Therapeutic Drug Monitoring Flashcards

1
Q

What is therapeutic drug monitoring?

A
  • The use of drug measurements in body fluids to aid the management of drug therapy for the cure, alleviation, or prevention of disease
  • Aims to aid clinicians in choice of drug dosing to provide optimum treatment for the patient and avoiding iatrogenic toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

When is the use of Therapeutic Drug Monitoring valid?

A
  • Poor correlation between dose and effect
  • Good correlation between plasma drug concentration and effect
  • Narrow concentration interval between therapeutic and toxic effects
  • Absence of good clinical/laboratory markers of effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Why is Poor correlation between dose and effect important for TDM?

A
  • For many drugs, dose is the primary determinant of effect. Standard doses can be used to titrate and TDM not required
  • TDM of benefit where there is poor correlation between dose and effect i.e. where there is wide inter-individual pharmacokinetic (movement of drugs within the body) variation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Why is good correlation between plasma drug concentration and effect important in TDM?

A

Essential requirement for valid TDM! Concentration must give accurate information about the biological effect/toxicity of the drug

A close concentration-effect relationship requires:

  • Little between-individual pharmacodynamic variation such as relationship between the concentration of the drug at its site of action and its effect
  • No active metabolites that contribute to effect that are not measured by the assay (e.g. carbamazepine)
  • Reversible mode of action at the receptor site (e.g. valproate)

Kinetic homogeneity”: Assumption that pharmacological response of drug is correlated to its concentration at the site of action, and that this is in turn related to its plasma concentration

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

Why is Narrow concentration interval between therapeutic and toxic effects important in TDM?

A

Therapeutic index = the margin between the therapeutic dose and the toxic dose – the higher (or wider) the better

  • Many drugs where TDM is used have a low (narrow) therapeutic index – margin between desirable and toxic dose is small
  • Symptoms of disease and of toxicity are hard to distinguish
  • Efficacy of the drug is hard to assess clinically

TDM unnecessary for drugs with a high therapeutic index as its is safe to use at much higher doses than required

Essential that a well-defined therapeutic range exists!

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

Why is Absence of good clinical/laboratory markers of effect important for TDM?

A
  • No point undertaking TDM if desired/toxic effects of drugs can be assessed via clinical assessment/laboratory tests
  • Examples: Antihypertensives (blood pressure), Statins (Lipids), Oral hypoglycaemic drugs (Glucose/HbA1c), Warfarin (INR)
  • Rare circumstances where TDM may be useful if drug not providing expected clinical effect e.g compliance issues or poor absorption/receptor dysfunction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are indications for Therapeutic Drug Monitoring?

A
  • Avoidance of toxicity: Toxicity is a clinical syndrome, not diagnosed by TDM but knowledge of drug concentration in plasma often valuable especially where the drug and the condition give similar features
  • Dosage adjustment: Especially where pharmacokinetics changing
  • Compliance checking: With care (the pharmacokinetic variability that makes TDM useful may provide a range of results )
  • Establishing a baseline: To ensure sufficient drug is reaching receptor to produce desired effect and relating this dose to a plasma concentration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are some patient information requirment for Therapeutic Drug Monitoring?

A
  • Patient: Age, Gender, Renal/hepatic function, Symptoms
  • Clinical indication: I.e. compliance/control/toxicity
  • Drug: Dose, Length of treatment, Time of dose, Other drugs
  • Sample: Time collected
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the appropriate samples used for TDM?

A

Serum/plasma samples most commonly used in TDM

Timing of sample of critical importance

  • Steady state concentrations of drugs most informative
  • Often trough (pre-dose) samples used
  • Exception = suspected overdose/toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are some factors that infleunce Acucurate analysis fo drugs?

A
  • Requirement for specificity is crucial: Should ideally detect all pharmacologically active substances
  • ‘Older’ drugs tend to be measured by immunoassay. Homogenous immunoassays have rapidly increased use of TDM such as FPIA, EMIT, CEDIA, KIMS etc
  • ‘Newer’ drugs often measured by chromatography such as GC-MS, HPLC, LC-MS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

How can results for TDM be interperated?

