Clinical PK
Clinical PK
By:
Clinical Pharmacy Department Staff
Department of Clinical Pharmacy
College of Pharmacy
KFS University
Contents:
Defmition of PK
•!• The science that study the rates of the transfer process associated with absorption,
distribution, metabolism, and excretion of drug through the body
•!• How the body affects the drug
Phannacodynamics
•!• Pharmacodynamics refers to the relationship between the drug at the site of action
(receptor) and pharmacologic response
•!• How the drug affect the body
Importance of Clinical PK
ADME
•!• Therapeutic and toxic effects of drugs depend on drug concentration at receptor site
(impractical to be measured, so the solution is plasma concentration curve)
®
•!• Plasma concentration is the simplest widely used method to evaluate the therapeutic and
toxic effect ofthe drug
•!• Absorption increase plasma concentration while metabolism and excretion decrease
plasma concentration
•!• Distribution in most cases have no significant effect of plasma concentration (one
compartment model)
Some drugs are administered IV, so these agents have no absorptive phase associated with their
pharmacokinetic profile.
IV administration of drugs is followed by distribution and elimination, and thus the time-amount
profile ofthe drug in the body is a decreasing function as shown below.
The drug a moun t-time profile in th e body is a n
in c reasin g, a nd th e n decreasi n g f"un ctio n as sh own
below.
o; 300
-=
j
200
i
-<
0 3 6 9 12 15 18
Tim e (hi')
•!• Some drugs are administered IV, so these agents have no absorptive phase associated
with their pharmacokinetic profile.
•!• IV administration of drugs is followed by distribution and elimination, and thus the time-
amount profile of the drug in the body is a decreasing function as shown below.
The tirne-arnount profile of the drug in the body is a
decreasing function
20
16
12
40
0 2 4 6 8 10 12
Time {hr)
iv
po
Routs of administration
bioavailability means the percent of the drug dose which reach systemic circulation (as
indicated by plasma concentration)
It is considered 100% in case of IV administration
Bioavailability is changed by factors affecting drug absorption
The phenomena where the concentration of a drug is greatly reduced before it reaches the
circulatory system reducing drug bioavailability.
The gut and liver metabolizes many drugs to such an extent that only a small amount of
active drug reach the rest of the circulatory system from the liver.
Sublingual route shows no first pass effects
Drug
Enterohepatic circulation
This refers to the circulation of drugs from the liver to the bile, then to the small
intestine, followed by reabsorption again through the enterocyte (often by the bacterial
help) and transport back to circulation and liver.
Drugs may be toxic as they reach unexpectedly high concentrations.
Enterohepatic circulation increase plasma concentration of drugs like oral contraceptive,
estrogen
Secondary peak is found in plasma concentration curve
P glycoprotein
Found in:
P<>.rt:ed ._ein
• P-glycoprotein
•
Inhibitor
Distribution of drugs
After absorption to blood stream, drug is rapidly distributed into the interstitial and
intercellular fluids
Liver, kidney, brain, and other well-perfused organs receive most of the drugs
Plasma proteins bound to many drugs. This bounded drugs have no action at receptor site
and only unbounded fraction has a pharmacological action
Displacement of highly bounded drug from plasma protein by other drugs can affect the
pharmacological action greatly ex: warfarin
Drug distribution determine the model of drug kinetics (one compartment or two
compartment)
®
- -
=J
=
--
.....-==
-
Metabolism
Excretion
Kidney is the main organ for excretion of drugs and their metabolites
elimination from lung is important mainly for the elimination of anesthetic drugs
Some drugs may be excreted in saliva and sweetings
Orders of reactions
A pharmacist weighs exactly 10 g of a drug and dissolves it in 100 mL of water. The solution is
kept at room temperature, in order to measure the stability and samples are removed periodically
and assayed for the drug. The pharmacist obtains the following data:
A= -k,t +A,
ie 60
0
Slope=- Ko
40
20
0
0 3 6 9 12 15
Ti me ( hr)
A pharmacist dissolves exactly 10 g of a drug into 100 mL of water. samples are removed
periodically and assayed for the drug. The data:
Drug Cone (mg/mL) Time(hr) Log Cone 100.00 0 2.00
50.00 4 1.70
25.00 8 1.40
12.50 12 1.10
6.25 16 0.80
20
16
bi 5
..<s
"S 12
e
""
<C
8
40
0 :z 6 8 10 1:Z
Time (h.-)
A= Ao e-kt
By integratio n
Ln A= -Kt+ In Ao
2.5 LogAO
0.2
t>.O
Slope = -K/2.303
_, 1.
5
1.0
0.5
0.
0 0 2 4 6 8 10 12
Time (hr)
Compare between first and zero order kinetics by completing the following table
First Zero
Log A= -Kt/2.303 + log Ao
A=-kot+Ao
Nature of The rate Cone or amount Amount independant
change dependant
0
y
0.001
®
1 0.02
3 0.08
7 10
10 400
.., 10 +
•
""C
....
....
aJ
+
""C + +
c: 8
:::l
:I: +
+
.... +
+
y 6 +
+
+ ..
t
+
4 + .. +
t +
+
2 + .. +
+ .. +
0
0 5 X 10 15
10000
1000 t
+
y
100
10
• I
1 t t t
0.1
•
0.01 t
0.001
0 5 10 15
Phannacokinetic Models:
•!• A model is a hypothesis enables using mathematical terms to simulate the rate processes
of drug absorption, distribution, and elimination (metabolism and excretion)
®
•!• Compartment models are based on linear assumptions using linear differential equations
•!• It enables Predict plasma, tissue, and urine drug levels with any dosage regimen
Before Aftet'"
Administ ration Administrat ion
Immediately After
Administration
Before Administration After o istlibution
Equilibrium
Model
I
One-compartment model
•!• In this case the plasma and all perfused organs deal as one unit (the central compartment)
•!• The drug is both added to and eliminated from a central compartment.
Oral d ose
eliminat ion
IV d ose
Two-compartment model
•!• In a two-compartment model, drug can move between the central ( plasma compartment and
highly perfused organ) to and from the tissue compartment(peripheral compartment).
•!• In this model, in most cases, there is no elimination from tissue compartment and the drug need to
be transported again to the plasma in order to be eliminated
absorpt ion
Penpheral
Central compartment
compartment
Oral dose
l
elimination
Central Peripheral
compartment compartment
IV dose
elimination
!-Concentration response plot: increase concentration increase response till reaching maximal response (
Most drugs)
Maxsmal
efficacy
Poten
Dose (log1o scaJe)
Hysteresis loops
The response not related to the dose, the cause may be:
Example
Three new drug for treating diabetes were introduced in the market. The following table represent the
relation between the plasma concentration and the drug effects as indicated by the decrease in blood
glucose level. For each drug determine the pharmacokinetic and pharmacodynamic interface after plotting
the concentration effect graph
drug A drug B drug C
5 2 2 2
10 4 4 4
20 8 8 8
30 16 12 10
20 18 8 6
15 12 G 4
10 10 4 2
5 6 2 1
c c
.Q Parameters of Interest .Q Parameters of Interest
MIC MIC
nme nme
Single IV dose
A= Ao e-kt
elimination
IV dose
Slope= K
Volume of distribution
Vd=dose/Cpo
t = ln2 0.693
k k
Clearance (CI)
Clearance (Clot TBC) is the volume of plasma that is completely cleared of the drug per unit time
Cl= K Vd
•!• The TBC is the product of the first-order rate constant (K) and the Vd, and has dimensions of
volume/time.
•!• Cl is constant for the same drug
•!• The total body clearance will be equal to the renal clearance + hepatic clearance + lung clearance.
•!• Cl= renal clearance+ liver clearance
The integral of plasma cone with respect to time after an IV dose is the area under the plasma cone-time
curve (AUC). Which has units of mass-time per volume (mg.hr/liter).
* Since the TBC of a drug in a given subject is constant, the AUC is proportional to the given dose.
Dose
AUC TBC
Cpo
AUC K
Single oral dose
Plasma concentration curve
c
---
---r-------
Th erap
· eutic
MTC
--- I:
.52
c
c
0
(.)
Dose= SOO mg
e
-= 400 Amount of drug I.n the GIT
s::
::::1
0
200
100
0
0 2 4
absorption elimination
Oral dose
Equation for single oral dose
FDKa
A ka -k
_ FDKa
con e Vd(ka-k)
ln ka -ln k
t max ka- k
F (bioavalability)
A[ / Coral
F = AUCiv
AUCiv .Dose
Cl
FDuse
A [ .7Coral = Cl
---------------------------------,
300
Post absorptivephasl:!
