline because they alter their cell membrane to make it impermeable to the drug

289
Q

What are 3 patterns of emergence of antibacterial resistance?

A
  1. Local selection (e.g. in a hospital)
  2. Clonal dissemination (e.g. around the country)
  3. Global spread
290
Q

What are some of the main antibiotic resistant organisms?

A

o Methicillin Resistant Staphylococcus Aureus (MRSA)
o Glycopeptide Intermediate susceptibility Staphylococcus Aureus (GISA)
o Glycopeptide Resistant Enterococci (GRE)
o Extended Spectrum Beta Lactamase enterobacteriaceae (ESBLs)
o Extensively Drug Resistant Klebsiella Pneumoniae (XDR-KP)

291
Q

What steps are taking place to try and avoid antibacterial resistance?

A

Antimicrobial Stewardship- measures to ensure appropriate usage of antibiotic
o Right antibiotic
o Right time
o Right dose, frequency and duration (Pharmacokinetics – ADME)
o Right route
Infection Control
o Prevent the spread of recognised resistant bacteria
• Isolation or cohorting
• Hand hygiene
• Decolonisation of patients
o Prevent bacterial exposure to antibiotics
• Minimise risk of infection
• Monitor and control antibiotic prescribing

292
Q

What are the pharmacokinetics of antibacterials?

A

o Administration- Oral or IV (immunocompromised)
o Distribution
o Metabolism/Elimination- Renal or Hepatic

293
Q

What are the pharmacodynamics of antibacterials?

A

Time dependent killing- B lactams- e.g. Vancomycin
o Works by having a prolonged presence at site of infection
o But antibiotics not at high concentration

Concentration dependent killing – Aminoglycosides
o Works by having a high antibiotic concentration at the site of infection, thus eradicating microorganisms
o But short duration

294
Q

What is MIC?

A

Minimum Inhibitory Concentration – MIC
Lowest concentration of an antibiotic that will inhibit the visible growth of a microorganism after overnight incubation.
A MIC is generally regarded as the most basic laboratory measurement of the activity of a microbial agent against an organism
Related to pharmacodynamics of antibacterials