You are on page 1of 9

CLINICAL PHARMACY & Chapter 3: Practical Pharmacokinetic

PHARMCOTHERAPY 1 Ms. Neri Margaret Fabillo


AY 2022-2023 MM/DD/YYYY
2nd Semester

OUTLINE
I. General Applications: Response, Loading dose, etc.
II. Application to Therapeutic drug
III. Basic Concepts: Volume of distribution, Elimination,
Absorption, peak and toug levels, consent. data, etc.
IV. Clinical Applications: Estimation of creatinine clearance,
Digoxin, theophylline, gentamicin, etc.

KEY NOTES
•Pharmacokinetics can be applied to a range of clinical
situations with or without therapeutic drug monitoring (TDM).
•TDM can improve patient outcomes but is only necessary for
drugs with a low therapeutic index, where there is a good
concentration response relationship, and where there is no
easily measurable physiological parameter.
•Sampling before steady state is reached or before distribution
is complete, leads to erroneous results. B. NEED FOR A LOADING DOSE
• The volume of distribution can be used to determine the ● The same type of information can be used to determine whether
loading dose. the loading dose of a drug is necessary
• The elimination half-life determines the time to steady state → drugs with longer half-lives are more likely to require loading
and the dosing interval. doses for acute treatment.
• Kinetic constants determine the rate of absorption and
elimination.
C. DOSAGE ALTERATIONS
•Clearance determines the maintenance dose.
•Creatinine clearance can be reliably estimated from
● Clinical pharmacokinetics can be useful in determining dosage
population values.
alteration if the route of elimination is impaired through end
• Use of actual blood level data wherever possible to
organ failure (e.g. renal failure) or drug interaction.
assist dose adjustment is advisable. However, population
● Using the fraction that should be excreted unchanged ( fe
pharmacokinetic values can be used for digoxin, theophylline
value), quantitative dosage changes can be estimated.
and gentamicin.
•Once daily dosing of gentamicin is a realistic alternative to
multiple dosing. D. CHOOSING A FORMULATION
•TDM is essential in the dose titration of lithium and
phenytoin, but of little value for valproate, or the newer ● An understanding of the pharmacokinetics of absorption may
anticonvulsants. also be useful in evaluating the appropriateness of particular
Note: If no learning objectives were given during the lecture, either formulations of a drug in a patient.
use the ones in the handout given or delete this portion altogether
II. APPLICATION TO THERAPEUTIC DRUG
I. GENERAL APPLICATIONS MONITORING

● Clinical Pharmacokinetics III. BASIC CONCEPTS


→ may be defined as the study of the time course of the
absorption, distribution, metabolism and excretion of drugs ELIMINATION
and their corresponding pharmacological response.
→ can be applied in daily practice to drugs with a low Drugs may be eliminated from the body by a number of
therapeutic index, even if drug level monitoring is not routes. The primary routes are:
required. → Excretion of the unchanged drug in the kidneys
→ Metabolism (usually in the liver) into a more water soluble
A. TIME TO MAXIMAL RESPONSE compound for subsequent excretion in the kidneys,
→ Combination of both.
● By knowing the half-life of a drug, the time to reach a steady
state may be estimated, and also when the maximal therapeutic ● Clearance (CL)
response is likely to occur, irrespective of whether drug level → The main pharmacokinetic parameter describing elimination.
monitoring is needed. → It is defined as the volume of plasma completely emptied of
drug per unit time.

● Rate of Elimination
Eq. (3)

Where:
CLINPHARM Group 2 - 2A : Andres, Balando, Bucane, Canillas, Galangue,Gonzales, Llarenas, Mengote, Muncada, Pabia, Pazon, Rosales, Samante, Severino, Vierras, Yee 1 of 9
1
→ A constant rate of elimination irrespective of plasma
▪ Rate of Elimination = Rate of drug disappearing from the
concentration
body (𝑔/ℎ𝑟)
▪ CL = Clearance (𝐿/ℎ𝑟) ● For drugs undergoing first-order elimination, there are two other
useful pharmacokinetic parameters in addition to the volume of
▪ C = Plasma drug concentration (𝑔/𝐿) distribution and clearance. These are the elimination rate
constant and elimination half-life.
● Total Body Elimination
→ Is the sum of the metabolic rate of elimination and the renal ● Elimination Rate Constant (𝐾𝑒)
rate of elimination. → is the fraction of the amount of drug in the body (A)
eliminated per unit time.
Eq. (5)

→ If the fraction eliminated by the renal route is known (𝐹𝑒),


then the effect of renal impairment on total body clearance
▪ Combining Eqs. (3) and (5) gives,
can be estimated.

