You are on page 1of 804

F

Basic Pharmacokinetics
Michael C. Makoid, Ph.D.
Professor of Pharmaceutical Sciences
Creighton University School of Pharmacy
and Allied Health Sciences,
Omaha, Nebraska

Phillip J. Vuchetich
Pharm.D. Candidate
Creighton University School of Pharmacy
and Allied Health Sciences,
Omaha, Nebraska

Umesh V. Banakar, Ph.D.


Professor of Pharmaceutical Sciences
St. Louis College of Pharmacy
St. Louis, Missouri

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-1

Copyright 1996-1999 The Virtual University Press


All rights reserved.

ISBN 0-000-000000-0

ABCDEFGHIJ-DO-89

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-2

Acknowledgement
When I first started teaching, I had the good fortune to work with another new
Ph.D., John Cobby. We struggled through our first five years on the otherside of
the podium together and learned many of the tenents upon which this book is
based, not content but process. First and formost, it was his belief that students are
bright, enthusiastic and hardworking. We should tell them what to do and get out
of their way. We both prepared extensive handouts complete with even more
extensive practice problems so that the student could experience the scientific
method as a detective might solve a murder mystery. The idea was to make learning pharmaceutical science interesting and fun. Through the years, as the methods
became more refined, student perceptions and performance improved dramatically.
John ultimately abandoned academe to go to work in the real world of industry,
clearly their gain and our loss. I approached him some years ago to co-author this
text. He declined believing himself to be too far removed from the cutting edge of
this discipline. That may be true (I doubt it!), but what can not be argued is that he
was a major contributor to this book in his philosophy and class notes. Over the
years, the explainations were rewritten and revised. Many new problems were
added and some were suplanted. These teaching aides have evolved but their origins are clear. Using the industry standard regarding authorship, which defines an
author as one whose contributions significantly alters the content of the paper, Dr.
Cobby is an author of this book.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-3

CHAPTER 1

Introduction
Basic Pharmacokinetics 1-2
Course Objectives: 1-3
Course Arrangement: 1-3
Learn the tools; get the pharmacokinetic parameters from patient information. 1-3
Learn the modifications of the pharmacokinetic parameters which result from illness. 1-5
Apply the tools; use the pharmacokinetic parameters to develop dosage regimens. 1-5
Apply the tools in specialized drug classes. 1-5

Exams

1-7

Library Assignment in Pharmacokinetics 1-7

Blooms taxonomy for the Hierarchy of Educational Objectives


Course Contract 1-10
Computers in the course 1-14
Survival Kit 1-15

1-9

Things for your Survival Kit! 1-15


What you will gain: (your goals) 1-16

Tentative Schedule

1-17

Study Group 1: Learn the tools - obtain pharmacokinetic parameters from data. 1-17
Study group 2: Learn how the parameters are modified. 1-19
Study Group 3: Apply the tools in compromised patients. 1-20
Study Group 4: Apply the tools in special cases. 1-20

Competency Statements Related To Pharmacokinetics

1-22

Specific Competency Statements addressed in this course

1-22

Pharmacokinetic Symbolism

1-25

Amount Terms (unit: mass) 1-25


Concentration terms (units mass/volume) 1-26
Volume Terms (unit: volume) 1-26
Time Terms (unit: time) 1-27
Rate Constant Terms (unit: reciprocal time (*), mass/time (**) 1-27
Clearance Terms (units: volume/time) 1-28
Rate Terms (units: mass/time (*), mass/time, volume (**), volume/time (***) 1-28
Other Terms 1-29
Subscripts 1-30
Superscripts 1-30

First Lesson in Pharmacokinetics

CHAPTER 2

1-32

Mathematics Review
Concepts of Mathematics 2-2
Mathematical Preparation 2-3
Zero and Infinity 2-3
Expressing Large and Small Numbers 2-3
Significant Figures 2-4
Rules of Indices 2-4
Logarithms 2-6
Natural Logarithms 2-6
Negative Logarithms 2-9

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-4

Using Logarithmic and Anti-logarithmic Tables 2-10


Dimensions 2-11
Dimensional Analysis 2-12

Calculus

2-14

Differential Calculus 2-14


Non-linear Graphs 2-14
Slope of Non-linear Graph 2-15
Value of the Slope 2-15
Differentiation from First Principles
Rule of Differentiation 2-17
Three Other Derivatives 2-17
A Seeming Anomaly 2-18
Integral Calculus 2-19
Rule of Integration 2-19
The Constant of Integration 2-20
The Exception to the Rule 2-20
A Useful Integral 2-20
Example Calculations 2-21

Graphs

2-16

2-24

Graphical Conventions 2-25


Straight Line Graphs 2-26
The Slope of a Linear Graph (m) 2-28
Linear Regression: Obtaining the slope of the line
Parallel lines 2-31
Graphical Extrapolations 2-32
Significance of the Straight Line 2-32
Graphical Honesty 2-32
Axes with Unequal Scales 2-33
Graphs of Logarithmic Functions 2-34
Semilogarithmic Coordinates 2-34
Log - Log Coordinates 2-38
Pitfalls of Graphing: Poor Technique 2-38
Graphical analysis 2-40

Pharmacokinetic Modeling

2-29

2-44

Making a Model 2-45


One Compartment Open Model 2-47

The LaPlace Transform

2-48

Table of LaPlace Transforms 2-49


Symbolism 2-49
Conventions used in drawing pharmacokinetic schema. 2-50
Steps for Integration Using the LaPlace Transform 2-53
Example Integration Using the LaPlace Transform 2-54
Second Example Integration Using the LaPlace Transform 2-56
Third Example Integration Using the LaPlace Transform 2-57
Conclusions 2-58
Table of LaPlace Transforms 2-60
LaPlace Transform Problems 2-62
LaPlace Transform Solutions 2-63

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-5

CHAPTER 3

Pharmacological Response
Pharmacological Response

3-2

The Hyperbolic Response Equation 3-2


Interrelationships between concentration, time and response 3-3

Change in Response with Time

3-4

One-Compartment Open Model: Intravenous Bolus Injection 3-4


One-Compartment Open Model: Oral Administration 3-4
Duration of Effective Pharmacological Response 3-4
Pharmacokinetic Parameters from Response Data 3-5
Delayed Response 3-5
Response of active metabolite: 3-6

Therapeutic Drug Monitoring

3-7

Therapeutic monitoring: Why do we Care?

Problems
Answers:
Answers:
Answers:

CHAPTER 4

3-9

3-11
Oxpranolol 3-18
Minoxidil 3-21
Propranolol 3-23

I.V. Bolus Dosing


I.V. Bolus dosing of Parent compound

4-2

Plasma 4-2
Iv bolus, parent compound, plasma Problems
Urine 4-47

Metabolite

4-7

4-51

Plasma 4-51
Urine 4-56

CHAPTER 5

I.V. Infusion
Parent compound

5-2

Plasma 5-2

Problems

5-10

CHAPTER 6

Biopharmaceutical Factors

CHAPTER 7

Oral Dosing
Oral dosing

7-3

Valid equations: ( oral dosing, plasma) 7-3


Utilization 7-3

CHAPTER 8

Bioavailability, Bioequivalence, and Drug Selection


Bioavailability, Bioequivalence and Drug Product Selection

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-2

-6

Relative and Absolute Bioavailability 8-2


Factors Influencing Bioavailability 8-4
Methods of Assessing Bioavailability 8-11
Study Design 8-15
In-vitro Dissolution and Bioavailability 8-15
In-vitro / in-vivo correlation studies- 8-18

Bioequivalence

8-20

Bioequivalence Regulations 8-23


Study Design 8-26
Assessment of bioequivalence 8-29
Controversies and Concerns in Bioequivalence 8-31
Generic Drugs and Product Selection 8-35
The Orange Book 8-38
Therapeutic equivalence 8-38
Therapeutic equivalence evaluation codes- 8-39

Drug Product Selection

8-44

Considerations in selecting a manufacturer 8-45


Special Cases 8-50

Summary

8-54

Questions 8-55
Answers to Questions

8-57

Bioavailibility Equations
Problems 8-60
Solutions 8-80

8-58

Caffeine on page 61 8-80


Cefetamet Pivoxil on page 62 8-83
Cefixime on page 63 8-86
Ceftibuten on page 64 8-87
Cimetidine on page 65 8-88
Diurnal Variability in Theophylline Bioavailability on page 66 8-89
cis-5-Fluoro-1-[2-Hydroxymethyl-1,3-Oxathiolan-5-yl] Cytosine (FTC) on page 67 8-90
Hydromorphone on page 68 8-91
Isosorbide Dinitrate on page 69 8-92
Ketanserin on page 70 8-93
Methotrexate on page 71 8-94
Moclobemide on page 72 8-95
Nalbuphine on page 73 8-96
Nefazodone on page 74 8-97
Ondansetron on page 75 8-98
Omeprazole on page 76 8-99
Paroxetine on page 77 8-100
Ranitidine on page 78 8-101
Sulpiride on page 79 8-102

References

CHAPTER 9

8-103

Clearance
Equations 9-2
Definitions and Terms

9-3

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-7

Measurement of Creatinine Clearance


Model Correlations 9-5

9-4

Renal Clearance 9-5


Systemic Clearance and Metabolic Clearance
Use in Pharmacokinetic Equations 9-6

Physiological Factors Affecting Clearance

9-5

9-7

Intrinsic Clearance 9-7


Extraction Ratio (E) 9-8

Hepatic Function and Clearance

9-10

Alterations in Hepatic blood Flow 9-10


Alterations in Hepatic Intrinsic Clearance
Tabulated or Graphical Alterations 9-11

9-10

Renal Function and Clearance 9-12


General Equations for Changes in Clearance

9-14

Plasma/Blood ratio 9-14


Half life and elimination rate constant in relationship to clearance 9-16
Effects of alterations in protein binding on clearance 9-16

Problems

CHAPTER 10

9-17

Dosage Regimen (Healthy, Aged, and Diseased Patients)


Therapeutic Drug Monitoring

10-2

Therapeutic Range 10-2


Therapeutic monitoring: Why do we Care?
Steady State 10-5

10-4

Diseases - Dosing the Compromised Patient


Problems 10-12
Answers 10-36

CHAPTER 11

10-10

Multicompartment Modeling
Executive Summary 11-2
Equations 11-3
PHARMCOKINETICS: MAMMILLARY MODELS
Begin 11-90
Problems 11-91

11-4

Two-compartment Model Equations 11-119


Answers 11-120

CHAPTER 12

Protein Binding

CHAPTER 13

Nonlinear (Michaelis-Menton) Kinetics


Problems 13-2
Nonlinear Equations

13-14

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-8

Answers 13-14

CHAPTER 14

Practice Exams: Section 1


Nifedipine - Section 1

Nifedipine Data 3
Nifedipine Questions 4
Nifedipine Solutions 6

Enalapril - Section 1 11
enalapril data 11
Enalapril Questions 13
enalapril Solutions 14

Ciprofloxacin Section 1 19
Ciprofloxacin data 19
Ciprofloxacin Questions 20
Ciprofloxacin Solutions 21

Methylphenidate Section 1

23

methylphenidate data 23
MethylPhenidate Questions: 24
methylPhenidate Solutions 25

Adinazolam - Section 1

27

ADINAZOLAM DATA 27
Adinazolam Questions 28
Adinazolam Solutions 30

Labetalol - Section 1

32

labetalol data 32
labetalol Questions 33
labetalol Solutions 34

Zidovudine Section 1

36

zidovudine data 36
Zidovudine Questions 38
Zidovuldine Solutions 40

Fosinopril Section 1

42

fosinopril data 42
Fosinopril Questions 44
Fosinopril Solutions 46

Omeprazole

49

Omeprazole Data 49
Omeprazole Questions 50
Omeprazole solutions 53

EXP3312, an Experimental Drug

57

EXP3312 DATA 57
EXP3312 & M1 Questions: 59

Graph Paper

CHAPTER 15

62

Practice Exams: Exam 2


Nifedipine: Exam 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-9

Nifedipine Questions: 3

Valproate: Exam 2

Valproate Questions 7
Valproate Solutions 10

Methyl phenidate

11

Methyl phenidate Questions: 12


Methyl phenidate Solutions: 13

Verapamil

15

Verapamil Questions 16
Verapamil Solutions 17

Hydromorphone hydrochloride

19

Hydromorphone hydrochloride Questions 20

Fosinopril Sodium

23

Fosinopril Questions 24
Fosinopril Sodium Solutions

Remoxipride

26

28

Remoxipride Questions 29
Remoxipride Solutions 31

Naproxen

33

Naproxen Questions 34
Naproxen Solutions 36
38

CHAPTER 16

Exam 3
Pharmacokinetics Final Exam

16-2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-10

CHAPTER 1

Introduction

Author: Michael Makoid and John Cobby


Reviewer: Phillip Vuchetich

OBJECTIVES
At the completion of this chapter, the successful student shall be able to:
1.

define pharmacokinetics

2.

state the overall objectives of the course

3.

state the major themes of the course

4.

state the course organizational structure with respect to study sections

5.

state the objectives of each study section

6.

state the examination structure and objectives

7.

state student performance expectations

8.

state the schedule and timeline

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-1

Introduction

1.1 Basic Pharmacokinetics


What is pharmacokinetics?

Pharmacokinetics is the mathematics of the time course of Absorption, Distribution, Metabolism, and Excretion (ADME) of drugs in the body. The biological,
physiological, and physicochemical factors which influence the transfer processes
of drugs in the body also influence the rate and extent of ADME of those drugs in
the body. In many cases, pharmacological action, as well as toxicological action, is
related to plasma concentration of drugs. Consequently, through the study of
pharmacokinetics, the pharmacist will be able to individualize therapy for the
patient.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-2

Introduction

1.2 Course Objectives:


The Roman numerals refer to the cognitive complexity as described in Bloom's
Taxonomy of Educational Objectives described elsewhere in this document. At the
completion of this course, the successful student will be able to:
What will you be able to
do?

Calculate (III) patient and drug specific pharmacokinetic parameters from patient data,
Predict (calculate - III) the changes in relevant pharmacokinetic parameters in the patient with
selected diseases,

Utilize the above parameters to individualize patient therapy (devise a dosage regimen - V),
Communicate his/her therapy recommendations to another competent health professional (write
a consult - V).

1.2.1

COURSE ARRANGEMENT:
Two courses are described below. The first, a two credit (Creighton University
required) and the second, a three credit (CU optional) version. The two credit
course will consist of major themes one through three and exams one and two,
while the three credit course will add theme four and exam three. The four major
themes are entitled:

How is the course


arranged?

Learn the tools; get the pharmacokinetic parameters from patient information.
Learn the modifications of the pharmacokinetic parameters which result from illness.
Apply the tools; use the pharmacokinetic parameters to predict patient response and develop
dosage regimens for the normal as well as for the compromised patient.

Apply the tools in specialized drug classes.

Each major theme of the course is further broken down into study sections, each
with their own set of general objectives as shown below:

1.2.2

LEARN THE TOOLS; GET THE PHARMACOKINETIC PARAMETERS


FROM PATIENT INFORMATION.
A. Basic Mathematical skills objectives:

What will I be required


to be able to do? How
will examination questions be written for this
material?

1.
2.
3.
4.

Given a data set containing a pair of variables, the student will properly construct (III) various graphs of the data.
Given various graphical representations of data, the student will calculate (III) the slope
and intercept by hand as well as using linear regression.
The student shall be able to interpret (V) the meaning of the slope and intercept for the
various types of data sets.
The student shall demonstrate (III) the proper procedures of mathematical and algebraic
manipulations.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-3

Introduction

5.

The student shall demonstrate (III) the proper calculus procedures of integration and differentiation.
6. The student shall demonstrate (III) the proper use of computers in graphical simulations
and problem solving.
7. Given the assumptions for the model, the student will construct (III) models of the ADME
processes using Laplace Transforms.
8. The student shall develop (V) integrated equations associated with the above models.
9. The student shall generate a pharmacokinetic model based on given information.
10. The student shall interpret a given model mathematically.
11. The student shall predict changes in the final result based on changes in variables throughout the model.

B. Pharmacological Response objectives:


1.

2.

Given patient data of the following types, the student will be able to properly construct
(III) a graph and compute (III) the slope: response (R) vs. concentration (C), response (R)
vs. time (T), concentration (C) vs. time (T)
Given any two of the above data sets, the student will be able to compute (III) the slope of
the third.

C. IV one compartment model, plasma and urine objectives:


1.

2.

Given patient drug concentration and/or amount vs. time profiles, the student will calculate (III) the relevant pharmacokinetic parameters available ( V d , K, k m , k r , AUC , Clearance, MRT) from IV data.
Given the appropriate pharmacokinetic parameters, the student shall simulate (III)
I.V. bolus/infusion dosing for parent compounds
Plasma concentration vs. time profile analysis

3.
4.
5.

Rate vs. time profile analysis


Given patient specific pharmacokinetic parameters, the student shall provide professional
communication regarding IV bolus/infusion information
The student shall utilize computer aided instruction and simulation
Given patient drug concentration and/or amount vs. time profiles, the student will calculate (III) the relevant metabolite (active vs. inactive) pharmacokinetic parameters available ( V d , K, k m , k r , AUC , Clearance, MRT) from IV data.

D. Biopharmaceutical factor objectives: the student shall be able to discuss:


1.
2.
3.
4.
5.

physiology and mechanisms of absorption


effects of diffusion, cardiac output / blood perfusion, physical properties of the drug and
body on distribution
biotransformation, first pass effect, and clearance
renal, biliary, mammary, salivary, other forms of excretion.
the effects of physiological changes with age, sex, and disease on the absorption, distribution, metabolism, and excretion of a drug.

E. Oral one compartment model objectives:


1.

Given patient drug concentration and/or amount vs. Time profiles, the student will calculate (III) the relevant pharmacokinetic parameters ( V d , K, k m , k r , k a , AUC , Clearance,
MRT, MAT) available from oral data.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-4

Introduction

F. Bioavailability objectives:
1.

Given sufficient data to compare an oral product with another oral product or an IV product, the student will estimate (III) the bioavailability (compare AUCs) and judge (VI) professional acceptance of the product with regard to bioequivalence (evaluate (VI) AUC, T p
and ( C p )max ).

2.

1.2.3

The student will write (V) a professional consult using the above calculations.

LEARN THE MODIFICATIONS OF THE PHARMACOKINETIC


PARAMETERS WHICH RESULT FROM ILLNESS.
G. Clearance objectives:
1.

1.2.4

Given patient information regarding organ function, the student will calculate (III)
changes in clearance and other pharmacokinetic parameters inherent in compromised
patients.

APPLY THE TOOLS; USE THE PHARMACOKINETIC PARAMETERS


TO DEVELOP DOSAGE REGIMENS.
H. Dosage regimens objectives:
1.
2.
3.
4.

1.2.5

Given population average patient data, the student will devise (V) dosage regimens which
will maintain plasma concentrations of drug within the therapeutic range.
Given specific patient information, the patient will justify (VI) dosage regimen recommendations.
Given patient information regarding organ function, the student will devise (V) and justify
(VI) dosage regimen recommendations for the compromised patient.
The student will write (V) a professional consult using the above calculations

APPLY THE TOOLS IN SPECIALIZED DRUG CLASSES.


I. Two Compartment Model objectives:
1.

Given patient Concentration and/or Amount of Drug vs. Time, profiles the student will
calculate (III) the relevant pharmacokinetic parameters( V d1 , Alpha, A 1 , Beta, B 1 ,
k 10 , k 12 , k 21 , AUC , Clearance, compartmental amount ratios) available from two com-

2.
3.
4.
5.

partment data.
Given population average patient data, the student will devise (V) a dosage regimen which
will maintain plasma concentrations of drug within the therapeutic range.
Given specific patient information, the patient will justify (VI) the optimal dosage regimen.
Given patient information regarding organ function, the student will devise (V) and justify
(VI) the optimal dosage regimen for the compromised patient.
The student will write (V) a professional consult using the above calculations.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-5

Introduction

J. Protein Binding objective:


1.
2.
3.
4.

Given population average patient data, the student will devise (V) dosage regimens which
will maintain plasma concentrations of unbound drug within the therapeutic range.
Given specific patient information, the patient will justify (VI) the optimal dosage regimen.
Given patient information regarding organ function, the student will devise (V) and justify
(VI) dosage regimens for the compromised patient.
The student will write (V) a professional consult using the above calculations.

K. Non-linear kinetics objective:


1.
2.
3.
4.

Given population average patient data, the student will devise (V) a dosage regimen which
will maintain plasma concentrations of drug within the therapeutic range.
Given specific patient information, the patient will justify (VI) the optimal dosage regimen.
Given patient information regarding organ function, the student will devise (V) and justify
(VI) dosage regimens for the compromised patient.
The student will write (V) a professional consult using the above calculations.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-6

Introduction

1.3 Exams
How are the exams
made?

1.3.1

Exams will consist of problems which will be linked directly back to an objective
(above) and a library assignment in which you will be asked to evaluate a research
article with the tools available to you by the time of the exam as discussed below.

LIBRARY ASSIGNMENT IN PHARMACOKINETICS


L. Library Assignment Objectives

What do I have to do in
the library?

1.
2.

Given a suitable primary research article in the area of pharmacokinetics, the student shall
calculate the pharmacokinetic parameters from the data using the tools learned in class.
The student shall communicate in writing the results of such calculations with suitable
commentary regarding differences and interpretations.

Format of the paper:


How should the paper
look?

1.

Tell me what type of paper you have chosen to evaluate:

The problem sets show what data you need for each of these.
First Exam
What content should I
look for in the paper and
what is its relative
worth?

IV Bolus Parent compound

10 pts

IV Bolus Parent metabolite

12.5 pts

IV Infusion

12.5 pts

Pharmacological Response

15 pts

Second Exam
Oral Dosing / Bioavailability

10 pts

Third Exam (2 credit course)


Multiple dosing

10 pts

Clearance and disease

12.5 pts

Dosage Regimen

15 pts

Third Exam (3 credit course)

2.

Two compartment model

10 pts

Protein Binding and Disease

12.5 pts

Non-linear kinetics and disease

15 pts

Include a Xerox copy of the entire paper. I need to evaluate it, too.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-7

Introduction

3.
Enlarge the graph by successive Xeroxes so that you can accurately evaluate the data.
4.

Do analysis of data by hand and by PKAnalyst, or Scientist.

5.

Compare your work with the authors (short paragraph).

6.
Comment on any differences of parameter calculation or interpretation. See
objectives above (Paragraph).
7.
Write an exam question to obtain pharmacokinetic parameters. You know
from the first exam what they should look like.
Why do I need to do this
library assignment?

Each of the above sections is designed to bring the student an understanding of the
information and the processes necessary to operate as a competent professional in
the area of pharmacokinetic evaluation and consulting. Consequently, the
course will evolve from a quantitative, manipulative mathematics course to a
course which stresses communication skills. Consults will be graded not only on
content (the proper dosage regimen for the patient) but also grammar, punctuation,
spelling, organization and neatness. You may have the best medical information in
the world, but if it is poorly executed, it will be ignored.

Can I cram the night


before?

This course will probably be one of the more rigorous ones that you will have
experienced in your college career to date. It will be one of the first ones which
attempt to show some clinical relevance. The course can be successfully completed with your current skills and background. It is not difficult IF (and that is a
big IF) taken slowly, in small bites. Its just like eating an elephant - you can't do it
all in one sitting. Some of you may try to get it all the night before the exam,
regardless of my admonitions and those of your upper-class friends (ask them!). In
many cases, that has been more than sufficient to get A's and B's on exams in previous courses. Past experience tells many of you that you can do it. I suggest that
the requirements and expectations of a professional school are considerably more
than your undergraduate experience and it most likely will not work in many
courses which require assimilation of the information presented, as is expected in a
professional program.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-8

Introduction

1.4 Blooms taxonomy for the Hierarchy of Educational Objectives


Blooms taxonomy for the Hierarchy of Educational Objectives describes the
expectations of a course in increasing order of complexity as:
What is cramming good
for? Lowest level of cognitive skills.

I.
To Know: means to memorize (recognize, recall) (Many college courses
require only this level of cognitive effort, hence the extensive experience with
multiple guess exams).
II.
To Comprehend: means to translate; to be able to put information into your
own words. (Essay exams routinely call for this level of effort on the part of the
student).

This is where we begin.

III.
To Apply: means to be able to use knowledge, rules and principles in an
unfamiliar situation. (This is the lowest level of skill necessary to function at a
technician level).

This is where we need


to be in school.

IV.
To Analyze: means to be able to critically examine a body of knowledge
and to be able to identify the relationships. (This is where a B.S. graduate should
operate. Education obviates the need for teachers.)

This is where we need


to be as graduates.

V.
To Synthesize: means to put together information, not necessarily previously so organized, in order to get a new piece of information. (This is the beginning level of professional judgment).
VI.
To Evaluate: means to be able to judge the worth of an idea, form hypotheses and do problem solving, research, invent new knowledge. (This is the doctoral
level of participation in the area).

Cant I just do it the


same way that I have
always studied?

A professional routinely operates at level IV and V with occasional forays into


level VI. This is where you will operate in this course and in most subsequent
courses in the professional curriculum. You will note that each of the objectives for
the course contains specific action words followed by the level in the taxonomy at
which you will operate. These are the standard descriptive terms for use in instructional objectives. You will be asked to do critical thinking, not simply recite or
recognize the right answer. Problems challenge thinking skills and demand the
synthesis of material into concepts. To facilitate this transition we both must work
very hard.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-9

Introduction

1.5 Course Contract


I Will:
What will the teacher do
in this class? Act as a
facilitator.

1.
Provide individualized learning methods: Some students learn by hearing
and others by seeing (auditory or visual learners). I have designed the course to
accommodate both types of learners. In class, I will provide you with executive
summaries of what you read. I will provide group leaders with detailed reviews of
materials for which they are responsible. I will tell you what I'm going to say, say
it, and then tell you what I said. I will also attempt to write it out and draw appropriate graphs, charts and pictures as well as appropriate visual aids in class and
with the homework problems. I will provide you with ample examples of the types
of manipulations that you will be expected to do. I will provide you with ample
problem sets so that you may practice those manipulations. I will provide you
with computer simulations so that you may see these manipulations in action and
begin to get a feel for the numbers and their magnitude. Feedback and interaction
is encouraged. If I am not meeting your perceived needs, you must tell me. Some
students might feel too intimidated to ask questions. To obviate this problem, you
will elect a group team leader, an ombudsman, whose job it will be to carry your
questions, concerns, and comments to me. It is your job and his responsibility to
see that the group interaction facilitates the learning process. This is not to prevent
you coming to see me but offered as another avenue of communication.