Absorption pha se
0------------------------------
0 6 9 12 15
nm e (hrl
FDKa
Ln c= In VdJw-k) kt
Continuous IV infusion
(l)IV infusion allows precise control of plasma drug concentrations to fit the individual needs of the
patient and the duration of drug therapy may be maintained or terminated as needed using IV infusion
(2)For drugs with a narrow therapeutic window (eg, heparin), IV infusion maintains an effective constant
plasma drug concentration by eliminating wide fluctuations between the peak (maximum) and trough
(minimum) plasma drug concentration.
(3)the IV infusion of drugs, such as antibiotics, may be given with parenteral nutrition that include
electrolytes and nutrients.
(4) Reduce toxicity on body as incase of immune reaction and vein damage by push IV or intermittent IV
administration
Steady-state Wash-out
Cone
nme
Steady state
Time
R=KAss
Ass= R/K
Cpss= Rl k Vd = R/Cl
Mathematically, the time to reach true steady-state drug concentration, C SS, would take an infmite time.
The time required to reach the steady-state drug concentration in the plasma is dependent on the
elimination rate constant of the drug for a constant volume of distribution.
cone
Steady state
Time
sameTime
Loading Dose
The loading dose, DL, or initial bolus dose of a drug, is used to obtain desired concentrations (steady
state) as rapidly as possible.
Cpo = Css
so:
DL=CssVd or DL=R/K
cone
Steady state
--y----=<,
I I
I
T Time
•••• ••
-
DIAMICRON l.tfk 60 mg
,.«tfNd,....- Release
Multiple-Dosage Regimens:
There are two main parameters that can be adjusted in developing a dosage regimen:
- "' -- ------1----------------
"'J- \j \j \j _
30T-------------------------------------------------
6 12 18 24 30 36
Time (hr)
A max= D/ ( 1-e-kr) A
min =A max e -kr
Dose= Amax-Amin
20
c:
0
· 15
E
c»
<.>
c:
0 10
<.>
Ill
E
1/)
Ill
a: 5
Time (h)
Dosing regimens
•!• Is the drug has minimum effective concentration (I\.1EC) that have to be achieved in order to
produce the therapeutic response?
•!• Is the drug has a minimum toxic concentration has to be maintained below in order to avoid toxic
effects?
®
•!• Is the difference between the MEC and the MTC is large or small (therapeutic index)? (if small
therapeutic index exists, frequent administration of small doses will be necessary to avoid large
fluctuation in plasma concentration)
•!• Is there a compliance problem with the drug? So, reducing frequency may be recommended
z
0
a:
w
, '--------......-----'
0
z Steady-State Concentrations
0 •Proportional to dose/dosage interval
0
•Proportional to F/CL
Fluctuations -------------------'
•Proportional to dose intervaVhalf-time
•Blunted by slow absorption
0------- ---- --- ---- --- ---
0 1 2 3 4 5 6
TIME (multiples of elimination half-time)
Loading dose
If need to rapidly reach the desired concentration, Loading dose may be used
LD = Cp. Vd
The average concentration is not the mean value of the maximum and the minimum plasma concentration
Cp Avg= FD/K Vd r
AUC=FD/KVd
Cp Avg=AUC/r
Presentation of IV Multiple dose curve
25
:._J, ' 20
Cl
sc:
.2 15
1§
E
Cl)
(.)
c:
0
(.)
10
E
"'
.!!!
(l. 5
8 16 24 32 56 64 72 88 96
Time(h)
dist ribution
K21
Central Peripheral
compartment compartment
K12
elimina tion
1
IV dose tvvo compa rtment
1ooor--------------------------------------------------------------,
-
+=
c
.....
(I
c
8
(U
.,
E
(U
a:
10
0 2 6 8 10
Time (hr)
-at
Cp==Ae
A the intercept for the distribution phase
B the intercept for the elimination phase
a the slope of the distribution phase
B the slope of the elimination phase
Hybrid first-order rate constants
K K
10 21
ap
K + K + K =(a+ P)
21 10 12
Chapter 3: Aminoglycosides TDM
Introduction
concentration
time
Following repeated administration of these short IV infusion, the drug will accumulate in the body until
steady state is achieved.
At steady state, the maximum and minimum plasma concentration will be constant if the dose, infusion
duration, dosing interval are kept constant.
10
Time (h)
10
Cssmi n=trough
Time (h)
Loading dose
The concentration at steady state depend only on the administration rate (infusion dose, t', -r-)
assuming the TBC is constant in this patient.
Similarly, it will take 5-6 half life to achieve the steady state.
Administration of loading dose can lead to immediate reaching the steady state without affecting
the concentration at steady state. As the nature of AG is concentration dependent. Cmax is used
for Loading dose calculation
Loading Dose:
LD = Cssrnax x Vd
3
I
. - . -.-.- . -. --!ll C
0
D
0
.
Time (Hou rs)
At steady state
Increasing the infusion dose, will result in proportional increase in Cmax and Cmin at steady
state.
Increasing the infusion time of the same infusion dose will result in decreasing the Cmax and
slight higher Cmin at steady state as larger amount of the drug is eliminated during the longer
infusion time.
Increasing the interval time (same dose and infusion time), will result in lower Cmax and Cmin at
steady sate.
J
a
...=.
g nt::amicin 120
-=
c:::::
-ce:
6
4
mg gi-v
Bc:::::
8
2 -----·--
<>
10
Aminoglycoside pharmacokinetic
In practice, the traditional infusion time for aminoglycoside (AG) 1s 0.5 or 1 hour.
ol--- -- ==
0 2 4 6 8 10 12
Time (h)
Conc.-dependent bactericidal effect (Cmax value is most important than time above MIC)
Not absorbed (poor bioavailability), may be given orally for local effect on GIT (neomycin,
streptomycin).
IM bioavailability: Almost 100% except in obese patients
Plasma protein binding is< 10% (insignificant)
Almost completely eliminated unchanged by the kidney
Removed by hemodialysis.
Contraindicated if CrCl< 20ml/min or rapidly changing renal function
PHARMACOKINETICS: Distribution
2
0
0
-r
.. ... -r,-
Despite the extremely high peak concentrations obtained during extended-interval dosing of
aminoglycosides, increased nephro/ oto-toxicity is not seen in these patients.
Monitoring AG toxicity
Steps:
0Patient: JM
0 Age: 50 Gentamicin Dose Per Pharmacy
0Height: 5 ft I 0 in
OWeight: 70 Kg
O Gender: M
O[Cr]: 0.9 mg/dl (stable for 5 days)
Olndication: gram-negative Pneumonia.
CIPatient: J M
CIAge: 20
CIHeigh't: 6 ft 3in [i obran ci'n Dose Per Pizarinacy
CIWeigh't: 76 Kg
CIGender: M
Cl[Cr]: 1.1mg/dl (stable for 5 days)
Cllndication: gram-negative Pneumonia.
*extended interval also include 36 hr and 48 hr depending on Crcl. The recommended interval can be
calculated by Hartford nonogram using 7 mg/kg dose
"1 4
=r
"1 2
.a,
"10
V>
Dose every
<0 24 hours
Ci..
.s a
,§
<0 6
.l:::o
liS
<..>
c: 4
8
2
6 7 a 9 "10 "I "I "1 2 "13 "1 4
Using Hartford nomogram recommend the dosing interval for extended interval gentamycin if the patient
weight 70 kg height 70 inches, the sample taken after 10 hours gentamycin concentration is 7 mg/1
10
< I
- -k(t-t')
Cpss,min -Cpss,max e
1-measure: At steady state, take two samples (for peak and trough) and measure AG
concentrations.
2-determine: using the measured peak and trough value, determine K and Vd for this
patient
• 3- decide: decide the required peak and trough
• 4-claculate the required Ko, T for this patient: adjust the dose if required based on
patient specific parameters
In TDM, monitor for AG cone. occur every 1-3 days until stabilization then every week.
• Monitoring is not require ifthe expected length oftherapy <3 days (e.g. UTI)
• Later on, the pharmacist measured two plasma concentration at steady state for
gentamycin, the patient peak concentration and trough concentration were 9.5 mg/L
and 2.2 mg/L. Based on patient PK parameter,calculate the required dosing rate and
interval.
Apply on clincalc.
• After the end of infusion of the first dose according to population parameters, Three
sample of AG is obtained in the period after the end of drug infusion and analyzed for
AG concentration
• These concentrations are presented graphically, from the obtained straight line
determine the K (elimination rate constant) from slope ofthe curve and Tl/2
graphically.