A

Requires knowledge of pharmacokinetics/dynamics of individual drugs from both the biochemist and the clinician.

Needs to be in response to a well formulated question

The aim of TDM is NOT to get drug concentrations into a nominal ‘target’ range

  • Ranges are adjunctive to clinical judgement and do not replace it
  • Optimum drug levels vary from patient to patient

Treat the patient and not the concentration

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

What are the indications, dose, mode of action, measurement and sampling time for Lithium?

A
  • Indication: Prophylaxis/treatment of bipolar disorder and mania and Refractory depression
  • Dose: Orally as lithium salts (carbonate) (1/2 day)
  • Mode of action: Unclear – modulates neurotransmission
  • Measurement: Direct ISE or colorimetric assays
  • Sampling time: 12 hours post dose in steady state
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is the pharmacokinetics of Lithium?

A
  • Renally excreted
  • Not bound to plasma proteins
  • Evenly distributed throughout body water
  • Accumulates in brain, kidney, thyroid
  • Thiazide diuretics and NSAIDs reduce clearance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the target range for Lithium?

A
  • Levels up to 1.2 mmol/L in acute therapy
  • 0.6 – 0.8 suggested NICE target in long-term treatment
  • 0.4-1.0 mmol/L as standard target

Lower levels in elderly and higher in younger or acute cases

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

How can Lithium lead to diabetes insipidus, hypothyroidism and hyperparathyroidism/hypercalcaemia?

A

Diabetes Mellitus

  • Due to lithium accumulation in renal tubules – leads to ADH resistance
  • Common renal SE of lithium tx

Hypothyroidism

  • Inhibits release for thyroid hormones – mechanism poorly understood
  • Often associated goitre (~40%) and hypothyroidsim (~20%)
  • Hypothyroidism is often mild/subclinical
  • Less commonly associated with hyperthyroidism

Also (rarely) hyperparathyroidism/hypercalcaemia

  • Likely due to an increase at which calcium supresses PTH release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are side effects of Lithium?

A
  • Toxicity
  • Chronic lithium therapy associated with hypothyroidism and diabetes insipidus (DI)
  • Also (rarely) hyperparathyroidism/hypercalcaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How can lithium be toxic?

A

Highly toxic and has narrow therapeutic range

  • Acute toxicity: Ataxia, confusion, tremor, muscle weakness, seizures, slurred speech, renal damage.
  • Levels >1.5 mmol/L require urgent review
  • Levels >3.0 mmol/L can be fatal even in patients maintained on Lithium
  • Treated with dialysis if severe

Chronic accumulation worse than acute toxicity (TDM essential)

  • Declining renal function leads to greatest danger of toxicity.
  • Lithium deposits in brain, thyroid, kidneys.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are the indications, dose, mode of action, measurement and sampling time for digoxin?

A
  • Indication: Chronic cardiac failure and atrial flutter/fibrillation
  • Dose: Orally (typical maintenance dose 125ug) or IV for emergency loading
  • Mode of action: Cardiac glycosid. It binds Na-K ATPase pump of cardiac myocytes – to inhibit it. Increased intracellular Na concentration causes increased intracellular Ca2+, which increases cardiac contractility
  • Measurement: Immunoassay
  • Sampling time: 6 hours post-dose
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the target range for Digoxin?

A
  • 0.9 – 2.0 ug/L typical reference range
  • 0.5 – 1.0 ug/L recommended by UK Pathology Harmony
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are Pharmacokinetics of Digoxin?

A
  • Mostly renal excretion
  • Around ¼ protein bound
  • High Vd and renal excretion means long half-life
  • Several drug interactions that affect absorption, binding, clearance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are factors that that are related to toxicity of digoxin?

A

Toxic effects digoxin related to

  • Concentration – more likely >2ug/L
  • Age
  • Degree of heart failure
  • Electrolyte imbalances (hypokalaemia potentiates digoxin toxicity, also hypercalcaemia, hypomagnesaemia)
  • Care in elderly and impaired renal function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What are the toxic effects of Digoxin?