Will I learn anything relevant in this course?

2.
Provide clinical relevance to the practice of pharmacy. This will be stressed
at all times. I will also relate real clinical experiences; virtually all of the problems
come from real patients. Some educators believe examples must fit the theory
exactly. This gives the student a false set of reality parameters. Consequently,
when the data does not fall on the line the student rejects relevant information.
You will become familiar with real data, and the problems associated with real
data.

How will I know how Im


doing?

3.
Give adequate feedback: Evaluation of your performance will be available
to you at all times. A running evaluation, updated weekly will be on my door for
your review. You may check any thing with me at any time. I expect that you will
see me outside of class time either individually if you need help or in supervised
review sessions. You must see me for assistance if your performance is unsatisfactory.

What will the teacher be


doing? Engaging you in
an active learning process.

4.
Teach: As an operational definition this means: clarifying what you read,
demonstrating how and why things work as they do, and unifying the material attempting to generate the A - HA! syndrome. The correlate of teach from the student view is learn. Neither is a passive process. I can not open your head and pour
the knowledge in. A saying in education is: Knowledge maketh a bloody
entrance. You must expend the effort necessary for you to learn.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-10

Introduction

What must I do in this


active learning process?
You MUST participate in
class and in your
assigned groups!!!

5.
Facilitate Learning. You received objectives (above) and a summary for
each study section (chapters in this text), of exactly what is expected of you with
examples in the problem sets at the end of each chapter. We will have ample time
during class to field questions generated by the correlated reading and problem
sets, as well as homework assignments. I will not be duplicating any book's efforts.
Student participation in class is required. You will answer (as well as ask) questions, do problems in class. You will sound things out and get feedback from me
and your fellow colleagues. Remember - the class is to help you learn. It is not the
sole means of learning, nor am I the source of all knowledge. Its only reason for
being is to help you organize and summarize what you learn. It has a relatively
simple plan with multiple examples. From these examples you will develop concepts which will obviate the need for memorizing individual facts (or actually me
entirely). I will assist you in the formation of these concepts. It is patently obvious
that I can not give you every possible example of every type of question that you
will be asked during your professional career. For one thing I don't know what
questions you will be asked nor problems you will encounter. Going from the specific to the general forms concepts which will allow you to go from the general to
the specific, even if you have never been there before. The total medical knowledge is now doubling at a rate of every 4 years. I can not teach you the content necessary to operate 5 years in the future, let alone 40. You must learn to learn.
Hence, if you plan to become a competent professional, you must operate at least
in Bloom's level V.

How do I get in touch


with the teacher?

6.
Be available: I do not have office hours. I believe them to be restrictive
from your view point. What I do have is a schedule prepared two weeks in advance
of when I am NOT available. You may set an appointment, at least a 1/2 day in
advance to guarantee that I see it, any other time. Of course, appointments are not
necessary if I'm in my office, but you take the chance of my not being there or
someone else being there ahead of you if you do not sign up. You may contact me
by e-mail: makoid@creighton.edu, or by phone: 402-280-2952.

How can I tell the


teacher how things are
going?

7.
Be responsive: After each study section, you will be asked to provide me
with a one minute summary of the topic consisting answering the following questions:
a.

What was the main thrust of the study section?

b.

What was clear about the study section? What was done well?

c.

What was unclear? How could it be done better?

This will provide me with a running monitor of my effectiveness as well as a


framework of what to stress and what to change.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-11

Introduction

Do I have any say in the


examination questions?

After each exam, in addition to working out the problems, we will decide whether
any individual question was not covered by the objectives. Note: Not that it was
tough, not that you got it wrong, not that it didn't allow you to tell me what you
knew, but did I tell you that I was going to ask you to do it? (Was it covered by the
objectives?)

How are the exams


graded?

8) Evaluate your performance fairly and honestly: Quite simply, I'm going to tell
you what I expect that you will do. I will show you how to do it. I will provide you
with practice in doing it. I will provide you with an exam which tests your ability
to do it. The exams, as well as the whole course, will use real data and/or pharmacokinetic parameters for real drugs in real patient settings, much like the state
board exams (and hopefully real life). Like both of these situations, all answers are
interconnected. What that means is, if you improperly calculate a parameter which
is needed to make another calculation which is used to make a third, etc. ALL are
wrong. Conversely, if you can't get a particular calculation by one method or equation, try another. That's simply the way it is. You probably wouldn't get much sympathy if you calculated a dosage regimen properly based on a wrong elimination
rate constant and ended up killing your patient.
You Will:

What do I have to do?


How much work is really
expected?

1) Come prepared to participate in class. This is your full time job. If you are
working full time, it is usually 40 to 60 hours per week. If you go to college 15 to
18 credits and prepare/study 2 hours for each credit, you work 45 to 55 hour per
week - you have a full time job. Your commitment is the 45 to 50 hour week not
just the contact hours and a night for each exam. This specifically means for each 1
hour class, I expect no less than 2 hours of preparation on your part. Each of you
will be assigned to a study group. You will work the problems together and teach
each other both in and out of class. We will have group discussion of class as well
as group problem solving. It will be your responsibility that every member of
your group be adequately prepared to answer for the group during recitation. There
will be a grade for group participation. Part of your grade will be based on quizzes,
how well your group performed both on the material for which you were responsible as well as overall and your peer evaluation.

Do I have to read the


text?

2) Read the text. When you read, read critically. Do you understand each idea?
Place a <+> (in pencil) in the left side of each paragraph after you read and understand it. If you don't get it, place a <?> and come prepared to ask about it in class.

Why do I have to do the


problem sets?

3) Work the problems. Check the answers. These come from old exams, so they
are the type that you are likely to see. Work them in your study groups so that
everyone can see your thought processes. Bring them to class if you can not do
them or come and see me privately. Be prepared to show me your attempts at solving the problem. I will show you how to get started and give direction to your
thought. I will not work the problem for you. You would not learn if I did it for
you. It is crucial that you work the problems. Each has a specific objective. Over-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-12

Introduction

all, they contribute to your gaining facility in the processes that a pharmacokineticist must know how to do.
Can I just coast
through?

4) Do not delude yourself with respect to your performance. If you received a


grade that was less than satisfactory for you, do not simply console yourself by
saying I knew the stuff, I just made a little error. Can you get it right consistently? That's when you know the stuff. That is not a laudable goal. That's what a
professional does. There have been several students in the past that knew that
stuff right up till the time that they had to repeat the course (and sometimes
beyond).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-13

Introduction

1.6 Computers in the course


Can I get through with
just paper and pencil?

Computer literacy is necessary in this field. Consequently computers will pervade


the course. The homework problems (above) are to done both by hand and
checked with the computer. This will help your understanding of pharmacokinetics
in general and that homework objective in particular. Computers are natural
adjunctive tools in the teaching of pharmacokinetics. The are able to simulate the
concentration vs. time profiles and do difficult repetitive calculations which allow
the student to get a broad view of the processes involved.

What are the programs


that I will be using?

Programs that are currently being used in the course are The Scientist and PKAnalyst both from MicroMath Scientific Software, P.O. Box 71550, Salt Lake City, UT
84171-0550; or http://www.micromath.com. A working student version of the software is available free for the downloading for your own work at home. A full
working version is on the Pharmacy Server.
In addition to the above course objectives, there are specific objectives for the use
of computers in the course. They are:

What will I be expected


to do with the computers?

1.
Simulation. The student will construct a graph of the drug time course
using classical pharmacokinetics. The student will demonstrate effects of changes
in pharmacokinetic parameters on the ADME processes and correlated pharmacological / therapeutic response.
2.
The student shall statistically evaluate models with regard to fit of data
using both linear and nonlinear regression analysis. The student will calculate
pharmacokinetic parameters which best describe the processes of ADME.

How can I use the computers in the homework


and library questions?

These objectives will be met in a variety of ways. Clearly, the most direct method
is the solution of the problem sets by computer. First, I expect that you would do
the problem by hand, complete with graphs and other supporting calculations followed by computer simulation and data analysis. Just how close did you come to
the best fit? Next, a portion of each exam will be a library exercise in which you
will find and evaluate a published article according to the principles that you
learned in class utilizing the computer facilities. How close did you come the
authors numbers? Do you, in fact, even agree with the authors? You will prepare a
short consult in which you describe the patient and what the authors did along with
your support (or non-support) of the authors conclusions.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-14

Introduction

1.7 Survival Kit


1.7.1

THINGS FOR YOUR SURVIVAL KIT!

What do I need to buy


RIGHT NOW?

1.
You will need a good calculator - One with
have it ASAP. You will use it in class.

and

ln x

functions. You must

2.
You will also need 2 cycle semilog paper and a clear straight edge ruler for
use in class. These are available in the book store or at an office supply store.
3.
You will need access to a computer (486 DX or higher). Micromath has
made available a student version of the programs for a nominal fee (This software
is pre-loaded in the Criss computer lab; you may purchase a copy for home use)
4.
You will need a 3 D three-ring binder for collecting and maintaining all
the pages in this book as well as your class notes.
What do I need to do in
and out of class?

Work in your study groups. You never learn it so well as when you teach it to
someone else. Everyone benefits from a well run prepared study group. You are
not in competition with your fellow classmates. If everyone earns an A, then
everyone will receive one.

How can I organize this


material?

Organize and label your study notes. This is basic survival. This is one strategy
that I find works well. I recommend it highly. Good study notes are formatted on
loose-leaf in a three ring binder. The individual pages have a line drawn down
about 1/3 the way in. The notes are taken on the right (2/3) of the page, while
labels go in the left. The labels on the left are often written as questions, which are
answered in the text on the right. Loose leaf binders allow for the incorporation of
reading summaries as well as relevant problems and homework to be organized
with a divider all in one place. You should write intelligently, with proper punctuation and spelling as if you were preparing a consult for a physician. Organization is
the key.
Remember: you may have all the information in the world at your fingertips; be
able to solve the most difficult therapy problem and no one will listen to you if you
can't communicate intelligently. Chapters in the book will be organized as above.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-15

Introduction

1.7.2

WHAT YOU WILL GAIN: (YOUR GOALS)

How is the course to be


graded?

1.
At the lowest level, a decent grade for a significant course. Specific grades
will be earned by attaining the following averages:

A 90 and above
B+ 85 to 89
B 80 to 84
C+ 75 to 79
C 70 to 74
D 60 to 69
F 59 and below

The number of exams and point distribution will be determined in class.


2.
At the next higher level, I will guarantee that if you comprehend this material at level V, you will have no trouble passing any state board anywhere with
regard to pharmacokinetics.
3.
You will gain a useful skill that will make you an integral part of the health
care team.
Do I really need a
teacher to learn?

4
You will learn to learn. There is an old proverb which goes: Give a man a
fish and you feed him for a day. Teach a man to fish and you feed him for a lifetime. The B.S. Degree is designed to eliminate teachers. An educated man is one
who has learned to how to learn, not one who memorized a page in a book. That
is what you need to be a professional. The total medical knowledge is doubling at a
rate of every 3-4 years. That means that you will be out of date shortly after graduation (if not before) if you simply memorized content and don't learn to learn and
continue to learn throughout your career.

What about cheating?

One last piece of information: Neither you nor I will not tolerate any academic
misconduct. Anyone caught will minimally receive an F for their efforts and I
will recommend dismissal from the program. The profession has no room for
unprofessional behavior. I will prosecute.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-16

Introduction

1.8 Tentative Schedule


1.8.1

STUDY GROUP 1: LEARN THE TOOLS - OBTAIN PHARMACOKINETIC


PARAMETERS FROM DATA.
A: Introduction
1. Texts
2. Literature
3. Grading Policy
4. Course Philosophy
B: Math review
1. Numbers and exponents
2. Graphs and reaction order
3. Calculus
4. Laplace transform
5. Computer Introduction
6. Computer simulation and problem sets
C: Pharmacokinetic modeling
1. What a model is and what it isn't.
2. Why we model
3. Philosophy of modeling
D: Pharmacological Response
1. Michaelis - Menton Mass balance equation
2. Interrelationships between Concentration, time and response.
E: I.V. Bolus dosing
1. Parent compound
I. Plasma
a. Plasma concentration vs. time profile analysis
b. Computer aided instruction and simulation
c. Problem sets
d. Professional communication.
II Urine
a. Amount vs. time profile analysis
b. Rate vs. time profile analysis
c. Computer aided instruction and simulation
d. Problem sets
e. Professional communication.
2. Metabolite

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-17

Introduction

I. Plasma
a. Plasma concentration vs. time profile analysis
b. Computer aided instruction and simulation
c. Problem sets
d. Professional communication.
II. Urine
a. Rate vs. time profile analysis
b. Computer aided instruction and simulation
c. Problem sets
d. Professional communication.
F: I.V. infusion
1. Parent compound
I. Plasma
a. Plasma concentration vs. time profile analysis
b. Computer aided instruction and simulation
c. Problem sets
d. Professional communication.
II. Urine
a. Amount vs. time profile analysis
b. Rate vs. time profile analysis
c. Computer aided instruction and simulation
d. Problem sets
e. Professional communication.
1. Metabolite
I. Plasma
a. Plasma concentration vs. time profile analysis
b. Computer aided instruction and simulation
c. Problem sets
d. Professional communication.
II. Urine
a. Amount vs. time profile analysis
b. Rate vs. time profile analysis
c. Computer aided instruction and simulation
d. Problem sets
e. Professional communication.
End of Material for first exam (six weeks for semester, two weeks for summer session)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-18

Introduction

1.8.2

STUDY GROUP 2: LEARN HOW THE PARAMETERS ARE MODIFIED.


G: Biopharmaceutical factors
1. Absorption
I. Physiology
II. Mechanisms
III. Physiological changes with age, sex, disease
2. Distribution
I. Diffusion
II. Cardiac output / blood perfusion
III. Physical properties of the drug
IV. Physical properties of the body
V. Physiological changes with age, sex, disease
3. Metabolism
I. Biotransformation methods
II. First pass effect
III. Clearance
IV. Physiological changes with age, sex, disease
4. Excretion
I. Renal
II. Biliary
III. Mammary
IV. Salivary
V. Misc.
VI. Physiological changes with age, sex, disease
H: Oral dosing
1. Parent compound
I. Plasma
a. Plasma concentration vs. time profile analysis
b. Computer aided instruction and simulation
c. Problem sets
d. Professional communication.
II. Urine
a. Amount vs. time profile analysis
b. Rate vs. time profile analysis
c. Computer aided instruction and simulation
d. Problem sets
e. Professional communication.
2. Metabolite
I. Plasma
a. Plasma concentration vs. time profile analysis
b. Computer aided instruction and simulation

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-19

Introduction

c. Problem sets
d. Professional communication.
II. Urine
a. Amount vs. time profile analysis
b. Rate vs. time profile analysis
c. Computer aided instruction and simulation
d. Problem sets
e. Professional communication.
I: Bioavailability, Bioequivalence, Drug product selection
1. Relative and Absolute Bioavailability
2. Factors Influencing Bioavailability
3. Methods of Assessing Bioavailability
I. in vivo
II. in vitro
III. Correlation
4. Bioequivalence
5. Bioavailability
6. Drug Product Selection

1.8.3

STUDY GROUP 3: APPLY THE TOOLS IN COMPROMISED PATIENTS.


J: Dosage regimen (Healthy, aged and diseased patients)
1. Multiple dose kinetics
2. Optimization of dosage regimen
3. Computer aided instruction
4. Computer simulation and problem sets
5. Computer aided consultation
6. Professional consultation process
End of Material for two credit course (six weeks semester - 2 weeks summer)

1.8.4

STUDY GROUP 4: APPLY THE TOOLS IN SPECIAL CASES.


K: Multicompartment Modeling
1. Parent compound plasma vs. time profile analysis
2. Multiple dose considerations
3. Computer aided instruction
4. Computer simulation and problem sets
5. Computer aided consultation
6. Professional consultation process
L: Protein Binding (healthy, aged and diseased patients)
1. Mass balance considerations / drug interactions
2. Effects of protein binding on pharmacokinetic parameters

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-20

Introduction

3. Computer aided instruction


4. Computer simulation and problem sets
5. Computer aided consultation
6. Professional consultation process
M: Non - linear (Michaelis - Menton) kinetics
1. Computer aided instruction
2. Computer simulation and problem sets
3. Computer aided consultation
4. Professional consultation process
End of material for three credit course (4 weeks semester, 2 weeks summer)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-21

Introduction

1.9 Competency Statements Related To Pharmacokinetics


The profession of pharmacy has determined that there are minimum, entry level
abilities necessary for a pharmacist. These form the Standards of Practice for the
profession of pharmacy, as written by The National Association of Boards of Pharmacy (who make the NAPLEX, coincidentally). It is important to note that these
abilities are not thought up by some faculty member who sits in his ivory tower
saying what he thinks is important. These are what pharmacists do. They have
been promulgated as competency statements They are also the basis for the state
board exams as well as the basis far your coursework while in the School of Pharmacy. They are broken down into three general areas:
Area 1: Manage Drug Therapy to Optimize Patient Outcomes (Approximately
50% of Test)
Area 2: Assure the Safe and Accurate Preparation and Dispensing of Medications
(Approximately 25% of Test)
Area 3: Provide Drug Information and Promote Public Health (Approximately
25% of Test)
For a complete listing of competency statements please refer to the National Association of Boards of Pharmacys web site, www.nabp.net

1.9.1

SPECIFIC COMPETENCY STATEMENTS ADDRESSED IN THIS


COURSE
Area 1: Manage Drug Therapy to Optimize Patient Outcomes (Approximately 50% of Test)
1.1.0 Evaluate the patient and/or patient information to determine the presence of a
disease or medical condition, to determine the need for treatment and/or referral,
and to identify patient-specific factors that affect health, pharmacotherapy, and/or
disease management.
1.1.1 Identify and/or use instruments and techniques related to patient assessment
and diagnosis.
1.1.2 Identify and define the terminology, signs, and symptoms associated with
diseases and medical conditions.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-22

Introduction

1.1.4 Identify patient factors, biosocial factors, and concurrent drug therapy that
are relevant to the maintenance of wellness and the prevention or treatment of a
disease or medical condition.
1.2.0 Assure the appropriateness of the patient's specific pharmacotherapeutic
agents, dosing regimens, dosage forms, routes of administration, and delivery systems.
1.2.1 Identify drug products by their generic, trade, and/or common names.
1.2.3 Evaluate drug therapy for the presence of pharmacotherapeutic duplications
and interactions.
1.2.5 Identify physicochemical properties of drug substances that affect their solubility, pharmacokinetics, pharmacologic actions, and stability.
1.2.6 Interpret and apply pharmacokinetic principles to calculate and determine
appropriate drug dosing regimens.
1.2.7 Interpret and apply biopharmaceutic principles and the pharmaceutical characteristics of drug dosage forms and delivery systems, to assure bioavailability and
enhance patient compliance.
1.3.0 Monitor the patient and/or patient information and manage the drug regimen
to promote health and assure safe and effective pharmacotherapy.
1.3.2 Evaluate patient information to determine the safety and effectiveness of
pharmacotherapy.
1.3.5 Identify and remedy interactions or contraindications with diagnostic or
monitoring tests or procedures.
Area 2: Assure the Safe and Accurate Preparation and Dispensing of Medications (Approximately 25% of Test)
2.1.0 Perform calculations required to compound, dispense, and administer medication.
2.1.1 Calculate the quantity of medication to be compounded or dispensed; reduce
and enlarge formulation quantities and calculate the quantity or ingredients needed
to compound the proper amount of the preparation.
2.1.3 Calculate the rate of drug administration.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-23

Introduction

2.1.4 Calculate or convert drug concentrations, ratio strengths, and/or extent of


ionization.
2.2.0 Select and dispense medications.
2.2.3 Interpret and apply pharmacokinetic parameters and quality assurance data to
determine equivalence among manufactured drug products, and identify products
for which documented evidence of inequivalence exists.
2.2.5 Identify and describe the use of equipment and apparatus required to administer medications.
Area 3: Provide Drug Information and Promote Public Health (Approximately 25% of Test)
3.1.0 Access, evaluate, and apply information to promote optimal health care.
3.1.1 Identify the typical content and organization of specific sources of drug and
health information.
3.1.2 Interpret and evaluate data presented in textual, tabular, or graphic form.
3.1.3 Evaluate the suitability, accuracy, and reliability of information from reference sources by explaining and evaluating the adequacy of experimental design
and by applying and evaluating statistical tests and parameters.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-24

Introduction

1.10 Pharmacokinetic Symbolism


Pharmacokinetics was developed in several locations simultaneously. Because of
this, the symbols used in the literature are not consistent. Provided each symbol is
rigorously defined prior to use, this inconsistency should not prove an insurmountable difficulty when assessing the literature. In this book, the symbolism below
will be generally used, though, as an illustration of the variety, some deviation may
be anticipated on occasions.

1.10.1

AMOUNT TERMS (UNIT: MASS)


ARE

amount remaining to be eliminated (excreted)

dose (or maintenance dose)

DL

loading dose

Xa

amount of drug remaining to be absorbed at any time

amount of unchanged drug in body at any time

Xm

amount of metabolite in body at any time

Xu

cumulative amount of unchanged drug excreted into urine up to any time

X mu

cumulative amount of metabolite excreted into urine up to any time

X max maximum amount of unchanged drug in body


X min

minimum amount of unchanged drug in body

average amount of unchanged drug in body (also Laplace transform)

X eff minimum amount of unchanged drug in body necessary for pharmacological response

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-25

Introduction

1.10.2

CONCENTRATION TERMS (UNITS MASS/VOLUME)


Cb

concentration of drug in blood at any time

Cp

concentration of drug in plasma at any time

Cm

Concentration of metabolite in plasma (or blood) at any time

( C p )ss average steady-state concentration of drug in plasma during a dosing


interval
( C p )max maximum concentration of drug in plasma
( C p )min minimum concentration of drug in plasma
Cp

average concentration of drug in plasma

KA

dissociation constant of drug-protein complex

KM

Michaelis-Menton rate constant

KR

dissociation constant of drug-receptor complex

MEC minimum effective concentration of drug or metabolite


MTC minimum toxic concentration of drug or metabolite

1.10.3

VOLUME TERMS (UNIT: VOLUME)


Vd

apparent volume of unchanged drug distribution in compartment

Vm

apparent volume of metabolite distribution in compartment

Vw

physiological volume of plasma water

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-26

Introduction

1.10.4

1.10.5

TIME TERMS (UNIT: TIME)


t

time since administration of dose

duration of zero-order input

t'

time since cessation of zero-order input

t0

lag time

mean time during sampling interval

elimination half-life (biological half-life)

t 0.5

time for 50% removal

t max

time when maximum amount or concentration occurs

t dur

duration of effective pharmacological response

dosing interval (greek theta)

time variable used in association with zero-order input

RATE CONSTANT TERMS (UNIT: RECIPROCAL TIME (*), MASS/TIME


(**)
K, k e ,K d, K i apparent first-order rate constant for elimination, Summation of all
the ways the drug is eliminated (*)
ka

apparent first-order rate constant for absorption (*)

k u, k r apparent first-order rate constant for urinary (renal) excretion of unchanged


drug (*)
km

apparent first-order rate constant for metabolism of unchanged drug (*)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-27

Introduction

1.10.6

1.10.7

k mu

apparent first-order rate constant for excretion of metabolite (*)

k ij

apparent first-order transfer rate constant (*)

k0

zero-order input rate constant (**)

zero-order infusion rate constant (**)

rate constant for decline in pharmacological effect (usual units:%/time)

hybrid first-order rate constant (*) (greek alpha)

hybrid first-order rate constant (*) (greek beta)

CLEARANCE TERMS (UNITS: VOLUME/TIME)


Cl

total body clearance (TBC)

Cl r

renal clearance (RC)

Cl m

metabolic clearance (MC)

Cl cr

creatinine clearance

Cl H

hepatic clearance (HC)

RATE TERMS (UNITS: MASS/TIME (*), MASS/TIME, VOLUME (**),


VOLUME/TIME (***)
dX
------dt

instantaneous rate of change of amount of unchanged drug (*)

X
---t

measured rate of change of amount of unchanged drug (*)

RH

rate of plasma flow through the liver (***)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-28

Introduction

1.10.8

Rr

rate of plasma flow through the kidney (***)

VM

theoretical maximum rate of a process (**)

OTHER TERMS
AUC area under the plasma concentration-time curve (units: time * mass/volume)
AUMC area under the first moment of the plasma concentration-time curve (units:
2

time mass volume

MRT Mean Residence Time (units:

time )

MAT Mean Absorption Time (units:

time )

MDT Mean Dissolution Time (units:

time )

intensity of pharmacological effect

EH

steady-state hepatic extraction ratio

Er

steady-state renal extraction ratio

E max maximum intensity of pharmacological effect


F

fraction of administered dose ultimately absorbed

FRE

fraction remaining to be eliminated (excreted)

hematocrit (fractional volume of erythrocytes in whole blood)

number of elimination half-lives in a dosing interval

accumulation factor

intercept

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-29

Introduction

1.10.9

fraction of drug that is free (unbound)

f ss

fraction of steady-state

slope (sometimes specifically for log dose-response plot)

number of doses

Laplace operator

[ ]

indicates molar concentration

SUBSCRIPTS
0

at time zero

at time infinity

ss

during steady-state

at time t

at time T

following dose n

diff

difference between extrapolated and observed

int

intrinsic

index (i.e., 1,2,3)

index (i.e., 1,2,3)

1.10.10 SUPERSCRIPTS
x

extrapolated

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-30

Introduction

last measured value

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-31

Introduction

1.11 First Lesson in Pharmacokinetics


It should be intuitively obvious to the most casual observer that the relative bioavailability of 2
simultaneous I.V. bolus doses of a drug is equal to the following:
(EQ 1-1)

1 1
1 1
2
2
1
ln lim ( [ x ] [ x ] )! + --- + ( sin q ) + ( cos q ) =

cosh p 1 ( tanh p )

------------------------------------------------n
2

(EQ 1-2)

n=0

given that 100% bioavailability of a single I.V. bolus dose is equal to 1, and both
doses contain an equal mass of active drug.
For the struggling pharmacokinetics student, we would like to show the veracity of
this statement. Of course, it is obvious that; the reverse of the transpose is equal to
the transpose of the inverse in matrix theory. i.e.:
1 1

[x ]

1 1

= [x ]

(EQ 1-3)

Also, it should be obvious that:


0! = 1

(EQ 1-4)

Consequently,
1 1

([x ]

1 1

[ x ] )! = 1

(EQ 1-5)

which means that:

2
2
1
ln lim 1 + --- + ( sin q ) + ( cos q ) =

cosh p 1 ( tanh p ) ------------------------------------------------n


2
n=0

(EQ 1-6)

By definition,

e = 1 + 1---

(EQ 1-7)

and
1 = cosh p 1 ( tanh p )

(EQ 1-8)

Thus:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-32

Introduction

----n-

ln e + ( sin q ) + ( cos q ) =

(EQ 1-9)

n = 02

Also,

2 =

----n1

(EQ 1-10)

n = 02

and
1 = ln e

(EQ 1-11)

and
2

1 = ( sin q ) + ( cos q )

(EQ 1-12)

So, as we observed in equation 1,


1+1 = 2

(EQ 1-13)

under the stated conditions, two I.V. bolus doses given simultaneously will have
twice as much available drug as a single I.V. bolus dose.
You will agree, however, equation 1-1 is obvious and therefore is more easily
understood by a pharmacokineticist!