• Then by extrapolation ofthe straight line determine the concentration at the end of
infusion of the first dose (Point A)
t:he:n from the follovvi:ng equat:io:n det:errrrine the "Vd for this
patient:
Cp = Ko (1- -kt:')
KVd e
-
Concent:ra t:l on
-
A
.> time
Chapter 4: Phenytoin TDM
Concepts for non-linear kinetics
•!• Some drugs do not follow first order kinetics specially in elimination process . So, these
drug will have elimination that changes as the concentration change.
•!• In this case, the steady state concentration will not be proportional to the rate of dosing.
•!• This is called nonlinear kinetics or capacity limited kinetics or kinetics of saturation
Clinical Example
•!• After 2 weeks on 300 mg/day of phenytoin, the measured cone. of phenytoin is 6 mg/L.
(Target therapeutic range is 10-20 mg/L)
•!• The physician doubled the dose rate to 600 mg/day to double the concentration achieved.
•!• 2 weeks later the patient calls the pharmacy with complaints of double-vision and
difficulty walking. A concentration is drawn = 36 mg/L(Explain the case ?)
•!• Linearity: the direct relation of elimination to the concentration of the drug in plasma.
The dose change result in propotional change in cone and AUC
•!• Non-linearity: there no direct relation between drug concentration and the elimination of
drugs. The dose change result in non-propotional change in cone and AUC
•!• Common examples ofthese drug: valproic acid, phenytoin
•!• Non-liner pharmacokinetics can occur in any ADME process but capacity limited
elimination (most common)
•!• Bin case oflow drug concentration (both linear and non-linear)
•!• A in case of large drug concentration in non Linear
•!• C in case of large drug concentration in linear pkarmacokinetics
•!• Michaelis- Menten enzyme kinetics expresses the non linear kinetic of some drugs.
•!• Enzyme (E) react with substrate(S) to form enzyme-substrate complex (ES) then the
product (P) is formed and the enzyme go back to react again with another substrate.
Ks kc..l
E + S ES----+ E + P
V= VmaxS
Km + S
Graphical presentation
0.3G
0.30 vmax
-
0.20
.$
0.20
0
:u
ro 0.15
Q)
a:::
0.10
Km
0.05
0.00
!/
0 1000 2000 3000 4000
Concentration of substrate
•!• V= (Vmax/km) x Cp
•!• The reaction appear similar to first order kinetics
•!• This means that Km is the constant which equal the substrate or plasma concentration at
which the process rate is half maximal
Compare linear and capacity limited kinetic After IV bolus dose
K Vmax
Central Central
compartment compartment Km
Vtnax C
R = pss
Km + CPss
At steady state of multiple administration
rate in = rate out
FD/T= _Vm_ax Cpss
Km+CPss
Vmax
TBC= Km + CPss
Half life and clearance for non-linear kinetics
TBC= v_ , _a.,.x
Krn+lP.s.s
T 1 2= 0.693Vd/TBC
0.693Vd
T1 r ?- (krn + Cps s )
V tnax
Phenytoin pharmacokinetics
Vd = 0.7 L/kg*
Extensive binding ( -90%) to albumin.
Free fraction is 10°/o.
95°/o elimination by liver
* f or obese
Vd = 0. 7 Ukg [IBW + 1.33 (TBW- IBW)]
•!• A steady-state trough total phenytoin serum concentration should be measured after
steady-state is attained in 7-14 days. Sample serum levels 7 to 10 days following each
dosage change to assess.
•!• Half-life: 7 to 42 hours (average= 24 hours).
•!• The maximum single oral dose should not exceed 400 mg. Divided doses increase
bioavailability as well as decrease potential for GI side effects.
•!• The maintenance dose is started 18-24 hours after the loading dose.
•!• maintenance dose normally given in divided doses q8-12h
•!• Total phenytoin concentration (cheap and fast) is the mainstream ofTDM when plasma
protein-binding abnormalities are absent.
•!• Free concentration is required in case of disease affecting the albumin amount in body as
in case of:
•!• 1-hypoalbuminaia (liver disease)
•!• 2-ESRD or dialysis
•!• 3-geriatric
Case Study
• A patient with ESRD has a reported phenytoin-related adverse drug reaction
and the total phenytoin concentration i s 15 mg/L.
Question 1:
A patient with ESRD has a reported phenytoin-related adverse drug reaction and the total
phenytoin concentration is 15 mg/L. serum albumin concentration was 3 g/dl.
Question 2:
LM is an epileptic patient being treated with phenytoin. He has hypoalbuminemia (albumin = 2.2
g/dL) and decreased renal function (creatinine clearance= 50 mL/min). His total phenytoin
concentration is 7.5 mg/L.
B- after 3 days, more decrease in renal function occur (creatinine clearance = 10 mUmin).
Compute an new estimated normalized phenytoin concentration for this patient.
Initial dosage detennination for phenytoin
Phenytoin adjusted body weight= IBW + 1.33 (TBW- IBW) is used only for loading dose in
obese
AdjBW = IBW + 0.4 (TBW-IBW) may be used for Maintenance dose in obese.
Question 3
TD is a 50-year-old, 75-kg (height = 5 ft 10 inch) male with simple partial seizures who requires
therapy with oral phenytoin capsule. He has normal liver and renal function. Suggest an initial
phenytoin sodium capsule dosage regimen designed to achieve a steady-state phenytoin
concentration equal to 12 mg/L.
Answer 3
Question 4
UO is a 10-year-old, 40-kg, 145 em male with simple partial seizures who requires therapy with
oral phenytoin suspension. He has normal liver and renal function. Suggest an initial phenytoin
dosage regimen designed to achieve a steady-state phenytoin concentration equal to 12 mg/L.
Answer4
•!• With only one steady state serum level available, you can calculate a value for Vmax if
you assume a value for Km (Note: references assume a value of 4 mg/L for Km)
•!• Vmax =daily dose(mg/day) x (Km/Css + 1)
•!• MD/Vmax (old)=MD/Vmax (new)
•!• This will be required for modification in dose in the Css is out of therapeutic range after
initial dosing.
•!• Changes in the daily maintenance dose should be made in small increments (30-100 mg
maximum)
Graphical method
14
RATE OF 12
ELIMINATI ON
R.Vm
<mo/k o/dovl 10
24 20 16 12 0 4 8 12 16
C55 (m/ 111od K m (rJ't9/liter )
Question 5
After 14 days of dose initiation, the plasma cone was 8 mg/L. the physician need dose
adjustment to reach the plasma cone of 12
Both require determination of two steady state concentration corresponding to different two
dosage regimen
1-Linear transformation method
3.0
2.5
D/T
Intercept = Vmax/SF
0.2
1.5
1.0
0.5
0.0 0 12
•!• This method require the knowledge of two different dosing rate and their corresponding
steady state concentration.
•!• Present the steady state concentration points (Cpss) on the left side of the X axis and the
dosing rate "daily dose" on the Y axis
•!• By plotting the two lines between each of the dosing rates and their corresponding steady
state concentration. The two lines will cross at a point.
•!• The value on X axis will be Km and the Value on Y axis will be Vmax
Graph presentation of The direct linear plot method
30
Phonyto•n C"- (f'ng/ L)
Question 6: A 74 kg, 28 year old man is receiving phenytoin for the treatment of seizures. When this
patient was taking a daily oral dose of 250 mg phenytoin tablet, his steady state plasma concentration was
7.2 mg!L. because phenytoin plasma concentration was well below the therapeutic range, the patient daily
dose was increased to 450 mg which resulted in steady state plasma concentration of 30 mg/L. phenytoin
bioavailability is 100% and the Vd is 50 L.
Question 7: A 25 kg, 14 year old female was admitted to the hospital because of frequent episodes
seizures. She was started on IV phenytoin with loading dose of 5 mg/kg followed by 80 mg phenytoin IV
every 12 hr. at steady state the avg plasma concentration was 8 mg!L. the phenytoin dose increased to 100
mg IV every 12 hr and the steady state concentration was 13.3 mg/L. later on, the phenytoin IV was
replaced by oral solution 225 mg at bed daily of phenytoin which has 100 bioavailability. The Vd is 0.8
Llkg.
Question 8: Mr SG is a 58 y/o, 72 inch, 70 kg male, seen recently with symptoms of a lower Respiratory
infection. At this time he had a lesion noted on his Chest X-ray. A lung biopsy revealed a carcinoma with
metastasis to The head, demonstrated via aCT scan. His albumin was 4 gm/1 and serum creatinine 1.0
.Phenytoin therapy was initiated prophylactically at a dosage of 400 mg daily. Two weeks later, the
patient had a grand mal seizure and the phenytoin Cp was5 mg/1. The dosage was increased to 500 mg qd
and 3 weeks later, The phenytoin concentration was 7 mg/1.
