flashcards 2

(50 cards)

1
Q

What is the primary purpose of Therapeutic Drug Monitoring (TDM)?

A

To measure drug concentrations in body fluids (e.g., blood, urine) to optimize treatment, ensuring efficacy while avoiding toxicity.

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

Since when has TDM been routinely performed in laboratories?

A

TDM has been routine in analytical laboratories since the 1970s.

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

Why might doubling a dose not necessarily improve a drug’s clinical effect?

A

Because the clinical response is proportional to the drug concentration at the site of action, not simply the administered dose; exceeding the therapeutic window can cause toxicity.

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

What factors affect a drug’s availability at its site of action?

A

Absorption (including first-pass metabolism), distribution (lipid vs. water solubility, volume of distribution), metabolism, excretion, and plasma protein binding.

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

What is meant by ‘first-pass metabolism’ in the context of TDM?

A

First-pass metabolism refers to the initial metabolism of an orally administered drug by gut and liver enzymes before reaching systemic circulation, reducing bioavailability.

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

Define the terms MTD and MED in relation to the therapeutic window.

A

MTD is the Maximum Tolerated Dose (threshold above which toxicity occurs), and MED is the Minimum Effective Dose (threshold below which therapeutic effect is inadequate).

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

When is the ideal time to take a blood sample for TDM to assess steady-state concentration?

A

Immediately before the next scheduled dose (trough level), when the drug concentration is at its lowest in the dosing interval.

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

What are ‘peaks’ and ‘troughs’ in a dosing regimen?

A

‘Peaks’ are maximum drug concentrations after a dose; ‘troughs’ are minimum concentrations just before the next dose.

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

Why is patient compliance critically considered in TDM?

A

Because nonadherence (e.g., missed doses, incorrect timing) can lead to subtherapeutic or toxic concentrations, misleading TDM interpretation.

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

How can physiological differences affect TDM outcomes?

A

Variations in body mass, organ function (hepatic/renal), age, and disease states can alter pharmacokinetics, changing drug absorption, distribution, metabolism, and excretion.

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

What role do drug–drug interactions play in TDM?

A

Concomitant medications can induce or inhibit metabolic enzymes (e.g., CYP450), altering clearance and resulting in unexpectedly high or low drug levels.

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

In pharmacodynamics, why is receptor status important for TDM?

A

Because the relationship between drug concentration and effect depends on drug binding to its receptor; receptor polymorphisms or disease states can modify response.

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

What characterizes a drug exhibiting zero-order kinetics?

A

Zero-order kinetics occurs when the metabolism rate is at or near enzyme capacity (e.g., phenytoin, ethanol), so a constant amount—not a constant fraction—is metabolized per unit time.

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

Which pharmacokinetic parameter describes the fraction of an administered dose that reaches systemic circulation?

A

Bioavailability (F), reflecting absorption and first-pass metabolism for non-intravenous routes.

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

What is ‘volume of distribution’ (Vd)?

A

Vd is a theoretical volume representing how extensively a drug distributes into tissues versus remaining in plasma; it helps infer tissue binding and storage.

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

Why is plasma protein binding relevant in TDM?

A

Highly protein-bound drugs have less free (active) concentration; changes in binding (e.g., due to hypoalbuminemia) can alter the free fraction and require dose adjustments.

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

What is the difference between clearance and elimination rate?

A

Clearance is the volume of plasma cleared of drug per unit time (mL/min or L/hr), while elimination rate describes the change in drug concentration over time (e.g., half-life).

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

Describe the relationship between dose and plasma concentration for most drugs.

A

Most drugs exhibit first-order kinetics, meaning plasma concentration changes proportionally with dose, as a constant fraction is eliminated per unit time.

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

What is internal quality control (QC) in a TDM laboratory?

A

Procedures using known control samples to monitor day-to-day analytical performance, detect drift, and ensure reproducibility.

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

What is external quality assurance (EQA) in TDM?

A

An independent program (e.g., UKNEQAS) that sends blind samples to laboratories to verify accuracy and maintain accreditation.

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

What is a Levey-Jennings chart used for in QC?

A

To plot control sample results over time against the mean and ±2 standard deviation limits, identifying trends or shifts in assay performance.

22
Q

In QC terminology, what defines a ‘shift’?

A

Six or more consecutive QC results on one side of the mean, indicating a sudden change in assay accuracy.

23
Q

In QC terminology, what defines a ‘trend’?

A

Six or more consecutive QC results moving steadily upward or downward, indicating gradual drift in assay performance.

24
Q

What does ALTM stand for, and how is it calculated?

A

All Laboratory Trimmed Mean: the average QC result after removing the top and bottom 5% of values to set a target value.