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1-33

CHAPTER 2

Mathematics Review

Author: Michael Makoid, Phillip Vuchetich and John Cobby


Reviewer: Phillip Vuchetich

BASIC MATHEMATICAL SKILLS OBJECTIVES


1.

Given a data set containing a pair of variables, the student will properly construct
(III) various graphs of the data.

2.

Given various graphical representations of data, the student will calculate (III) the
slope and intercept by hand as well as using linear regression.

3.

The student shall be able to interpret (V) the meaning of the slope and intercept
for the various types of data sets.

4.

The student shall demonstrate (III) the proper procedures of mathematical and
algebraic manipulations.

5.

The student shall demonstrate (III) the proper calculus procedures of integration
and differentiation.

6.

The student shall demonstrate (III) the proper use of computers in graphical simulations and problem solving.

7.

Given information regarding the drug and the pharmacokinetic assumptions for
the model, the student will construct (III) models and develop (V) equations of the
ADME processes using LaPlace Transforms.

8.

The student will interpret (IV) a given model mathematically.

9.

The student will predict (IV) changes in the final result based on changes in variables throughout the model.

10.

The student will correlate (V) the graphs of the data with the equations and models so generated.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-1

Mathematics Review

2.1 Concepts of Mathematics


Pharmacokinetcs is a challenging field involving the application of mathematical
concepts to real situations involving the absorbtion, distribution, metabolism and
excretion of drugs in the body. In order to be successful with pharmacokinetics, a
certain amount of mathematical knowledge is essential.
This is just a review.
Look it over. You should
be able to do all of these
manipulations.

This chapter is meant to review the concepts in mathematics essential for understanding kinetics. These concepts are generally taught in other mathematical
courses from algebra through calculus. For this reason, this chapter is presented as
a review rather than new material. For a more thorough discussion of any particular concept, refer to a college algebra or calculus text.
Included in this section are discussions of algebraic concepts, integration/differentiation, graphical analysis, linear regression, non-linear regression and the LaPlace
transform. The Scientist and PKAnalyst are the computer programs used in this
course.

Something new LaPlace transforms.


Useful tool.

A critical concept introduced in this chapter is the LaPlace transform. The LaPlace
transform is used to quickly solve (integrate) ordinary, linear differential equations. The Scientist by Micromath Scientific Software, Inc.1 is available for working with the LaPlace transform for problems throughout the book.

1. MicroMath Scientific Software, Inc., P.O. Box 21550, Salt Lake City, UT 84121-0550,
http://www.micromath.com

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-2

Mathematics Review

2.2 Mathematical Preparation


2.2.1

ZERO AND INFINITY


Any number multiplied by zero equals zero. Any number multiplied by infinity
( ) equals infinity. Any number divided by zero is mathematically undefined.
Any number divided by infinity is mathematically undefined.

2.2.2

EXPRESSING LARGE AND SMALL NUMBERS


Large or small numbers can be expressed in a more compact way using indices.

How Does Scientific


Notation Work?

316000 becomes 3.16 10

Examples:

0.00708 becomes 7.08 10


In general a number takes the form:
A 10

Where A is a value between 1 and 10, and n is a positive or negative integer


The value of the integer n is the number of places that the decimal point must be
moved to place it immediately to the right of the first non-zero digit. If the decimal
point has to be moved to its left then n is a positive integer; if to its right, n is a
negative integer.
Because this notation (sometimes referred to as Scientific Notation) uses indices, mathematical operations performed on numbers expressed in this way are subject to all the rules of indices; for these rules see Section 2.2.4.
A shorthand notation (AEn) may be used, especially in scientific papers. This may
n

be interpreted as A 10 , as in the following example:


4

2.28E4 = 2.28 10 = 22800

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-3

Mathematics Review

2.2.3

SIGNIFICANT FIGURES
A significant figure is any digit used to represent a magnitude or quantity in the
place in which it stands. The digit may be zero (0) or any digit between 1 and 9.
For example:
TABLE 2-1. Significant

Value

Figures

Significant
Figures

Number of
Significant
Figures

(a)

572

2,5,7

(b)

37.10

0,1,3,7

(c)

10.65 x 104

0,1,6,5

(d)

0.693

3,6,9

(e)

0.0025

2,5

How do I determine the


number of significant
figures?

Examples (c) to (e) illustrate the exceptions to the above general rule. The value 10
raised to any power, as in example (c), does not contain any significant figures;
hence in the example the four significant figures arise only from the 10.65. If one
or more zeros immediately follow a decimal point, as in example (e), these zeros
simply serve to locate the decimal point and are therefore not significant figures.
The use of a single zero preceding the decimal point, as in examples (d) and (e), is
a commendable practice which also serves to locate the decimal point; this zero is
therefore not a significant figure.

What do significant figures mean?

Significant figures are used to indicate the precision of a value. For instance, a
value recorded to three significant figures (e.g., 0.0602) implies that one can reliably predict the value to 1 part in 999. This means that values of 0.0601, 0.0602,
and 0.0603 are measurably different. If these three values cannot be distinguished,
they should all be recorded to only two significant figures (0.060), a precision of 1
part in 99.
After performing calculations, always round off your result to the number of significant figures that fairly represent its precision. Stating the result to more significant figures than you can justify is misleading, at the very least!

2.2.4

RULES OF INDICES

What is an index?

An index is the power to which a number is raised.


n

Example: A where A is a number, which may be positive or negative, and n is


the index, which may be positive or negative.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-4

Mathematics Review

Sometimes n is referred to as the exponent, giving rise to the general term, Rules
of Exponents. There are three general rules which apply when indices are used.
(a) Multiplication
n

A A = A
A B

n+m

n
n+m
= A
--- B
B

(b) Division
n

A - = An m
-----m
A
n

A - = A
n Bn m
------- B
m
B
(c) Raising to a Power
n m

(A )

= A

nm

There are three noteworthy relationships involving indices:


(i) Negative Index
A

1- As n tends to infinity ( n ) then A n 0 .


= ----n
A

(ii) Fractional Index


1
--n

A =

(iii) Zero Index


0

A = 1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-5

Mathematics Review

2.2.5

LOGARITHMS

What is a logarithm?

Some bodily processes, such as the glomerular filtration of drugs by the kidney,
are logarithmic in nature. Logarithms are simply a way of succinctly expressing a
number in scientific notation.
In general terms, if a number (A) is given by
A = 10

then
log ( A ) = n
where log signifies a logarithm to the base 10, and n is the value of the logarithm
of (A).
5

Example: 713000 becomes 7.13 10 ,


and 7.13 = 10

0.85

, thus 713000 becomes 10

0.85

10 = 10

( 5 + 0.85 )

= 10

5.85

and log ( 713000 ) = 5.85


Logarithms to the base 10 are known as Common Logarithms. The transformation
of a number (A) to its logarithm (n) is usually made from tables, or on a scientific
calculator; the reverse transformation of a logarithm to a number is made using
anti-logarithmic tables, or on a calculator.
What is the characteristic? the mantissa?

2.2.6

The number before the decimal point is called the characteristic and tells the placement of the decimal point (to the right if positive and to the left if negative). The
number after the decimal is the mantissa and is the logarithm of the string of numbers discounting the decimal place.

NATURAL LOGARITHMS

What is a natural logarithm?

Instead of using 10 as a basis for logarithms, a natural base (e) is used. This natural
base is a fundamental property of any process, such as the glomerular filtration of a
drug, which proceeds at a rate controlled by the quantity of material yet to undergo
the process, such as drug in the blood. To eight significant figures, the value of the
transcendental function, e, is
e = 2.7182818 ....

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-6

Mathematics Review

Strictly speaking,

e = 1+

---x!
1

x=1

Where x is an integer ranging from 1 to infinity ( ) ,

denotes the summation from x = 1 to x = , and

x=1

! is the factorial (e.g., 6! = 6x5x4x3x2x1= 720)


n

In general terms, if a number (A) is given by A = e , then by definition,


ln ( A ) = n
Where, ln signifies the natural logarithm to the base e , and n is the value of the
natural logarithm of A .
Natural logarithms are sometimes known as Hyperbolic or Naperian Logarithms;
again tables are available and scientific calculators can do this automatically. The
anti-logarithm of a natural logarithm may be found from exponential tables, which
n

give the value of e for various values of n.


How are natural logarithms ln x and common
logarithms log x related?

Common and natural logarithms are related as follows:


ln ( A ) = 2.303 log ( A ) , and
log ( A ) = 0.4343 ln ( A )
Because logarithms are, in reality, indices of either 10 or e , their use and manipulation follow the rules of indices (See Section 2.2.4).
(a) Multiplication:
n

To multiply N M , where N = e and M = e ; NM = e e

= e

n+m

By definition,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-7

Mathematics Review

ln ( NM ) = n + m ;
but
n = ln ( N )
and
m = ln ( M ) ,
hence
ln ( NM ) = ln ( N ) + ln ( M )
Thus, to multiply two numbers (N and M) we take the natural logarithms of each,
add them together, and then take the anti-logarithm (the exponent, in this case) of
the sum.
(b) Division
N- = ln ( N ) ln ( M )
ln ---M
(c) Number Raised to a Power
m

ln ( N ) = m ln ( N )
There are three noteworthy relationships involving logarithms:
(i) Number Raised to a Negative Power
ln ( N

1-
) = m ln ( N ) = m ln --N

As m tends to infinity ( m ) , then ln ( N

(ii) Number Raised to a Fractional Power

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-8

Mathematics Review

ln ( m

--m
1
N ) = ln N = ---- ln ( N )
m

(iii) Logarithm of Unity


ln ( 1 ) = log ( 1 ) = 0

2.2.7

NEGATIVE LOGARITHMS
The number 0.00713 may be expressed as:
3

7.13 10 , or
10
10

0.85

10 , or

2.15

Hence, log ( 0.00713 ) = 2.15 , which is the result generated by most calculators.
However, another representation of a negative logarithm (generally used by referencing a log table):
log (0.00713) = 3.85
The 3 prior to the decimal point is known as the characteristic of the logarithm; it
can be negative (as in this case) or positive, but is never found in logarithmic
tables. The .85 following the decimal point is known as the mantissa of the logarithm; it is always positive, and is found in logarithmic tables.
In fact 3 is a symbolic way of writing minus 3 (-3) for the characteristic. In every
case the algebraic sum of the characteristic and the mantissa gives the correct
value for the logarithm.
Example: log (0.00713) = 3.85
Add -3 and 0.85
Result is -2.15, which is the value of log ( 0.00713 )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-9

Mathematics Review

The reason for this symbolism is that only positive mantissa can be read from antilogarithmic tables, and hence a positive mantissa must be the end result of any logarithmic manipulations. Note that while there are negative logarithms (when N <
1), they do not indicate that number itself is negative; the sign of a number (e.g., N) is determined only by inspection following the taking of anti-logarithms.

2.2.8

USING LOGARITHMIC AND ANTI-LOGARITHMIC TABLES


Though the preferred method to using logarithms is with a calculator or computer,
the understanding of how the number is being manipulated may be important in
understanding the use of logarithms. (See the end of this chapter for Logarithm
tables).
(a) Find the log of (62.54)
1

62.54 = 6.254 10

Look up the mantissa for 6254 in a table of logarithms: it is 7962.


1

Hence, 6.254 10 = 10

0.7962

10 = 10

1.7962

and log ( 62.54 ) = 1.7962

(b) Find the log of (0.00329)


3

0.00329 = 3.29 10

The mantissa for 329 is 5172 Hence, log(0.00329) = 3.5172.


Note that in both examples the value of the characteristic is the integer power to
which 10 is raised when the number is written in scientific notation.
How do I multiply using
logarithms?

(c) Multiply 62.54 by 0.00329


log (62.54) = 1.7972
log (0.00329) = 3.5172
log (62.54 + log (0.00329) = 1.7962+3.5172 = 1.7962-3+ 0.5172=-0.6866
0.6866=1.3134

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-10

Mathematics Review

(d) We wish to find anti-log (1.3134) Look up the anti-log for the 0.3134 (mantissa) in a table: it is 2058.
1

Antilog (1.3134) = 2.058 10

Hence, antilog (1.3134) = 0.2058


How do I divide using
logarithms?

log (62.54) - log (0.00329) = 1.796 - 3.5172=1.796 +3 - 0.5172=4.2788


antilog 4.2788 = 19002

2.2.9

DIMENSIONS

What is a unit?

There are three fundamental dimensions which are used in various combinations to
express the properties of matter. Each of these dimensions has been assigned a definite basic unit, which acts as a reference standard.
TABLE 2-2 Dimensions

Dimension

Dimensional
Symbol

Unit

Unit Symbol

Length

meter

Mass

gram

Time

second

sec

How are units made bigger and smaller?

In the metric system, which emerged from the French Revolution around 1799,
there are various prefixes which precede the basic units and any derived units. The
prefixes indicate the factor by which the unit is multiplied. When the index of the
factor is positive the prefixes are Greek and have hard, consonant sounds. In contrast, when the index is negative, the prefixes are Latin and have soft, liquid
sounds. (see Table 2-3).

How big is big?

Examples: An average adult male patient is assumed to have a mass of 70 kilograms (70 kg). An average adult male patient is assumed to have a height of 180
centimeters (180 cm). A newly minted nickel has a mass of 5.000 g. Doses of
drugs are in the mg (10-3 g) range (occasionally g) never Kg (103 g) or larger. Students have told me that the dose that they have calculated for their patient is 108 g
(converting to common system - ~ 100 tons). I doubt it. Get familiar with this system. Note that the plural of Kg or cm is Kg or cm; do not add an s. In pharmacy
there are two derived units which are commonly used, even though they are
related to basic units. The Liter (L) is the volume measurement and is a cube 10
cm on a side (1L = (10cm)3 = 1000 cm3 ) while the concentration measurement
and has the units of Mass per Volume.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-11

Mathematics Review

Why should I use units?

Whenever the magnitude of a measured property is stated, it is imperative to state


the units of the measurement. Numbers are useless by themselves.
Example: The procainamide concentration range is 4-8 g/ml; stating the range
without units may lead to a potentially lethal error in which procainamide is
administered in a sufficient dose to attain a range of 4-8 mg/ml, which is 1000
times too large and would give rise to cardiac arrest.

TABLE 2-3 Scale

Name

Symbol

exa-

peta-

tera-

giga-

mega-

kilo-

hecto-

deca-

da

of Metric system and SI


Multiplication Factor
10
10
10

Name

Symbol

18

deci-

15

centi-

12

milli-

micro-

nano-

pico-

femto-

atto-

10
10
10
10
10

Multiplication Factor
10
10
10
10
10
10
10
10

1
2
3
6
9

12
15
18

TABLE 2-4

2.2.10

Dimension

Dimensional
Symbol

Volume

liter

Concentration

grams/liter

g/l

Unit

Unit
Symbol

DIMENSIONAL ANALYSIS

How are units useful?

It is a general rule that the net dimensions (and units) on the two sides of any equation should be equal. If this is not so, the equation is necessarily meaningless.
Consider the following equation which defines the average concentration of a drug
FDin blood after many repeated doses, ( C b ) = ---------VK

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-12

Mathematics Review

Where:

F is the fraction of the administered dose ultimately absorbed (Dimensions: none),

D is the mass of the repeated dose (Dimension: M),


V is the apparent volume of distribution of the drug (Dimension: V = L )
K is the apparent first-order rate constant for drug elimination (Dimension: T

),

and is the dosing interval (Dimension: T )

Writing the dimensions relating to the properties of the right-hand side of the equation gives:
M
M
------------------------ = ----1
V
VT T
Thus ( C b ) has the dimensions of M
----- , which are correctly those of concentration.
V
Sometimes dimensional analysis can assist an investigator in proposing equations
which relate several properties one with the other. If the units cancel, and you end
up with the correct unit of measure, you probably did it right. If you obtain units
that do not make sense, its guaranteed sure that you did it wrong.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-13

Mathematics Review

2.3 Calculus
What is Calculus?

Calculus concerns either the rate of change of one property with another (differential calculus), such as the rate of change of drug concentrations in the blood with
time since administration, or the summation of infinitesimally small changes (integral calculus), such as the summation of changing drug concentrations to yield an
assessment of bioavailability. In this discussion a few general concepts will be provided, and it is suggested an understanding of graphical methods should precede
this discussion.

2.3.1

DIFFERENTIAL CALCULUS

2.3.2

NON-LINEAR GRAPHS
Consider the following relationship: y = x
TABLE 2-5 x,

y sample data

27

64

As can be seen from the graph (Figure 2-1), a non-linear plot is produced, as
expected.
FIGURE 0-1.

y=x3

70
60
50
40
30
20
10
0
1

(Question: How could the above data be modified to give a linear graph?)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-14

Mathematics Review

2.3.3

SLOPE OF NON-LINEAR GRAPH


As with a linear graph,
y2 y1
y
---------------- = -----x2 x1
x
Where y is the incremental change in y and x is the incremental change in x
But, as can be seen (Figure 1), the slope is not constant over the range of the graph;
it increases as x increases. The slope is a measure of the change in y for a given
change in x. It may then be stated that:
the rate of change of y with respect to x varies with the value of x.

2.3.4

VALUE OF THE SLOPE


3

We need to find the value of the slope of the line y = x when x = 2 (See Figure
1). Hence, we may choose incremental changes in x which are located around
x 2.
FIGURE 0-2. y / x

when x 2

x1

x2

y1

y2

y
-----x

64

64

16.000

27

26

13.000

1.5

2.5

1.0

3.375

15.625

12.250

12.250

1.8

2.2

0.4

5.832

10.648

4.816

12.040

1.9

2.1

0.2

6.859

9.261

2.042

12.010

1.95

2.05

0.1

7.415

8.615

1.200

12.003

As may be seen, the value of the slope

-----y-
x

tends towards a value of 12.000 as the

magnitude of the incremental change in x becomes smaller around the chosen


value of 2.0. Were the chosen incremental changes in x infinitesimally small, the
true value of the slope (i.e., 12.000) would have appeared in the final column of
the above table.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-15

Mathematics Review

Calculus deals with infinitesimally small changes. When the value of x is infinitesimally small it is written dx and is known as the derivative of x. Hence,
dy
------ = f ( x )
dx
Where dy/dx is the derivative of y with respect to x and f ( x ) indicates some function of x.

2.3.5

DIFFERENTIATION FROM FIRST PRINCIPLES


Differentiation is the process whereby the derivative of y with respect to x is
found. Thus the value of dy/dx, in this case, is calculated.
(a) Considering again the original expression:
y = x

(b) Let the value of y increase to y + dy because x increases to x + dx .


Hence,
y + dy = ( x + dx )

(EQ 1-14)

Multiplying out:
3

y + dy = x + 3x ( dx) + 3x ( dx ) + ( dx )

(EQ 1-15)

(c) The change in y is obtained by subtraction of the original expression from the
last expression. (i.e., Eq. 2 - Eq. 1)
2

dy = 3x ( dx ) + 3x ( dx ) + ( dx )

(EQ 1-16)

Dividing throughout to obtain the derivative,


2
2
dy
------ = 3x + 3x ( dx ) + ( dx )
dx

When dx is infinitesimally small, its magnitude tends to zero ( dx 0 ) . The limiting value of this tendency must be dx = 0 . At this limit,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-16

Mathematics Review

2
dy
------ = 3x
dx

(EQ 1-17)

Hence the derivative of y with respect to x at any value of x is given by 3x .


(d) In section 2.3.4 we saw how the true value of the slope (i.e., dy/dx) would be
12.0 when x = 2 . This is confirmed by substituting in Equation 1-16.
2
2
dy
------ = 3x = 3 ( 2 ) = 12
dx

2.3.6

RULE OF DIFFERENTIATION
Although the rate of change of one value with respect to another may be calculated
as above, there is a general rule for obtaining a derivative.
Let x be the independent variable value, y be the dependent variable value, A be a
constant, and n be an exponential power.
The general rule is:
If y = Ax

then
n1
dy
------ = nAx
dx

The Rules of Indices may need to be used to obtain expressions in the form
y = Ax

(e.g., if y =

2.3.7

x)

THREE OTHER DERIVATIVES


0

(a) If y = Ax ,
then y = A (i.e., y is constant)
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-17

Mathematics Review

dy
Hence, ------ = 0
dx
Thus the derivative of a constant is always zero.
(b) Accept that if y = ln ( x )
dy
1
then ------ = --- .
dx
x
This derivative is important when considering apparent first-order processes, of
which many bodily processes (e.g., excretion of drugs) are examples.
(c) Accept that if y = Be
Ax
dy
base then ------ = ABe
dx

Ax

where B and A are constants, and e is the natural

This derivative will be useful in pharmacokinetics for finding the maximum and
minimum concentrations of drug in the blood following oral dosing.

2.3.8

A SEEMING ANOMALY
Consider the following two expressions:
n

(a) If y = Ax , then
n1
dy
------ = nAx
dx
n

(b) If y = Ax + A ,
n1
n1
dy
+ 0 = nAx
then ------ = nAx
dx

Both of the original expressions, although different, have the same derivative. This
fact is recognized later when dealing with integral calculus.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-18

Mathematics Review

2.3.9

INTEGRAL CALCULUS
Generally integral calculus is the reverse of differential calculus. As such it is used
to sum all the infinitesimally small units (dy) into the whole value (y).
Thus,

dy
2.3.10

= y , where

is the symbol for integration.

RULE OF INTEGRATION
The derivative expression may be written:
n
dy
------ = Ax , or
dx
n

dy = Ax dx
To integrate,
y =

dy

Ax

dx = A x dx

A general rule states:


n+1

n
Ax - + A
A x dx = --------------n+1

Where A is the constant of integration However, there is one exception - the rule
is not applicable if n = 1
2
Example: If dy
------ = 3x (See section 2.3.5),
dx
2+1

then y = 3x
--------------- + A , and
2+1
3

y = x +A

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-19

Mathematics Review

2.3.11

THE CONSTANT OF INTEGRATION


There has to be a constant in the final integrated expression because of the seeming
3

anomaly referred to in section 2.3.8. As mentioned, both y = x and y = x + A


2
will give, on differentiation, dy
------ = 3x .
dx
So whether or not a constant is present and, if so, what is its value, can only be
decided by other knowledge of the expression. Normally this other knowledge
takes the form of knowing the value of y when x = 0 .
In the case of our graphical example we know that when x = 0 , then y = 0 . The
integrated expression for this particular case is:
3

y = x + A , therefore
3

0 = 0 + A , thus A = 0
In some examples, such as first-order reaction rate kinetics, the value of A is not
zero.

2.3.12

THE EXCEPTION TO THE RULE


It occurs when n = 1
1
1
y = A x dx = A --- dx
x

Upon integration, y = A ln ( x ) + A
This is the reverse of the derivative stated in section 2.3.10 (b).

2.3.13

A USEFUL INTEGRAL
Accept that if,
Ax
dy
------ = Be
dx

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-20

Mathematics Review

then,
Ax

Be + A
y = ----------A
This integral will be useful for equations which define the bioavailability of a drug
product.

2.3.14

EXAMPLE CALCULATIONS
(a) Consider,
2

c = 3t ( t 2 ) + 5
Where c is the drug concentration in a dissolution fluid at time t .
Then, multiplying out,
3

c = 3t 6t + 5
The rate of dissolution at time t is
2
dc
------ = 9t 12t
dt

So at any time, the rate may be calculated.


2
dc
(b) Consider, ------ = 3t ( t 4 ) = 9t 12t
dt

Then rearranging,
2

dc = 9t dt 12t dt
The integral of c is:
c =

dc

= 3t + A 6t + B

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-21

Mathematics Review

where B is a second constant.


Adding the two constants together,
3

c = 3t = 6t + D
where D = A + B
We know, from previous work, that when t = 0 , then c = 5
Substituting 5 = D , the final expression becomes:
2

c = 3t + 6t + 5
Which is the initial expression in example (a) above.
(c) Following administration of a drug as an intravenous injection,
dC p
------------- = KC p
dt
Where C p is the plasma concentration of a drug at time t
K is the apparent first-order rate constant of elimination.
Rearranging,
1- dC
K dt = ----p
Cp
Kt = K dt =

- dC p
----Cp

This integral is the exception to the rule (see section 2.3.12).


Kt = ln ( C p ) + A
We know that when t = 0 , C p = ( C p ) 0 .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-22

Mathematics Review

Substituting, 0 = ln ( C p )0 + A
Or,
A = ln ( C p )0
Hence
Kt = ln ( C p ) ln ( C p ) 0
or,
ln ( C p ) = ln ( C p ) 0 Kt
or,
Cp = ( C p )0 e

Kt

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-23

Mathematics Review

2.4 Graphs
Why do we graph?

We would like to agonize the chaotic world around us so that we can predict (see
into the future) and retrodict (see into the past) what will happen or has happened.
Our recorded observations are collectively known as data. We make a theory
about what we think is happening and that theory is expressed in an equation. That
determines our paradigm of how we see the world. This paradigm is expressed as
a graph. The language of science is mathematics and graphs are its pictures.

TABLE 2-6

English

What is a graph?

Science

Observations

Data

Theory

Equations

Paradigms (pictures)

Graphs

A graph is simply a visual representation showing how one variable changes with
alteration of another variable. The simplest way to represent this relationship
between variables is to draw a picture. This pictorializing also is the simplest way
for the human mind to correlate, remember, interpolate and extrapolate perfect
data. An additional advantage is it enables the experimenter to average out small
deviations in experimental results (non-perfect, real data) from perfect data. For
example:
TABLE 2-7 Perfect

vs. Real data

Perfect

Real

-3

-5

-4.6

-2

-3

-3.4

-1

-1

-0.6

+1

+0.8

+1

+3

+3.4

+2

+5

+4.4

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-24

Mathematics Review

FIGURE 0-3.