•!• a-Using the Ludden method graph, estimate the Vmax and Km for this patient.
•!• b-estimate a dose necessary to achieve a phenytoin concentration of 15 mg/1
Chapter 5: Dosage adjustment in renal & hepatic disease state
Introduction
TBC is the sum of metabolic clearance (kmVd), the renal clearance (KeVd) and other clearance if
found (KxVd).
There is situations in which the drug clearance may change (like age,drug interaction,diseases,
genetic variations).
The change in drug clearance will depend on drug nature:
If the drug is rna inly cleared by metabolism,renaI dysfunction will have no effect on TBC
If the drug is rna inly cleared by excretion,renal dysfunction will have effect on TBC
Metabolic clearance
Variable affecting MC
Age
Liver disease
Drug interactions affecting metabolism
Plasma protein binding of drug (transient effects)
Hormonal effects
Renal dysfunction
Age
Drugs interaction on renal tubules
As the renal clearance depend on GFR on its value, so any changes in renal function (known by changes
in GFR) in will cause a parallel effect on the drug elimination for drug excreted unchanged by kidney
Liver disorder (like hepatitis C,bilharzia isis,alcoholic fatty liver) will affects drug metabolism for
drug mainly excreted by metabolism.
There is no direct relation between the degree of liver dysfunction and the decrease in drug
metabolism.
Sever liver damage must be present before significant change in drug metabolism occur.
Not a II liver diseases affect the pharmacokinetics of the drugs to the same extent.
Oral drug bioavailability may be increased by liver disease due to decrease first-pass effects, or
decrease activity for prodrug by decreased drug activation
Child-pugh classification
Enc:cph.a.lopa.thy None 1
Moderate 2
Se-vere 3
A.set tes Ahent ]
Slight 2
Moderat-e 3
BiJicubin (mg/d.L) <2 1
2.1-3 2
>3 3
Albumin (g/d.L) >3.5 1
2 8-3 5 2
<2.8 3
Prothroo:nbin Tir:n e 0-3.9 J
(seconds ::;. coottol) 4-6 2
>6 3
Cefoperazone Chlordiazepoxide
Chloramphenicol Diazepam
Erythromycin Hexobarbital
Metronidazole Lidocaine
Meperidine Metoprolol
Pentazocine Propranolol
Tocainide Theophylline
Verapamil Promazine
Hormonal Influence
For some drugs,both the drug and the metabolite contribute to the overall therapeutic
response of the patient to the drug. The concentration of both the drug and the metabolite in
the body should be known.
(1) when the drug is more potent than the metabolite,the overall pharmacologic activity will increase in
the hepatic-impaired patient because the parent drug concentration will be higher
(2) when the drug is less potent than the metabolite,the overall pharmacologic activity in the hepatic
patient will decrease because less of the active metabolite is formed.
Table 3. Renally Excreted Drugs and Drugs That Require Dosage Adjustmentsa
• ACEis and ARBs • Hyp:lglycemic drugs (e.g., acarbose, metformin,m litol,
• Mbfungals (e.g.,amphoterictn,fluconazole, nateghnkle,repaglrnlde,frrst·generabon sulfonyklreas
ftucytosine, itraconazole, vooconazOie) and selecled second·generabon suHonytureas
• Mbgout drugs (e.g.,allopurinol,roldl ne) [ghmeJ)ride, gl)turide)l
• Mtrtubercutosls drugs (e.g.,etllambutol,INH,nfampln) • Most penlcinins and cephalosporin anbbiotJcs
• MWiraldrugs (e.g.,acyciOVtr.amantadrne. foscarnet • NSAJOs (e.g.,ibuprofen.ketorOiaC. naproxen)
ir ,rimanladine, valacyclovi • Opiold drugs (e.g., code1ne, meperidine,morphine,
• Chemotherapeutic drugs propoxyphene)
• DigOxin • SuHamelhoxazolelttfmethoprim
• Auoroquinolone antibtotJcs (e.g.,ciprotloxactn, • vancomycin IV
levofloxacin) • Others, including cetiril•ne,duloxetine,enoxaparin,
• H2 anlagooisiS (e.g.,e metidine,famobdine, fexofenadine,gabapenbn, HMG·CoA reductase inhlbilors
rallltrdrne) (statins),lllllrum.metoelopram de,methOtrexate,
• Hydrophil( bela·blockers (e.g.,alenolol,solalol) nitrofurantoin,tetracyclme,tirofiban,lramadol
Calculation of (KF)
Creatinine clearance
A- 24 hr urine collection:
UcrX24hrvolurnern l / mtn
.
CrCI=
PcrX60X24
Twenty-four hour urine collection to assess creatinine clearance is no longer widely performed, due to
difficulty in assuring complete specimen collection as need to calculates the amount of creatinine
excreted over a 24-hour period
Ideally a 24 hour urine collection and a mid-point serum creatinine should be obtained. This
method is the most accurate clinical measure of creatinine clearance.
Schwartz equation:
(Patient population: infants over 1week old through adolescence (18 years old))
Cleao rCJonc:•
,:..;:
(...-. L,/ n
100
00
..,;>
.:>0
4U
30
The nomogram method estimates Crcl on the basis of age, weight, and serum creatinine
To use the nomogram,connect the patient's weight on the second line from the left with the patient's
age on the fourth line with a ruler. Note the point of intersection on Rand keep the ruler there. Turn the
right part of the ruler to the appropriate serum creatinine value and the left side will indicate the
clearance in ml/min.
The methods for renal dose adjustment discussed in the previous sections all assume that Vd is
unchanged. These assumptions are convenient and hold true for many drugs.
However,if Vd is changed, Wagner method can be used for calculation of patient half-life.
®
The method assume a linear relationship between Ke and Crcl for all drug after collecting data
from populations
K = a + b (Crcl)
The value for a and bin the equation will depend on the drug type. The values of a and bare
determined statistically for each drug from pooled data on uremic patients.
Dialysis is required in ESRD for removal of waste from body. It may be required from once every
2 days to 3 times a week, with each treatment period lasting 2 to 4 hours.
Dosing of drugs in patients receiving hemodialysis is affected greatly by the frequency and type
of dialysis machine used and by the physicochemical and pharmacokinetic properties of the
drug.
Dialysis clearance
Q(
Cl0
Where:
Special consideration:
The loading drug dose is based on Vd of the patient. In renaI dysfunction, Vd is not altered significantly,
and therefore that the loading dose of the drug is the same as in subjects with normal renal function.
The maintenance dose in multiple drug administration will be changed by (1) decreasing the
maintenance dose,(2) increasing the dosage interval.
Chapter 6: Vancomycin TDM
•!• Following repeated administration of these short IV infusion, the drug will accumulate in
the body until steady state is achieved.
•!• At steady state, the maximum and minimum plasma concentration will be constant ifthe
dose, infusion duration, dosing interval are kept constant.
lO
... 37.
"'"
=
25.0
(Q
12 5
c::
0
0
0.0
0 10 20 30 ..10
Tome (nours)
Fonnulation and stability
Routine dosing
•!• In adjusting the dose make approximation to the nearest available bottle in the market for
vancomycin it is 500 mg, 750 mg, 1 gm, 125omg, 1.5 gm, 1750 mg, 2 gm, 2.5 gm.
6 r- §gEt
:J:
4r o o£
J2 r- l:J'S
010------ --------IT
100
0 I I
10
I
100
I
1000
O '--'1----LI----LI----1'--
0 25 50 75
..1-.J
100
1 24·Hour AUC/MIC Pcook/MIC Time obovo MIC
colony-forming unit (CFU or cfu) is a mea sure of viable bacterial or fungal cells. In direct
microscopic counts where all cells,dead and living, are counted,but CFU measures
only viable cells.
I..LIC
.....c.
Vancomycin pharmacokinetics
Full-term neonates 7
Infants e1month - <1 year) 4.1
Children (2.5-11 years) 5.6 ± 2.1
Adults (16-65 Years) 7.0 ± 1.5 --_-
Adults with renalImpairment 32 ± 19
(CrCI10-60 ml/min)
Geriatrics (>65 years) 12 ± 0.8
Major body burns (>30%-40% BSA) 4
Vancomycin dosing guidelines
•!• A loading dose of25-30 mg/kg may be considered (should be rounded to the nearest 250
mg) specially for critically ill patient.
or
•!• LD = Cmax x Vd
•!• The usual intravenous dosage ofvancomycin is 10 mg/kg per dose given every six hours.