A
  • Gastrointestinal: Abdominal pain, nausea, vomiting, diarrhoea
  • Neurotoxicity: Headaches, fatigue, insomnia, confusion
  • Cardiac: Bradycardia, AV block, arrhythmias
  • Visual disturbances: blurred vision, coloured casts/halos

TDM essential

23
Q

What is the treatment for Digoxin toxicity?

A

Severe toxicity/poisoning can be treated with DigiBind/DigiFab

  • Digoxin specific antibody fragments us in cases of life-threatening toxicity. Severe toxicity often digoxin >3-4 ug/L and may be associated with hyperkalaemia
  • Decision to treat is clinical and not on the basis of digoxin or K+ levels
  • Hypokalaemia may develop rapidly
24
Q

What are the assay specific interferences for Digoxin?

A

DLIS (Digoxin-like immunoreactive substances)

  • Usually cause false elevations in digoxin immunoassays
  • Increased in elderly/pregnant patients, and in renal/liver failure
  • Assay-specific degree of interference

DigiBind/DigiFab will cross-react in immunoassays

  • Digoxin cannot be measured until DigiBind is cleared (15-20h t1/2)

Some drugs (e.g. spironolactone) may interfere

25
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Phenytoin?

A
  • Indication: Tonic-clonic, Grand mal, Focal, Partial seizures, Status epilepticus (now rarely)
  • Dose: Oral or IV
  • Mode of action: Inhibits voltage-gated Na channels to reduce spread of excitation from epileptic foci
  • Measurement: Immunoassay
  • Sampling time: Usually trough levels
  • Target range: 5-20mg/L (UK Pathology harmony), but wide variation
26
Q

What are the paharcokinetic properties of Phenytoin?

A
  • Absorption can be variable
  • Saturation kinetics (aka zero order kinetics)
  • >90% protein bound; total concentration will be affected by plasma protein level – competition for binding from other drugs i.e. valproate, hypoalbuminaemia, pregnancy, liver/renal failure
  • Metabolism is via hepatic CYP450 enzyme CYP2D9 – enhanced and indibited by a range of other drugs
27
Q

What are some drug interactions of Phenytoin?

A

Increase Metabolism

  • Folic acid, Dexamethasone, Phenobarbital, Diazepam, Rifampin, Methadone, Estrogens

Decrease metabolism

  • Valproic acid, Carbamazepine, Warfarin, Cimetidine, Omeprazole, Ibuprofen, Metronidazole, Fluconazole, Fluoxetine, Risperidone, Amiodarone
28
Q

What are side effects of Phenytoin?

A

Widely used; poor side effect profile means that not 1st line

  • Cosmetic; coarse faces/acne/gingival hyperplasia/hirsutism
  • Depression and behavioural changes
  • Osteomalacia and folate deficiency in long-term use

Neurotoxic effects increasing likely where phenytoin >20mg/L

  • Nystagmus/ataxia/vomiting/nausea/tremor
  • Paradoxical seizures clinically indistinguishable from epileptic seizures
  • Prolonged toxicity may lead to irreversible cerebellar/nerve
29
Q

What are first and zero order elimination kinetics?

A
  • First-order: Elimination of a constant fraction per time unit of the drug quantity present. The elimination is proportional to the drug concentration.
  • Zero-order: Elimination of a constant quantity per time unit of the drug quantity present. The elimination is independent of the drug concentration.
30
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Carbamezepine?

A
  • Indication: Tonic-Clonic (Grand mal) and partial (focal seizures), Bipolar disorder/mania/depression, Trigeminal neuralgia
  • Dose: Oral
  • Mode of action: Inhibits voltage-gated Na channels to reduce spread of excitation from epileptic foci
  • Measurement: Immunoassay
  • Sampling time: Trough
  • Target range: 4-12 mg/L (UK Pathology Harmony)
31
Q

What are implications of zero order kinetics?

A
  • Most drugs exhibit 1st order kinetics – clearance proportional to concentration.
  • Zero order kinetics occur above a point where metabolism/clearance mechanisms are saturated - therefore becomes independent of concentration
  • Effect is that small increases in drug doses or change to metabolism (drugs/illness) can have major impact on plasma concentration and half life of the drug
32
Q

What are pharmacokinetic properties of Carbamezepine?

