Drugs Flashcards

1
Q

What needs to be considered when prescribing a drug?

A

Mode of action

Efficacy

Safety

Patient suitability

Formulation

Dose

Route

Frequency

Duration

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

What is efficacy?

A

Power to produce an effect- drugs ability to elicit a response when it binds to receptor

Prescribers normally choose drugs w/ greatest efficacy

May be appropriate, however, to compromise on efficacy if other drugs are more convenient, safer to use or less expensive.

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

how does formulation of drug affect prescribing?

A

Some drugs- choice of formulation- some are easier to ingest particularly by children

Formulation important when writing repeat prescriptions for drugs w/ low there-tunic index that come in different formulations
- Even if prescribed dose is same, an alternative formulation may differ in absorption & bioavailability- thus plasma drug conc.

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

What factors influence route of drug administration? What routes are possible?

A

Different routes:

  • IM- intramuscular
  • INH- inhalation
  • IV- intravenous
  • PR-per rectum
  • SC- subcutaneous
  • SL- sublingual
  • Orally

Factors influencing route:

  • Comfort- morphine SC rather than IV
  • Ease of access- diazepam PR, adrenaline- IM
  • Direct access to site of action
  • Poor absorption
  • Rapid action- haloperidol IM rather than oral
  • Vomiting
  • Certainty of effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

How does dosage differ depending on drug & patient?

A

Prescribers should start w/ low dose & slowly increase as necessary
-Important if patient is more sensitive to adverse pharmacodynamic effects e.g. delirium or hypotension in elderly.

However, some drugs must achieve therapeutic conc quickly because of clinical cirumstance g.g. antibiotics, flu corticoids
- When early effect is important but there may be delay in achieving steady state due to drug’s long half-life, a loading dose is given prior to establishing maintenance dose

If adverse effects occur, dose should be reduced or alternative drug should be prescribed
- lower drug may suffice if combined w/ another synergistic drug e.g. immunosuppressant azathioprine reduces glucocorticoid requirements in patients w/ inflammatory disease

Higher doses may produce little added therapeutic effects & might increase chances of toxicity- dose-response curve

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

How does frequency of drug doses differ?

A

Frequency of doses dictated by manufacturer’s recommendation

  • Less frequent doses more convenient for patients but result in greater fluctuation in drug conc- transitions from peaks & troughs more frequent
  • Issue if peaks associated w/ adverse effects e.g. dizziness w/ antihypertensives, or troughs associated w. loss of effects e.g. anti-parkinsonian drugs

Problems can be tackled by splitting dose or by employing modified release formulation.

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

How does duration of a drug differ between drugs & patients?

A

Some drugs require single dose e.g. thrombolysis post myocardial infarction

Others, the duration of course of treatment is certain at outset e.g. antibiotics
- duration will depend on prescriber’s discretion & will depend on response & disease progression e.g. antidepressants & analgesics.

Many treatments are long-term e.g. insulin, antihypertensives.

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

How does patient suitability affect what drugs are prescribed?

A

Age:

  • drug metabolism is low in newborns, enhanced in young people & becomes less effective w/ age
  • Drug excretion calls w/ age-related decline in renal function

Sex:
- women have higher body fat than men- increases vol of distribution & half-life of lipid-soluble drugs

Body weight:
- obesity increases vol of distribution & half life of lipid-soluble drugs

Smoking:
- tar in tobacco smoke stimulates oxidation of some drugs

Alcohol:

  • consumption stimulates liver enzyme synthesis
  • binge drinking may temporarily inhibit drug mechanism

Take into account patient’s

  • features of disease- meds should be based on known or suspected sensitivity of infective organism
  • Co-existing disease- may be either an indication or contraindication to therapy- hypertensive patients might be prescribed b-blockers if they also have left ventricular impairment but not have asthma.

Patient adherence to therapy - prescribers should choose drugs w/ a simple dosing schedule or easier administration e.g. ACE inhibitor lisinopril once daily rather than captopril 3 times daily for hypertension

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

How does patient safety determine which drugs are prescribed?

A

Prescribers should be wary of choosing drugs that are likely to cause adverse effect (e.g. cephalosporins rather than alternatives for patients allergic to penicillin) or worsen coexisting conditions (e.g. b-blockers as treatment for angina in patients w/ asthma)

Prescribers should avoid giving combo of drugs that might interact, directly or indirectly

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

Definition of pharmacodynamic?

A

What the drug does to the body.

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

Definition of agonist?

A

Bind to receptor to produce conformational change that is coupled to a biological response

As agonist conc increases, so does proportion for receptors occupied & thus biological effect.