Each dose should be administered over a period of at least 60 minutes.
Infusion time
Infusion-related events "red man syndrome" are related to both concentration and rate of
administration ofvancomycin. Concentrations of no more than 5 mg/mL and rates of no more
than 1gm/hr are recommended in adults. Also minimized by rotating the sites of infusion.
Response to therapy
Adverse Reactions:
•!• Nephrotoxicity
•!• Ototoxicity.
•!• Infusion reactions (e.g. Red Man Syndrome).
•!• The exact mechanism of vancomycin-induced nephrotoxicity (VIN) has not been fully
elucidated.
•!• Reversible nature suspected if an increase in the SCr level of >0.5 mg/dL or 50 percent
from baseline.
Risk factors:
•!• Vanco. trough and [Cr] at least weekly in hemodynamically stable patients.
•!• More frequent monitoring (daily) in high risk patients.
Empiric I
Dose by level
Peak Trough
UTI, Skin infection 25-40 10-15
Steps:
OPatient: FG
D Age: 89 Vancoinycin Dose Per Pharinao/
D Height: 160 em
O Weight: 49.2 Kg
O Gender: F
0 [Cr]: 0.9 mg/dl
D indication: HCAP
Case 1: Answer
K= 0.0317 h- 1
Tl/2= 21.85 hr
I:JPatient: JM
I:JAge: SO Vancon:1ycin Dose Per Pharn:1acy
I:JHeight: Sft 11i
n I:JWeight: 70
Kg I:JGender: M
I:J[Cr]: 0.9 mg/dl (stable for 5 days)
Dlndication: Cellulitis
Case 2: answer
•!• 97 ml/min
The prescribed maintenance dose would be:1000 mg every 12 hours over 1 hr infusion
•!• 1-measure: At steady state, take two samples (for observed peak and observed trough)
and measure concentrations and determine the time in between.
•!• 2-calculate the true peak and true trough concentration
•!• 2-determine K and Vd for this patient
•!• 3- decide: decide the required peak and trough
®
•!• 4-claculate the required Ko, T for this patient: adjust the dose if required bases on patient
specific parameters
Dose by level
•!• Two samples at steady state (before the 4 dose) are taken one for peak (1 hr or more after
the end of infusion to allow adequate time for drug distribution) and the other for trough
(30 min or more before the dose administration).
•!• K= ln(Cl!C2)/L1 t
•!• C1 =observed peak
•!• C2 = observed trough
•!• L1 t =difference in time between C1 and C2 within the same dosing interval
•!• Then determine true peak and true trough
•!• C1 = Cpeak x e<· K x t2)
•!• where Cpeak =true peak (end ofvanco infusion).
•!• Ctrough = C2 x e( ·K x t3)
•!• where Ctrough = true trough.
•!• t2 = time between peak drawn and the end of the infusion
•!• t3 =time between trough drawn and true trough
Example
•!• AJ has HCAP with Height= 5'3", Weight= 130 lbs, SCr = 0.9 mg/dL
•!• AJ receives vancomycin 1250 mg once daily at 10:00 AM. A peak level drawn on day 4
oftherapy at 2:00PM was 30.5 mg/L and a trough level drawn on day 5 of therapy at
8:30AM was 13.0 mg/L.
•!• Make dose adjustment required to achieve the desired trough cone above 15 mg/L.
Answer:
•!• Ll t=18.5
•!• K= 0.046 hr-1
•!• True peak= 33.44 mg/L
•!• Trough trough= 12.13 mg/L
•!• Vd (using Cmax equation) = 54.6 L
•!• Then decide Cmax =37.5 and Cmin=17.5 mg/L
•!• Calculate 1: and Ko
•!• 750 mg infused over 60 min repeated every 12 hr may be advised
Chapter 7: Bioavailability & Bioequivalence
Introduction:
A multisource drug product is a drug product that contains the same active drug substance in the
same dosage form and is marketed by more than one pharmaceutical manufacturer. Single-
source drug products are drug products for which the patent has not yet expired or has certain
exclusivities so that only one manufacturer can make it. Single-source drug products are usually
brand-name (innovator) drug products. After the patent and other exclusivities for the brand-
name drug expires, a pharmaceutical firm may manufacture a generic drug product that can be
substituted for the branded drug product. Since the formulation and method of manufacture of
the drug product can affect the bioavailability and stability of the drug, the generic drug
manufacturer must demonstrate that the generic drug product is bioequivalent and therapeutically
equivalent to the brand-name drug product.
Drug product selection and generic drug product substitution are major responsibilities for
physicians, pharmacists, and others who prescribe, dispense, or purchase drugs. To facilitate
such decisions, the U.S. Food and Drug Administration (FDA) publishes annually, in print and
on the Internet, Approved Drug Products with Therapeutic Equivalence Evaluations, also known
as the Orange Book (www.fda.gov/cder/orange/default.htm). The Orange Book identifies drug
products approved on the basis of safety and effectiveness by the FDA and contains therapeutic
equivalence evaluations for approved multisource prescription drug products. These evaluations
serve as public information and advice to state health agencies, prescribers, and pharmacists to
promote public education in the area of drug product selection and to foster containment of
health care costs. The following definitions are from the 2003 Orange Book, Code of Federal
Regulations.
Defmitions
Bioavailability. Bioavailability means the rate and extent to which the active ingredient or active
moiety is absorbed from a drug product and becomes available at the site of action. For drug
products that are not intended to be absorbed into the bloodstream, bioavailability may be
assessed by measurements intended to reflect the rate and extent to which the active ingredient or
active moiety becomes available at the site of action.
Bioequivalence requirement. A requirement imposed by the FDA for in-vitro and/or in vivo
testing of specified drug products, which must be satisfied. Bioequivalent drug products. This
term describes pharmaceutical equivalent or pharmaceutical alternative products that display
comparable bioavailability when studied under similar experimental conditions. For systemically
absorbed drugs, the test (generic) and reference listed drug (brand-name) shall be considered
bioequivalent if: (1) the rate and extent of absorption of the test drug do not show a significant
difference from the rate and extent of absorption of the reference drug when administered at the
same molar dose of the therapeutic ingredient under similar experimental conditions in either a
single dose or multiple doses; or (2) the extent of absorption of the test drug does not show a
significant difference from the extent of absorption of the reference drug when administered at
the same molar dose ofthe therapeutic ingredient under similar experimental conditions in either a
single dose or multiple doses and the difference from the reference drug in the rate of
®
absorption of the drug is intentional, is reflected in its proposed labeling, is not essential to the
attainment of effective body drug concentrations on chronic use, and is considered medically
insignificant for the drug.
When the above methods are not applicable (eg, for drug products that are not intended to be
absorbed into the bloodstream), other in-vivo or in-vitro test methods to demonstrate
bioequivalence may be appropriate. Bioequivalence may sometimes be demonstrated using an
in-vitro bioequivalence standard, especially when such an in-vitro test has been correlated with
human in-vivo bioavailability data. In other situations, bioequivalence may sometimes be
demonstrated through comparative clinical trials or pharmacodynamic studies.
Bioequivalent drug products may contain different inactive ingredients, provided the manufacturer
identifies the differences and provides information that the differences do not affect the safety
or efficacy of the product.
Brand name. The trade name of the drug. This name is privately owned by the manufacturer
or distributor and is used to distinguish the specific drug product from competitor's products (eg,
Tylenol, McNeil Laboratories).
Chemical name. The name used by organic chemists to indicate the chemical structure of
the drug (eg, N-acetyl-p-aminophenol).
Generic name. The established, nonproprietary, or common name of the active drug in a
drug product (eg, acetaminophen).
For unmarketed drugs that do not have full NDA approval by the FDA, in-vitro and/or in-vivo
bioequivalence studies must be performed on the drug formulation proposed for marketing as a
generic drug product. Furthermore, the essential pharmacokinetics of the active drug ingredient
or therapeutic moiety must be characterized. Essential pharmacokinetic parameters, including the
rate and extent of systemic absorption, elimination half-life, and rates of excretion and
metabolism, should be established after single- and multiple-dose administration. Data from
these in-vivo bioavailability studies are important to establish recommended dosage regimens
and to support drug labeling.
®
In-vivo bioavailability studies are also performed for new formulations of active drug ingredients
or therapeutic moieties that have full NDA approval and are approved for marketing. The
purpose of these studies is to determine the bioavailability and to characterize the
pharmacokinetics of the new formulation, new dosage form, or new salt or ester relative to a
reference formulation.