A
  • Hepatic metabolism by CYP3A4 to active 10,11-epoxide
  • Mostly protein bound
  • Short half-life
  • Metabolism is induced by phenobarbital and phenytoin
  • Metabolism is inhibited by valproate and lamotrigine
33
Q

What are side effects of Carbamezepine?

A
  • Blurred vision
  • Dizziness
  • Ataxia
  • Erythematous rash in 3-5% patients
  • SIADH
  • Leucopenia (rare)
34
Q

Why does Carbamezepine require TDM?

A
  • Narrow therapeutic range
  • Poor dose-concentration relationship
  • Drug interaction
  • But presence of an active metabolite not measured in most methods
  • TDM used to establish baseline and where seizures are hard to control
35
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Valproate?

A
  • Indication: Myoclonic and absence seizures
  • Dose: Oral
  • Mode of action: Enhances post-synaptic GABA response
  • Measurement: Immunoassay
  • Sampling time: Trough
  • Target range: 50 – 100 mg/L as guide only
36
Q

What are phamacokinetic properties of Valproate?

A
  • Protein bound in concentration dependent manner
  • Hepatic metabolism via variety of mechanisms
  • Clearance increased by other anticonvulsants
  • Short half live but metabolites have slower clearance rates
37
Q

What side effects of Valproate?

A
  • Lack of CNS side effects and sedation make it a 1st line drug especially in children
  • Rarely associated with severe hepatotoxicity and hyperammonaemia in children
38
Q

Why is Valproate not candidate for routine TDM?

A
  • Concentration-effect correlation uncertain; therapeutic effects continue after drug cleared from circulation
  • Few concentration dependent toxic effects
  • Intra-individual variation
  • Lack of evidence-based reference range
39
Q

What are features of ‘Newer’ anticonvulsants and their TDM?

A
  • Often have wider therapeutic ranges than older ACDs and fewer serious adverse effects
  • Reference intervals often not established
  • Routine monitoring not usually recommended at this time
  • Analysis tends to be limited to chromatographic assays in specialist centres
40
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Theophylline?

A
  • Indication: Asthma and COPD
  • Dose: Oral for chronic treatment or IV (aminophylline) for acute exacerbations of asthma
  • Measurement: Immunoassay
  • Sampling time: Trough
  • Target range: 8-20mg/L
41
Q

What is the mode of actions of Theophylline?

A
  • Inhibition of phosphodiesterase PDE3 and PDE4
  • Theophylline and caffeine are methylxanthines; drugs that relax smooth muscle. Can thus relieve or prevent bronchoconstriction
  • No longer 1st line therapy for asthma + COPD
  • Bronchodilation & anti-inflammatory effects superseded by other drugs but still has role in some patient groups
42
Q

What are pharmacokinetics properties of Theophylline?

A
  • Modified release tablets often used
  • Hepatic metabolism via hydroxylation and demethylation: ~50-60% protein bound
  • Highly variable rate of metabolism
  • Concentrations increased in heart failure and cirrhosis
  • Concentrations decreased in smokers, chronic alcoholics and by enzyme-inducing drugs
43
Q

What are side effects of Theophylline?

A
  • Common within target range such as Nausea/Headaches/Jitteriniess
  • Toxicity occurs at concentrations >20mg/L. Leads to. Arrhythmias/seizures/tremors/palpitations
  • Varied metabolism = poor correlation between dose & concentration
44
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Ciclosporin and Tacrolimus?

A

Indication

  • Used to prevent solid organ graft rejection
  • Autoimmune diseases – RA/UC/Psoriasis/Dermatitis

Dose

  • Oral (liquid or capsules) or IV
  • Oral maintenance doses following kidney transplant: Ciclosporin 10-15mg/kg/day/Tacrolimus 0.1-0.2mg/kg/day

Mode of action

  • Calcineurin inhibitors – inhibit cytokine production and T-cell activation. Tacrolimus 10-100x more potent

Measurement: Immunoassay or Chromatography

Sampling time:

  • Trough (C0) or Peak (C2) for ciclosporin
  • Trough for tacrolimus

Target range: Vary with time after transplant/transplant type/other drugs

45
Q

What are the pharmacokinetic properties of Ciclosporin and Tacrolimus?