Plot of Perfect vs. Real data

-2

-4

-6
-3

-2

-1

Simply looking at the columns x and y (real) it might be difficult to see the relationship between the two variables. But looking at the graph, the relationship
becomes apparent. Thus, the graph is a great aid to clear thinking. For every graph
relating variables, there is an equation and, conversely for every equation there is a
graph. The plotting of graphs is comparatively simple. The reverse process of finding an equation to fit a graph drawn from experimental data is more difficult,
except in the case of straight lines.

2.4.1

GRAPHICAL CONVENTIONS

How are graphs made?

Certain conventions have been adopted to make the process of rendering a data set
to a graphical representation extremely simple.
The y variable, known as the dependent variable, is depicted on the vertical axis
(ordinate); and the x variable, known as the independent variable, is depicted on
the horizontal axis (abscissa). It is said that y varies with respect to x and not
x varies with y.
A decision as to which of the two related variables is dependent can only be made
be considering the nature of the experiment. To illustrate, the plasma concentration
of a drug given by IV bolus depends on time. Time does not depend on the plasma
concentration. Consequently, plasma concentration would be depicted on the y
axis and time on the x axis.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-25

Mathematics Review

Any point in the defined space of the graph has a unique set of coordinates: 1) the
x value which is the distance along the x axis out from the y axis and always
comes first; and 2) the y value which is the distance, along the y axis up or
down from the x axis, and always comes second. Several points are shown in
Figure 2. For example, (0,1) is on the line and (1,0) is not.
The intersection of x and y axis is the origin with the coordinates of (0,0). In two
dimensional spaces, the graph is divided into 4 quadrants from (0,0), numbered
with Roman numerals from I through IV. It should be readily apparent that the
coordinates for all points within a particular quadrant are of the same sign type i.e.,
TABLE 2-8 Quadrants

on a cartesian graph

Quadrant II (-x, +y)

Quadrant I (+x, +y)

Quadrant III (-x, -y)

Quadrant IV (+x, -y)

A line (or curve) on a graph is made up of an infinite number of points, each of


which has coordinates that satisfy a given equation. For example, each point on the
line in Figure 2 is such at its coordinates fit the equation y = 2x + 1 . That is for
any value of x (the independent variable), multiplying the x value by 2 and adding
1 results in the y value (the dependent variable).

2.4.2

STRAIGHT LINE GRAPHS

What is a straight line?

A graph is a straight line (linear) only if the equation from which it is derived has
the form
y = mx + b
Where:
y = dependent variable
x = independent variable
m = slope of the straight line =

y
-----x

b = the y intercept (when x = 0)


or if the equation can be linearized, e.g.,
y = be

mx

is not linear

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-26

Mathematics Review

However.
ln y = ln b + mx
is of the same general form as:
y = b + mx
and consequently a plot of ln y (the dependent variable) versus x (the independent variable) will yield a straight line with a slope of m and an intercept of ln b .
Expressions of any other form are non linear. For example:
An expression relating the plasma concentration of a drug ( C p ) over time ( t ) .
C p = C p0 e

Kt

this relationship put in linear perspective yields:


ln C p = ln C p0 Kt , which is in the form
y = b + mx
The graphs that yield a straight line are the ones with the ordinate being ln C p0 ,
and the abscissa being t .
Any other combination of functions of C p and t will be non-linear, e.g.,
C p versus t
C p versus ln t
ln C p versus ln t
The appropriate use of a natural logarithm in this case serves to produce linearity.
However, the use of logarithms does not automatically straighten a curved line in
all examples. Some relationships between two variables can never be resolved into
a single straight line, e.g., y = k 0 + k 1 x

( n m)

+ k2 x

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(n m + 1)

+ + ( k n )x

2-27

Mathematics Review

where n 2 ;n = m + 1
or
K a FD
K t
Kt
C p = ------------------------ (e e a )
V ( Ka K )
(It is possible to resolve this equation into the summation of two linear graphs
which will be shown subsequently.)

2.4.3

THE SLOPE OF A LINEAR GRAPH (M)

What is the slope of a


straight line?

From the equation a prediction may be made as to whether the slope is positive or
negative. In the previous example, the slope is negative, i.e: m = K
TABLE 2-9 Sample

data of caffeine elimination

t (min)

g-
C p ------ mL

ln C p

12

3.75

1.322

40

2.80

1.030

65

2.12

0.751

90

1.55

0.438

125

1.23

0.207

173

0.72

-0.329

The differences in both the y-values and the x-values may be measured graphically
to obtain the value of the slope, m. Then knowing the value of m, the value of K
may be found.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-28

Mathematics Review

FIGURE 0-4.

Plasma Concentration

( C p ) of caffeine over time

Caffeine Concentration (

g/mL)
u

101

2.4.4

100

10-1
0

50

100

150

200

Time (min)

LINEAR REGRESSION: OBTAINING THE SLOPE OF THE LINE


The equation for a straight line is:
y = mx+b
Where y is the dependent variable
x is the independent variable
m is the slope of the line
b is the intercept of the line
The equation for the slope of the line using linear regression is:
( ( x ) ( y ) ) ( n ( x y ))
m = -------------------------------------------------------------------2
2
[ ( x ) ] ( n ( x ) )
And the intercept is
b = y (m x)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-29

Mathematics Review

TABLE 2-10

Linear Regression for data in table 2-9


2

XY

12

1.322

144

15.864

40

1.030

1600

41.2

65

0.751

4225

48.815

90

0.438

8100

39.42

125

0.207

15625

25.875

173

-0.329

29929

-56.917

X = 505

Y = 3.239

X = 59623

XY = 114.257

( X ) = 255025
x
x = ------ = 4.167
n

y
y = ------ = 0.5398
n

Using the data from table 2-10 in the equation for the slope of the line

( 505 3.239 ) ( 6 114.257 )


m = --------------------------------------------------------------------- = 0.01014
255025 ( 6 59623 )

and the intercept would be b =


ln C .
In oder to find the
b

Cpo = e = e

1.4229

0.5398 ( 0.01014 4.167 ) = 1.4229 .


C p0 ,

the anti-ln of

Note that this is


must be taken. i.e.

= 4.15

It is important to realize that you may not simply take any two data pairs in the
data set to get the slope. In the above data, if we simply took two successive data
pairs from the six data pairs in the set, this would result in five different slopes
( x y ) ranging from -0.0066 to -0.0125 as shown in table 2-11. Clearly, this is
unacceptable. Even to guess, you must plot the data, eyeball the best fit line by
placing your clear straight edge through the points so that it is as close to the data
as possible and look to make sure that there are an equal number of points above
the line as below. Then take the data pairs from the line, not the data set.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-30

Mathematics Review

TABLE 2-11 Sample

2.4.5

slope data from figure 0-4

Time (x)

ln Conc. (y)

y
-----x

12

1.322

-28

0.292

-0.0104

40

1.030

-25

0.28

-0.0112

65

0.751

-25

0.312

-0.0125

90

0.438

-35

0.231

-0.0066

125

0.207

-48

0.536

-0.0112

173

-0.329

PARALLEL LINES
Two straight lines are parallel if they have the same slope.
Calculating for the intercept of a linear graph (b):
(a) Not knowing the value of m;
The graph may be extrapolated, or calculations performed, at the situation where
t = 0 . In this case b = ln C p0 .
(b) Knowing the value of m;
There are two ways: for any point on the graph:
y 1 = mx 1 + b
b = y 1 mx 1
Hence, b may be calculated from a knowledge of y 1 and x 1 . Secondly, the graph
may be extrapolated or calculations performed, at the situation where t = 0 . In
this case,
b = ln C p

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-31

Mathematics Review

2.4.6

GRAPHICAL EXTRAPOLATIONS

How far can I predict?

It is dangerous to extrapolate on non-linear graphs, and it is unwise to extrapolate


too far on linear graphs. Most often extrapolation is used to find the value of y at a
selected value of x.
If the size of the graph does not permit physical extrapolation to the desired value,
the required result may be obtained by calculation. The values of m and b must be
found as shown above. Then:
y' = mx' + b ,
where x' is the selected value of x, and
y' is the new calculated value for y.

2.4.7

SIGNIFICANCE OF THE STRAIGHT LINE


The more closely the experimental points fit the best line, and the higher the number of points, the more significant is the relationship between y and x. As you may
expect, statistical parameters may be calculated to indicate the significance.

What good is a straight


line?

By using all the experimental data points, calculations may be made to find the
optimum values of the slope m, and the intercept, b. From these values the correlation coefficient (r).and the t-value may be obtained to indicate the significance.
Exact details of the theory are available in any statistical book, and the calculations
may most easily be performed by a computer using The Scientist or PKAnalyst in
this course.
The advantage of computer calculation is that it gives the one and only best fit to
the points, and eliminates subjective fitting of a line to the data.

2.4.8

GRAPHICAL HONESTY

How many points are


needed?

Any graph drawn from 2 points is scientifically invalid. Preferably, straight-line


graphs should have at least 3 - 5 points, and non-linear graphs a few points more.

Can I discard points that


dont fit?

As a graph is a visual representation which enables the experimenter to average


out the small deviations in results from the perfect result, no one result can be
unjustifiably ignored when the best fitting line is drawn. Still, an errant point
may be justifiably ignored if there were unusual experimental circumstances
which may have caused the deviation. Thus it is not justifiable to omit a point
solely because it does not fit.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-32

Mathematics Review

2.4.9

AXES WITH UNEQUAL SCALES


In mathematical studies, the scales of the x and y are almost always equal but very
often in plotting chemical relations the two factors are so very different in magnitude that this can not be done. Consequently, it must be borne in mind that the relationship between the variables is given by the scales assigned to the abscissa and
ordinate rather than the number of squares counted out from the origin.
FIGURE 0-5. y = 0.1 x

0.35
0.30
0.25
0.20
0.15
0.10
0.05
0.00
0

10

10

10
8
6
4
2
0

For example (shown in Figure 0-5), these two parabolic curves represent the same
equation the only difference is the scales are different along the y axis.
Frequently it is not convenient to have the origin of the graph coincide with the
lower left hand corner of the coordinate paper. Full utilization of the paper with
suitable intervals is the one criteria for deciding how to plot a curve from the
experimental data. For example, the curve below (Figure 0-6) is poorly planned,
where the following (Figure 0-7) is a better way of representing the gas law
PV = nRT

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-33

Mathematics Review

FIGURE 0-6.

Poorly presented graph

50
40
30
20
10
0
0

FIGURE 0-7.

12

16

20

Well arranged graph

25
20
15
10
5
0
1

9 10

Pressure (atm)

2.4.10

GRAPHS OF LOGARITHMIC FUNCTIONS


2

Previously variables were raised to constant powers; as y = x . In this section


x

constants are raised to variable powers; as y = 2 . Equations of this kind in which


the exponent is a variable are called (naturally) exponential equations. The most
x

important exponential equation is where e is plotted against x .

2.4.11

SEMILOGARITHMIC COORDINATES
Exponential or logarithmic equations are very common in physical chemical phenomenon. One of the best ways of determining whether or not a given set of phenomenon can be expressed by a logarithmic or exponential equation is to plot the
logarithm of one property against another property. Frequently a straight line is
obtained and its equation can be readily found. For example:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-34

Mathematics Review

In the following table the plasma concentration ( C p ) of the immunosuppressant


cyclosporine was measured after a single dose (4mg/kg) as a function of time.
TABLE 2-12 Plasma

concentration of cyclosporine
Concentration

Time (hours)
0.25

1900

.75

1500

1.5

1300

900

600

390

ng
-----ml

Dmello et al., Res. Comm. Chem. Path. Pharm. 1989: 64 (3):441-446

These can be illustrated in three different ways (Figures 0-8, 0-9, 0-10),
A. Concentration vs. time directly
B. Log concentration vs. time directly
C. Log concentration vs. time with concentration plotted directly on to logarithmic
scale of ordinates.
FIGURE 0-8.

Concentration (ng/ml) vs. time (hr)

2000
1800
1600
1400
1200
1000
800
600
400
200
0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-35

Mathematics Review

FIGURE 0-9.

Log Concentration vs. time

3.200
3.100
3.000
2.900
2.800
2.700
2.600
2.500
2.400
2.300

Log Concentration - Time Curve

FIGURE 0-10.

Log concentration (on log scale) vs. time

10000

1000

100
0

Graphing is much easier because the graph paper itself takes the place of a logarithmic table, as shown in Figure 1-10.
Only the mantissa is designated by the graph paper. Scaling of the ordinate for the
characteristic is necessary. The general equation y = Be ax can be expressed as a
straight line by basic laws of indices.
ax

ln y = ln B + ln ( e ) ln y = ln B + ax
ln y = ax + ln B

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-36

Mathematics Review

One axis is printed with logarithmic spacing, and the other with arithmetic spacing. It is used when a graph must be plotted as in the example (Figure 1-4)
y = log [ C p ] and x = t .
In this example, the vertical logarithmic axis is labelled Plasma concentration of
cyclosporine and the values plotted are the ordinary values of [ C p ] . Thus, there is
no need to use logarithmic tables, because the logarithmic spacing is responsible
for obtaining a straight line.
Two problems may occur when graphing on a logarithmic mantissa:
a) there are not enough cycles to incorporate all the data
b) obtaining the value of the slope is difficult. In this instance the slope is given by:
ln [ C p ] 2 ln [ C p ] 1
y2 y1
m = --------------- = -------------------------------------------x2 x1
t2 t1
Hence, before calculating the value of m, the two selected values of [ Cp ] 1 and [ Cp ] 2
must be converted, using a calculator, to ln [ Cp ] 1 and ln [ C p ] 2 in order to satisfy the
equation. The same problem may arise in obtaining the intercept value, b.
The two problems may be avoided by plotting the same data on ordinary paper, in
which case the vertical axis is labelled log plasma concentration. However, in
this instance the ordinary values of [ C p ] must be converted to ln [ Cp ] prior to plotting. It is the ln [ Cp ] values which are then plotted.
The calculation of the slope is direct in this case, as the values of
read from the graph.

y1

and

y2

may be

Hence, one must consider the relative merits of semilogarithmic and ordinary
paper before deciding which to use when a log plot is called for.
In the case of semilog graphs the slope may be found in a slightly different manner,
i.e., taking any convenient point on the line ( y 1 ) we usually take the as the second
point, ( y 2 ) one half of ( y 1 ) . Thus,
y1
1 -
ln -------------------
ln --------
ln y 1 ln ( ( 1 2 )y 1 )
( 1 2 )y 1
1 2
ln 2 - = 0.693
= ------------------------------= --------------------- = ------------------------m = ----------------------------------------------t 1 t2
t1 t2
t1 2
t 1 t2
t 1 t2
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-37

Mathematics Review

(in which case, t 2 t 1 is called the half-life t ). Since


because

2.4.12

0.693
m = ------------- = k
t1 2

and

t1 < t2 ,

then

t1 t2 = t1 2

0.693
k = ------------- .
t1 2

LOG - LOG COORDINATES


a

Functions of the type y = Bx give straight lines when plotted with logarithms
along both axis.
i.e., equation in logarithmic form is:
log y = log B + a log x or
log y = a log x + log b which is in the form
y = mx + b
This is directly applicable to parabolic and hyperbolic equations previously discussed (see Figure 1-5).

2.4.13

PITFALLS OF GRAPHING: POOR TECHNIQUE


The utility of these procedures requires proper graphing techniques. The picture
that we draw can cause formation of conceptualizations and correlations of the
data that are inconsistent with the real world based simply on a bad picture. Consequently the picture must be properly executed.
The most common error is improper axes labelling. On a single axis of rectilinear
coordinate paper (standard graph paper), a similar distance between two points
corresponds to a similar difference between 2 numbers. Thus,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-38

Mathematics Review

FIGURE 0-11.

Graphing using standard number spacing

40
30
20
10
0
0

FIGURE 0-12.

10

15

20

25

30

Nonstandard (incorrect) graph

40
30
20
10
1
0
2

10

20

30

Obviously, the distance (Time) on the graph 12 between 0 and 2 hours should not
be the same as the distance between 10 and 20 hours. It is, and therefore Figure 012 is wrong.
Similarly, the use of similar paper may result in some confusion. With logarithms
the mantissa for any string of numbers, differing only by decimal point placement,
is the same. What differentiates one number from another, in this case, is the characteristic. Thus,
TABLE 2-13 Logarithmic

graphing

Number

Mantissa

Characteristic

Log

234

.3692

2.3692

23.4

.3692

1.3692

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-39

Mathematics Review

TABLE 2-13 Logarithmic

graphing

Number

Mantissa

Characteristic

Log

2.34

.3692

0.3692

0.234

.3692

-1

1.3692

The paper automatically determines the relationship between strings of numbers


(mantissa) by the logarithmic differences between the numbers on the axis within a
cycle. The student must determine the order of magnitude (characteristic) to be relegated to each cycle.
FIGURE 0-13.

Logarithmic mantissa
Logarithmic Plot
103
234

102

Y axis (units)

23.4

101
2.34

100
0.234
10-1
1.0

1.5

2.0

2.5

3.0

3.5

4.0

X axis (units)

Thus, we see, in Figure 0-13, the cycle on the semilog paper to relate to orders of
magnitude (e.g., 1, 10, 100, 1000, etc.) and consequently the characteristic of the
exponent.
The third common problem is labelling the log axis as log y. This is improper. It
is obvious from the spacing on the paper that this function is logarithmic, and thus
the axis is simply labelled y.
There are almost as many different errors as there are students and it is impossible
to list them all. These few examples should alert you to possible problems.

2.4.14

GRAPHICAL ANALYSIS
We will look at several different types of plots of data:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-40

Mathematics Review

FIGURE 0-14.

Straight line going down on semi-log paper

Find the slope by taking any two values on the Y axis such that the smaller value is
one half of the larger. The time that it takes to go from the larger to the smaller is
the half-life. Dividing 0.693 by the half-life yields the rate constant.
Extrapolating the line back to t = 0 yields the intercept.

FIGURE 0-15.

Curved line which plateaus on semi-log paper.

10

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

12

2-41

Mathematics Review

FIGURE 0-16.

Curved line which goes up and then straight down on semi-log paper.

10

15

20

25

Find the terminal slope by taking any two values on the Y axis such that the
smaller value is one half of the larger. The time that it takes to go from the larger to
the smaller is the half-life. Dividing 0.693 by the half-life yields the rate constant.
Plot type one is reasonably easily evaluated. There are 2 important things that can
be obtained: Slope and Intercept. However, the slope and intercept have different
meanings dependent on the data set type plotted. The slope is the summation of all
the ways that the drug is eliminated, -K.

TABLE 2-14 Plot

type 1

Data Type

Y axis

X axis

Slope

Intercept

IV Bolus Parent

Drug Conc. parent compound

Time

-K

dose
C p0 = ----------Vd

IV Bolus Parent

dXu
---------- urine rate of excretion
dt
parent compound

Time
(mid)

-K

Kr X 0

IV Bolus Parent

Xu Xu Cumulative urine

Time

-K

kr
-------------K X0

data

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-42

Mathematics Review

Plot type two is not usually evaluated in its present form as only the plateau value
can be obtained easily. But again it has different meanings dependent on the data
plotted.
TABLE 2-15 Plot

Type 2
Y axis

X axis

Plateau Value

IV Bolus Parent

Data Type

Xu Cumulative urine
data parent compound

Time

kr
Xu = -------------K X0

IV Infusion
Parent

Drug concentration
parent compound

Time

Q - = ---Q
( C p )ss = ----------KV
cl

Usually urine data of this type (parent compound - IV bolus) is replotted and evaluated as plot 1 (above). Infusion data can be replotted using the same techniques,
but usually is not.
Plot type 3 must be stripped of the second rate constant from the early time points,
thus:
There are 3 things that can be obtained from the plot: the terminal slope (the
smaller rate constant), the slope of the stripped line (the larger rate constant) and
the intercept. The rate constants obtained from a caternary chain (drug moving
from one box to another in sequence in compartmental modeling) are the summation of all the ways that the drug is eliminated from the previous compartment and
all the ways the drug is eliminated from the compartment under consideration. See
LaPlace Transforms for further discussion.
Again, dependent on the data set type being plotted they will have different values.

TABLE 2-16 Plot

Data
Type

Type 3
Y axis

X axis

S1

S2

Intercept

IV Bolus
Parent

Metabolite conc.

Time

-Ksmall

-Klarge

km X 0
------------------------------------------------------( K l arg e K small ) V dm

IV Bolus
Parent

dXmu
--------------- excretion
dt
rate of metabolite
into urine

Time
(mid)

-Ksmall

-Klarge

k mu k m X 0
-----------------------------------K l arg e K small

Oral

Drug conc.

Time

-Ksmall

-Klarge

k a fX 0
--------------------------------------------------( K l arg e K small ) V d

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-43

Mathematics Review

2.5 Pharmacokinetic Modeling


It has been observed that, after the administration of a drug, the concentration of
the drug in the body appear to be able to be described by exponential equations.
Thus, it appears that, even though the processes by which the drug is absorbed. distributed, metabolized and excreted (ADME) may be very complex, the kinetics
(math) which mimics these processes is made up of relatively simple first order
processes and is called first order pharmacokinetics. A second observation is that
the resulting concentration is proportional to dose. When this is true, the kinetics is
called linear. When this math is applied to the safe and effective therapeutic management of an individual patient, it is called clinical pharmacokinetics. Thus, in
clinical pharmacokinetics, we monitor plasma concentrations of drugs and suggest
dosage regimens which will keep the concentration of drug within the desired therapeutic range. Pharmacodynamics refers to the relationship between the drug concentration at the receptor and the intensity of pharmacological (or toxicological)
response. It is important to realize that we want to control the pharmacological
response. We do that indirectly by controlling the plasma concentration. In order
for this to work, we assume kinetic homogeneity, which is that there is a predictable relationship between drug concentration in the plasma (which we can measure) and drug concentration at the receptor site (which we can not measure). This
assumption is the basis for all clinical therapeutics.
Models are simply mathematical constructs (pictures) which seem to explain the
relationship of concentration with time (equations) when drugs are given to a person (or an animal). These models are useful to predict the time course of drugs in
the body and to allow us to maintain drug concentration in the therapeutic range
(optimize therapy). The simplest model is the one used to explain the observations.
We model to summarize data, to predict what would happen to the patient given a
dosage regimen, to conceptualize what might be happening in disease states and to
compare products. In every case, the observations come first and the explanation
next. Given that a data set fits a model, the model can be used to answer several
different types of questions about the drug and how the patient handles the drug
(its disposition), for example: if the drug were to be given by an oral dose, how
much is absorbed and how fast? Are there things which might affect the absorption, such as food or excipients in the dosage form itself. What would happen if the
drug were to be given on a multiple dose regimen? What if we increased the dose?
etc.
You should be able to:
be facile in the use of the equations. You should be able to graphically manipulate data sets and
extract pharmacokinetic parameters, applying the appropriate equations or variations of them.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-44

Mathematics Review

define all new words used in this section. e g.: Succinctly define, stating rigorously the meaning
of any symbols used and the dimensions of measurement.

compare and contrast new concepts used in this section. e. g.: rate and rate constant, zero and
first order kinetics, bolus and infusion methods, excretion and elimination, the assumptions
made in pharmacokinetic models with physiological reality. Why can these assumptions be
made?

pictorially represent any two variables (graph) one vs. the other. e.g. for each of the following
pairs of variables (ordinate against abscissa), draw a graph illustrating the qualitative profile of
their relationship. Where appropriate, indicate the nature of important slopes, intercepts, and
values. Unless you specifically indicate on your plot that semi-log paper is being considered
(write S-L), it will be assumed that rectilinear paper is being considered. Graphs are for a
drug given by IV Bolus where applicable.

2.5.1

MAKING A MODEL

ka

The differential equations used result from the


model which is our conceptualization of what is
happening to the drug in the body.

The box (compartment) is the area of interest. We


want to find out how the mass of drug, X , changes
with time in that compartment, the rate, and how the rates change with time, the
differential equations.
How do we make a differential equation?

The picture that we build is made up of building blocks, consisting of the arrow
and what the arrow touches. The arrow demonstrates how quickly the mass of
drug, X , declines. The arrow times the box that the arrow touches = the rate. Rates
can go in, i.e. arrows pointing to a box mean drug is going in (+ rate). Rates can go
out, i.e. arrows pointing away means drug is going out (- rate). Rate = rate constant
(arrow) times mass of drug (box). So the arrow and box really is a pictorial representation of a rate where the rate is the rate constant on the top of the arrow times
what the tail of the arrow touches.
Again, the rate constant, k , tells the magnitude of the rate,

kX.

Consider the following simple chain:

X1

k12

X2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

k23

X3

2-45

Mathematics Review

The building blocks are k 12 X1 and k 23 X2 . Every arrow that touches the compartment of interest becomes part of the differential equation. If the arrow goes to the
box, its positive; if it goes away from the box, its negative.
To find dX 1 dt (the rate of change of X 1 with time), we simply add up all of the
rates which affect X1 (all of the arrows that touch X1 )
dX 1
--------- = k 12 X 1
dt
and thus:
dX 2
--------- = k 12 X 1 k 23 X2
dt
dX 3
--------- = k 23 X 2
dt
(Note: the first subscript of the rate constant and the subscript of the box from
which it originates are the same.)
You should be able to develop the series of interdependent differential equations
which would result from any model. The integration of those equations by use of
the Laplace Table is done by transforming each piece of the equation into the
Laplace domain (looking it up on the table and substituting). The algebra performed solves for the time dependent variable: put everything except the variable
(including the operator, s) on the right side and put the variable on the left. Find the
resulting relationship on the left side of the table. The corresponding equation on
the right side of the table in the integrated form.
You should be able to integrate any differential equation developed from any
model (within reason) that we can conceptualize.
(Note: Each subsequent variable is dependent on the ones that precedes it. In fact,
the solutions to the preceding variables are substituted into the differential to
remove all but one of the time dependent functions - the one that we are currently
attempting to solve.)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-46

Mathematics Review

2.5.2

ONE COMPARTMENT OPEN MODEL


A simplified picture (mathematical construct) of the way the body handles drug is
one where the body can be conceived to be a rapidly stirred beaker of water (a single compartment). We put the drug in and the rate at which the drug goes away is
proportional to how much is present (first order). Thus the assumptions are:

Body homogeneous (one compartment)


Distribution instantaneous
Concentration proportional to dose (linear)
Rate of elimination proportional to how much is there. (First order)

It is important to note that we know some of these assumptions are not true. It is of
little consequence, as the data acts as if these were true for many drugs. The visual
image which is useful is one of a single box and a single arrow going out of the
box depicting one compartment with linear kinetics. The dose is placed in the box
and is eliminated by first order processes. In many cases, more complicated models (more boxes) are necessary to mathematically mimic the observed plasma versus time profile when one or more of these assumptions are not accurate. For
example, the two compartment (or multi-compartment) model results when the
body is assumed to not be homogeneous and distribution is not instantaneous.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-47

Mathematics Review

2.6 The LaPlace Transform


Why do we need to
know the LaPlace transform?