Partial agonists activate receptor but cannot produce maximal signalling effect like full agonist, even when all receptors occupied.

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

Definition of antagonists?

A

Bind to a receptor but do not produce the conformational change that initiates an intracellular signal.

Competitive antagonist- competes w/ endogenous ligands to occupy receptor-binding sites w/ resulting antagonism depending on conc of drug & ligand.

Non-competitive inhibit effect of agonist y mechanisms other than direct competition for receptor binding w/ agonist.

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

Definition of potency?

A

The strength of a drug at a particular dosage.

Concentration/ dosage required to produce 50% of maximal effect.

  • Drugs of different potencies will require different doses to elicit drug response required
  • very potent drug= only small amount needed to achieve full effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Definition of therapeutic index?

A

The ratio of the ED50 for therapeutic efficacy & for a major adverse effect.
- Usually based on adverse effects that might require dose reduction or discontinuation.

ED50- dose of a medication that produces a desired pharmacologic effect in 50% of the studied patient population that takes the medication

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

What are the mechanisms by which drugs ?

A

Pharmacokinetic

Pharmacodynamic

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

How does pharmacodynamics work?

A

When drug enters body, it interacts w/ receptor & creates signal
- This signal results in biological effect e.g. it can tell DNA to stop replicating.

Receptors can be divided into 4 types:

  • Ligand-gated ion channels
  • G protein-couples receptors
  • enzyme-linked receptors
  • intracellular receptors

Drugs interact w/ receptors which determines the effects of drugs:

  • Selectivity- describes the propensity for a drug to bind to 1 target rather than another
  • Affinity- describes propensity for a drug to bind to a receptor & is related to the molecular fit & strength of the chemical bond. Some drug-receptor interactions are irreversible, either due because affinity is so strong or because drug modifies structure of its molecular target.
  • Agonist
  • Antagonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How do ligand-gated channel receptors work? Example of drugs that use this receptor?

A

ligand- molecule or ion

channel has Wigan binding site
-when ligand binds to it, channel opens - briefly
allows sodium, potassium, calcium

E.g. GABA receptor, nicotinic acetylcholine receptor

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

How do g-coupled receptors work? Examples of drugs w/ this receptor?

A

Pass through cell membrane 7 times

Composed of 3 sub-units: alpha, beta, gamma- together known as G-protien

  • in inactive form, alpha subunit is attached to GDP
  • When ligand attaches to receptor, affinity for GTP increases, so GTP replaced GDP
  • This causes alpha subunit to dissociate from beta- game sub unit- both complex go on to interact w/ other enzymes & proteins (regulate), leading to some response.

3 types of G-protein:

  • Gs
  • Gi
  • Gq

Gs:

  • stimulative G- protein
  • activates enzyme adenylyl cyclase- which produces cAMP from ATP
  • cAMP (2nd messenger)- very important.

Gi:

  • Inhibitory g-protein
  • inhibits adenyl cyclase, thus lowering levels of cAMP in cell

Gq:

  • activates enzymes called PLC
  • PLC produces 2 second messengers- DAG & IP3
  • DAG leads to different responses through activation of protein kinases
  • IP3 produces responses by mediating intra-ceullar release of calcium

Have ability to amplify signals they receive e.g. 1 stimulated G-protein receptor can activate many adenylyl cyclase- results in more cAMP produced, thus amplifying response.

E.g. Acetylcholine receptors, B-adrenoceptors, dopamine receptors, opioid receptors

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

How do enzyme-linked receptors work?

A

Have extracellular binding sites (ligands) which hormones & growth factors bind to stimulate enzyme activity in cell.

Most enzyme-linked receptors are of tyrosine kinase type- means display kinase activity & amino acid tyrosine involved.

When ligand binds to 2 receptors, causes conformational change that results in aggregation of both receptors

  • Dimer is formed & tyrosine regions get activated & cause ATP to become ATP- results in auto-phosphorylation of receptors.
  • Once each tyrosine picks up phosphate group, different inactive intracellular proteins come up & attach to phosphorylated tyrosine
  • this causes conformational change in attached protein leading to cascade of activations that produce cellar response
20
Q

How do transcription factor receptors (intracellular receptors) work?

A

Located inside the cell rather on cell memorable like others
- ligand has to cross lipid membrane to bind to receptor

Once bonded, wigan-receptor complex moves to nucleus to bind to DNA & regulate gene expression
- lead to synthesis of specific proteins

Each cells DNA has code that synthesise proteins from which different receptors are assembled
-once assembled, receptors embedded into cell membrane & receive & respond to signalling molecules.