In summary, clinical studies are useful in determining the safety and efficacy of drug products.
Bioavailability studies are used to define the effect of changes in the physicochemical properties
of the drug substance and the effect of the drug product (dosage form) on the pharmacokinetics
of the drug. Bioequivalence studies are used to compare the bioavailability of the same drug
(same salt or ester) from various drug products. Bioavailability and bioequivalence can also be
considered as performance measures of the drug product in-vivo. If the drug products are
bioequivalent and therapeutically equivalent (as defined above), then the clinical efficacy and the
safety profile of these drug products are assumed to be similar and may be substituted for each
other.
..
<
-
"' c.....
c:: i1
-... ......
G.astrk; f luod "'
a :=!.:
I»
In this graph p..-oduct B,which has lower F than p..-oduct A,may not have
any phannacological effect.
200r---------------------------------------------------,
MEC
"E'
..m
c..
Time (llarl
Relative and Absolute Availability
Relative (apparent) availability is the availability of the drug from a drug product as compared to a
recognized standard formulation, usually a solution of the pure drug evaluated.
The area under the drug concentration-time curve (AUC) is used as a measure of the total
amount of unaltered drug that reaches the systemic circulation. The AUC is dependent on the
total quantity of available drug, FD 0, divided by the elimination rate constant, k, and the
apparent volume of distribution, V D· F is the fraction of the dose absorbed. After IV
administration, F is equal to unity, because the entire dose enters the systemic circulation.
Therefore, the drug is considered to be completely available after IV administration. After oral
administration of a drug, F may vary from a value of 0 (no drug absorption) to 1 (complete drug
absorption).
AUCa
Relative availability=
AUCb
• In order to exert a pharmacological effect, drug cone in the plasma must exceed its
minimum effective cone (MEC).
• If the plasma cone exceeds the MEC the duration of effect will normally last until the
cone falls below the MEC.
In this graph product B, which has lower F than product A, may not have
any pharmacological effect.
200 r-----------------------------------------------
Time (hr)
Tmax
•!• When comparing drug products for bioequvalence, t max can be used as an approximate
indication of drug absorption rate. The value for tmax will become smaller (indicating
less time required to reach peak plasma concentration) as the absorption rate for the drug
becomes more rapid
Cmax
•!• C max provides indications that the drug is sufficiently systemically absorbed to provide
a therapeutic response (intensity).
•!• In addition, C max provides warning of possibly toxic levels of drug. The units of C max
are concentration units (eg, mg/mL, ng/mL).
•!• Although not a unit for rate, C max is often used in bioequivalence studies as a measure
for the rate of drug bioavailability
AUC
•!• The area under the plasma level-time curve, AUC, is a measurement of the extent of drug
bioavailability. The AUC reflects the total amount of active drug that reaches the systemic
circulation
•!• The AUC can be determined by a numerical integration procedure, such as the
trapezoidal rule method. The units for AUC are concentration time (eg, g.hr/mL).
Peak Conc ent ration
Time (hr")
a b
w
The total AUC is found by summing all trapezoids, a nd adding the
arca under the tail of Lhc curve.
8
C2
:::;- 6
.......
.."§".
r::
0
·.;:::: 4
.s
r::
C1J
u
r::
8
E2
;;";':' The tail
of the
curve
co
OL- ---- -- -- =----- ---- --
0.0 1.0 2.0 3.0 4.0
Time (hr)
c:
AUCttast = CtastiA z
0
._
cQ)
(.)
c:
0
<....>
•!• Where Clast is the last measured concentration and K is elimination rate constant.
•!• The total area under the curve is the sum of all trapezoids plus the area of the tail.
•!• Urinary drug excretion data is an indirect method for estimating bioavailability. The drug
must be excreted in significant quantities as unchanged drug in the urine in significant
amount. In addition, timely urine samples must be collected and the total amount of
urinary drug excretion must be obtained.
•!• The total cumulative amount of drug excreted in urine is directly proportional to the total
amount of drug absorbed.
Parameters included are:
•!• Acou, is related directly to the total amount of drug absorbed. Experimentally, urine
samples are collected periodically after administration of a drug product.
•!• A graph is constructed that relates the cumulative drug excreted to the collection-time
interval. The relationship between the cumulative amount of drug excreted in the urine
and the plasma level-time curve is shown in . When the drug is almost completely
eliminated (point C), the plasma concentration approaches zero and the maximum
amount of drug excreted in the urine, A co u, is obtained.
•!• Because most drugs are eliminated by a first-order rate process, the rate of drug
excretion is dependent on the first-order elimination rate constant k and the
concentration of drug in the plasma C p.
•!• When urine sample are collected pe1iodically, the urinary drug excretion rate time
profile should have the same shape of the plasma concentration time profile. where ,
the maximum rate of drug excretion, (dA u/dt)max, is at point B, corresponding to
C max in plasma concentration time curve
•!• The time for maximum urinary excretion will correspond to the Tmax in plasma
concentration time curve
•!• As shown in the following graph point D will be similar to Tmax
Pharmacodynamic Effects
increased cardiac output
decreased pulmonary capillary wedge pressure
increased ejection fraction.
Clinical Indications
Heart failure
Atrial fibrillation
Pharmacokinetics: Absorption
Formulation F
el i xir
tablet
capsule
Pharmacokinetics: Distribution
--
:z:
...
••
07.5 -e Dt O.OllliN Y
lt
.....
::z::
·
--
"'-"
:z.
>C
0
c:J
a
•
0..0.
DIS'IUtl 8U1111'10.. P HAS'E EU MIN All" ION PHASE
...,. Qo .. .. • .,
HOURS
••
2 compartmental model:
< 20% metabolism by the liver (stepwise cleavage of the sugar moiety and lactone ring
reduction).
No significant role of Cytochrome P450 enzymes.
Digoxin metabolites have minor cardioactive effect compared to the parent drug.
Pharmacokinetics: Excretion
non-renal clearance
= 20 ml/min for patients with heart failure
= 40 ml/min for patients without heart failure
Therapeutic Concentration
CHF 0.7-1.2
Sampling:
--- --.
0 t2 1 2
. hr
MS is a 47-year-old male to be initiated on intravenous digoxin for atrial fibrillation. Her weight
is 70 kg and height is 68 inches. Her CrCl is estimated to be at 95 mL/min. Calculate his digoxin
loading dose for a desired plasma concentration of 1 ng/mL.
72 74
G.2S mg dolly 71 7J
70 72
69 71
6e70i
67 09'
661!1115"
6S 67
0.12Smgd.oil)<
64 «1
6l OS
67 64
61 63
so 60 62
S9 61
2. PK Equation
where CL is the clearance of digoxin, Css is the steady state concentration, '( is the dosing
interval in days and F is the bioavailability ofthe formulation.
Maintenance
A more patient-specific maintenance dose can be determined using the patient's dosing history
and the observed digoxin concentrations to derive a patient-specific drug clearance.
CL = (MD)(F)/(Css)l"[)
Assuming MS (from Example 2) was initiated on 0.375 mgtablets daily and in 14 days a steady-
state serum digoxin concentration was obtained right before the next scheduled dose. The Css
was measured to be 2.8 ng/mL. Calculate his true CL and a new dosing regimen to attain a new
desired Css of 1.5 ng/mL.
CL = (375 mcg)(0.7)/(2.8 ng!mL) (l day)
= 93.8 L/day (Note: Her true CL is significantly
different from the estimated 229.3 L/day.)
Population PK changes
Drug Interactions
pgp inhibitors: They are expected to increase digoxin exposure by more than 25%. e.g:
®
amiodarone, captopril, carvedilol, clarithromycin, conivaptan, cyclosporine, diltiazem,
dronedarone, felodipine, itraconazole, lopinavir and ritonavir, quinidine, ranolazine,
ticagrelor, and verapamil.
pgp inducers: They are expected to decrease digoxin AUC by greater than 20%. e.g.
phenytoin, rifampin, St. John's wort, and tipranavir/ritonavir.
CASE 1:
A 47-year-old female, IBW 63.5 kg, is to be initiated on digoxin for atrial fibrillation. Calculate
an appropriate digoxin loading dose and maintenance dose if the patient is to be started on an
intravenous loading and oral maintenance dose using digoxin tablets. Assume her CrCl is 80
mL/min and her target Css is 1 ng/mL
Case2:
Self-study examples:
1. UV is a 75-year-old, 62-kg (height= 5 ft 9 in) male with atrial fibrillation. His current
serum creatinine is 1.3 mg/dL, and it has been stable since admission. Compute an
intravenous loading and maintenance digoxin dose for this patient to provide a steady-
state concentration of 1.5 ng/mL..