One of the important facets of biopharmaceutics is a familiarity with the principles


of pharmacokinetics. This latter discipline describes the study of the dynamic processes by which the body handles or disposes of an administered drug. These
processes (absorption, distribution, metabolism, and excretion (ADME) are
dynamic in that they represent the time-dependent changes occurring to the drug.
Thus, in pharmacokinetics the time course of these changes, which overall
describe the fate of the administered drug, is described mathematically. If the
mathematical principles are understood, it is then possible to use pharmacokinetics
in clinical practice, such as the design of rational dosage regimens (T.S. Foster and
D.U.A. Bourne, Amer. J. Hosp. Pharm., 34, 70-75 (1977). Understanding
(Blooms level 5) is not simply memorizing (Blooms level 1) nor calculating
using a memorized equation (Blooms level 3). The authors believe that the proper
conceptualizing of the process and the subsequent derivation of the appropriate
equations will lead to an understanding of the mathematical principles, and thus, a
better, more optimal dosing regimen.
Since a mathematical description of the time-dependent ADME processes is
required, it becomes necessary to deal with their corresponding rate equations.
Inevitably this will involve calculus (mainly integral calculus). However, the
LaPlace Transform provides a method whereby calculus can be performed with
minimal trauma. If a conscientious effort to learn the method is made and applied,
a potentially serious obstacle (the fear of calculus) to the understanding and appreciation of biopharmaceutics will be removed. Indeed, many students will find they
no longer fear integration and are thus free to comprehend the principles underlying pharmacokinetics, which, after all, is the primary aim. So, the LaPlace Transform is a tool which is of great assistance in pharmacokinetics; its utility and
importance should not be lightly disregarded.

The LaPlace Transform: What Is It?

There is, of course, a theoretical background to the LaPlace Transform. However,


it can be used without recourse to a complete theoretical discussion, though appropriate pharmaceutical use of the method is found in the following references:
M. Mayersohn and M. Gibaldi, Amer. J. Pharm. Ed., 34, 608-614 (1970).
M. Gibaldi and D. Perrier, Pharmacokinetics, Marcel Dekker, pp. 267-272
(1975).
Basically, the LaPlace Transform is used to solve (integrate) ordinary, linear differential equations. In pharmacokinetics such equations are zero and first-order rate
equations in which the independent variable is time. For instance, if a differential
equation describing the rate of change of the mass of drug in the body with time is

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-48

Mathematics Review

integrated, the final equation will describe the mass of drug actually in the body at
any time.
The procedure used is to replace the Independent variable (time) by a function
containing the LaPlace Operator, whose symbol is s. In doing so we have
replaced the time domain by a complex domain. This is analogous to replacing a
number by its logarithm. Once in the complex domain, the transformed function
may be manipulated by regular algebraic methods. Then the final expression in the
complex domain is replaced by its equivalent in the time domain, yielding the integrated equation. This ultimate process is analogous to taking an antilogarithm.

2.6.1

TABLE OF LAPLACE TRANSFORMS

A table of useful LaPlace


transforms is given in
Section 2.7. Page 2-56.

The replacement of expressions in one domain by their equivalents in another is


accomplished by reference to tables. One column shows time domain expressions,
stated as f ( t ) , and second column shows the corresponding complex domain
expressions, stated as the LaPlace Transform. Note that f ( t ) simply means
at

some function of time. For example, when f ( t ) is Be , then the LaPlace


Transform is B ( s + a ) , where B is a constant and a is a rate constant
For example, when the LaPlace Transform is

2.6.2

As

, then f ( t ) is At .

SYMBOLISM
For simplicity in writing transformed rate expressions (and to distinguish them
from untransformed (time domain) expressions), the following symbolism will be
employed:
a bar will be placed over the dependent variable which is being transformed.
Example:
If X is the mass of unchanged drug in the body at any time, then X is the LaPlace
Transform of this mass.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-49

Mathematics Review

2.6.3

CONVENTIONS USED IN DRAWING PHARMACOKINETIC SCHEMA.


When drugs enter the body, they will encounter many different fates. It is important to set up the possible fates of the drug by creating a well thought out flow
chart or scheme in order to follow all the events that are occurring in the body as
described by the pharmacokinetic description of the drug. For example, a drug
may be excreted unchanged or may undergo hepatic metabolism to yield active or
inactive metabolites. All of these components are part of pharmacokinetics, which
by definition, includes ADME (the Absorption, Distribution, Metabolism and
Excretion of drugs), and must be considered. This flow chart becomes the backbone or the framework upon which to build the equations which describe the pharmacokinetics of the drug. The differential equations result as a direct consequence
of the flow chart. Using Laplace transforms, the integration of these differential
equations are simplified and provide the pharmacokineticist to (easily?) keep track
of all of the variables in the equation. If the drug scheme or flow chart is set up
incorrectly, this would have a definite negative impact or the expected equations
(as well as the answers and your grade). Below are two examples of how to construct a flow chart. Note that not all drugs follow the same flow chart and it is
quite possible that you will need only to use a portion of these examples when construction your own.
In general, schema are relatively consistent in the placement of the compartments
in relationship to one another. You might consider, for example a drug, given by
IV bolus, which is metabolized and both the metabolite and the parent compound
are excreted unchanged as shown below:
Feces

Body

Urine

Dose
kf

Parent Compound
Xf

ku
X

Xu

km
kmf
Metabolite

Xmf

kmu
Xm

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

Xmu

2-50

Mathematics Review

Using the pharmacokinetic symbolism from chapter one, the compartments are
named and placed: metabolites below (or above the plane of the parent compound): compounds going into the urine, to the right; and compounds going into
the feces, to the left of the compounds in the body. The rate constants connecting
the compartments also follow the symbolism from chapter one. In the above flow
chart, K1, the summation of all the ways that X is removed from the body, is ku +
kf + km while K2, the summation of all the ways that Xm is removed from the
body, is kmu +kmf.
Only those compartments are used which correspond to the drugs pharmacokinetic description, thus when a drug is given by IV bolus and is 100% metabolized
with the metabolite being 100% excreted into the urine the model would look like
this:
Dose
X
km
kmu
Xm

Xmu

Thus in this flow chart, K1, the summation of all the ways that X is removed from
the body, is km while K2, the summation of all the ways that Xm is removed from
the body, is kmu.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-51

Mathematics Review

Drugs sometimes are metabolized to two (or more) different metabolites. In the
first case, the drug is metabolized by two separate pathways resulting in this flow
chart:
Xmf1

kmf1

Xm1

Dose

Xf

kf

kmu1

Xmu1

km1
ku

Xu

km2
Xmf2

kmf2

kmu2

Xm2

Xmu2

In this flow chart, K1, the summation of all the ways that X is removed from the
body, is ku + kf + km1 + km2 while K2, the summation of all the ways that Xm1 is
removed from the body, is kmu1 +kmf1 and K3, the summation of all the ways
that Xm2 is removed from the body, is kmu2 + kmf2.
While in a second case, the drug is metabolized and the metabolite is further
metabolized resulting in this flow chart:
Dose
Xf

kf

ku

Xu

km1
Xmf1

kmf1

Xm1

kmu1

Xmu1

km2
Xmf2

kmf2

Xm2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

kmu2

Xmu2

2-52

Mathematics Review

In this flow chart, K1, the summation of all the ways that X is removed from the
body, is ku + kf + km1 while K2, the summation of all the ways that Xm1 is
removed from the body, is kmu1 +kmf1+ km2 and K3, the summation of all the
ways that Xm2 is removed from the body is kmf2 + kmu2.
Both of these flow charts result in very different end equations, so it is imperative
that the flow charts accurately reflect the fate of the drug.

2.6.4

STEPS FOR INTEGRATION USING THE LAPLACE TRANSFORM


Draw the model, connect the boxes with the arrows depicting where the drug goes.
The building blocks of the differential rate equations are the arrows and what the tail touches.
Write the differential rate equation for the box in question. The box is on the left side of the
equal sign and the building blocks are on the other. If the arrow goes away from the box, the
building block is negative, if it is going towards the box, the building block is positive.

Take the LaPlace Transform of each side of the differential rate equation, using the table where
necessary.

Algebraically manipulate the transformed equation until an equation having only one transformed dependent variable on the left-hand side is obtained.

Convert the transformed expression back to the time domain, using the table where necessary to
yield the Integrated equation.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-53

Mathematics Review

2.6.5

EXAMPLE INTEGRATION USING THE LAPLACE TRANSFORM


Following an intravenous injection of a drug (bolus dose), its excretion may be
represented by the following pharmacokinetic scheme:

(Scheme I)

ku

Xu

Where X is the mass of unchanged drug in the body at any time.


X u is the cumulative mass of unchanged drug in the urine up to any time, and k u is
the apparent first-order rate constant for excretion of unchanged drug.
Consider how the body excretes a drug
a. The building block is the arrow and what it touches. This first box (compartment) of interest is [ X ] . The arrow ( k u ) is going out, therefore, the rate is going out
and is negative, thus
dX
------- = k u X
dt

(EQ 1-18)

The negative sign indicates loss from the body.


Taking the LaPlace Transform of each side of equation 1-18:
sX X0 = k u X

(EQ 1-19)

Note that because the independent variable (time) did not appear on the right-hand
side of equation 1-18, neither did the LaPlace Operator, s, appear there in equation
1-19. All that was necessary was to transform the dependent variable ( X ) into X .
Hence, the table was only required for transforming the left-hand side of equation
1-19.
Manipulating the transformed equation:
1. Get only one variable which changes with time
2. Get

(X)

on the left and everything else on the right.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-54

Mathematics Review

sX + k u X = X0
X ( s + k u ) = X0
X0
X = ------------s + ku
LetX 0 = A

Letk u = a

(EQ 1-20)

A
X = ---------------(s + a)

(EQ 1-21)

Note that X is the only transformed dependent variable and is on the left-hand side
of equation 1-20.
Converting back to the time domain:

X = X0e

ku t

(EQ 1-22)

A
Note that the right-hand side of equation 1-22 was analogous to ---------------- in the
(s + a)
table, because X0 is a constant (the initial dose administered). The left-hand side
of equation 1-22 could be converted back without the table.
The final expression is the familiar first-order integrated expression.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-55

Mathematics Review

2.6.6

SECOND EXAMPLE INTEGRATION USING THE LAPLACE


TRANSFORM
Look at Scheme I again. Consider how the drug goes from the body into the urine.
The next box of interest is Xu . The arrow is coming in, therefore the rate is coming in and is positive. thus,
dX u
--------- = k u X
dt

(EQ 1-23)

(b) Taking the LaPlace Transform of each side of equation 1-23:


sX u ( X u ) 0 = k u X

(EQ 1-24)

But, at zero time, the cumulative mass of unchanged drug in the urine was zero:
that is ( Xu ) 0 = 0 .
sX u = k u X

(EQ 1-25)

(c) Manipulating the transformed equation:


ku X
Xu = -------s

(EQ 1-26)

Note that there are two transformed dependent variables. One of them ( X ) can be
replaced by reference to equation 1-20.
ku X0
Xu = -------------------s(s + ku)
Let ( k u X0 ) = A

Let ( k u ) = a

(EQ 1-27)

A
X = -----------------s(s + a)

(EQ 1-28)

(d) Converting back to the time domain:


k t

kuX0 ( 1 e u )
X u = ---------------------------------------ku

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-56

Mathematics Review

Where k u X 0 and k u are analogous to A and a respectively in the table. Simplifying,


Xu = X0 ( 1 e

2.6.7

kut

(EQ 1-29)

THIRD EXAMPLE INTEGRATION USING THE LAPLACE TRANSFORM


During the intravenous infusion of a drug, its excretion may be represented by the
following pharmacokinetic scheme:
(Scheme II)

Infusion

ku

Xu

Where Q is the zero-order infusion rate constant (the drug is entering the body at a
constant rate and the rate of change of the mass of drug in the body is governed by
the drug entering the body by infusion and the drug leaving the body by excretion).
The drug entering the body does so at a constant (zero-order) rate.
dX
------- = Q k u X
dt

(EQ 1-30)

(b) Taking the LaPlace Transform of each side of equation 1-30:


Q
sX X 0 = ---- k u X
s
Note that because Q is a rate, and is therefore a function of the independent variable (time), its transformation yields the LaPlace Operator. In this case, Q was
analogous to A in the table. But, at zero time, the mass of unchanged drug in the
body was zero: that is, X0 = 0
Q- k X
sX = --u
s

(EQ 1-31)

(c) Manipulating the transformed equation:


Q X = -------------------s ( s + ku )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 1-32)

2-57

Mathematics Review

Let ( Q = A )

A
X = ------------------s( s + a)

Let ( k u = a )

(EQ 1-33)

(d) Converting back to the time domain:


kut

Q(1 e )
X = ----------------------------ku

2.6.8

(EQ 1-34)

CONCLUSIONS
The final integrations (Eqs. 24, 24, and 28) are not the ultimate goal of pharmacokinetics. From them come the concepts of:
1.

(a) elimination half-life

1.

(b) apparent volume of drug distribution

2.

(c) plateau drug concentrations

These, and other concepts arising from still other equations, are clinically useful.
Once the method of LaPlace Transforms is mastered, it becomes easy to derive
equations given only the required pharmacokinetic scheme. Under such circumstances, it no longer becomes necessary to remember a multitude of equations,
many of which, though very similar, differ markedly in perhaps one minute detail.
As with any new technique, practice is required for its mastery. In this case, mastery will banish the calculus blues.
It is also possible to see certain patterns which begin to emerge from the derivation
of the equations. For example, for a drug given by IV bolus the equation is
monoexponential, with the exponent being K1, summation of all the ways that the
drug is removed form the body. A graph of the data (Cp v T on semi-log paper)
results in a straight line the slope of which is K1, always. If the drug is entirely
metabolized K1 = km. If the drug is entirely excreted unchanged into the urine,
K1 = ku. If the drug is metabolized and excreted unchanged into the urine, K1 =
km +ku. thus K1 can have different meanings for different drugs, depending on
how the body removes the drug. Following the drug given by IV bolus a second
example of a pattern would be that of the data of the metabolite of the drug. From
the LaPlace, the equation for the plasma concentration of the metabolite of the
drug has in it K1 and K2, the summation of all the ways that the metabolite is
removed from the body, always. K2 would have different meanings depending on
how the metabolite is removed from the body.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-58

Mathematics Review

After several years teaching, I was fortunate to have a resident rotate through our
pharmacokinetic site. She had come with a strong Pharmacokinetcs background
and during our initial meeting, she had told me that she had a copy of John Wagners new textbook on pharmacokinetcs. She was excited that, finally, there was a
compilation of all the equations used in pharmacokinetics in one place.
There are over 500 equations in the new book and I know every one, she said.
Im not sure which one goes with which situation, though. OOPS!
Throughout this text and on each exam, each equation is derived from first principles using scientific method, modeling and LaPlace Transforms in the hopes that
memorization will be minimized and thought (and consequently proper interpretation) would be maximized.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-59

Mathematics Review

2.6.9

TABLE OF LAPLACE TRANSFORMS

TABLE 2-17 Table

A, B

are constants

a, b, c

are rate constants ( a b c )

is the LaPlace Operator

is a variable, dependent on time ( t )

is a power constant

of LaPlace Transforms
Time Function, F ( t )

LaPlace Transform, f ( s )

A
--s

At

A
---2
s

A
( m! -)
-------------m+1
s

at

A
----------s+a

m at

A
-------------------------m+1
(s + a)

At
Ae

At e

at

A
------------------s (s + a )

A(1 e )
-------------------------a
at

A( 1 e )
At
----- -------------------------2
a
a

A
--------------------2
s (s + a)

at

As B-----------------s (s + a )

Ae

at

B( 1 e )
-------------------------a
at

e - Bt
A + B
--- 1--------------------
a a
a

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

As B
--------------------2
s (s + a)

2-60

Mathematics Review

TABLE 2-17 Table

of LaPlace Transforms
Time Function, F ( t )
bt

LaPlace Transform, f ( s )

at

A
-------------------------------(s + a)(s + b)

A( e e )
--------------------------------(a b)
A
e - 1---------------- e -
1-------------------------------

(a b)
b
a

A
-----------------------------------s( s + a )( s + b )

1 e bt 1 e at
At
A
------ ---------------- ------------------ ------------------
ab ( a b ) b2 a2

A
-------------------------------------2
s ( s + a) ( s + b)

1 [ ( B + Aa )e at ( B + Ab )e bt ]
---------------( a b)

As B -------------------------------(s + a)(s + b)

( 1 e bt ) ( 1 e at )
bt
at
1
---------------- A ( e e ) B ---------------------- ----------------------
(a b)
b
a

As B
-----------------------------------s( s + a )( s + b )

( 1 e bt )
( 1 e at )
1
B
B
Bt
---------------A
+
-----------------------
A
+
--

---------------------- -----(a b)
b
b
a
a
ab

As B
-------------------------------------2
s ( s + a) ( s + b)

bt

ct

at

bt

at

e
e
e
A -------------------------------- + --------------------------------+ --------------------------------(a c)(b c) (a b )(c b) (b a )(c a)
ct

bt

at

(1 e )
(1 e )
(1 e )
A ------------------------------------ + ------------------------------------- + ------------------------------------c( a c )( b c ) b( a b)( c b ) a( b a )( c a)

A
------------------------------------------------(s + a)(s + b)(s + c)
A
---------------------------------------------------s( s + a )( s + b )( s + c )

(1 e )
(1 e )
(1 e )
At- A -----------------------------------------+ --------------------------------------- + --------------------------------------2
2
2
bc
c ( a c) ( b c ) b ( a b) ( c b) a ( b a )( c a)

A
------------------------------------------------------2
s ( s + a) ( s + b ) ( s + c )

dX
------dt

sX X 0

ct

bt

at

(m + 1 )

A
----m
s

At
------------------(m + 1)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-61

Mathematics Review

2.6.10

LAPLACE TRANSFORM PROBLEMS


By means of the LaPlace Transform, find the equation for:
1.

The amount of drug in the body when the drug is given by IV Bolus (assume no metabolism).

2.

The amount of drug in the urine when the drug is given by IV Bolus (assume no metabolism).

3.

The amount of metabolite in the body when the drug is given by IV Bolus (assume no parent
drug excretion)

4.

The amount of metabolite of the drug in the urine when the drug is given by IV Bolus (assume
no parent drug excretion)

5.

The amount of metabolite of the drug in the urine when the drug is given by IV Bolus (assume
both parent drug and metabolite excretion)

6.

The amount of drug in the body when the drug is given by IV infusion (assume no metabolism).

7.

The amount of drug in the urine when the drug is given by IV infusion (assume no metabolism).

8.

The amount of metabolite in the body when the drug is given by IV infusion (assume no parent
drug excretion).

9.

The amount of metabolite in the urine when the drug is given by IV infusion (assume no parent
drug excretion).

10.

The Rate of excretion of the metabolite into the urine for a drug given by IV bolus when
km+ku=kmu.

11.

The amount of the principle metabolite (Xm1) when the drug is eliminated by several pathways
(Xu, Xm1,Xm2,Xm3,etc)

12.

X- , in the body when the drug is given orally by a delivery system


The concentration of drug, ----Vd

which is zero order. What is the concentration at equilibrium ( T ).


13.

The amount of metabolite of a drug in the body when the drug is given by IV Bolus and concomitant IV infusion.

14.

Disopyramide (D) is a cardiac antiarrythmic drug indicated for the suppression and prevention
of ectopic premature ventricular arrythmias and ventricular tachycardia. It appears that disopyramide is metabolized by a single pathway to mono-dealkylated disopyramide (MND). In a
recent study, the pharmacokinetics of disopyramide were attempted to be elucidated by means
of a radioactive tracer. Since both D and MND would be labeled by the tracer, any equations
showing the time course of the label would show both the D and MND. By means of the
laPlace transform, find the equation for the rate of appearance of tracer into the urine if the drug
were given by IV Bolus.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-62

Mathematics Review

2.6.11

LAPLACE TRANSFORM SOLUTIONS

1. The amount of drug in the body when the drug is given by IV bolus (assume no metabolism).

Dose
X

ku

Xu

X = Xo At time zero, all of the IV bolus is in the compartment.


Here K1 = ku

dX
------- = k u X
dt
sX X0 = kuX
sX + kuX = X o
X ( s + k u )= Xo
Xo
X = ------------s + ku
kut
A
Let ( X0 ) = A , k u = a ,X = ---------------X = Xo e
(s + a)

2. the amount of a drug in the urine when the drug is given by IV bolus (assume the drug is NOT metabolized

Dose
X

ku

Xu

Again K1 = ku

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-63

Mathematics Review

dX
--------u- = k u X
dt

sX Xo = k m X
sX + k m X = Xo

sX u Xuo = k u X

X ( s + k m ) = Xo

sX u = k u X

Xo
X = -------------s + km

X o ku
sX u = -----------------(s + ku)

A
Let ( X 0 ) = A , k m = a , X = ---------------(s + a)

Xo ku
X u = -------------------s(s + ku)

X = Xo e

A
Let ( X0 k u ) = A , k u = a , X u = -----------------s( s + a )

kmt

dX m
---------- = k m X k mu X m
dt

kut

ku X o ( 1 e
)
X u = -----------------------------------ku
Xu= Xo ( 1 e

kut

sX m Xm0 = k m X kmu X m
km X 0
sX m = ------------- k mu Xm
s + km

3.
the amount of metabolite of a drug in the body
when the drug is given by IV bolus (assume no parent
drug excretion).

km Xo
X m = ----------------------------------------( s + k mu ) ( s + k m )
Let ( k m X 0 ) = A , k m = a = K1 , k mu = b = K2
A
X m = --------------------------------(s + a)(s + b )

Dose

( km X o )
k t
k t
X m = ------------------------ ( e mu e m ) or
( k m k mu )

( km Xo )
K 2t
K 1t
X m = ------------------------ (e
e
) in general terms.
( K1 K2 )

km
Xm

kmu

Xmu

NOTE: We could also:

Let ( k m X 0 ) = A , k m = b , k mu = a
Here K1 = km and K2 = kmu

and then

dx
------ = k m X
dt

( km X o )
k t
k t
X m = ------------------------ ( e m e mu ) or
( k mu k m )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-64

Mathematics Review

( km Xo )
K 1t
K2t
(e
e
)
X m = ------------------------( K2 K1 )

dX mu
------------- = k mu Xm
dt

Both of those equations are identical.

sX mu Xom = k mu X m

4.
the amount of metabolite of a drug in the urine
when the drug is given by IV bolus (assume the parent
compound is not excreted).

Remember at time zero,

X om = 0

Here, again, K1 = km and K2 = kmu

sX mu = k mu X m

Dose

( k mu ) ( k m X o )
sX mu = ----------------------------------------( s + k mu ) ( s + k m )
( k mu ) ( k m ) ( X o )
X mu = -------------------------------------------s ( s + kmu ) ( s + k m )

k mu k m Xo 1 e kmt 1 e kmut
X mu = -------------------- --------------------- -----------------------
k mu k m k m
k mu

km
Xm

kmu

Xmu
5.
the amount of metabolite of a drug excreted in
the urine when both the parent and metabolite are
excreted.

dXm
----------- = k m X k mu X m
dt

Here K1 = km + ku and K2 = kmu

sX m Xom = k m X kmu X m

Dose
sX m + k mu Xm = k m X
X
s + km

o
substitute previously solved X = --------------

Xm

km X o
X m = ----------------------------------------( s + k mu ) ( s + k m )
kmt

kmut

Xu

kmu

Xmu

km

km Xo
X m ( s + k mu ) = -------------s + km

km Xo ( e
e
)
X m = -------------------------------------------------( k mu k m )

ku

dX
------- = k u X k m X
dt
sX Xo = k u X k m X

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-65

Mathematics Review

sX + k u X + k m X = X o
X ( s + ku + km ) = Xo

ku

Xu

( k u + k m ) = K1
Xo
X = --------------s + K1
X = Xo e

K1t

dXm
------------ = k m X k mu Xm = k m X K2Xm
dt
sXm X om = k m X K2Xm

At time zero, all the drug is still in the IV bag, therefore


there is no drug in the body. X0 = 0
Here K1 = ku

dX
------- = Q k u X
dt
---- K u X
sX X0 = Q
s

sX m + K2Xm = k m X

Q
sX + k u X = --s

km Xo
X m = ----------------------------------------( s + K2 ) ( s + K1 )

Q X = -------------------s ( s + ku )

dX mu
------------ = k mu Xm
dt

Q- ( 1 e k u t )
K1t
Q
X = ---)
or X = ------- ( 1 e
ku
K1

sX mu Xom = k mu X m = K2Xm
sX mu = K2X m
( k mu ) ( kmX o )
sX mu = ----------------------------------------( s + K2 ) ( s + K1 )
( k mu ) ( k m ) ( Xo )
X mu = -------------------------------------------s ( s + K2 ) ( s + K1 )
k mu k m X o 1 e K1t 1 e K2t
X mu = --------------------- --------------------- --------------------K2 K1 K1
K2

7.
the amount of drug in the urine when the drug is
given by infusion (assume the drug is NOT metabolized).

ku

Xu

Here K1 = ku

dX
------- = Q k u X
dt
6. The amount of drug in the body from a drug given by
IV infusion (assume no metabolism).