25
How is 'bias' defined in QC for TDM assays?
Bias is the percentage deviation of the laboratory’s measured value from the target (ALTM) value for a control sample.
26
What is 'rolling bias' in QC parameters?
The mean of current and previous five QC results, indicating short-term assay accuracy trends.
27
What is 'rolling variability' in QC?
The average deviation (ignoring direction) of the current and previous five QC results, reflecting assay precision over time.
28
Define 'Bias Index Score (BIS)' in QC.
BIS compares each QC result’s deviation from the target to assess overall laboratory performance across multiple analytes.
29
Why is TDM indicated in cases of suspected toxicity?
To determine if high drug or metabolite levels (due to overdose, impaired metabolism/excretion, or interactions) are causing toxicity.
30
Give three examples of drugs requiring TDM due to narrow therapeutic index.
Digoxin (cardiac glycoside), Lithium carbonate (psychiatric), and Carbamazepine (anticonvulsant).
31
What laboratory method is commonly used to measure Digoxin levels?
Immunoassay techniques (e.g., immunofluorescence or immunoenzymatic assays).
32
Which drugs can elevate Digoxin concentrations via CYP450 interactions?
Amiodarone, Quinidine, and Verapamil can inhibit Digoxin clearance, raising serum levels.
33
Why is TDM critical for patients with renal or hepatic impairment?
Because reduced metabolism (hepatic) or excretion (renal) can lead to drug/metabolite accumulation and toxicity.
34
Name three patient populations for whom TDM is especially important.
Pediatric (especially neonates), geriatric, and patients undergoing dialysis or hemofiltration.
35
Why is TDM generally not required for drugs with a broad therapeutic index?
Because dosage variations are less likely to cause toxicity or loss of efficacy, and clinical endpoints can often be monitored functionally.
36
What defines a 'hit-and-run' drug for which TDM is not typically needed?
Drugs with rapid onset and offset of action, minimal toxicity risk, and easily measurable functional effects (e.g., omeprazole, MAO inhibitors).
37
Which class of antibiotics often requires TDM and why?
Aminoglycosides (e.g., Gentamicin, Streptomycin) require TDM due to nephrotoxicity and ototoxicity risks at high concentrations.
38
Describe the pharmacokinetic properties of aminoglycoside antibiotics relevant to TDM.
Poor oral absorption, wide distribution (Vd ≈ 0.25 L/kg), negligible plasma protein binding, renal excretion via glomerular filtration, and a half-life of ~2–3 hours.
39
What toxic effects are associated with aminoglycoside accumulation?
Nephrotoxicity (tubular cell damage via phospholipid metabolism disruption) and reversible ototoxicity (inner ear free radical generation).
40
Name two anti-epileptic drugs that require TDM and their metabolic considerations.
Phenytoin (zero-order kinetics near enzyme capacity) and Phenobarbital (administered as prodrug Primidone, hepatic metabolism).
41
Which psychoactive drug requiring TDM is used for mood stabilization?
Lithium carbonate, due to its narrow therapeutic window and risk of renal toxicity.
42
Why is Cyclosporine monitored in transplant patients?
Because Cyclosporine has a narrow therapeutic index and can cause nephrotoxicity; dosing is complicated by variable absorption and metabolism.
43
What anti-neoplastic agent commonly undergoes TDM and why?
Methotrexate, since it inhibits DNA synthesis and can cause severe toxicity if plasma levels are too high; clearance varies with renal function.
44
How long after a single marijuana use can THC metabolites be detected in urine?
Approximately 3–5 days, using GC/MS methods.
45
Which analytical techniques are used to detect amphetamines in body fluids?
Liquid chromatography (LC) and gas chromatography (GC).
46
Why are anabolic steroids monitored in athletes?
Because they can enhance muscle mass and performance; illicit use is detected via GC or GC/MS in urine.
47
What are common methods to detect opioids in suspected abuse cases?
Gas chromatography–mass spectrometry (GC/MS) assays of urine or blood.
48
In a TDM context, why is sampling time critical when measuring trough levels?
Because sampling immediately before the next dose ensures the lowest steady-state concentration is measured, reflecting true maintenance dosing.
49
How does renal hemofiltration affect drug clearance and the need for TDM?
Hemofiltration can increase elimination of renally cleared drugs, necessitating dose adjustments and close TDM to maintain therapeutic levels.
50
What key information must be considered to interpret Amiodarone levels correctly?
Patient’s dosing history, time since last dose (half-life ~50 days), concurrent medications, and liver/thyroid function, since Amiodarone distributes extensively and has complex metabolism.