E.g. steroid receptors, thyroid hormone receptors, vitamin D receptors, retinoid receptors.

21
Q

How do cells regulate themselves?

A

Cells can down-regulate receptors to prevent damaged (signal overload)
- they can be removed from membrane & recycled so fewer number are expressed- decreased sensitivity to signalling molecules

Cell up-regulates when
most receptors get blocked & cell receives weak signals
- means more receptors are inserted into membrane thus increasing sensitivity to signalling molecules.

22
Q

What is the relationship between dose & response (graph)?

A

Plotting drug dose against response produces signmodial dose-response curve.

Increase in drug dose (which is proportional to plasma drug conc) produce increasing response but only w/in narrow range of dose
- further increase in dose beyond this range produces little effect.

Full agnostic produces maximum response that receptor is capable of
- partial agonist at same receptor will have lower efficacy

More potent drugs produce biological effects at lower doses- have lower ED50
- lessphortent drug can still have same efficacy if given at higher dose.

23
Q

Definition of desensitisation?

A

When biological response to a drug diminishes when given continuously or repeatedly
- possible to restore response by increasing the dose of drug

24
Q

Definition of Tachyphylaxis

A

desensitisation that occurs rapidly, sometimes w/ initial dose, Implies depletion of chemicals that may be necessary for pharmacological actions of drug or receptor phosphorylation.

25
Q

Definition of tolerance?

A

Graudual loss of response to a drug that occurs over days or weeks.
- slower change implies changes in receptors numbers or development of counter-regulatory physiological changes that offset the actions of drug.

26
Q

Definition of drug resistance?

A

Describing the loss of effectiveness of an antimicrobial or cancer chemotherapy drug

27
Q

What is meant by withdrawal?

A

When drugs induce chemical, hormonal & physiological changes that offset their actions, discontinuation may allow these changes to cause rebound withdrawal effect

Glucocorticoids w/drawal symptoms:

  • weakness
  • fatigue
  • decreased appetite
  • nausea
  • weight loss
  • vomiting
  • Signs- hypotension & hypoglycaemia

Opioids- w/drawal symptoms:

  • Sneezing
  • yawning
  • abdominal & leg cramping
  • nausea, diarrhoea, vomiting
28
Q

What are the stages of pharmacokinetics?

A
  1. Absorption
  2. Distribution
  3. Metabolism
  4. Excretion
29
Q

What is drug absorption? What does it depend on?

A

Absorption is the process by which drug molecules gain access to blood stream

Rate & extent of drug absorption depend on route of administration & drug solubility

Lipophilic- fat soluble
hydrophilic- water soluble
Lipid soluble- pass through gut much quicker

30
Q

Examples of enteral administration?

A

These routes involve administration via gastrointestinal tract

Oral- used by patients to self-administer

  • complex process that depends on drug surviving exposure to gastric acid, being absorbed across small bowel muscle into portal venous system & surviving metabolism by gut or liver enzymes after being swallowed.
  • Absorptions frequently incomplete
  • bioavailability describes proportion of dose that reaches systemic circulation intact

Sublingual (SL)- under tongue.

  • rapid absorption into systemic circulation w/out uncertainties w/ oral administration
  • E.g. opioid analgesics

Rectal (PR)
- rectal mucosa used as site of administration when oral route is compromised (vomiting or unconsciousness)

31
Q

Examples of parenteral administration?

A

Routes avoid absorption via gastrointestinal tract & first-pass metabolism in liver

Intravenous (IV)- into veins

  • all dose enters systemic circulation reliably, w/out concerns about absorption or first pass metabolism
  • rapidly achieves high plasma conc
  • Ideal for ill patients

Intramuscular (IM)-into muscles

  • easier to achieve than IV route
  • absorption less predictable & depends on muscle blood flow\
  • painful-less common
  • e.g. adrenaline for acute anaphylaxis

Subcutaneous (SC)- injected into fat layers

  • ideal for low oral bioavailability drugs- absorbed well from subcutaneous fat
  • e.g. insulin, heparin

Transdermal

  • Patch- enables drug to be absorbed through skin into circulation
  • E.g. oestrogen, nicotine, nitrates
32
Q

What are other routes of administration?

A

Topical|:

  • application of drug involves direct administration to site of action e.g. ears, skin, eye
  • achieves sufficient conc at site whilst minimising systemic exposure & risk of adverse effects elsewhere

Inhaled (INH)-

  • allows drugs to e delivered directly to a target in respiratory tree usually small airways
  • some of inhaled dose may be absorbed from lung or swelled & can reach systemic circulation
33
Q

What is meant by drug distribution?