Q- k X
sX Xo = --u
s
QsX + k u X = --s

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-66

Mathematics Review

QX (s + k u ) = --s

dX
------- = Q k m X
dt

Q
X = ------------------------s (s + ku )

Q- k X
sX Xo = --m
s

dX
--------u- = k u X
dt

sX + k m X = Q
---s

sX u Xo = k u X

QX ( s + k m ) = --s

sX u = k u X

Q
X = --------------------s ( s + km )

ku Q
sX u = ------------------------s (s + ku)

kmt

(1 e
)
X = Q -------------------------km

ku Q
X u = --------------------------2
s ( s + ku )

dX m
---------- = k m X k mu X m
dt

e kut
k u Qt
X u = ---------- Qk u 1-------------------
ku
k2

sX m Xo = k m X k mu Xm

kut

K1t

(1 e
)
(1 e
)
X u = Qt Q ------------------------- or Xu = Qt Q -------------------------ku
K1

8.
the amount of metabolite of a drug in the body
from a drug given by IV infusion (assume no parent drug
excretion) Here K1 = km and K2 = kmu

sX m = k m X k mu Xm
km Q
X m = -------------------------------------------s ( s + k mu ) ( s + k m )
A
X m = ---------------(a b)

bt

at

e 1 e -
1---------------- b - ----------------a

1 e k m t 1 e k mut
km Q
X m = ------------------------ -------------------- --------------------- or
( k mu k m ) k m k mu
K1t
K2t
kmQ
e - 1------------------- e -
1-------------------X m = ------------------------( K2 K1 ) K1 K2

X
km
Xm

kmu

Xmu

9.
the amount of metabolite of a drug in the urine
from a drug given by IV infusion (assume that the parent
compound is not excreted). Here K1 = km and K2 = kmu

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-67

Mathematics Review

sX mu Xomu = k mu X m substitute X m

k mu k m Q
X mu = ---------------------------------------------2
s ( s + k m ) ( s + k mu )

k mu k m Qt k mu k m Q ( 1 e kmut ) ( 1 e kmt )
X mu = --------------------- --------------------- ----------------------------- --------------------------
2
2
k m k mu k m k m u

k
k

km

mu

Xm

kmu

Xmu

or

k mu k m Qt k mu k m Q ( 1 e K2t ) ( 1 e K1t )
X mu = --------------------- -------------------- -------------------------- --------------------------
2
2
K1 K2 K1 K2

K2
K1

dX
------- = Q k m X
dt
Q
sX Xo = ---- k m X
s
sX + k m X = Q
---s
X ( s + km ) = Q
---s
Q X = --------------------s ( s + km )

10.
the rate of excretion of the metabolite into the
urine for a drug given by IV bolus when

k m + k u = k mu
In this case, K1 = ku +km and K2 = kmu and thus K1 =
K2. This is not normal but could happen. The problem
arises when we get to the LaPlace that assumes the rate
constants are different (i.e. a b ) because for this special
case a = b .

dX m
---------= k m X k mu X m
dt

Dose

sX m Xo = k m X k mu Xm
X

sX m + k mu X m = k m X
km Q
X m ( s + k mu ) = --------------------s ( s + km )

dX mu
------------ = k mu Xm
dt

Xu

kmu

Xmu

km
Xm

km Q
X m = -------------------------------------------s ( s + k m ) ( s + k mu )
k m Q 1 e kmut 1 e kmt
X m = -------------------- ----------------------- ---------------------
k m k mu k mu k m

ku

dX

. ------- = k u X k m X
dt

sX Xo = k u X k m X
sX + k u X + k m X = X o

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-68

Mathematics Review

the principal metabolite ( X m 1 ) when the drug is

X ( s + ku + km ) = Xo

11.

Xo
X = -----------------------------( s + k u + km )

In this case K1 = km1 + km2 + ku, K2 = kmu1 and K3 =


kmu2

cleared by several pathways ( X u, X m 1, X m 2 )

K1 = ( k u + k m )
k mu = K2

Xm1

Xo
X = -------------------( s + K1 )
X = Xo e

Dose

kmu1

Xmu1

km1

K1t

ku

Xu

dX m
---------= ( k m X k mu X m )
dt
km2

sX m Xom = ( k m X K2X m )

Xm2

sX m + K2X m = k m X
km X o
X m ( s + K2 ) = -----------s+K
km Xo
X m = ----------------------------------------( s + K2 ) ( s + K1 )

X m = k m Xo te

Xmu2

dX
------- = k u X k m1 X k m2 X
dt
(remember- K2 = K1)

km Xo
X m = ----------------------------------------( s + K1 ) ( s + K1 )
km Xo
X m = ---------------------2
( s + K1 )

kmu2

sX Xo = k u X k m1 X k m2 X
sX + k u X + k m1 X + k m2 X = X o
X ( s + k u + k m1 + k m2 ) = Xo Let K1 = ku + km1 + km2

(kmX0 = A)

K1t

dX mu
------------ = k mu Xm
dt
dX mu
K1t
------------ = k mu k m Xo te
dt

Xo
X = -------------------( s + K1 )
K1 = ( k u + k m1 + k m2 ) and K2 = k mu1
dX m1
------------- = k m1 X K2X m1
dt
sX m1 Xm1o = k m1 X K2X m1
X m1o = 0
k m1 Xo
sX m1 + K2X m1 = -------------------( s + K1 )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-69

Mathematics Review

k m1 Xo
X m1 ( s + K2 ) = -------------------( s + K1 )

Xo= 0
ka Q
sX + K1X = -------------------s ( s + ka )

k m1 X o
X m1 = ----------------------------------------( s + K1 ) ( s + K2 )

ka Q
X ( s + K1 ) = -------------------s ( s + ka )

k m1 Xo K1t K2t
(e
e
)
X m1 = -------------------K2 K1

ka Q
X = -----------------------------------------s ( s + k a ) ( s + K1 )
12.
the concentration of drug X Vd in the body
when the drug is given orally by a delivery system which
is zero order. What is the concentration in the body at
equilibrium ( t )
Here K1 = ku

ka

Xa

ku

Xu

k a Q ( 1 e K1t ) ( 1 e kat )
X = ---------------------- --------------------------- -------------------------
ka
K1
( k a K1 )

ka Q
( 1 e K1t ) ( 1 e kat )
C = ----------------------------- --------------------------- -------------------------
( k a K1 )Vd
K1
ka

k Q

1
1
a
If t= , then e kt = 0 , thus C = ----------------------------- ------- -----
( k a K1 )Vd K1

ka

Q
simplified yields: C = --------------

dX
--------a- = Q k a X a
dt
Q
sX a Xao = ---- k a Xa
s

K1Vd

13
the metabolite of a drug in the body Xm given
by IV infusion and concomitant IV bolus dose.
Infusion:
Here K1 = km and K2 = kmu

X a0 = 0

Dose

sX a + k a Xa = ( Q s )
X a ( s + ka ) = ( Q s )
Q X a = -------------------s(s + ka)

X
km

kat

Q( 1 e )
X a = ----------------------------ka

Xm

kmu

Xmu

dX
------- = k a X a K1X
dt
sX Xo = k a Xa k1X

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-70

Mathematics Review

dX
------- = Q K1X
dt

dX m
---------- = k m X K2X m
dt

sX Xo = Q
---- K1X
s

sX m Xo = k m X K2X m

sX + K1X = Q
---s

sX m + K2X m = k m X

X ( s + K1 ) = Q
---s
Q
X = ----------------------s ( s + K1 )
dX m
---------= k m X K2X m
dt

km X o
X m ( s + K2 ) = --------------s + K1
km Xo
X m = ----------------------------------------( s + K1 ) ( s + K2 )
km Xo
K2t
K 1t
X m = ------------------------(e
e
)
( K1 K2 )
Thus,

sX m Xo = k m X K2Xm
sX m + K2X m = k m X
km Q
X m ( s + K2 ) = ----------------------s ( s + K1 )
km Q
X m = -------------------------------------------s ( s + K1 ) ( s + K2 )
k m Q ( 1 e K1t ) ( 1 e K2t )
X m = ------------------------- -------------------------- --------------------------
( K2 K1 )
K1
K2

km X o
K2t
K 1t
(e
e
) + below
X m = ------------------------( K1 K2 )
k m Q ( 1 e K2t ) ( 1 e K1t )
-------------------- -------------------------- --------------------------
K2
K1
K1 K2

14.
By means of the LaPlace transform, find the
equation for the rate of appearance of the tracer in the
urine if the drug were given by IV bolus.
Here K1 = ku + km and K2 = kmu

IV Bolus:

dX
------- = K1X
dt

Dose

sX Xo = K1X

ku

Xu

kmu

Xmu

sX + K1X = X o
km

X ( s + K1 ) = Xo

Xm

Xo
X = --------------s + K1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-71

Mathematics Review

dX
------- = k u X k m X
dt
sX Xo = k u X k m X
sX + k u X + k m X = X o
X ( s + ku + km ) = Xo
( k u + k m ) = K1
Xo
X = -------------------( s + K1 )
X = Xo e

K1t

dX
K1t
--------u- = k m X = k u ( Xo e
)
dt
dX m
---------= k m X K2X m
dt
sX m Xo = k m X k2Xm
k m Xo
sX m + K2X m = -------------------( s + K1 )
km X o
X m ( s + K2 ) = --------------s + K1
km Xo
X m = ----------------------------------------( s + K2 ) ( s + K1 )
km Xo
K1t
K2t
X m = ------------------------{e
e
}
( K2 K1 )
k mu k m Xo K1t K2t
dX mu
------------ = k mu Xm = ------------------------{e
e
}
dt
( K2 K1 )
k mu k m X o
dX u dX mu
K1t
{ e K1t e K2t }
--------- + ------------- = k u ( X o e
) + ------------------------
( K2 K1 )

dt
dt

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

2-72

CHAPTER 3

Pharmacological Response

Author: Michael Makoid and John Cobby


Reviewer: Phillip Vuchetich

OBJECTIVES
After completing this chapter, the student will be able to:
1.

Given patient data of the following types, the student will be able to properly construct (III) a graph and compute (III) the slope using linear regression: response
(R) vs. concentration (C), response (R) vs. time (T), concentration (C) vs. time (T)

2.

Given any two of the above data sets, the student will be able to compute (III) the
slope of the third by linear regression.

3.

Give response vs. time and response versus concentration data, the student will be
able to compute (III) the terminal (elimination) rate constant and half life of the
drug.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-1

Pharmacological Response

3.1 Pharmacological Response


Drug must get into blood
and blood is in contact
with receptor.

3.1.1

One theory (A.J. Clarke) on the mechanism of action of drugs is the occupation
theory. It suggests that the intensity of a pharmacological response (E) is proportional to the concentration of a reversible drug-receptor complex

THE HYPERBOLIC RESPONSE EQUATION


A mathematical description of the occupation theory, assuming complete and
instantaneous drug distribution, yields
[ D ]E max
E = ---------------------KR + [D ]

(EQ 1-35)

where E is the intensity of the pharmacological response, Emax is the maximum


attainable value of E , [ D ] is the molar concentration of free drug at the active complex and K R is the dissociation constant of the drug-receptor complex.

PKAnalyst Plot
1.0

0.8

0.6

If

0.4

is plotted against

[D]

a hyperbolic curve will result; the asymptote will be

E max .

0.2

0.0
0.0

0.8

1.6

2.4

3.2

4.0

a. If linear pharmacokinetics hold, the molar concentration of free drug at the


active site is proportional to the plasma concentration of the drug once equilibrium
has been established. Hence, a plot of E against Cp will also be hyperbolic.
b. Because the mass of drug in the body is
hyperbolic.

X = V Cp ,

a plot of

against

will be

c. For a series of doses the value of X at the same given time after dosing is proportional to the dose (D). Thus, a plot of E against D will also be hyperbolic at a specific time.
d. Any hyperbolic curve, if plotted on reverse semilogarithmic paper (i.e., abscissa
is logarithmic), has a sigmoid shape. If we plot E against Cp (of X , or D ) in this
manner, the plot is virtually linear in the range E Emax = 0.2 0.8 ; and if this is the
clinical range of responses, linear equations may be written. For example,

1.0

Response

0.8
0.6
0.4
0.2
0.0
10 -810-710 -610-510 -410 -310-210 -1

E = m ln x + b

Conc.

where

is the slope

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-2

Pharmacological Response

Plot of Response vs.


Ln(C) is a straight line in
the middle (if you
squint), but only
between 20% and 80%
maximum response

3.1.2

This example equation shows that, in the clinical range, the intensity of a pharmacological response is proportional to the logarithm of the administered dose, providing response is measured at a consistent time after dosing. The proportionality
constant (slope, m ) is a function of the affinity of the drug for the receptor. In fact,
equation yields a log-dose response plot. Note that doubling the dose does not
double the response.

INTERRELATIONSHIPS BETWEEN CONCENTRATION, TIME AND


RESPONSE
Pharmacological Response (R), Concentration (C), and Time (t) are interrelated.
The response and concentration relationship is studied in pharmacology. The concentration and time relationship is studied in pharmacokinetics. The response and
time relationship is applied in therapeutics.

Remember: Use only


the data between 20%
and 80% of maximum
response for the straight
part of both response
vs. Ln(c) and response
vs. t.

You should know what the various graphical relationships look like. Response vs.
natural log of concentration is sigmoidal. (S shaped). We are interested in the middle almost straight part. The slope is dR d ln c .
Response vs. time is a straight line. The slope is

dR dt .

Natural log of concentration vs. time (drug given by IV bolus) is a straight line.
The slope is d ln c dt .
You should be able to obtain the slope of each of these relationships from data sets.
You should be able to obtain the third slopes relationship given the other two (or
data sets with which to get the other two).
dR
dR d ln c
------- = ----------- ----------dt
d ln c dt
dR
dR dt
----------- = -------------------d ln c
d ln c dt

NOTE: Only between


20% and 80% of maximum response!!!!!!

(EQ 1-36)

(EQ 1-37)

d ln c
dR dt
(EQ 1-38)
----------- = ---------------------dt
dR d ln c
You should be able to apply the equation y = mx + b to each of the above relationships. Given the slope (or having obtained the slope) and two of the three variables
(y, x, b), you should be able to find the third.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-3

Pharmacological Response

3.2 Change in Response with Time


3.2.1

ONE-COMPARTMENT OPEN MODEL: INTRAVENOUS BOLUS


INJECTION
X = X0 e

Kt

= De

Kt

(EQ 1-39)

or
Ln ( X ) = Ln ( D ) Kt

(EQ 1-40)

Substituting twice from eq. once at time t and once at zero time
E
b = E
0b
------------------------- Kt E = E0 Rt
m
m

(EQ 1-41)

Hence a plot of the intensity of the pharmacological response at any time ( E )


against time declines linearly. The slope is R = ( K m ) and the intercept is E0 (the
initial intensity).

3.2.2

ONE-COMPARTMENT OPEN MODEL: ORAL ADMINISTRATION

Response follows
plasma profile.

3.2.3

Because E is proportional to ln x at any time, a plot of E against t will be analogous to a plot of ln x against t . Hence E will rise at first and then decline with time.
When t is large, the terminal slope will be R .

DURATION OF EFFECTIVE PHARMACOLOGICAL RESPONSE ( t dur )

Duration of action is
related to how long
plasma concentration is
above Minimum Effective Concentration.

Once equilibrium has been established, there is a minimum plasma concentration


below which no pharmacological response is seen; this concentration is ( C p ) eff or
MEC . For an intravenous bolus injection, the time to reach ( C p ) eff is t dur .
( C p )eff = ( C p ) 0 e

Kt dur

multiplying by the volume of distribution we obtain


ln ( X eff ) = ln ( D ) Kt dur

(EQ 1-42)

Rearranging,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-4

Pharmacological Response

t dur

D
ln -------X
eff
= ------------------K

(EQ 1-43)

The duration of effective pharmacological response is proportional to the (natural)


logarithm of the dose. A second rearangement of equation 1-42 results in :
ln ( Dose ) ln ( Xeff )
t dur = ----------------------- -----------------K
K

(EQ 1-44)

Thus a plot of duration of action vs ln dose would result in a straight line with a
slope of 1/K and an x intercept of

3.2.4

ln ( X eff )
.
------------------K

PHARMACOKINETIC PARAMETERS FROM RESPONSE DATA

How can I get the elimination rate constant


from pharmacological
data? Use this cookbook.

The measurement of pharmacological effect provides a non-invasive means of


obtaining the value of t 1 2 (but not V ).

Remember: Use only


the data between 20%
and 80% of maximum
Response for both of
these plots.

b. Find the slope

3.2.5

a. Obtain a log dose-response plot (Eq. 1-37). The response must always be measured at the same time after administering the dose.
(m)

of this plot.

c. Obtain a response against time plot for a single dose (Eq. 1-36).
d. Find the terminal slope
e. Calculate

R
K = ---- .
m

f. Calculate

0.693
t 1 2 = -------------
K

( R )

of this plot.

DELAYED RESPONSE

Two compartment
model - biophase is in
second compartment.

If a drug does not distribute instantaneously to all the body tissues (including the
active site), the pharmacological response will not always parallel the drug concentrations in the plasma. In such a situation the response may parallel the mass of
drug presumed to be in a second compartment ( X 2 ) , and hence seem delayed.
Eventually, however, once equilibrium is attained, the response will parallel
plasma concentrations. In such a case, E is proportional to ln X 2 .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-5

Pharmacological Response

Thus a plot of E against X1 (or E against Cp ) will show a hysteresis loop with time,
most noticeably during an intravenous infusion.

3.2.6

RESPONSE OF ACTIVE METABOLITE:

Parent compound (inactive) yields active


daughter compound.

In the case of an inactive prodrug yielding an active metabolite, the response


curves will mirror the active metabolite plasma profile (assuming the biophase is
the plasma) and not the prodrug plasma profile.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-6

Pharmacological Response

3.3 Therapeutic Drug Monitoring


Part of Pharmaceutical
Care!

The pharmacokinetics of a drug determine the blood concentration achieved from


a prescribed dosing regimen. During multiple drug dosing, the blood concentration
will reflect the drug concentration at the receptor site; and it is the receptor site
concentration that determines the intensity of the drugs effect. Therefore, in order
to predict a patients response to a drug regimen, both the pharmacokinetics and
pharmacological response characteristics of the drug must be understood. Pharmacological response is closely related to drug concentration at the site of action.
We can measure plasma concentration and assume that the site of action is in rapid
equilibrium with the plasma since we usually do not measure drug concentration in
the tissue or at the receptor site. This assumption is called kinetic homogeneity
and is the basis for clinical pharmacokinetics.

Need to keep plasma


concentration in the
therapeutic range to
optimize therapy.

There exists a fundamental relationship between drug pharmacokinetics and pharmacologic response. The relationship between response and ln-concentration is
sigmoidal. A threshold concentration of drug must be attained before any response
is elicited at all. Therapy is achieved when the desired effect is attained because
the required concentration has been reached. That concentration would set the
lower limit of utility of the drug, and is called the Minimum Effective Concentration (MEC). Most drugs are not clean, that is exhibit only the desired therapeutic response. They may also exhibit undesired side effects, sometimes called toxic
effects at a higher, (hopefully a lot higher), concentration. At some concentration,
these toxic side effects become become intolerable/and or dangerous to the
patient.. That concentration, or one below it, would set the upper limit of utility
for the drug and is called the Maximum Therapeutic Concentration or Minimum
Toxic Concentration (MTC). Patient studies have generated upper (MTC) and
lower (MEC) plasma concentration ranges that are deemed safe and effective in
treating specific disease states. These concentrations are known as the therapeutic
range for the drug (Table 2-18).
When digoxin is administered at a fixed dosage to numerous subjects, the blood
concentrations achieved vary greatly. Clinically, digoxin concentrations below 0.8
ng ml will elicit a subtherapeutic effect. Alternatively, when the digoxin concentration exceeds 2.0 ng ml side effects occur (nausea and vomiting, abdominal pain,
visual disturbances). Drugs like digoxin possess a narrow therapeutic index
because the concentrations that may produce toxic effects are close to those

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-7

Pharmacological Response

required for therapeutic effects. The importance of considering both pharmacokinetics and pharmacodynamics is clear.
TABLE 2-18 Average

therapeutic drug concentration

DRUG

RANGE

digoxin

0.8-2.0 ng ml

gentamicin

2-10 g ml l

lidocaine

1-4 g ml

lithium

0.4-1.4 mEq L

phenytoin

10-20 g ml

phenobarbitol

10-30 g ml

procainamide

4-8 g ml

quinidine

3-6 g ml

theophylline

10-20 g ml

Note that drug concentrations may be expressed by a variety of units.


Pharmacokinetic factors that cause variability in plasma drug concentration are:

drug-drug interactions
patient disease state
physiological states such as age, weight, sex
drug absorption variation
differences in the ability of a patient to metabolize and eliminate the drug

If we were to give an identical dose of drug to a large group of patients and then
measure the highest plasma drug concentration we would see that due to individual
variability, the resulting plasma drug concentrations differ. This variability can be
attributed to factors influencing drug absorption, distribution, metabolism, and
excretion. Therefore, drug dosage regimens must take into account any disease
altering state or physiological difference in the individual.
Therapeutic drug monitoring optimizes a patients drug therapy by determining
plasma drug concentrations to ensure the rapid and safe drug level in the therapeutic range.
Two components make
up the process of
therapeutic drug
monitoring:

Assays for determination of the drug concentration in plasma


Interpretation and application of the resulting concentration data to develop a safe and effective
drug regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-8

Pharmacological Response

The major potential advantages of therapeutic drug monitoring are the maximization of therapeutic drug benefits and the minimization of toxic drug effects. The
formulation of drug therapy regimens by therapeutic drug monitoring involves a
process for reaching dosage decisions.

3.3.1

THERAPEUTIC MONITORING: WHY DO WE CARE?


The usefulness of a drugs concentration vs. time profile is based on the observation that for many drugs there is a relationship between plasma concentration and
therapeutic response. There is a drug concentration below which the drug is ineffective, the Minimum Effective Concentration (MEC), and above which the drug
has untoward effects, the Minimum Toxic Concentration (MTC). That defines the
range in which we must attempt to keep the drug concentration (Therapeutic
Range).
The data in Table 2-18 are population averages. Most people respond to drug concentrations in these ranges. There is always the possibility that the range will be
different in an individual patient.
For every pharmacokinetic parameter that we measure, there is a population average and a range. This is normal and is called biological variation. People are different. In addition to biological variation there is always error in the laboratory assays
that we use to measure the parameters and error in the time we take the sample.
Even with these errors, in many cases, the therapy is better when we attempt to
monitor the patients plasma concentration to optimize therapy than if we dont.
This is called therapeutic monitoring. If done properly, the plasma concentrations
are rapidly attained and maintained within the therapeutic range throughout the
course of therapy. This is not to say all drugs should be monitored. Some drugs
have a such a wide therapeutic range or little to no toxic effects that the concentrations matter very little. Therapeutic monitoring is useful when:

a correlation exists between response and concentration,


the drug has a narrow therapeutic range,
the pharmacological response is not easily assessed, and
there is a wide inter-subject range in plasma concentrations for a given dose.

In this era of DRGs, where reimbursement is no longer tied to cost, therapeutic


monitoring of key drugs can be economically beneficial to an institution. A recent
study (DeStache 1990) showed a significant difference with regard to length of
stay in the hospital between the patients on gentamicin who were monitored (and
their dosage regulated as a consequence) vs. those who were not. With DRGs the
hospital was reimbursed a flat fee irrespective of the number of days the patient
stayed in the hospital. If the number of days cost less than what the DRG paid, the

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-9

Pharmacological Response

hospital makes money. If the days cost more than the hospital loses money. This
study showed that if all patients in the hospital who were on gentamicin were monitored, the hospital would save $4,000,000. Thats right FOUR MILLION per year.
I would say that would pay my salary, with a little left over, and that is only one
drug!
The process of
therapeutic monitoring
takes effort.

First the MD must order the blood assays.


Second, someone (nurse, med tech, you) must take the blood.
Someone (lab tech, you) must assay the drug concentration in the blood.
You must interpret the data.
You must communicate your interpretation and your recommendations for dosage regimen
change to the MD. This will allow for informed dosage decisions.

You must follow through to ensure proper changes have been made.
You must continue the process throughout therapy. Therapeutic drug monitoring, in many cases,
will be part of your practice. It can be very rewarding.

Thus, if we have determined the therapeutic range, we could use pharmacokinetics


to determine the optimum dosage regimen to maintain the patients plasma concentration within that range.
Selected References

1. Nagashima, R., OReilly, RA., and Levy, G, Kinetics of pharmacologic effects in man: the anticoagulant action of warfarin. Clin.
Pharm. Therap, 10 22-35 (1969).
2. Wagner, J.G, Relations between drug concentration and response. J. Mond. Pharm., 4, 279-310 (1971).
3. Gibaldi M. and Levy, G. Dose-dependent decline of pharmacologic effects of drugs with linear pharmacokinetics characteristics. J.Pharm.Sci, 61, 567-569 (1972).
4. Brunner, L., Imhof, P., and Jack, D. Relation between plasma concentrations and cardiovascular effects of oral oxprenolol in
man. Europ. J. Clin. Pharmacol., 8, 3-9 (1975).
5. Galeazzi, R.L., Benet, L.Z., and Sheiner, L.B. Relationship between the pharmacokinetics and pharmacodynamics of procainamide. Clin. Pharm. Therap., 20, 67-681 (1976).
6. Joubert, P., et al. Correlation between electrocardiographic changes, serum digoxin, and total body digoxin content. Clin.
Pharm. Therap., 20, 676-681 (1976).
7. Amery, A., et al. Relationship between blood level of atenolol and pharmacologic effect. Clin. Pharm. Therap., 21, 691-699
(1977).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-10

Pharmacological Response

3.4 Problems
What to do.---> We want to get pharmacokinetic data (elimination rate constant) from pharmacological response data (Response vs concontration and
Response vs time graphs) .
Response vs Time Graph
1. Plot Response vs Time on Cartesian (regular) Graph Paper.
2. Use Response data between 20 and 80 percent of maximum (Pick the straight part) to do the linear regression on. (Rule of thumb: Connect first and last data point with a straight line. If all
the points fall on one side of the line, its not straight!

1.0

Response

0.8

0.6

3. Find the slope of the straight line, dR


------- , (eyeball the rise over the run or use linear regression as
dT

0.4

0.2

0.0
10

10

10
Time

10

10

10

required). Important: you must determine the best fit line through all of the points that you will
use. Eyeball method: Get the line as close to the points as possible placing as many points
above the line as below the line. Take two points on the line (not data points) to calculate the
change in Y over the change in X.

Response vs Ln(Concentration) Graph


1. Turn semi-log paper on its side so that the numbers are on the top.

0.6
0.4
0.2
0.0
10 -810-710 -610-510 -410-310 -210 -1

Response

Response

10010

10

0.8

10 10

10010

1.0

1010

Conc.

Concentration

10

What we are attempting to do is get the logarithm part of the paper on the x axis and have the
numbers get bigger as you go from left to right.
2. Plot concentration on the x axis and response on the y.
3. Find the slope of the line plotted this way by the rise over the run method.
Run is change in ln(C).
If you take any two concentrations such that C2 = 2*C1 then the run is (ln(C2) - ln(C1)).
Using rules of logs, when two logs are subtracted, the numbers are devided, thus: = ln(C2/C1).
If C2 = 2*C1 then ln(C2/C1) = ln(2) = 0.693.
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-11

Pharmacological Response

Rise = change in Response.