2. Patient UV (see problem 1) was prescribed digoxin 200 11g/d intravenously, and this dose
has been given for 2 weeks. A steady-state digoxin concentration was 2.4 ng/mL. Compute
a revised digoxin dose for this patient to provide a steady-state concentration of
1.5 ng/mL.
3. Patient UV (see problems 1 and 2) had a dosage change to digoxin 125 Jlg/d
intravenously which produced a steady-state concentration equal to 1.4 ng/mL. Compute
an oral tablet digoxin dose for this patient that will provide about the same steady-state
drug concentration as that found during intravenous therapy..
4. SD is a 35-year-old, 75-kg (height = 5 ft 7 in)) female with NYHA Class IV heart failure
secondary to viral cardiomyopathy. Her current serum creatinine is 3.7 mg/dL, and it has
been stable since admission. Compute oral digoxin loading and maintenance doses using
tablets for this patient to provide a steady-state concentration of 1 ng/mL.
Chapter 9: Lithium TDM
Clinical Pharmacology
Pharmacodynamic Effects
competition with other cations at receptor and tissue sites,
dopamine-receptor supersensitivity blockage,
decreased stimulation of -receptor-induced adenylate cyclase, and enhanced sensitivity
to serotonin (5-HT), acetylcholine, and GABA.
Clinical Indications
Pharmacokinetics: Absorption
PK parameter
Pharmacokinetics: Distribution
0
E
g
c:
1
--
Ot or distributi on phase
-e 0. 1
or elimination phase
E
Q.>
g
8
0.01
0 10 20 30 40 so
Time (h)
Multicompartment model.
Insignificant plasma protein binding.
Significant inter-individual variations (average Vd= 0.9 L/kg).
Pharmacokinetics: Elimination
Lithium is eliminated almost completely (>95%) unchanged in the urine with elimination tl/2 of
24h.
filtered freely at the glomerulus, and subsequently 60%-80% of the amount filtered is
reabsorbed(along with Na+) by the proximal tubule of the nephron.
consistent relationship between lithium clearance and creatinine clearance with a ratio of 20%
(lithium clearance-creatinine clearance).
Monitoring of renal function is necessary
Pharmacokinetics: Clearance
Therapeutic Concentration
These therapeutic ranges are based on steady-state lithium serum concentrations obtained 12 hours
after a dose
1.5-3.0: confusion,
Pharmacology agitation,nystagmus,
hypertonia, ataxia.
Therapeutic Range
0.6-1.2 mEqll j >1.6 mEqtL j • 1.2- 1.5:NVD, hand
tremors, reduced memory
!10-1.2 mEq/L I• TOXICITY
ACUTE MANIA
Sampling
®
When lithium serum concentration monitoring is anticipated for an individual, the patient needs
to understand that it is important to:
take their medication as instructed for 2-3 days before the blood sample is obtained,
to have the blood sample withdrawn 12 ± 0.5 h after the last dose,and
to report any discrepancies in compliance and blood sampling time to their care provider.
Therapeutic Monitoring
Onset of action for lithium is 1-2 weeks,and a 4- to 6-week treatment period is required to
assess complete therapeutic response to the drug.
where Cl is the clearance of lithium (L/day), Css is the steady state concentration (mg/L),Tis the
dosing interval in days and F is the bioavailability of the formulation (100% for all Lithium
formulations).
Example 1
MJ is a 50-year-old,70-kg (height= 5 ft 10 in) male with bipolar disease. He is not currently experiencing
an episode of acute mania. His serum creatinine is 0.9 mg/dl. Compute an oral lithium dose for this
patient for maintenance therapy
Example 2
MJ is a 50-year-old,70-kg (height= 5 ft 10 in) male with bipolar disease. He is not currently experiencing
an episode of acute mania. His serum creatinine is 3.5 mg/dl. Compute an oral lithium dose for this
patient for maintenance therapy
Dosage Adjustment
Example 3
YC is a 37-year-old, 55-kg (height= 5 ft 1in) female with bipolar disease. She is currently not
experiencing an episode of acute mania and requires prophylactic treatment with lithium. Her serum
creatinine is 0.6 mg/dl. The patient is receiving 900 mg of lithium carbonate at 0800 H, 1400 H,and
2000 H,and her 12-hour pos-tdose steady-state lithium serum concentration equals 1.1mmoi/L.
Compute a new lithium dose to achieve a steady state concentration of 0.6 mmoi/L
Drug Interactions
Practice Cases
1. PG is a 67-year-old, 72-kg (height= 6ft 1in,serum creatinine= 1.2 mg/dl)male with bipolar disease
requiring maintenance therapy with oral lithium. Suggest an initial lithium carbonate dosage regimen
designed to achieve a steady-state lithium concentration equal to 0.6 mmoi/L.
2. Patient PG (see problem 1) was prescribed lithium carbonate 900 mg orally every 12 hours. The
current 12-hour post-dose steady-state lithium concentration equals 1.0 mmoi/L. Compute a new
lithium carbonate dose that will provide a steady-state concentration of 0.6 mmoi/L.
4. Patient DU (see problem 3) was prescribed lithium carbonate 600 mg orally at 0800 H, 1400 H,and
2000 H. The current 12-hour post-dose steady state lithium concentration equals 0.6 mmoi/L. Compute
a new oral lithium dose that will provide a steady-state concentration of 1mmoi/L.
Zero order reaction Cp Versus time plot after a single oral
𝑪𝟐 − 𝑪𝟏 dose
𝒔𝒍𝒐𝒑𝒆 = −𝒌 ∘=
𝒕𝟐 − 𝒕𝟏 𝑭 𝑫 𝑲𝒂
𝒀 𝒊𝒏𝒕𝒆𝒓𝒆𝒑𝒕 =
𝑨∘ 𝑽𝒅 (𝑲𝒂 − 𝒌)
𝒕𝟏 =
𝟐𝑲 ∘
𝟐
The area under the curve (AUC)
𝑨 = 𝑨 ∘ −𝑲𝒕
𝑭 . 𝑫𝒐𝒔𝒆 𝑭 . 𝑫𝒐𝒔𝒆
𝑨𝑼𝑪{𝒕=∞
𝒕=𝟎 = =
First order reaction 𝑻𝑩𝑪 𝑲 𝑽𝒅
𝒀 𝜟𝑨𝒆
𝒍𝒐𝒈 𝟐⁄𝒀 = 𝒌𝒆 . 𝑨 = 𝒌𝒆 . 𝑽𝒅 . 𝑪𝒑𝒕 𝒎𝒊𝒅 = 𝒌𝒆 . 𝑨 ∘ . 𝒆−𝒌 𝒕𝒎𝒊𝒅
K= 𝟏
× 𝟐. 𝟑𝟎𝟑 𝜟𝒕
𝑿𝟐 −𝑿𝟏