A

Process by which drug molecules transfer into & out of blood stream

Influenced by

  • drug’s molecular size & lipid solubility
  • Extent to which it binds to proteins in plasma
  • susceptibility to drug transporters expressed on cell surfaces
  • its binding to its molecular target & to other cellular proteins
34
Q

How do most drugs distribute through the body? How does drug dose affect it?

A

Diffuse passively across capillary walls down conc gradient into interstitial fluid until conc of free drug molecules in interstitial fluid is equal to that in plasma

As drug molecules in blood are removed by metabolism or excretion, plasma conc falls, drug molecules diffuse back from tissue compartment into blood & annually all will be eliminated

This reverse movement of drug from tissues will be prevented if further drug doses are administered & absorbed in plasma.

35
Q

What is meant by volume of distribution?

A

The volume into which a drug appears to have distributed following IV injection

Drugs highly bound to plasma proteins may have volume of distribution below 10L e.g. warfarin & aspirin

Drugs that diffuse into interstitial fluid but do not enter cells due to low lipid solubility may have volume of distribution between 10 & 30L e.g. amoxicillin.

Drugs w/ larger volume of distribution have longer half-lives- take longer to reach steady state on repeated administration & eliminated more slowly from body after discontinuation

36
Q

What is meant. by metabolism? What are the different phases?

A

Process by which drugs are chemically altered from a lipid-soluble for absorption & distribution to a water soluble form that is necessary for excretion

Prodrugs- inactive form when administered but converted to active metabolite in vivo

Phase I

  • involves oxidation, reduction or hydrolysis to make drug more suitable for phase II reactions or excretion
  • Oxidation most common- involves cytochrome P450 of membrane-bound enzymes in endoplasmic reticulum of hepatocytes

Phase II:

  • involves combining phase I metabolites w/ substrate to form inactive conjugate that is more water-soluble
  • reactions include glucuronidation, sulphation, acetylation, methylation
  • necessary for renal excretion, otherwise lipid-soluble metabolites will diffuse back into body after glomerular filtration.
37
Q

What is meant by excretion? Process of excretion?

A

Process by which drugs & their metabolites removed from body.

Usual route for elimination of drugs that are low molecular weight & water-soluble to avoid reabsorption from renal tube

  • drugs bound to plasma proteins are not filtered by glomeruli
  • pH of urine is more acidic that plasma so some drugs become un-ionised & reabsorbed.
  • Alkalination of urine can hasten excretion
  • For some drugs, active secretion into proximal tubule lumen, rather than glomerular filtration- predominant mechanism of excretion- penicillin

Faecal excretion is main route of elimination for drugs w/ high molecular weight, including those that are excreted in bile after conjugation w/ glucuronide in liver & drugs not absorbed after enteral administration

  • those excreted in bile enter small intestine where, if lipid-soluble, are reabsorbed through gut wall & return to liver via portal vein
  • this recycling between liver, bile, gut & portal vein is called enterohepatic circulation & prolongs residency pf drugs in body
38
Q

What is meant by elimination kinetics? How does it work for different drugs?

A

Net removal of drug from circulation results from combination fo drug metabolism & excretion- described as clearance
- I.e vol of plasma that is completely cleared of drug per unit time

Elimination is high-capacity process- does not become saturated even at high dosage

  • rate of elimination therefore directly proportional to drug conc
  • higher drug conc will drive faster metabolic reactions & support higher renal filtration rates
  • results in first-order kinetics- when constant fraction of drug remaining in circulation is eliminated in a given time & the decline in con over time is exponential- i.e. drugs half life
  • Importance- effect of increasing doses on plasma conc is predictable- double dose leads to doubled conc

For other drugs e.g. alcohol, elimination capacity is exceeded w/in usual dose range- zero-order kinetics
- Importance- if rate of administration exceeds maximum rate of elimination, drug will accumulate progressively, leading to toxicity

39
Q

What are the 4 mechanisms of drug interactions?