Take the difference of the two responses coresponding to the concentrations picked. (R2-R1).
R 2 R1
4. The slope of the line is m = rise
-------- = -----------------run

0.693

Ln(Concentration) vs Time Graph (Pharmacokinetic Data)


If you have concentration vs Time data:
1. Plot Concentration vs time on semi-log paper (Y axis is concentration this time)

Concentration

10
100

1010

110
0

Time

2. Find the slope as before, using semi log paper (Remeber the log is on the Y axis this time, so you
find two concentrations such that c2 = 2*c1 and put it in the rise this time. Thus the slop of the
0.693 = 0.693
line is m = rise
-------- = -------------------------- = k
run

t2 t1

t1
-2

If you have pharmacological response data:


1. Divide the slope of the Response vs Time graph by the slope of the Response vs ln(C) graph:
dR
------slope of r vs t
dT
dln(C)
------------------------------------------ = --------------- = --------------- = m = k
slope of r vs ln(c)
dR
dT
--------------dln(C)

Both methods should be equivalent.


Additional problems are available in chapter 14, practice exams.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-12

Pharmacological Response

Oxpranolol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 0 - 1)

AHFS 00:00.00
GPI: 0000000000

Brunner et al, Europ. J. Clin. Pharmacol., 8, 3-9 (1975).

In humans, the pharmacological response to oxpranolol (a beta blocker) is a decrease in beats per minute (bpm) compared to placebo during physical exercise. The following approximate mean data is from 7 healthy volunteers: beats per
minute (bpm) altered with time (t) after oral administration of three doses (D).
TABLE 2-19

Response vs
Concentration

Response vs
time

BPM

Dose (mg)

BPM

Time (hr)

10

40

17.6

13.5

60

13.9

16

80

10.2

19

120

6.6

21

160

TABLE 2-20 Oxpranolol

plasma concentration following 160 mg IV dose

Time (min)

C p ng
------
ml

30

699

60

622

120

413

150

292

240

152

360

60

480

24

1. Calculate the half life ( t 1 2 ) of oxpranolol from the pharmacological response table.
2. Plot plasma concentration data on Cartesian graph paper directly as well as transforming Cp into ln C p .
3. Plot plasma concentration data on semilog paper. Use linear regression to find the rate constant of elimination of
oxpranolol.
4. Calculate the half life obtained from the concentration data and compare it with the half life calculation based on the
pharmacological response.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-13

Pharmacological Response

Minoxidil
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 0 - 2)

AHFS 00:00.00
GPI: 0000000000

Shen et al. Clin. Pcol. Ther 17:593-8 (1975)

Minoxidil is a potent antihypertensive which lowers the mean arterial blood pressure (MAP) in certain patients.
PROBLEM TABLE 0 - 2. Minoxidil

Initial decrease in MAP ( mmHg )

Dose ( mg )

17

2.5

40

5.0

53

7.5

63

10.0

76

15

PROBLEM TABLE 0 - 2. Minoxidil

25 mg I.V. Bolus yielded:


Decrease in MAP ( mmHg )

Time ( hr )

75

20

66

30

56

40

48

50

From the preceding information, determine the following:

dR
d ln C

1. Graph and find ------------ (slope of (R)esponse vs. ln(C)oncentration graph).

dR
dt

2. Graph and find ------- (slope of (R)esponse vs. (T)ime graph).

dR-----dt
3. Find the ln(C)oncentration vs. (T)ime slope : ------------- : Note that your slope m = K . If you are having problems
dR
-----------d ln C
understanding this, refer to Sections 2.4.2 -2.4.4. K is the elimination rate constant.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-14

Pharmacological Response

------------- .
4. Calculate t 1 2 = 0.693
K

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-15

Pharmacological Response

Propranolol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 0 - 3)

AHFS 00:00.00
GPI: 0000000000

Citation?

Beta blockers can be considered first line drugs of choice in the treatment of hypertension in certain patients. The following data was obtained regarding Propranolol used to treat hypertension in a group of patients.
PROBLEM TABLE 0 - 3. Propranolol

Fall in Systolic BP (mmHg)

Cp

20

50

16

40

11

30

20

PROBLEM TABLE 0 - 3. Propranolol

I.V. Bolus dose of Propranolol


Fall in Systolic BP (mmHg)

Time (hr)

24

20

19

From the preceding information, determine the following:

dR
d ln C

1. Graph and find ------------ (slope of (R)esponse vs. ln(C)oncentration graph).

dR
dt

2. Graph and find ------- (slope of (R)esponse vs. (T)ime graph).

dR
------dt
3. Find the ln(C)oncentration vs. (T)ime slope : ------------- : Note that your slope m = K . If you are having problems
dR
-----------d ln C
understanding this, refer to Sections 2.4.2 -2.4.4.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-16

Pharmacological Response

------------- .
4. Calculate t 1 2 = 0.693
K

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-17

Pharmacological Response

3.4.1

ANSWERS:

OXPRANOLOL

1. Calculate the half life ( t 1 2 ) of oxpranolol from the pharmacological response table

Oxpranolol

Response (BPM)

22
20
18
16
14
12
10
1

10

10

10

Dose (mg)

TABLE 3.

X
ln(Dose)

Dose

Y
Response

3.689

40

10

13.61

36.89

4.094

60

13.5

16.76

55.27

4.382

80

16

19.20

70.11

4.787

120

19

22.92

90.96

5.075

160

21

25.75

106.58

Y = 79.5

X = 98.25

X = 22.03

XY

XY = 359.82

( X ) = 485.23

( x ) ( y ) ) ( n ( x y ) )m = (-------------------------------------------------------------------2
2
[(x) ] ( n (x ))

y
y = ------ = 15.9
n

X
X = --------- = 4.41
n

Slope of the line from linear regression. Chapter 2.4.4

22.03 79.5 ) ( 5 359.82 )


m = (------------------------------------------------------------------- = 7.93
485.32 ( 5 98.25 )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-18

Pharmacological Response

dR = 7.93
----------the slope is equal to the linear regression of the change in response vs. ln concentration.
d ln c

OXPRANOLOL
18

Response (BPM)

16
14
12
10
8
6
1.0

1.5

2.0

2.5

3.0

3.5

4.0

Time (hr)

R R
T1 T 2

16 10
1.45 3.07

dR
dt

1
2
- = --------------------------- = 3.71 therefore, ------- = 3.71 .
The slope of this plot is m = -----------------

dR
------dt = d----------ln c = k = ------------3.71 = 0.4678hr 1
----------dR
dt
7.93
----------d ln c

ln 2 = -------------------------0.693 - = 1.48hr
t 1 2 = -------half life (89 min).
1
k
0.4678hr

2. Plot plasma concentration data on Cartesian graph paper directly as well as transforming Cp into ln C p .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-19

Pharmacological Response

Plasma concentration vs. Time

Oxpranolol

800
3

10

Concentration (ng/mL)

640

Concentration (ng/ml)

480

320

160

10

10

0
0

100

200

300

400

100

500

200

300

400

500

Time (min)

Time (min)

3. Plot plasma concentration data on semilog paper. Use linear regression to find the rate constant of elimination of
oxpranolol.
Using linear regression, as described above, the elimination rate constant is approximately
0.007797 min-1 * (60 min/hr) = 0.4678 hr-1
4. Calculate the half life obtained from the concentration data and compare it with the half life calculation based on the
pharmacological response.

0.693
t 1 2 = ------------------- = 90min = 1.5 hrs compared to 1.48 hours (89 min) from the pharmacological response
0.00763
method.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-20

Pharmacological Response

3.4.2

ANSWERS:

MINOXIDIL

NOTE: Answers will vary depending on whether linear regression is calculated


via calculator or using the formula such as observed in Problem 1. Either method
can be used. However, if you use the formula, you should be within 10% of the
calculated answer. A word of caution: if you choose to do linear regression via
calculator make sure you have valid data. This cannot be assured until you have
graphed all the data points given. Many a student has incorrectly calculated
parameters because he/she falsely assumes that all the points are valid. Blindly
choosing data points for linear regression will only lead to error. Every problem
in this manual has been derived from actual journal articles and will therefore be
real data. This real-world data is inexact.
dR (slope of (R)esponse vs. ln(C)oncentration graph).
1. Graph and find -----------mHg)

d ln C

R vs Ln(C)
80
60
40
20
0
10

10

10

Dose (mg)

dR = 32.96
-----------d ln C

dR
dt

2. Graph and find ------- (slope of (R)esponse vs. (T)ime graph).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-21

Pharmacological Response

mmH

R vs T
75
70
65
60
55
50
45
20

25

30

35

40

45

50

Time (hr)

dR
------- = 0.91
dt
dR-----dt
3. Find the ln(C)oncentration vs. (T)ime slope : ------------- : Note that your slope m = K .
dR
-----------d ln C
K = 0.028hr

------------0.91 = ( 0.028 )
32.96

4. Calculate t 1 2 = 0.693
------------- .
K

0.693 - = 24.75hr
t 1 2 = ---------------------- 1
0.028hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-22

Pharmacological Response

3.4.3

ANSWERS:

PROPRANOLOL

dR (slope of (R)esponse vs. ln(C)oncentration graph).


1. Graph and find -----------d ln C

R vs T
20
15
10
5
0
10

10

dR = 16.36
-----------d ln C
dR
dt

2. Graph and find ------- (slope of (R)esponse vs. (T)ime graph).

R vs T
25
20
15
10
5
0

dR
------- = 2.93
dt

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-23

Pharmacological Response

dR-----dt
3. Find the ln(C)oncentration vs. (T)ime slope : ------------- : Note that your slope m = K .
dR
-----------d ln C
K = 0.179hr

------------2.93 = 0.179
16.36

------------- .
4. Calculate t 1 2 = 0.693
K

0.693 - = 3.87hr
t 1 2 = ----------------------
1
0.179hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

3-24

CHAPTER 4

I.V. Bolus Dosing

Author: Michael Makoid and John Cobby


Reviewer: Phillip Vuchetich

OBJECTIVES
For an IV one compartment model plasma and urine:
1.

Given patient drug and/or metabolite concentration, amount, and/or rate vs. time
profiles, the student will calculate (III) the relevant pharmacokinetic parameters
available from IV plasma, urine or other excreta data: e.g.

V d, K, k m, k r, AUC, AUMC, CL, MRT, t 1 2


2.

The student will provide professional communication regarding the pharmacokinetic parameters obtained to patients and other health professionals.

3.

The student will be able to utilize computer programs for simulations and data
analysis.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-1

I.V. Bolus Dosing

4.1 I.V. Bolus dosing of Parent compound


4.1.1

PLASMA

Valid equations:
(Obtained from the
LaPlace transforms
derived from the
appropriate models
derived from the
pharmacokinetic
descriptions of the drug)

ln C p = K1 t + ln C p 0

(EQ 4-1)

ln X = K1 t + ln X0

(EQ 4-2)

C p = C p0 e

( )

AUC =

Cp dt

K1t

(EQ 4-3)

DC p 0 = ----Vd

(EQ 4-4)

0.693
t = ------------K

(EQ 4-5)

( Cp n + Cp n + 1 )
Cp last
t + -------------= ------------------------------------2
K1

(EQ 4-6)

0
( )

AUMC =

t C p dt =

Cp last

( t last Cp last )

(EQ 4-7)

K1

Utilization:
Can you determine the
slope and intercept from
a graph? Plot the data
in table 4 -1.on semi-log
graph paper. Extrapolate the line back to time
= 0 to get Cp0. Find the
half life. Calculate the
elimination rate constant.

( t n Cp n ) + ( t n + 1 Cp n + 1 )

- t + --------------- + --------------------------------- ------------------------------------------------------------------


2
2
K1

MRT = AUMC
-----------------AUC

(EQ 4-8)

Cl = K Vd

(EQ 4-9)

You should be able to plot a data set Concentration vs. time on semilog yielding a straight line
with slope = K1 and an intercept of C p0 .

TABLE 4-1.

Nifedipine 25 mg IV bolus

Time (hr)

Cp
(mcg/L)

139

65.6

31.1

14.6

FIGURE 4-1.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-2

I.V. Bolus Dosing

Does your Graph look


like this?

FIGURE 4-1.

Nifedipine IV Bolus (25 mg IV Bolus)

100

Concentration (ng/mL)

Concentration (mic/L)

10 3

Cp0 = 295 mic/L


-K1 = -0.375 hr -1

10 2

50

1.85 hr
10 1
0

Time (hours)

Time (hr)

You should be able to determine K1. A plot of the data in TABLE 4-1. results in FIGURE 41. Remember from high school algebra, the slope of any straight line is the rise over the run,
dy
------ , In the case of semi-log graphs dy is the difference in the logarithms of the concentrations.
dx

Thus, using the rules of logarithms, when two logs are subtracted, the numbers themselves are
divided. i.e. ln ( C1 ) ln ( C2 ) = ln C1
------- . Thus if we are judicious in the concentrations that we
C2
take, we can set the rise to a constant number. So, if we take any two concentrations such that
one concentration is half of the other (In FIGURE 4-1. above, we took 100 and 50), the time it
takes for the concentration to halve is the half life (in the graph above, 1.85 hr). Then
1
0.693
0.693
K1 = ------------- = ---------------- = 0.375 hr
t
1.85hr

You should be able to determine V d :. To do this, extrapolate the line to t = 0 . The value of Cp
when t = 0 is C p0 (in the graph above, C p0 = 295 mic
--------- which is equal to D V d for an IV bolus
L

dose only.
Dose
Dose
25mg 1000mic
Thus, Cp 0 = ------------ , V d = ------------- = ------------------ --------------------- = 85L
Vd

Cp 0

295mic
-----------------L

mg

The volume of distribution is a mathematical construct. It is merely the proportionality constant


between two knowns - the C p0 which results from a given D 0 . It is, however, useful because it
is patient specific and therefore can be used to predict how the patient will treat a subsequent
dose of the same drug. You should be able to obtain the volume of distribution from graphical
analysis of the data. Pay attention to the units! Make sure that they are consistent on both sides
of the equation. NOTE: the volume of distribution is not necessarily any physiological space.
For example the approximate volume of distribution of digoxin is about 600 L If that were a
physiological space and I were all water, that would mean that I would weigh about 1320
pounds. I'm a little overweight (I prefer to think that I'm underheight), but REALLY!

Given any three of the variables of the IV bolus equation, either by direct information (the volume of distribution is such and such) or by graphical data analysis, you should be able to find
the fourth.

You should be able to calculate Area Under the Curve (AUC) from IV Bolus data (Time vs. Cp).
From the above data in TABLE 4-1. the AUC is calculated using (EQ 4-6):

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-3

I.V. Bolus Dosing

()

AUC =

Cp dt
0

Cp n + Cp n + 1 Cp l
= --------------------------------+ -------- which in this case is:

K1
t

Cp 1 + Cp 2
Cp 2 + Cp 3
Cp 3 + Cp last
Cp last
Cp o + Cp 1
t last + -------------- ------------------------- t 1 + ------------------------- t 2 + ------------------------- t 3 + ------------------------------
K1
2
2
2
2

295 + 139
139 + 65.6
65.6 + 31.1
31.1 + 14.6
14.6 mcg
------------------------ 2 + ------------------------- 2 + --------------------------- 2 + --------------------------- 2 + ------------- ----------hr or
2
2
2
2
0.375 L

mcg
mcg
{ 434 + 204.6 + 96.7 + 45.7 + 38.9 } ----------hr = 819.9----------hr . In tabular format, the AUC calculation
L
L

is shown in TABLE 4-2.

TABLE 4-2 AUC


t
t1

AUC

AUC

t
0

TIME

Cp

295

139

434.0

434.0

65.6

204.6

638.6

31.1

96.7

735.3

14.6

45.7

781.0

38.9

819.9

The AUC of a plot of plasma concentration vs. time, in linear pharmacokinetics, is a number
which is proportional to the dose of the drug which gets into systemic circulation. The proportionality constant, as before, is the volume of distribution. It is useful as a tool to compare the
amount of drug obtained by the body from different routes of administration or from the same
route of administration by dosage forms made by different manufacturers (calculate bioavailability in subsequent discussions).
The AUC of a plot of Rate of Excretion of a drug vs. time, in linear pharmacokinetics, is the
mass of drug excreted into the urine, directly.

You should be able to calculate the AUMC from IV Bolus data (Time vs. Cp). The equation for
AUMC is equation 4-7:
( )

AUMC =

t C p dt =

( t n Cp n ) + ( t n + 1 Cp n + 1 )

Cp last

( t last Cp last )

- t + --------------- + ---------------------------------- which in the


------------------------------------------------------------------
2
2
K1
K1
0

data given in TABLE 4-1. is:


T0 C po + T1 C p1
T1 C p1 + T2 C p2
T 2 C p 2 + T3 C p3
---------------------------------------------- t 1 + ---------------------------------------------- t 2 + ---------------------------------------------- t 3 +
2
2
2
T 3 C p 3 + T last C p last
T last C p last Cp last
- + --------------- and thus,
--------------------------------------------------------- t last + -----------------------------2
2
K1
K1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-4

I.V. Bolus Dosing

295 + 2 139
2
65.6 + 6 31.1 2 mcg
139 + 4 65.6 2 + 4----------------------------------------- 2 + 2--------------------------------------- 0------------------------------------------------hr +
2
L
2
2

6 31.1 + 8 14.6
8 14.6
14.6 mcg 2
------------------------------------------ 2 + ------------------ + ---------------2- ----------hr or
2
0.375

0.375 L

mcg 2
{ 278 + 540.4 + 449 + 303.4 + 311.47 + 103.82 } = 1986.1----------hr
L

Thus in tabular format the AUMC for data given in TABLE 4-1. is TABLE 4-3 below.
TABLE 4-3 AUMC

Cp*T

AUMC t

AUMC

TIME

Cp

295

139

278

278.0

278.0

65.6

262.4

540.4

818.4

31.1

186.6

449.0

1267.4

14.6

116.8

303.4

1570.8

415.3

1986.1

t
0

The AUMC is the Area Under the first Moment Curve. A plot of T*Cp vs. T is the first
moment curve. The time function buried in this plot, the Mean Residence Time (MRT), can be
extracted using equation 4-8 below.
It is the geometric mean time that the molecules of drug stay in the body. It has utility in the fact
that, as drug moves from the dosage form into solution in the gut, from solution in the gut into
the body, and from the body out, each process is cumulatively additive. That means if we can
physically separate each of these processes in turn, we can calculate the MRT of each process.
The MRT of each process is the the inverse of the rate constant for that process.

You should be able to calculate MRT from IV Bolus data (Time vs. Cp) using equation 4-8
AUMC
1986.1
MRT = ------------------ = ---------------- = 2.42
AUC
819.9

Since there is only the process of elimination (no release of the drug from the dosage form, no
absorption), the MRT is the inverse of the elimination rate constant, K. Thus MRT = 1/K.
Flow Chart 2-1

IV Bolus

MRT(IV) = 1/K

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-5

I.V. Bolus Dosing

Suppose the drug were given in a solution. Then the drug would have to be absorbed and then
eliminated. Since the MRTs are additive, the MRT of the oral solution would be made up of the
MRTs of the two processes, thus:
Flow Chart 2-2

Oral Solution
Ka
Xa

K
X

MRT(os) = MAT(os)+MRT(IV)
MRT(os) =

1/Ka

+ 1/K

Consequently, if a drug has to be released from a dosage form for the drug to get into solution
which is subsequently absorbed, a tablet for example, the MRT of the tablet will consist of the
MRT(IV) and the MAT(os) and the Mean Dissolution Time (MDT), thus:
Flow Chart 2-3

Tablet

Kd

Ka

Xd

Xa

K
X

MRT(tab) = MDT

+ MAT(os)

+ MRT(IV)

MRT(tab) =

1/Kd

1/Ka

1/K

MRT(tab) = MAT(tab)

+ MRT(IV)

MRT(tab) = 1/Ka (apparent)

1/K

Normally, we dont have information from the oral solution, just IV and tablet. So in that case
the information obtained about absorption from the tablet is bundled together into an apparent
absorption rate constant consisting of both dissolution and absorption.
It should be apparent that this is a reasonably easily utilized and powerful tool used to obtain
pharmacokinetic parameters.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-6

I.V. Bolus Dosing

4.1.2

IV BOLUS, PARENT COMPOUND, PLASMA PROBLEMS

Equations used in this


section:

1.

K1 = slope from equation 4-3

2.

ln 2 equation 4-5
t 1 2 = -------K1

3.

1
AUMC
MRT = ------- ( estimate ) MRT = ------------------ equation 4-8
K1
AUC

4.

Cp 0 = the y-intercept of the line from equation 4-3

5.

Cp
Estimate for AUC = AUC = ---------0 which is
K1
()

AUC =

0 Cp dt

( Cp n + Cp n + 1 )
Cp last
Cp dt = ------------------------------------(
t
)
+
-------------

2
K1

Trapezoidal rule applied to equation 4-6


6.

Estimate for AUMC = AUMC = AUC MRT from equation 4-8


( )

AUMC

Cp dt =

( t n Cp n ) + ( t n + 1 Cp n + 1 )

Cp last

( t last Cp last )

- t n + --------------- + --------------------------------- ------------------------------------------------------------------


2
K1
2
0

K1

from equation 4-7


7.

V d = Dose
------------- from equation 4-4
Cp 0

8.

Cp 0 Dose Dose
Cl = K1 V d = ----------- ------------- = ------------AUC Cp 0
AUC

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-7

I.V. Bolus Dosing

Acyclovir
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 1)

AHFS 12:34.56 Antivirals


GPI: 0000000000 Antivirals

De Miranda and Burnette, Metabolic Fate and Pharmacokinetics of the Acyclovir Prodrug Valaciclovir in Cynomolgus Monkeys, Drug Metabolism and Disposition (1994): 55-59.

Acyclovir is an antiviral drug used in the treatment of herpes simplex, varicella zoster, and in suppressive therapy. In
this study, three male cynomolgus monkeys were each given a 10 mg kg intravenous dose. The monkeys weighed an
average of 3.35 kg each. Blood samples were collected and the following data was obtained:
PROBLEM TABLE 4 - 1.

Acyclovir
Serum concentration

Time (hours)

( g mL )

0.167

26.0

0.300

23.0

0.500

19.0

0.75

16.0

1.0

12.0

1.5

7.0

2.0

5.0

From the data presented in the Preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-8

I.V. Bolus Dosing

(Problem 4 - 1) Acyclovir:

CONCENTRATION (MIC/ML)

10

10

100
0.0

0.5

1.0

1.5

2.0

TIME (HR)
1

1.

k = 0.93hr

2.

t = 0.75hr .

3.

MRT = 1.08hr .

4.

( C p )0 = 30.4ug mL .

5.

AUC = 32.75ug mL hr .

6.

AUMC = 35.2ug mL hr .

7.

V d = 1.1L

8.

Cl = 1.02L hr .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-9

I.V. Bolus Dosing

Aluminum

(Problem 4 - 2)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Xu, Pai, and Melethil, "Kinetics of Aluminum in Rats. II: Dose-Dependent Urinary and Biliary Excretion", Journal of Pharmaceutical Sciences, Oct 1991, p 946 - 951.

A study by Xu, Pai, and Melethil establishes the pharmacokinetics of Aluminum in Rats. In this study, four rats with an
average weight of 375g, were given an IV bolus dose of aluminum (1 mg/kg). Blood samples were taken at various
intervals and the following data was obtained:
PROBLEM TABLE 4 - 2.

Aluminum

Time (hours)

ngSerum concentration, ------mL

0.4

19000

0.6

18000

1.4

15000

1.6

14500

2.3

12500

3.0

10500

4.0

8500

5.0

6500

6.0

5000

8.0

3250

10.0

2000

12.0

1250

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-10

I.V. Bolus Dosing

(Problem 4 - 2) Aluminum:

CONCENTRATION (NG/ML)

10

10

10

10

12

TIME (HR)

1.

k = 0.234hr

2.

t = 3hr .

3.

MRT = 4.3hr .

4.

( C p )0 = 21000ng mL .

5.

AUC = 89285ng mL hr .

6.

AUMC = 383926ng mL hr .

7.

V d = 17.86mL

8.

Cl = 4.18mL hr .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-11

I.V. Bolus Dosing

Amgen

(Problem 4 - 3)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Salmonson, Danielson, and Wikstrom, "The pharmacokinetics of recombinant human erythropoetin after intravenous
and subcutaneous administration to healthy subjects", Br. F. clin. Pharmac. (1990), p 709- 713.
Amgen (r-Epo) is a form of recombinant erythropoetin. Erythropoetin is a hormone that is produced in the kidneys and
used in the production of red blood cells. The kidneys of patients who have end-stage renal failure cannot produce
erythropoetin; therefore, r-Epo is being investigated for use in these patients in order to treat the anemia that results
from the lack of erythropoetin. In a study by Salmonson et al, six healthy volunteers were used to demonstrate that
both IV and subcutaneous administration of erythropoetin have similar effects in the treatment of anemia due to
chronic renal failure. The six volunteers were each given a 50 U/kg intravenous dose of Amgen. The average weight
of the six volunteers was 79 kg. Blood samples were drawn at various times and the data obtained is summarized
below:
PROBLEM TABLE 4 - 3.

Amgen

Time (hours)

--------Serum concentration, mU
mL

700

600

400

300

12

150

24

40

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-12

I.V. Bolus Dosing

(Problem 4 - 3) Amgen:

Con (mU/mL)

CONCENTRATION (MU/ML)

103

102

101
0

10

15

20

25

TIME (HR)

1.

k = 0.134hr

2.

t = 5.2hr .

3.

MRT = 7.46hr .

4.

( C p )0 = 900mU mL .

5.

AUC = 6945mU mL hr .

6.

AUMC = 49600 mU mL hr .

7.

V d = 4.44L

8.

Cl = 0.6L hr .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-13

I.V. Bolus Dosing

Atrial Naturetic Peptide (ANP)


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 4)

AHFS 00:00.00
GPI: 0000000000

Brier and Harding, "Pharmacokinetics and Pharmacodynamics of Atrial Naturetic Peptide after Bolus and Infusion Administration in the Isolated Perfused Rat Kidney", The Journal of Pharmacology and Experimental Therapeutics (1989), p 372 - 377.

A study by Brier and Harding a dose of 45 ng was given by IV bolus to rats. Samples of blood were taken at various
intervals throughout the length of the study and the following data was obtained:
PROBLEM TABLE 4 - 4.

Atrial Naturetic Peptide (ANP)

Time (minutes)

pg
Serum concentration, -------mL

380

10

280

20

170

30

130

40

100

50

70

60

50

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-14

I.V. Bolus Dosing

(PG/ML)
Con CONCENTRATION
(pg/mL)

(Problem 4 - 4) Atrial Naturetic Peptide (ANP):

ANP

103

102

101
0 10 20 30 40 50 60

Time (min)
1

1.

k = 0.0345min

2.

t = 20.09min .

3.

MRT = 28.95min .

4.

( C p )0 = 386.6pg mL .

5.

AUC = 11206.4pg mL min .