𝜟𝑨𝒆
𝒍𝒏 = 𝒍𝒏 𝒌𝒆 . 𝑨 ∘ −𝒌 . 𝒕𝒎𝒊𝒅
First order elimination 𝜟𝒕
𝜟𝑨𝒆 𝒌 . 𝒕𝒎𝒊𝒅
𝑪𝒑 = 𝑪𝒑 ∘ 𝒆−𝒌𝒕 𝒍𝒐𝒈 = 𝒍𝒐𝒈 𝒌𝒆 . 𝑨 ∘ −
𝜟𝒕 𝟐. 𝟑𝟎𝟑
𝒌𝒕
𝒍𝒐𝒈 𝑪𝒑 = 𝒍𝒐𝒈 𝑪𝒑 ∘ − 𝑻𝑩𝑪 = 𝑹 𝒄𝒍 + 𝑴. 𝒄𝒍
𝟐. 𝟑𝟎𝟑
𝜟𝑨
𝒍𝒏 𝑪𝒑 = 𝒍𝒏 𝑪𝒑 ∘ −𝒌𝒕
𝑹𝒆𝒏𝒂𝒍 𝒄𝒍 = 𝒌𝒆 . 𝑽𝒅 = 𝜟𝒕
𝟎. 𝟔𝟗𝟑 𝑪𝒑𝒕 𝒎𝒊𝒅
𝒕𝟏 =
𝟐 𝒌 𝑻𝑩𝑪 = 𝑲. 𝑽𝒅
−𝒌𝒕
𝑨=𝑨∘𝒆 𝑲𝒆 𝑨𝒆∞ 𝑨𝒆∞
𝑹𝒆𝒏𝒂𝒍 𝒄𝒍 = 𝑻𝑩𝑪 × = 𝑻𝑩𝑪 × =
𝒌 𝑭 . 𝑫 𝑨𝑼𝑪𝒊𝒗
𝒌𝒕
𝒍𝒐𝒈 𝑨 = 𝒍𝒐𝒈 𝑨 ∘ −
𝟐. 𝟑𝟎𝟑 𝑨𝒆∞ 𝒐𝒓𝒂𝒍
𝑨𝒃𝒔𝒐𝒍𝒖𝒕𝒆 𝑭 =
𝒍𝒏 𝑨 = 𝒍𝒏 𝑨 ∘ −𝒌𝒕 𝑨𝒆∞ 𝒊𝒗
𝑫𝒇𝒂𝒊𝒍 𝑫𝒏𝒐𝒓𝒎𝒂𝒍
For female
=
𝑪𝒍𝒇𝒂𝒊𝒍 𝑪𝒍𝑵𝒐𝒓𝒎𝒂𝒊𝒍 𝑰𝑩𝑾 = 𝟒𝟓 + (𝟐. 𝟑𝒙 𝒉𝒆𝒊𝒈𝒉𝒕 𝒊𝒏 𝒊𝒏𝒄𝒉𝒆𝒔 𝒎𝒐𝒓𝒆 𝒕𝒉𝒂𝒏 𝟓 𝒇𝒐𝒐𝒕)
𝑫𝒏𝒐𝒓𝒎𝒂𝒍 × 𝒕𝟏⁄
𝟐𝒏𝒐𝒓𝒎𝒂𝒍
= 𝑫𝒇𝒂𝒊𝒍 × 𝒕𝟏⁄
𝟐𝒇𝒂𝒊𝒍 Crcl for pediatric
𝒕𝒐𝒕𝒂𝒍 𝒂𝒎𝒐𝒖𝒏𝒕 𝒆𝒙𝒄𝒓𝒆𝒕𝒆𝒅 Schwartz equation:
𝒇=
𝑫𝒐𝒔𝒆
𝒍𝒆𝒏𝒈𝒕𝒉 (𝒄𝒎) × 𝒌
𝑲𝒆 𝑹𝒄𝒍 𝑹𝒄𝒍 𝑨𝒆∞ 𝑪𝒓𝑪𝒍 =
𝒇= = = = 𝑺𝑪𝒓
𝑲 𝑻𝑩𝑪 𝑲 . 𝑽𝒅 𝑭. 𝑫
𝟎.𝟒𝟓 𝒇𝒓𝒐𝒎 𝟏𝒕𝒐 𝟓𝟐 𝒘𝒆𝒆𝒌𝒔
𝟎.𝟓𝟓 𝒇𝒓𝒐𝒎 𝟏 𝒕𝒐 𝟏𝟑 𝒚𝒆𝒂𝒓𝒔
𝒌 =𝟎.𝟓𝟓 𝒇𝒓𝒐𝒎 𝟏𝟑 𝒕𝒐 𝟏𝟖 𝒚𝒆𝒂𝒓𝒔 𝑭𝒆𝒎𝒂𝒍𝒆
𝟎.𝟕 𝒇𝒓𝒐𝒎 𝟏𝟑 𝒕𝒐 𝟏𝟖 𝒚𝒆𝒂𝒓𝒔 𝒎𝒂𝒍𝒆
Where;
Dose adjustment in pediatric
𝑪𝒓𝑪𝒍 𝒐𝒇 𝒇𝒂𝒊𝒍𝒖𝒓𝒆 𝒄𝒂𝒔𝒆 𝑮𝑭𝑹𝑭𝒂𝒊𝒍𝒖𝒓𝒆
𝑲𝑭 = = BSA = 0.007184 × Wt 0.425 × Ht 0.725
𝑪𝒓𝑪𝒍 𝒐𝒇 𝒏𝒐𝒓𝒎𝒂𝒍 𝒄𝒂𝒔𝒆 𝑮𝑭𝑹𝒏𝒐𝒓𝒎𝒂𝒍
For
Fourth Level (Pharm D Program)
2024-2025
By:
Clinical Pharmacy Department
Department of Clinical Pharmacy
College Of Pharmacy
KFS University
Clinical pharmacokinetics Fourth Year
Syllabus
Topic Page
number
1 Revision 3
3 Phenytoin pharmacokinetics 10
5 Digoxin pharmacokinetics 15
6 Lithium pharmacokinetics 18
7 Aminoglycoside pharmacokinetics 23
8 Vancomycin pharmacokinetics 27
9 Bioavailability 31
2|Page
Clinical pharmacokinetics Fourth Year
Lab 1
3|Page
Clinical pharmacokinetics Fourth Year
A patient with liver failure and a patient with heart failure need to be treated
with a new antiarrhythmic drug. You find a research study that contains the
following information for Stopa beat in patients similar to the ones you need
to treat: • Normal subjects: clearance = 45 L/h, volume of distribution = 175
L • Liver failure: clearance = 15 L/h, volume of distribution = 300 L • Heart
failure: clearance = 30 L/h, volume of distribution = 100 L
4|Page
Clinical pharmacokinetics Fourth Year
After the first dose of gentamicin is given to a patient with renal failure, the
following serum concentrations are obtained:
Compute the half-life and the elimination rate constant for this patient
5|Page
Clinical pharmacokinetics Fourth Year
Lab 2
………………………………………………………………………………
………
………………………………………………………………………………
………
………………………………………………………………………………
………
………………………………………………………………………………
………
………………………………………………………………………………
………
………………………………………………………………………………
………
6|Page
Clinical pharmacokinetics Fourth Year
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
7|Page
Clinical pharmacokinetics Fourth Year
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
8|Page
Clinical pharmacokinetics Fourth Year
9|Page
Clinical pharmacokinetics Fourth Year
Lab 3
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
10 | P a g e
Clinical pharmacokinetics Fourth Year
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
……………………………………………………………………………………………
11 | P a g e
Clinical pharmacokinetics Fourth Year
12 | P a g e
Clinical pharmacokinetics Fourth Year
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………..
13 | P a g e
Clinical pharmacokinetics Fourth Year
14 | P a g e
Clinical pharmacokinetics Fourth Year
15 | P a g e
Clinical pharmacokinetics Fourth Year
Lab 5
16 | P a g e
Clinical pharmacokinetics Fourth Year
17 | P a g e
Clinical pharmacokinetics Fourth Year
2. Patient UV was prescribed digoxin 200 μg/d intravenously, and this dose
has been given for 2 weeks. A steady-state digoxin concentration was 2.4
ng/mL. Compute a revised digoxin dose for this patient to provide a steady-
state concentration of 1.5 ng/mL
18 | P a g e
Clinical pharmacokinetics Fourth Year
Lab 6
19 | P a g e
Clinical pharmacokinetics Fourth Year
Ali was prescribed lithium carbonate 900 mg orally every 8 hours. The
current 12-hour post dose steady-state lithium concentration equals 1.0
mmol/L. Compute a new lithium carbonate dose that will provide a
steady- state concentration of 0.6 mmol/L.
20 | P a g e
Clinical pharmacokinetics Fourth Year
21 | P a g e
Clinical pharmacokinetics Fourth Year
22 | P a g e
Clinical pharmacokinetics Fourth Year
Lab 7
23 | P a g e
Clinical pharmacokinetics Fourth Year
24 | P a g e
Clinical pharmacokinetics Fourth Year
25 | P a g e
Clinical pharmacokinetics Fourth Year
26 | P a g e
Clinical pharmacokinetics Fourth Year
………………………………………………………………………………………
……………………………………………………………………………………
………………………………………………………………………………………
……………………………………………………………………………………
………………………………………………………………………………………
……………………………………………………………………………………
………………………………………………………………………………………
……………………………………………………………………………………
………………………………………………………………………………………
……………………………………………………………………………………
………………………………………………………………………………………
……………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
………………………………………………………………………………………
27 | P a g e
Clinical pharmacokinetics Fourth Year
Lab 8
28 | P a g e
Clinical pharmacokinetics Fourth Year
29 | P a g e
Clinical pharmacokinetics Fourth Year
30 | P a g e
Clinical pharmacokinetics Fourth Year
31 | P a g e
Clinical pharmacokinetics Fourth Year
Lab 9
f-If the minimum effective concentration for this drug is about 12.5
mg/L. what will be the approximate duration of action in the four
formulations?
g- Can we calculate absorption rate constant of the three oral forms and
how? without applying the residual method?
32 | P a g e
Clinical pharmacokinetics Fourth Year
33 | P a g e
Clinical pharmacokinetics Fourth Year
34 | P a g e