● Clearance of most drugs remains constant for each individual. 𝐶𝐿 × 𝐶 = 𝐾𝑒 × 𝐴


However, it may alter in cases of drug interactions, changing ▪ And since,
end-organ function or autoinduction. 𝐴
𝐶 = 𝑉𝑑
‘As the plasma concentration changes so will the rate of ▪ Then,
elimination.” 𝐴
𝐶𝐿 × 𝑉𝑑
= 𝐾𝑒 × 𝐴
● Steady state
→ When the rate of administration is equal to the rate of ▪ Therefore,
𝑠𝑠 Eq. (6)
elimination and the plasma concentration is constant (𝐶 ) . 𝐶𝐿 = 𝐾𝑒 × 𝑉𝑑

● Elimination Half-Life (𝑇1/2)


● Rate of administration → Is the time it takes for the plasma concentration to decay by
→ At the beginning of a dosage regimen the plasma half.
concentration is low. Therefore, the rate of elimination from → In five half-lives the plasma concentration will fall to
Eq. (3) is less than the rate of administration, and approximately zero.
accumulation occurs until a steady state is reached. Eq. (7)
−𝑘𝑒×𝑡
Eq. (4) 𝐶2 = 𝐶1 × 𝑒
Where:
▪ C1 and C2 = Plama concentrations
● First-order elimination
→ As the plasma concentration falls (e.g. on stopping treatment ▪ t = time
or after a single dose), the rate of elimination also falls.
→ The plasma concentration time graph follows a non-linear ● Two ways of determining 𝑘 :
curve characteristic. 𝑒
→ Estimating the half-life and applying Eq. (8)
Figure 3.4 First-order elimination → Substituting two plasma concentrations in Eq. (7) and
applying natural logarithms:

In 𝐶 = In 𝐶1 − (𝑘𝑒 × 𝑡)
2

(𝑘𝑒 × 𝑡) =In 𝐶1 − In 𝐶2

𝐼𝑛 𝐶1− 𝐼𝑛 𝐶2
𝑘𝑒 = 𝑡

● It takes approximately five half-lives for a drug to accumulate to


the steady state on repeated dosing or during constant infusion.

Eq. (8)
𝑠𝑠 −𝑘𝑒×𝑡
𝐶 =𝐶 ⎡⎣1 − 𝑒 ⎤

Where:
▪ C = The plasma concentration at time t after the start of
● Zero-order Elimination
the infusion

CLINPHARM Practical Pharmacokinetics 2 of 9


1
𝑠𝑠 ● To determine a loading dose:
▪ 𝐶 = the steady state plasma concentration.
Eq. (11)
ABSORPTION

● Intravenous Route
→ In this route all of the administered drug is absorbed.
→ Most efficient; Other routes must be absorbed into the
● to determine maintenance dose or steady state plasma
bloodstream thus it may or may not be 100% efficient.
concentrations at the steady state:
● Bioavailability (𝐹) Eq. (12)
→ The fraction of the administered dose which is absorbed into
the bloodstream
→ For oral administration, the dose or rate of administration
must be multiplied by 𝐹
→ Bioavailability 𝐹 is determined by calculating the area under
Where:
the concentration time curve (AUC). ▪ 𝑇 = the dosing interval.

● Rate of Elimination PEAK AND TROUGH LEVELS


→ The fraction of the administered dose which is absorbed into
the bloodstream.
● For oral dosing and constant intravenous infusions, it is usually
adequate to use the term ‘average steady state plasma
𝑇ℎ𝑒 𝑎𝑚𝑜𝑢𝑛𝑡 𝑒𝑙𝑖𝑚𝑖𝑛𝑎𝑡𝑒𝑑 𝑖𝑛 𝑎𝑛𝑦 𝑜𝑛𝑒 𝑢𝑛𝑖𝑡 𝑜𝑓 𝑡𝑖𝑚𝑒 𝑑𝑡 = 𝐶𝑙 × 𝐶 × 𝑑𝑡
concentration’ (average C ss).
→ However, for some intravenous bolus injections it is
𝑇𝑜𝑡𝑎𝑙 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝑑𝑟𝑢𝑔 𝑒𝑙𝑖𝑚𝑖𝑛𝑎𝑡𝑒𝑑 = 𝐶𝑙 𝐶𝐿 × 𝐶 × 𝑑𝑡 sometimes necessary to determine peak and trough levels.