A
  • Low and variable oral bioavailability
  • Hepatic metabolism by CYP3A4 enzyme system
  • Highly lipophilic and concentrates in tissues
  • Many drug interactions
  • Almost completely plasma protein bound
46
Q

What is the Therapeutic Drug monitoring for ciclosporin and tacrolimus?

A
  • Wide variation in pharmacokinetics – many drug interactions
  • Narrow therapeutic index - overlap between efficacious levels and toxicity
  • Potential impact of under-replacement/non compliance
  • Measurement is in EDTA whole blood: Ciclosporin levels 2-3x higher in RBCs/Tacrolimus levels 20-30x higher in RBCs
  • Variation in exposure to ciclosporin in 1st 4 hours: C2 sample better correlation with AUC, reflecting exposure/ C2 measurement show to better reflect with efficacy and toxicity
47
Q

What are side effects of Tacrolimus and Ciclosporin?

A
  • Renal dysfunction – vasoconstriction effect may lead to chronic graft dysfunction
  • Increase blood pressure
  • Ciclosporin associated with hyperlipidaemia and adverse cosmetic effects
  • Tacrolimus associated with new-onset diabetes in 10% cases
  • Chronic over-immunosuppression related to infection and neoplasia
48
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Gentamicin?

A
  • Indication: Severe systemic infection by some gram-positive and many gram-negative organisms
  • Dose: Parenteral, usually once daily 5-7mg/kg
  • Mode of action: Interfere with protein synthesis in susceptible organisms and has Bactericidal effects
  • Measurement: Immunoassay
  • Sampling time: Trough or peak
  • Target range:
    • Trough = <1-2mg/L
    • Peak = 5-10mg/L
49
Q

What are pharmacokinetic properties for Gentamicin?

A
  • Must be given parenterally as poor oral bioavailability
  • Not bound to proteins
  • Not metabolised
  • Cleared renally
  • Short plasma half-life, but long elimination phase as drug concentrated in tissues
  • Post-antibiotic effect
50
Q

How is TDM conducted for Gentamicin?

A

Poor dose-concentration relationship. Varied absorption from IM dose/volume of distribution/half-life (renal clearance)

Narrow therapeutic index

  • Nephrotoxicity: A ‘vicious cycle’ as impaired renal function will reduce aminoglycoside excretion. Usually reversible
  • Ototoxicity

Target range is used to determine next dosing interval. Hartford nomogram may be used

  • Not applicable to children, pregnancy, ascites, endocarditis, CF, burns, neutropenia, severe renal impairment
  • Redosing often based on body weight in clinical practice ​
51
Q

What are the indications, dose, mode of action, measurement, target range and sampling time for Vancomycin?

A
  • Indication: Prophylaxis/treatment of serious infections caused by gram-positive bacteria (e.g MRSA)
  • Dose: IV
  • Mode of action: Inhibits cell wall synthesis in gram positive bacteria
  • Measurement: Immunoassay
  • Sampling time: Peak or trough levels
  • Target range:
    • Peak = 20-40mg/L
    • Trough = 10-20mg/L
52
Q

What are Pharmacokinetic Properties of Vancomycin?

A
  • Must be given parenterally as poor oral bioavailability
  • Not bound to proteins
  • Not metabolised
  • Cleared renally
  • Post-antibiotic effect
53
Q

What are Side Effects of Vancomycin?

A
  • As for aminoglycosides, nephrotoxicity and ototoxicity the major concerns
  • Risk of toxicity increases if aminoglycosides are also given
  • Flushing of upper body (‘red man syndrome’) if infusion too rapid/concn too high
54
Q

How is TDM conducted for Vancomycin?

A
  • Little role for peak concentration?
  • Trough levels more useful
  • Some support TDM in all patients treated for >2-3 days
  • Others target specific patient groups