A

Absorption interactions:

  • drugs that either delay or enhance gastric emptying influence the rate of plasma conc increase of other drugs but not the total amount of drug absorbed
  • drugs that bind to form insoluble complexes can reduce absorption

Distribution interactions:

  • co-administration of drugs that compete for protein binding in plasma can increase unbound drug conc
  • but effect is short lived due to increased elimination & restoration of pre-interaction equilibrium
  • E.g. diazepam

Metabolism interactions:

  • many drugs rely on metabolism by different isoenzymes of cytochrome P450 (CYP) in liver
  • CYP enzyme inducers reduce plasma conc of other drugs, but may enhance activation of prodrugs.
  • CYP enzyme inhibitors have opposite effect e.g. grapefruit juice

Excretion interactions:

  • affect renal excretion
  • e.g. drug-induced reduction in glomerular filtration rate can reduce clearance & increase plasma con of many drugs including those w/ low therapeutic index e.g. NSAID, ACE inhibitors, diuretic induced dehydration
  • Less commonly- interaction may be due to competition for common anion transporter e.g. methotrexate excretion may be inhibited by competition w/ NSAID
40
Q

How to avoid drug interactions?

A

Drug interactions increase in patients w/ polypharamcy

Avoid drug interactions by taking careful drug history before prescribing more drugs, only prescribing for clear indications & take special care when prescribing drugs w/ narrow therapeutic index (warfarin)

Sometimes interactions are unavoidable- need to anticipate potential risk & warn patients & arrange for monitoring

41
Q

Define medication error.

A

any preventable event that may lead to inappropriate medication use or patient harm while the medication is in control of the healthcare professional or patient

42
Q

What are the system factors that contribute to prescribing errors?

A

Working hours of prescribers

Patient throughout

Professional support & supervision by colleagues

Availability of info e.g. medical records

Distractions

Availability of decision support

Checking routines

Reporting & reviewing of incidents

43
Q

How can user contribute to prescribing errors?

A
Knowledge:
- Clinical pharmacology principles
• Drugs in common use
• Therapeutic problems commonly encountered
• Knowledge of workplace systems
Skills:
• Taking a good drug history
• Obtaining information to support prescribing
• Communicating with patients
• Numeracy and calculations
• Prescription writing

Attitudes:
Coping with risk and uncertainty
• Monitoring of prescribing
• Checking routines

44
Q

What is the human error theory?

A

Unitended errors occur:

  • because prescriber fails to complete the prescription correctly- a slip
  • E.g. writes dose in mg and not micrograms
  • Or forgets part of the action that is important for success- a lapse
  • E.g. forgets to co-presribe folic acid w/ methotrexate

Intended error occurs:

  • when prescriber acts incorrectly due to lack of knowledge- prescriber is unaware - a mistake
  • E.g. prescribes atenolol for patient w/ known severe asthma because not educated about contraindication
  • Violation- deliberate deviations from standard practise- presider is aware
45
Q

How to reduce, identify & correct medication errors?

A

Prevention of error must focus on training prescriber

All prescribers make errors

  • First duty is to protect patient’s safety- clinical review & taking steps that will reduce harm e.g. monitoring, recording event in notes, informing colleagues.
  • Patient should be informed if exposed to potential harm

For errors that do not reach patient- prescribers duty to report them so others can learn from experience & take opportunity to reflect on how similar incidents can be avoided in future

Most errors occur due to prescriber & health-service systems
- organisations encourage reporting of errors w/in ‘no-blame culture’ so they can be subject to ‘root cause analysis’ using human error theory

Prevention of errors targets the causes mentioned above

  • supported by prescribers communicating & cross-checking w/ colleagues
  • by health-care systems providing clinical pharmacist support (e.g. for checking the patient’s previous & current prescriptions)
  • electronic prescribing (which avoids errors due to illegibility or serious dosing mistakes)
46
Q

What are the main components of a medication history?

A

Check written sources of info e.g. drug list on referral letter or patient record

  • compare w/ patient’s own recollection of what they take
  • Use generic pharmaceutical names for patients understanding
  • Ask about prescribed drugs & over counter drugs, herbal remedies & vitamin/mineral supplements
  • Ask about inhalers, topical meds (as patients assume tablets only)
  • note drug names, doses, duration of treatment
  • if patients on drugs for addiction, as community pharmacy to confirm diagnosis & stop dispensing for duration of hospital admission

Half of patients do not take prescribed meds as directed

  • Adherence- take meds as prescribed
  • Concordance- implies patient & doctor has negotiated & reached agreement on management- means adherence is likely

Ask for allergies to meds
- ask for other allergies e.g. pollen & food

Ask if they use non-prescribed drugs (cannabis)- ask following questions:
-what drugs are you taking?
how often & how much?
- how long have u taken them?
- what symptoms do you have if you cannot get drug?
- do you ever inject? needles or syringe?

47
Q

What is medical reconciliation?

A
  • Lists the patients current medications
  • List the medications currently needed
  • Compare lists
  • Make new list based on comparison
  • comunicate w/ patient

Prescriptions are reviewed for optimal safe & appropriate therapy.