As previously explained, CL is constant. Therefore , ● At the steady state,


→ the change in concentration due to the administration of an
𝑇𝑜𝑡𝑎𝑙 𝑎𝑚𝑜𝑢𝑛𝑡 𝑒𝑙𝑖𝑚𝑖𝑛𝑎𝑡𝑒𝑑 = 𝐶𝐿 × Σ𝐶 × 𝑑𝑡 from start until 𝐶 intravenous dose will be equal to the change in concentration
is zero due to elimination over one dose interval:

● Σ𝐶 × 𝑑𝑡 from start until zero is the area under the plasma


concentration–time curve (Fig. 3.5)
● Within one dosing interval,
𝑠𝑠
→ the maximum plasma concentration (𝐶 ) will decay to the
𝑚𝑎𝑥
𝑠𝑠
minimum plasma concentration (𝐶 ) as in any first-order
𝑚𝑎𝑥
process.
𝑠𝑠 𝑠𝑠
→ Substituting 𝐶 for 𝐶 and 𝐶 for 𝐶 in Eq. (7):
𝑚𝑎𝑥 1 𝑚𝑖𝑛 2

𝑠𝑠 𝑠𝑠 −𝑘𝑒×𝑡
𝐶𝑚𝑎𝑥 = 𝐶𝑚𝑎𝑥 × 𝑒
Where:
▪ 𝑡 = the dosing interval.

− If this is substituted into the preceding equation:

𝑆 × 𝐹 × 𝑑𝑜𝑠𝑒 𝑠𝑠 𝑠𝑠 −𝑘𝑒×𝑡
𝑉𝑑
= 𝐶𝑚𝑎𝑥 − (𝐶𝑚𝑎𝑥 × 𝑒 )
DOSING REGIMENS

● To determine the change in plasma concentration ΔC Therefore,


immediately after a single dose:
Eq. (13)
Eq. (10) 𝑠𝑠 𝑆 × 𝐹 × 𝑑𝑜𝑠𝑒
𝐶𝑚𝑎𝑥 = −𝑘𝑒×𝑡
𝑉𝑑 ⎡⎣1−𝑒 ⎤

Eq. (14)
𝑠𝑠 𝑆 × 𝐹 × 𝑑𝑜𝑠𝑒 −𝑘𝑒×𝑡
Where: 𝐶𝑚𝑎𝑥 = −𝑘𝑒×𝑡 ×𝑒
𝑉𝑑 ⎡⎣1−𝑒 ⎤

▪ F = bioavailability

▪ S = salt factor (fraction of active drug when the dose is INTERPRETATION OF DRUG CONCENTRATION
administered as a salt) DATA

CLINPHARM Practical Pharmacokinetics 3 of 9


1
The following are the criteria that should be fulfilled before → usually used as a guide to the optimum concentration
undertaking therapeutic drug monitoring: → limits of these ranges should not be takes an absolute
→ the drug should have low therapeutic index → these ranges are only adjuncts to dose determination, which
→ there should be a good concentration-response relationship should always be done in the light of clinical response
→ there are no easily measurable physiological parameters
IV. CLINICAL APPLICATION
● Monitor Adherence or Confirm Toxicity
→ other justification for undertaking TDM in the absence of the A. ESTIMATION OF CREATININE CLEARANCE
criteria
● 24-h urine collection couped with a plasma creatinine
Factors needed to be considered when interpreting TDM data: measurement
→ usual method in estimating creatinine clearance
● Sampling Times
→ TDM as an aid to dose adjustment: concentration should be The rate of elimination is calculated from the measurement of the
at steady state total amount of creatinine contained in the 24-h urine sample
→ approximately five half-lives should elapse after initiation divided by 24, that is,
before sampling
→ EXCEPTION OF THE RULE: toxicity is suspected 𝑎𝑚𝑜𝑢𝑛𝑡 𝑜𝑓 𝑐𝑟𝑒𝑎𝑡𝑖𝑛𝑖𝑛𝑒
= 𝑟𝑎𝑡𝑒 𝑜𝑓 𝑒𝑥𝑐𝑟𝑒𝑡𝑖𝑜𝑛 (𝑚𝑔/ℎ)
→ Steady state has reached: sample at the correct time 24
→ whole process is cumbersome and there is an inevitable
Dosage Adjustment delay in obtaining results
- simple → biggest problem is the inaccuracy of the 24-h urine collection
- linear relationship exists between the dose and
concentration if a drug follows first-order elimination ● Estimate the rate of production of creatinine
→ alternative approach and has advantages
● Capacity limited clearance → does not involve 24-h urine collections and requires only a
→ If a drug is eliminated by the liver, it is possible for the single measure of plasma creatinine
metabolic pathway to become saturated.
→ Initial elimination: first-order Equations have been produced which are rearrangements of
→ Elimination of a saturated system: zero-order equation 4, that is,
𝑟𝑎𝑡𝑒 𝑜𝑓 𝑝𝑟𝑜𝑑𝑢𝑐𝑡𝑖𝑜𝑛
→ Majority of drugs eliminated by the liver, the chracteristic 𝑐𝑟𝑒𝑎𝑡𝑖𝑛𝑖𝑛𝑒 𝑐𝑙𝑒𝑎𝑟𝑎𝑛𝑐𝑒 = 𝐶𝑠𝑠
dose-concentration (seen in the graph Fig. 3.6B) is not seen → rate of production is replaced by a formula which estimates
at normal therapeutic doses. It only occurs at very high this from physiological parameters of age, weight and sex
supratherapeutic levels.
→ EXCEPTION: Phenytoin - the saturation of the enzymatic ● Cockcroft and Gault (1976) Equation
pathway occurs at therapeutic doses. → appears to be the most satisfactory
→ modified version using SI units
● Increasing Clearance
→ First-order elimination: clearance increases as the plasma
concentration increases (Fig. 3.6C)
→ Under normal circumstances, the plasma protein binding
sites available to a drug far outnumber the capacity of the where F = 1.04 (females) or 1.23 (males)
drug to fill those binding sites. → used in patients with moderate to severe renal failure
→ Valproate and Disopyramide: as the concentration
increases, the plasma protein binding sites become ● Modification of Diet in Renal Disease (MDRD) Formula
saturated and the ratio of unbound drug to bound drug → can be used to estimate glomerular filtration rate (eGFR)
increases → uses plasma creatinine, age, sex and ethnicity
- elimination of these drugs are dependent on the
unbound concentration

eGFR = glomerular filtration rate (mL/min per 1.73 m2).


→ should be used with care when calculating doses of drugs

Digoxin
● Action and uses
→ most widely used of the digitalis glycosides.
→ primary actions on the heart: ↑ the force of contraction and ↓
conduction through the atrioventricular node.
→ treatment of atrial fibrillation
→ also used in the treatment of heart failure in the presence of
sinus rhythm.

● Plasma concentration–response relationship:


Figure 3.6 Dose-concentration relationships: (A) first-order elimination, (B),
• <0.5 μcg/L: no clinical effect
capacity-limited clearance, (C) increasing clearance.
• 0.7 μcg/L: some positive inotropic and conduction
blocking effects
● Therapeutic range
• 0.8–2 μcg/L: optimum therapeutic range (0.5–0.9 μcg/L in

CLINPHARM Practical Pharmacokinetics 4 of 9


1
patients >65) → distribution time: approximately 40 min.
• 2–2.5 μcg/L: increased risk of toxicity, although tolerated ● Elimination
in some patients → first-order process primarily by hepatic metabolism to
• >2.5 μcg/L: gastro-intestinal, cardiovascular system and relatively inactive metabolites.
central nervous system toxicity. → population average clearance: 0.04L/h/kg
→ Neonates: 50% of theophylline is converted to
● Distribution caffeine.
→ widely distributed; extensively bound in varying → treatment of neonatal apnoea of prematurity range:
degrees to tissues throughout the body. lower therapeutic range (usually 5–10 mg/L)
= high apparent volume of distribution
→ VD can be estimated using the equation: ● Product formulation
7.3L/kg (ideal body weight (BWt)) Aminophylline
→ VD in patient with renal impairment: → the ethylenediamine salt of theophylline
Vd = 3.8 x ideal BWt + (3.1 x creatinine clearance → 80% theophylline
(mL/min))
→ distribution time: 6–8h ● Practical Implications
→ Clinical effects are seen earlier after intravenous doses → IV bolus doses of aminophylline
- need to be given slowly (preferably by short
● Elimination infusion) to avoid side effects
→ eliminated primarily by renal excretion of unchanged drug → Oral doses (sustained release preparations)
(60–80%); some hepatic metabolism occurs (20–40%) - may be assumed to provide 12h cover
→ population average value for digoxin clearance:
digoxin clearance (mL/min) = 0.8 BWt + (creatinine Gentamicin
clearance (mL / min))
→ patients with severe congestive heart failure have a reduced ● Clinical use
hepatic metabolism and a slight reduction in renal excretion of
digoxin: ➔ The spectrum of activity of gentamicin is similar to other
digoxin clearance (mL/min) = 0.33 BWt + (0.9 x aminoglycosides but its most significant activity is against
creatinine clearance (mL / min)) Psuedomonas aeruginosa.

● Absorption ● Therapeutic range


→ poorly absorbed from the gastro-intestinal tract ➔ Gentamicin has a low therapeutic index, producing dose
→ dissolution time affects the overall bioavailability.
related side effects of nephro- and ototoxicity.
→ Bioavailabilities of two oral formulations:
F (tablets) 0.65 ➔ The use of TDM to aid dose adjustment is essential
F (liquid) 0.8 ➔ It is generally accepted that the peak level should not
exceed 12 mg/L and the trough level should not exceed 2
● Practical Implications mg/L.
→ congestive heart failure, hepatic and renal diseases: ↓
elimination of digoxin.
● Distribution
→ hypothyroidism: ↑ plasma concentration, ↑ sensitivity of the
heart to digoxin ➔ Gentamicin is relatively polar and distributes primarily
→ Hyperthyroidism: Opposite of hypothyroidism into extracellular fluid.
→ Hypokalaemia, hypercalcaemia, hypomagnesaemia and ➔ the apparent volume of distribution is only 0.3 L/kg.
hypoxia: ↑ sensitivity of the heart to digoxin. ➔ it follows a two-compartment model with distribution
being complete within 1 h.

Theophylline
● Elimination
● is an alkaloid related to caffeine.
● Clinical effects: ➔ Elimination is by renal excretion of the unchanged drug.
→ mild diuresis ➔ Gentamicin clearance is approximately equal to
→ central nervous system stimulation creatinine clearance.
→ cerebrovascular vasodilatation ● Practical implications
→ ↑ cardiac output Initial dosage.
→ bronchodilatation (major therapeutic effect)

● Plasma concentration–response relationship: ➔ This may be based on the patient's physiological


• <5mg/L: no bronchodilatation1 parameters.
•5–10mg/L: some bronchodilatation and possible ➔ Gentamicin clearance may be determined directly from
anti-inflammatory action creatinine clearance, and the volume of distribution may
• 10–20 mg/L: optimum bronchodilatation, minimum side be determined from ideal body weight.
effects
Changing dosage
• 20–30mg/L: increased incidence of nausea, vomiting2
and cardiac arrhythmias ➔ This is not as straightforward as for theophylline or
• >30mg/L: cardiac arrhythmias, seizures. digoxin, since increasing the dose will increase the peak
and trough levels proportionately.
● Distribution
→ extensively distributed throughout the body
→ average VD: 0.48L/kg.
→ does not distribute very well into fat

CLINPHARM Practical Pharmacokinetics 5 of 9


1
Once daily dosing. ➔ there is a wide interindividual variation in clearance, and
the lithium half-life in the population varies between 8
➔ Aminoglycosides accumulate in the kidneys, and once and 35 h with an average of approximately 18 h.
daily dosing could reduce renal tissue accumulation.
➔ clinical trials comparing once daily administration of
● Practical implications
aminoglycosides with conventional administration have
➔ lithium should not be prescribed unless facilities for
shown less nephrotoxicity, no difference in ototoxicity,
monitoring plasma lithium concentrations are available.
and similar efficacy with once daily administration.
➔ Blood samples should be drawn 12 h after the evening
➔ Initial dosage for a once daily regimen is 5–7 mg/kg/day
dose, since this will allow for distribution and represent
for patients with a creatinine clearance of >60 mL/min,
the slowest excretion rate.
and this is subsequently adjusted on the basis of blood
➔ Population pharmacokinetic data (particularly the volume
levels.
of distribution) cannot be relied upon to make initial
➔ Once daily dosing has not been well studied in pregnant
dosage predictions, although renal function may give an
or breastfeeding women, patients with major burns, renal
approximate guide to clearance.
failure, endocarditis or cystic fibrosis; therefore, it cannot
➔ Blood level measurements are reported in SI units and
be recommended in these groups and multiple daily
therefore it is useful to know the conversion factors for
dosing should be used.
the various salts.
● 100 mg of lithium carbonate is equivalent to 2.7
mmol of lithium ions
Lithium
● Lithium is effective in the treatment of acute mania and in the ● 100 mg of lithium citrate is equivalent to
prophylaxis of manic depression. 1.1 mmol of lithium ions.
● Lithium is toxic, producing dose-dependent and dose-
independent side effects; thus, TDM is essential in assisting in
the management of the dosage. Phenytoin
● used in the treatment of epilepsy
● associated with dose-independent side effect:
● Dose-dependent effects - hirsutism, acne, coarsening of facial features, gingival
● <0.4 mmol/L: little therapeutic effect hyperplasia, hypocalcaemia and folic acid deficiency
● 0.4–1.0 mmol/L: optimum range for prophylaxis ● has a narrow therapeutic index and has serious
● 0.8–1.2 mmol/l: optimum range for acute mania concentration-related side effects
● 1.2–1.5 mmol/L: causes possible renal impairment ● Plasma concentration–response relationship
● 1.5–3.0 mmol/L: causes renal impairment, ataxia, •<5mg/L: generally no therapeutic effect
• 5–10mg/L: some anticonvulsant action with approximately
weakness, drowsiness, thirst, diarrhoea
50% of patients obtaining a therapeutic effect with
● 3.0–5.0 mmol/L: causes confusion, spasticity, concentrations of 8–10mg/L
dehydration, convulsions, coma, death. (Levels above • 10–20mg/L: optimum concentration for anticonvulsant effect
3.5 mmol/L are regarded as a medical emergency.) • 20–30mg/L: nystagmus, blurred vision
• >30mg/L: ataxia, dysarthria, drowsiness, coma.
● Dose-independent effects ● Distribution
➔ These include tremor, hypothyroidism, nephrogenic - follows a two-compartment model with a distribution time
of 30–60min
diabetes insipidus, gastro-intestinal upset, loss in bone
- apparent volume of distribution is 1L/kg
density, weight gain and lethargy. ● Elimination
- main route of elimination is via hepatic metabolism
● Distribution - can be saturated at normal therapeutic doses
➔ Lithium is unevenly distributed throughout the body, with - results in the characteristic non-linear dose/concentration
a volume of distribution of approximately 0.7 L/kg. curve
- instead of the usual first-order pharmacokinetic model, a
➔ Lithium follows a two-compartment model with a Michaelis– Menten model, used to describe enzyme
distribution time of 8 h activity, is more appropriate
- Using this model, the daily dosage of phenytoin can be
● Elimination described by
➔ Lithium is excreted unchanged by the kidneys. - (insert equation)
- Km is the plasma concentration at which metabolism
➔ Lithium clearance is approximately 25% of creatinine
proceeds at half the maximal rate.
clearance, since there is extensive reabsorption in the - varies greatly with age and race
renal tubules. - Vmax is the maximum rate of metabolism of phenytoin
➔ changes in renal function, dehydration, diuretics and is more predictable at approximately 7mg/kg/day.
(particularly thiazides), ACE inhibitors and NSAIDs - clearance changes with blood concentration, the halflife
(except aspirin and sulindac) all decrease lithium also changes
clearance. - as a rule of thumb, 1–2 weeks should be allowed to
elapse before sampling after a dosage change.
➔ aminophylline and sodium loading increase lithium
- In overdose, it can be assumed that metabolism of the
clearance drug is occurring at the maximum rate of Vmax
● Practical implications

CLINPHARM Practical Pharmacokinetics 6 of 9


1
- Care is needed when interpreting TDM data and making ▪ Blood samples should be taken after 2-4 weeks (steady
dosage adjustments steat must be achieved first)
- phenytoin is approximately 90% protein bound ▪ trough level must be measure when sampling
- in patients with a low plasma albumin/ uraemia, the free ▪ Rule of thumb: each 100mg dose will increase the plasma
fraction increases and therefore an adjusted total concentration at the steady state by approximately 1mg/L
phenytoin should be calculated or a free salivary level in adults
taken. ▪ Interactions: Other drugs (e.g. Phenytoin) given at the
- To adjust the observed concentration in same wime with carbamazepine affects metabolism and
hypoalbuminaemia the following equation can be applied: blood concent.
- (insert equation)
- Albumin concentration is in g/L.
- In ureamic patients with severe renal failure, the unbound Phenobarbital
fraction is approximately doubled, so the target ● Phenobarbital is in a class of medications called barbiturates. It
concentration needs to be half the normal concentration works by slowing activity in the brain.
or apply the adjusted concentration equation if albumin .
level is known. → Indication:
- oral formulations of phenytoin show good bioavailability. ▪ Used for treatment of all forms of epilepsy except
However, tablets and capsules contain the sodium salt (S absence seizures.
= 0.9), whereas the suspension and infatabs are
phenytoin base (S = 1). → Side effects
- Intramuscular phenytoin is slowly and unpredictably ▪ Sedation with continuous therapy
absorbed, due to crystallisation in the muscle tissue, and ▪ Tolerance can happen (Impartant to monitor plasma
is therefore not recommended. concent.)
- Fosphenytoin, a prodrug of phenytoin, is better absorbed
from the intramuscular site. Doses should be expressed → Distribution:
as phenytoin equivalent. Fosphenytoin sodium 1.5mg is ▪ Distributed most into body tissues
equivalent to phenytoin sodium 1mg ▪ protein bound drug (50%)
→ Detail ▪ Ave. VD is (0.7–1L/kg)
▪ Smaller Detail
→ Elimination:
- ▪ Metabolized by the the liver (80%), excreted (20%)
● unchanged in the urine
▪ Induces own metabolism or auto-induction
Carbamazepine ▪ First order Elimination (Elimination proportional to
● Carbamazepine is in a class of medications called concentration)
anticonvulsants. It works by reducing abnormal electrical ▪ Clearance in children (Faster) is twice the adult clearance
activity in the brain
. → Practical Implications:
→ Indication: ▪ Long half-life, given as single dose
▪ Used for treatment of partial and secondary generalized ▪ The pharmacokinetics of phenobarbital may be altered by
tonic-clonic seizures, primary generalized tonic-clonic liver and (less markedly) renal disease
seizures, trigeminal neuralgia, and prophylaxis ▪ not affected by the simultaneous administration of other
(Prevention) of bipolar disorder unresponsive to lithium anticonvulsants.

→ Side effects Primidone


▪ Different dermatologic reactions (e.g. rashes) ● Primidone is in a class of medications called barbiturates. It
▪ Aplastic anemia (bone marrow cannot make new RBC’s) works by slowing activity in the brain.
▪ Stevens–Johnson syndrome (disorder of the skin and .
mucous membranes) → Indication:
▪ Used for treatment of tonic–clonic and partial seizures.
→ Distribution: These are siezures that involve both tonic (stiffening) and
▪ Distributed widely, most concentrated in the kidneys clonic (twitching or jerking) phases of muscle activity.
▪ A highly protein bound drug (70%-80%)
▪ Ave. VD is (0.8–1.9L/kg) → Side effects
▪ sedation
→ Elimination: ▪ nausea
▪ Metabolized by the body, excreted 2% unchanged in the ▪ ataxia (clumsy voluntary movements due to poor muscle
urine control. It may impair walking, balance, speech,
▪ Induces own metabolism or auto-induction swallowing, and eye movement.)
▪ First order Elimination (Elimination proportional to
concentration) Valproate
▪ Half-Life = 35 hrs ● Sodium valproate as valproic acid is an anticonvulsant used to
control complex partial seizures and both simple and complex
→ Absorption: absence seizures.
▪ Slow (oral absorption) .
▪ Incomplete absorption → Indication:
▪ Bioavailability = 80% ▪ broad spectrum of anticonvulsant activity, being useful in
generalised absence, generalised tonic–colonic and
→ Practical Implications: partial seizures

CLINPHARM Practical Pharmacokinetics 7 of 9


1
→ Toxic Effects
→ Plasma concentration–response relationship ▪ nephrotoxicity
▪ Valproate has no concentration–response relationship, but ▪ hepatotoxicity
a range of 50–100mg/L is recommended, with 50% of ▪ gastro-intestinal intolerance
patients responding at levels above 80mg/L. ▪ hypertrichosis
▪ Above 100mg/L has no therapeutic value. ▪ neurological issues.

→ Toxic Effects → Plasma concentration–response relationship


▪ Valproate's rare hepatotoxicity is linked to plasma levels ▪ Therapeutic ranges are not absolute for all monitored
over 150mg/L. drugs.
▪ Since there are many factors, defining a ciclosporin
→ Distribution: therapeutic range is even harder.
▪ Highly protein bound bound drug (90-95%) 1. sampling matrix affects concentration (i.e.
▪ saturates protein binding sites at concentrations above whole blood or plasma).
50mg/L 2. it depends on whether the assay includes
▪ Ave. VD varies (0.1 to 0.5L/kg) metabolites or is ciclosporin-specific.
▪ Target concentrations vary by center, but are typically
→ Elimination: 100–200 ng/mL in the first 6 months after transplantation
▪ Elimination by the the liver/hepatic, excreted (5%) and 80–150 ng/mL after 6 months onwards.
eliminated by the kidneys. ▪ Graft rejection increases below this window.
▪ At higher concentrations, drug clearance increases due to ▪ Above-limit levels increase nephrotoxicity and
protein binding site saturation and increased free fraction. hepatotoxicity.
▪ Plasma concentration changes non-linearly with dose.

→ Practical Implications: → Distribution:


▪ Valproate levels should not be measured due to the lack ▪ Highly lipophilic
of a concentration–response relationship and variable ▪ protein binding (98%)
pharmacokinetics. ▪ Ave. VD varies (4–8L/kg)
▪ Clinical response determines dosage in most cases.
▪ Valproic acid takes weeks to become fully active, so dose → Elimination:
adjustments should be made slowly. ▪ Elimination by the the liver/hepatic
▪ Clearance (0.1–2L/h/kg)
Lamotrigine, vigabatrin, gabapentin, tiagabine, ▪ The milligram per kilogram dosage is 40% higher in
topiramate, pregabalin, lacosamide and children. Elderly and hepatic patients have lower
levetiracetam clearance rates.
● These medications are calles called anticonvulsants..
. → Practical Implications:
● Indication: ▪ In normal subjects, ciclosporin absorption is variable and
→ indicated for multiple epilepsy types and some with incomplete (F = 0.2–0.5).
additional indications. All are adjunctive (Another ▪ Bioavailability varies even more in transplant patients,
treatment used together with the primary treatment) to especially in the first few months after transplant.
other anticonvulsants, and some are monotherapy. ▪ Numerous drugs interact with ciclosporin.
▪ Due to wide inter-patient variation, population averages
● Plasma concentration–response relationship are not useful for dose prediction, but therapeutic drug
→ These newer anticonvulsants have no clear plasma monitoring will help select the best dose.
concentration-response relationship. ▪ TDM (monitoring) with ciclosporin requires practical
→ These preparations are usually added to other considerations:
anticonvulsants, which complicates theplasma − whole blood should be the sampling matrix for
concentratio-response relationship. ciclosporin varies between blood and plasma
− • Draw samples at steady state, 2–3 days after starting
● Practical Implications or changing dosage, to represent trough levels
→ These agents have narrow therapeutic indices and inter- (average half-life is 9 h).
and intra-individual pharmacokinetic variation, but there is
not enough evidence to support routine TDM (minitoring). − Postoperative ciclosporin concentration monitoring
→ Dosage should be titrated to clinical response. should be done every 2–3 days until the patient's
clinical condition is stable. Monitoring can continue
every 1–2 months.
− Since ciclosporin brands differ in bioavailability, TDM
Ciclosporin should be done when switching brands.
● . Immunosuppressive agent
.
→ Indication:
▪ Tolypocladium inflatum gams produces the neutral
REVIEW QUESTIONS (optional)
lipophilic cyclic endecapeptide cyclosporin.
▪ It reduces graft rejection after organ and tissue
1. Which of the following anti-convulsants is contraindicated in
transplantation as a potent immunosuppressant. patients with a history of Tricyclic antidepressants
▪ Has low therapeutic index (causes various toxicities) hypersensitivity?
▪ Concentration affects graft rejection and nephro- and A. Ethosuximide
hepatotoxicity. B. Carbamazepine
C. Phenobarbital

CLINPHARM Practical Pharmacokinetics 8 of 9


1
D. Phenytoin
E. Gabapentin
2. What are the most common adverse effects of anticonvulsive
drugs?
I. Alteration in cognition and mentation
II. Headaches and dizziness
III. Gastrointestinal symptoms
A. II only
B. II and III only
C. I, II and III only
D. III only
E. I and II only
3. Which of the following drugs may increase the level of
carbamazepine due to its ability to decrease carbamazepine
metabolism?
A. Valproic acid
B. Primidone
C. Phenytoin
D. Phenobarbital
E. Topiramate
4. Which of the following is/are known side effects of phenytoin?
I. Steven-Johnson Syndrome
II. Gingival hyperplasia
III. Ataxia
IV. Lupus erythematosus

A. II and III only


B. I, II and III only
C. II and IV only
D. II only
E. I, II, III and IV

Aswers: b, c, a, e

REFERENCES

Roger Walker and Cate Whittlesea Clinical Pharmacy and


Therapeutics Fifth Edition

CLINPHARM Practical Pharmacokinetics 9 of 9


1

You might also like