6.

AUMC = 324425.4pg mL min .

7.

V d = 116.4mL

8.

Cl = 4.02mL min .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-15

I.V. Bolus Dosing

Aztreonam

(Problem 4 - 5)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Cuzzolim et al., "Pharmacokinetics and Renal Tolerance of Aztreonam in Premature Infants", Antimicrobial Agents and Chemotherapy (Sept. 1991), p. 1726 - 1928.

Aztreonam is a monolactam structure which is active against aerobic, gram-negative bacilli. The pharmacokinetic
parameters of Aztreonam were established in a study presented in by Cuzzolim et al in which Aztreonam (100 mg/ kg)
was administered intravenously to 30 premature infants over 3 minutes every 12 hours. The group of neonates had an
average weight of 1639.6g. The following set of data was obtained:
PROBLEM TABLE 4 - 5.

Aztreonam

Time (minutes)

gSerum concentration, ------mL

40.50

34.99

29.99

23.88

22.20

19.44

16.55

14.99

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-16

I.V. Bolus Dosing

Aztreonam

Con (ug/mL)
CONCENTRATION (UG/ML)

(Problem 4 - 5) Aztreonam:

10 2

10 1
0

TIME (MIN)

1.

k = 0.144min

2.

t = 4.81min .

3.

MRT = 6.94min .

4.

( C p )0 = 45.75ug mL .

5.

AUC = 317.7ug mL min .

6.

AUMC = 2204.8ug mL min .

7.

V d = 3.58L

8.

Cl = 0.516L min .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-17

I.V. Bolus Dosing

Recombinant Bovine Placental Lactogen


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 6)

AHFS 00:00.00
GPI: 0000000000

Byatt, et. al., "Serum half-life and in-vivo actions of recombinant bovine placental lactogen in the dairy cow", Journal of Endocrinology (1992), p. 185 - 193.

Bovine placental lactogen (bPL) is a hormone similar to growth hormone and prolactin. It binds to both prolactin and
growth hormone receptors in the rabbit and stimulates lactogenesis in the rabbit. In a study by Byatt, et. al., four cows
(2 pregnant and 2 nonpregnant) were given IV bolus injections of 4 mg and the following data was obtained:
PROBLEM TABLE 4 - 6.

Recombinant Bovine Placental Lactogen

Time (minutes)

-----Serum concentration g
L

3.8

117

6.8

72

12.0

43

16.0

27

20.0

18

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-18

I.V. Bolus Dosing

(Problem 4 - 6) Recombinant Bovine Placental Lactogen:

(MIC/L)
ConCONCENTRATION
(ug/L)

103

102

101
0

10

15

20

Time (min)
1

1.

k = 0.113min

2.

t = 6.13min .

3.

MRT = 8.85min .

4.

( C p )0 = 167.8ug L .

5.

AUC = 1484.9ug L min .

6.

AUMC = 13141.1ug L min .

7.

V d = 23.84L

8.

Cl = 2.69L min .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-19

I.V. Bolus Dosing

Caffeine

(Problem 4 - 7)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Dorrbecker et. al., "Caffeine and Paraxanthine Pharmacokinetics in the Rabbit: Concentration and Product Inhibition Effects.",
Journal of Pharmacokinetics and Biopharmaceutics (1987), p.117 - 131.

This study examines the pharmacokinetics of caffeine in the rabbit. In this study type I New Zealand White rabbits
were given an 8 mg intravenous dose of caffeine. Blood samples were taken and the following data was obtained:
PROBLEM TABLE 4 - 7.

Caffeine

Time (minutes)

g
Serum concentration -------mL

12

3.75

40

2.80

65

2.12

90

1.55

125

1.23

173

0.72

243

0.37

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-20

I.V. Bolus Dosing

(MIC/ML)
ConCONCENTRATION
(ug/L)

(Problem 4 - 7) Caffeine:

101
100
10-1
0

50

100 150 200 250

Time (min)
1

1.

k = 0.00997min

2.

t = 69.51min .

3.

MRT = 100.3min .

4.

( C p )0 = 4.105ug mL .

5.

AUC = 411.7ug mL min .

6.

AUMC = 41293.5ug mL min .

7.

V d = 1.95L

8.

Cl = 19.44mL min .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-21

I.V. Bolus Dosing

Ceftazidime

(Problem 4 - 8)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Demotes-Mainard, et. al., "Pharmacokinetics of Intravenous and Intraperitoneal Ceftazidime in Chronic Ambulatory Peritoneal
Dyialysis", Journal of Clinical Pharmacology (1993), p. 475 - 479.

Ceftazidime is a third generation cephalosporin which is administered parenterally. In this study, eight patients with
chronic renal failure were each given 1 g of ceftazidime intravenously. Both blood samples were taken the data
obtained from the study is summarized in the following table:

PROBLEM TABLE 4 - 8.

Ceftazidime

Time (hours)

mg
Serum concentration ------L

50

45

38

24

21

36

14

48

11

60

72

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-22

I.V. Bolus Dosing

CONCENTRATION (MG/L)
Con (mg/L)

(Problem 4 - 8) Ceftazidime:

10 2
10 1
10 0
0

20

40

60

80

Time (hours)
1

1.

k = 0.0324hr

2.

t = 21.39hr .

3.

MRT = 30.86hr .

4.

( C p )0 = 47.57mg L .

5.

AUC = 1468.2mg L hr .

6.

AUMC = 45308.6mg L hr .

7.

V d = 21.02L

8.

Cl = 0.681L hr .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-23

I.V. Bolus Dosing

Ciprofloxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 9)

AHFS 00:00.00
GPI: 0000000000

Lettieri, et. al., "Pharmacokinetic Profiles of Ciprofloxacin after Single Intravenous and Oral Doses", Antimicrobial Agents and
Chemotherapy (May 1992), p. 993 -996.

Ciprofloxacin is a fluoroquinolone antibiotic which is used in the treatment of infections of the urinary tract, lower respiratory tract, skin, bone, and joint. In this study, twelve healthy, male volunteers were each given 300 mg intravenous
doses of Ciprofloxacin. Blood and urine samples were collected at various times throughout the day and the following
data was collected:
PROBLEM TABLE 4 - 9.

Ciprofloxacin

Time (hours)

mg
Serum concentration ------L

1.20

0.85

0.70

0.50

0.35

10

0.25

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-24

I.V. Bolus Dosing

CONCENTRATION (MG/L)
Con (mg/L)

(Problem 4 - 9) Ciprofloxacin:

101
100
10-1
0

10

Time (hours)
1

1.

k = 0.1875hr

2.

t = 3.7hr .

3.

MRT = 5.33hr .

4.

( C p )0 = 1.57mg L .

5.

AUC = 8.395mg L hr .

6.

AUMC = 44.74mg L hr .

7.

V d = 190.6L

8.

Cl = 35.74L hr .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-25

I.V. Bolus Dosing

The effect of Probenecid on Diprophylline (DPP)


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 10)

AHFS 00:00.00
GPI: 0000000000

Nadai et al, "Pharmacokinetics and the Effect of Probenecid on the Renal Excretion Mechanism of Diprophylline", Journal of
Pharmaceutical Sciences (Oct 1992), p. 1024 - 1027.

Diprophylline is used as a bronchodilator. A study by Nadai et al was designed to determine whether or not coadministration of Diprophylline with Probenecid affected the pharmacokinetic parameters of Diprophylline. In this study,
male rats (average weight: 300 g) were given 60 mg/kg of Diprophylline intravenously and a 3 mg/kg loading dose of
Probenecid followed by a continuous infusion of 0.217 mg/min/kg of Probenecid. The following set of data was
obtained for Diprophylline (DPP):

PROBLEM TABLE 4 - 10.

The effect of Probenecid on Diprophylline (DPP)

Time (minutes)

g
Serum concentration -------mL

16

40.00

31

27.00

60

13.00

91

6.50

122

3.50

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-26

I.V. Bolus Dosing

CONCENTRATION (MIC/ML)
Con (ug/mL)

(Problem 4 - 10) The effect of probenecid on diprophylline (DPP):

102

10 1

100
0

20

40

60

80

100

Time (min)
1

1.

k = 0.023min

2.

t = 30.13min .

3.

MRT = 43.48min .

4.

( Cp ) 0 =

5.

AUC = 2396.96ug mL min .

6.

AUMC = 104219.8ug mL min .

7.

V d = 326.5mL

8.

Cl = 7.5mL min .

55.13ug mL .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-27

I.V. Bolus Dosing

Epoetin

(Problem 4 - 11)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

MacDougall et. al., "Clinical Pharmacokinetics of Epoetin (Recombinant Human Erythropoetin", Clinical Pharmacokinetics
(1991), p 99 - 110.

Epoetin is recombinant human erythropoetin. Erythropoetin is a hormone that is produced in the kidneys and used in
the production of red blood cells. The kidneys of patients who have end-stage renal failure cannot produce erythropoetin; therefore, Epoetin is used in these patients to treat the anemia that results from the lack of erythropoetin. Epoetin
has also been used in the treatment of anemias resulting from AIDS. malignant disease, prematurity, rheumatoid arthritis, sickle-cell anemia, and myelosplastic syndrome. In a study by Macdougall et al, eight patients who were on peritoneal dialysis (CAPD) were given an IV bolus dose of 120 U/kg which decayed monoexponentially from a peak of 3959
U/L to 558 U/L at 24 hours. The following data was obtained:
PROBLEM TABLE 4 - 11.

Epoetin

Time (hours)

U
Serum concentration ---L

0.0

4000

0.5

3800

1.0

3600

2.0

3300

3.0

3000

4.0

2550

5.0

2350

6.0

2150

7.0

1900

From the data presented in the preceding table and assuming that the patient weighs 65 kg, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-28

I.V. Bolus Dosing

Con (U/L)
CONCENTRATION (U/L)

(Problem 4 - 11) Epoetin:

104

103
0

Time (hours)
1

1.

k = 0.107 hr

2.

t = 6.5 hr .

3.

MRT = 9.38 hr .

4.

( Cp) 0

5.

Units hr
AUC = 37775-----------------------.
L

6.

hr
AUMC = 354697Units
-------------------------- .
L

7.

V d = 1.9 L

8.

L
Cl = 0.2065 ----.
hr

= 4023 Units/L .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-29

I.V. Bolus Dosing

Famotidine

(Problem 4 - 12)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Kraus, et. al., "Famotidine--Pharmacokinetic Properties and Suppression of Acid Secretion in Pediatric Patients Following Cardiac Surgery", Clinical Pharmacokinetics (1990), p 77 - 80.

Famotidine is a histamine H2-receptor antagonist. The study by Kraus, et. al., focuses on the kinetics of famotidine in
children. In the study, ten children with normal kidney function and a body weight ranging from 14 - 25 kg, were each
given a single intravenous 0.3 mg/kg dose of famotidine. Blood and urine samples were taken providing the following
data:
PROBLEM TABLE 4 - 12.

Famotidine

Time (hours)

g
Serum concentration -----L

0.33

300

0.50

250

1.00

225

4.00

125

8.00

70

12.00

40

16.00

15

From the data presented in the preceding table, determine the following assuming that the patient weighs 17.2 kg:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-30

I.V. Bolus Dosing

(Problem 4 - 12) Famotidine:

(MIC/L)
ConCONCENTRATION
(ug/mL)

10 3

10 2

10 1
0

10

15

20

Time (hours)
1

1.

k = 0.17 hr

2.

t = 3.9 hr .

3.

MRT = 5.7 hr .

4.

( Cp ) 0 =

5.

g hrAUC = 1600 ---------------.


L

6.

g hr
AUMC = 9000-----------------.
L

7.

V d = 18 L

8.

Cl = 3.2L .

g
285-----.
L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-31

I.V. Bolus Dosing

Ganciclovir
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 13)

AHFS 00:00.00
GPI: 0000000000

Trang, et. al., "Linear single-dose pharmacokinetics of ganciclovir in newborns with congenital cytomegalovirus infections", Clinical Pharmacology and Therapeutics (1993), p. 15 - 21.

Ganciclovir (mw: 255.23) is used against the human herpes viruses, cytomegalovirus retinitis, and cytomegalovirus
infections of the gastrointestinal tract. In this study, twenty-seven newborns with cytomegalovirus disease were given
4 mg/kg of ganciclovir intravenously over one hour. Blood samples were taken and the data obtained is summarized in
the following table:

PROBLEM TABLE 4 - 13.

Ganciclovir

Time (hours)

Serum concentration

1.50

4.50

2.00

4.00

3.00

3.06

4.00

2.40

6.00

1.45

8.00

0.87

From the data presented in the preceding table and assuming the patient weighs 3.6 kg, determine the following :
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-32

I.V. Bolus Dosing

(Problem 4 - 13) Ganciclovir:

CONCENTRATION (MICMOLE/L)

10

10

10
0

TIME (HR)
1

1.

k = 0.288hr

2.

t = 2.4hr .

3.

MRT = 3.5hr .

4.

( Cp ) 0 =

5.

mole hr
AUC = 80 -------------------------.
mL

6.

mole hr AUMC = 280---------------------------.


mL

mole23 --------------.
mL

7.

8.

1000 g4 mg
------- 3.6kg -----------------kg
mg - = 2.45L
V d = Dose
------------- = -----------------------------------------------------------Cp 0 23 ---------------mole 255.23 --------------g
L
mole
L .
Cl = 0.7 ----hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-33

I.V. Bolus Dosing

Imipenem

(Problem 4 - 14)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Heikkila, Renkonen, and Erkkola, "Pharmacokinetics and Transplacental Passage of Imipenem During Pregnancy", Antimicrobial
Agents and Chemotherapy (Dec. 1992), p 2652 - 2655.

Imipenem is a beta-lactam antibiotic which is used in combination with cilastin and is active against a broad spectrum
of bacteria. The pharmacokinetics of Imipenem in pregnant women is established in this study. Twenty women (six of
which were non-pregnant controls) were given a single intravenous dose of 500 mg of imipenem-cilastin (1:1). Blood
samples were taken at various intervals and the data obtained is summarized in the following table:

PROBLEM TABLE 4 - 14.

Imipenem

Time (minutes)

mg
Serum concentration ------L

10

27.00

15

23.50

30

15.50

45

9.50

60

6.50

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-34

I.V. Bolus Dosing

(Problem 4 - 14) Imipenem:


2

CONCENTRATION (MG/L)

10

10

10

10

20

30

40

50

60

TIME (MIN)
1

1.

k = 0.029 min

2.

t = 24 min .

3.

MRT = 34.5 min .

4.

( Cp) 0=

5.

min
AUC = 1250mg
--------------------.
L

6.

min
AUMC = 43125mg
----------------------- .
L

36.2 mg
------- .
L

7.

8.

500mg = 13.8L
Vd = Dose
------------- = -----------------Cp 0 36.2mg
------L
L
Cl = 0.4 --------.
min

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-35

I.V. Bolus Dosing

Methylprednisolone
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 15)

AHFS 00:00.00
GPI: 0000000000

Patel, et. al., "Pharmacokinetics of High Dose Methylprednisolone and Use in Hematological Malignancies", Hematological
Oncology (1993), p. 89 - 96.

Methylprednisolone is a corticosteriod that has been used in combination chemotherapy for the treatment of hematological malignancy, myeloma, and acute lymphoblastic leukemia. In a study by Patel et. al., eight patients were given
1.5 gram intravenous doses of methylprednisolone from which the following data was obtained:

PROBLEM TABLE 4 - 15.

Methylprednisolone

Time (hours)

g
Serum concentration ------mL

0.5

19.29

1.0

17.56

1.8

15.10

4.0

9.98

5.8

7.10

8.0

4.70

12.0

2.21

18.0

0.71

24.0

0.23

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-36

I.V. Bolus Dosing

(Problem 4 - 15) Methylprednisolone:

CONCENTRATION (MIC/ML)

102
101

Con (ug/mL)

100
10-1
0

10

15

20

25

Time (hours)
1

1.

k = 0.188 hr

2.

t = 3.69hr .

3.

MRT = 5.3hr .

4.

( Cp) 0=

5.

g hrAUC = 112.5 ---------------.


mL

6.

g hr
AUMC = 598.4-----------------.
mL

7.

V d = 71L

8.

L
Cl = 13.3 ----.
hr

g
21.2 -------.
mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-37

I.V. Bolus Dosing

Omeprazole
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 16)

AHFS 00:00.00
GPI: 0000000000

Anderson, et. al., "Pharmacokinetics of [14C] Omeprazole in Patients with Liver Cirrhosis", Clinical Pharmacokinetics (1993), p.
71 - 78.

Omeprazole (mw: 345.42) is a gastric proton-pump inhibitor which decreases gastric acid secretion. It is effective in
the treatment of ulcers and esophageal reflux. In normal patients 80% of the omeprazole dose is excreted as metabolites in the urine and the remainder is excreted in the feces. In the study by Anderson, et. al., eight patients with liver
cirrhosis were given 20 mg, IV bolus doses of omeprazole. The patients had a mean body weight of 70 kg. Both blood
were taken at various intervals throughout the study and the following data was obtained:

PROBLEM TABLE 4 - 16.

Omeprazole

Time (hours)

mole
Serum concentration ---------------mL

0.75

3.49

1.00

3.25

2.00

2.46

3.00

1.86

4.00

1.40

5.00

1.06

6.00

0.80

7.00

0.61

8.00

0.46

10.00

0.26

12.00

0.15

From the data presented in the preceding table, determine the following :
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-38

I.V. Bolus Dosing

Omeprazole

CONCENTRATION (PICOMOLE/ML)
Con (umol/mL)

(Problem 4 - 16) Omeprazole:

101
100
10 -1
0

10 12

Time (hours)

1.

k = 0.280hr

2.

t = 2.5hr .

3.

MRT = 3.57hr .

4.

( Cp ) 0 =

5.

mole hr
AUC = 15.4 -------------------------.
mL

6.

mole hr AUMC = 55 ---------------------------.


mL

mole
4.3 ---------------.
mL

7.

8.

Dose- = -----------------------------------------------------------------------------------------------------------20mg
Vd = -----------= 13465L
Cp 0 4.3 -------------- mole
mmole
345.42mg
1000mL
- ------------------------- ------------------------ -------------------mL 109 mole mmole
L
L
Cl = 3.9 ----.
hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-39

I.V. Bolus Dosing

Pentachlorophenol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 17)

AHFS 00:00.00
GPI: 0000000000

Reigner, Rigod, and Tozer, "Absorption, Bioavailability, and Serum Protein Binding of Pentachlorophenol in the B6C3F1 Mouse",
Pharmaceutical Research (1992), p 1053 - 1057.

Pentachlorophenol (PCP) is a general biocide. That is, it is an insecticide, fungicide, bactericide, herbicide, algaecide,
and molluskicide, that is used as a wood preservative. Extensive exposure to PCP can be fatal. In a study by Reigner
et al, six mice (average weight: 27 g) were given 15 mg/kg of PCP by intravenous bolus. Blood samples were taken at
various intervals from which the following data was obtained:
PROBLEM TABLE 4 - 17.

Pentachlorophenol

Time (hours)

g
Serum concentration ------mL

0.083

38.00

4.000

22.00

8.000

14.00

12.000

7.90

24.000

1.30

28.000

0.75

32.000

0.60

36.000

0.40

From the data presented in the preceding table, determine the following :
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p ) 0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-40

I.V. Bolus Dosing

CONCENTRATION (MIC/ML)

(Problem 4 - 17) Pentachlorphenol:

102
101

Con (ug/mL)

100
10-1
0

10

20

30

40

Time (hours)
1

1.

k = 0.134 hr

2.

t = 5.2hr .

3.

MRT = 7.5hr .

4.

g- .
( C p ) 0 = 35.6 ------mL

5.

g hr
AUC = 281 ----------------- .
mL

6.

hr - .
AUMC = 2100g
-----------------mL

7.

V d = 11.4mL

8.

Cl = 1.5 ml
------ .
hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-41

I.V. Bolus Dosing

9-(2-phophonylmethoxyethyl) adenine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 18)

AHFS 00:00.00
GPI: 0000000000

Naesens, Balzarini, and Clercq, "Pharmacokinetics in Mice of the Anti-Retrovirus Agent 9-(2-phophonylmethoxyethyl) adenine",
Drug Metabolism and Disposition (1992), p. 747- 752.

9-(2-phophonylmethoxyethyl) adenine (PEMA) is an anti-retrovirus (anti-HIV) agent. The pharmacokinetics of


PEMA in mice were established in a study by . In this study there were three different PEMA doses given: 25 mg/kg,
100 mg/kg, and 500 mg/kg. Each of these doses was injected intravenously into male mice. The data obtained from
study using the 25 mg/kg dose is summarized in the following table:

PROBLEM TABLE 4 - 18.

9-(2-phophonylmethoxyethyl) adenine

Time (minutes)

g
Serum concentration -------mL

2.0

90.3

2.9

83.9

5.6

67.3

8.9

51.5

10.5

45.2

13.5

35.4

15.0

31.3

20.0

20.9

24.0

15.1

59.6

0.9

From the data presented in the preceding table, determine the following. (Assume that the mouse weighs 200g.)
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-42

I.V. Bolus Dosing

(MIC/ML)
ConCONCENTRATION
(ug/mL)

(Problem 4 - 18) Pema:

102
101
100
10 -1
0

10 20 30 40 50 60

Time (min)
1

1.

k = 0.08min

2.

t = 8.67min .

3.

MRT = 12.5min .

4.

g( C p ) 0 = 105 ------.
mL

5.

hr- .
AUC = 1300 g
---------------mL

6.

hr .
AUMC = 16250g
------------------mL

7.

V d = 47.6ml

8.

mL
Cl = 3.8 --------.
min

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-43

I.V. Bolus Dosing

Thioperamide
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 4 - 19)

AHFS 00:00.00
GPI: 0000000000

Sakurai, et. al., "The Disposition of Thioperamide, a Histamine H3-Antagonist, in Rats", J. Pharm. Pharmacol. (1994), p. 209 212.

Thioperamide is a histamine (H3) receptor-antagonist. In a study by Sakurai et al, rats were given 10 mg/kg intravenous injections of Thioperamide. The following data was obtained from the study:
PROBLEM TABLE 4 - 19.

Thioperamide

Time (minutes)

g
Serum concentration -------mL

3.7

3.1

7.5

2.8

13

2.4

45

1.1

60

0.74

120

0.16

From the data presented in the preceding table, determine the following:
1.

Find the elimination rate constant, k .

2.

Find the half life, t .

3.

Find MRT .

4.

Find ( C p )0 .

5.

Find the Area Under the Curve, AUC .

6.

Find the area under the first moment curve, AUMC .

7.

Find the volume of distribution, V d

8.

Find the clearance, Cl .

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-44

I.V. Bolus Dosing

(MIC/ML)
ConCONCENTRATION
(ug/mL)

(Problem 4 - 19) thioperamide:

101

100

10-1
0

20

40

60

80 100 120

Time (min)
1

1.

k = 0.0254min

2.

t = 27.3min .

3.

MRT = 39.4min .

4.

g
( C p )0 = 3.39 -------.
mL

5.

minAUC = 133.5 g
-------------------.
mL

6.

min AUMC = 5256g


---------------------.
mL

7.

10 mg
------Dose
kg
LVd = ------------- = ------------------------------------------------------------------- = 2.95 ----Cp 0 3.39 ------g- -----------------mg - 1000mL
kg
-------------------mL 1000g
L

8.

Cl = 0.0254min

L- -----------------1000ml = 75 ------------------mL 2.95 ----.


kg
L
min kg

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-45

I.V. Bolus Dosing

Cocaine

(Problem 4 - 20)

Khan,vM. et. al. Determination of pharmacokinetics of cocaine in sheep by liquid chromatography J. Pharm. Sci. 76:1 (39-43)
Jan 1987

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-46

I.V. Bolus Dosing

4.1.3

URINE
From the Laplace Transform of a drug given by IV bolus we find that :
ku
( K1 t )
X u = ------ X0 ( 1 e
)
K1

(EQ 4-10)

where Xu is the cumulative amount of drug in the urine at time t. Rearranging, we


get:
k
K1t
( X u ) X u = ------u- X0 e
K1

(EQ 4-11)

ku

-X
where the amount of drug that shows up in the urine at infinite time, ( Xu ) = -----K1 0 .
Thus a plot of ( Xu ) X u vs. time on semi-log paper would result in a straight line
with a slope of -K1 and an intercept of ( Xu ) .. and we can get ku from the intercept

and the slope. Rearranging the intercept equation, we get

( X u )
k u = K 1 -------------X0

This

method of obtaining pharmacokinetic parameters is known as the Amount


Remaining to be Excreted (ARE) method.
TABLE 4-4 Enalapril

urinary excretion data from 5 mg IV Bolus

Time (hr)

Cumulative
Enalapril in urine
(mg)

0.41

0.59

0.65

0.35

0.80

0.20

0.88

0.12

0.96

0.04

1.0

------

X u mg

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-47

I.V. Bolus Dosing

Utilizations: A.R.E.
Method

FIGURE 4-2.

10

Cumulative Enalapril in urine

Xu(inf) - Xu

0.2
0.1
-1

10

1.3 hr

half life

-2

10

Hours

You should be able to transform a data set containing amount of drug in the urine vs. time into
cumulative amount of drug in the urine vs. time and plot the ARE. (Amount Remaining to be
Excreted ->

{ ( Xu ) Xu ( cum ) } vs. time on semi-log yielding a straight line with a slope of

K1 = 0.533 hr

and an intercept of

ku X0
( X u ) = --------------= 1.0 mg
K1

You should be able to determine the elimination rate constant, K1, from cumulative urinary
excretion data. (Calculate the slope of the graph on SL paper.)

You should be able to determine the excretion rate constant, ku, from cumulation urinary excretion data. (Divide the intercept of the graph by X0 and multiply by K1.
( X u )
1 1.0 mg
1
k u = K 1 -------------= 0.53 hr ----------------- = 0.106 hr )
X0
5.0 mg

You should be able to determine k m . K = k u + k m


You should be able to calculate percent metabolized or excreted from a data set. Thus,
Percent metabolized =

km
ku
------ 100 and percent excreted unchanged = ------ 100 assuming
K1
K1

K = k u + km
A second method is to plot the rate at which the drug shows up in the urine over
time. Again, using the LaPlace transforms, we find that:

Utilization: Rate of
excretion method

dX u
K1 t
K1 t
--------- = k u X0 e
= R0 e
(EQ 4-12)
dt
Thus, a plot of the rate of excretion vs. time results in a straight line on semi-log
paper with a slope of -K1 and an intercept, R0 , of kuX0 . Rearranging the intercept

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-48

I.V. Bolus Dosing

equation yields
rate

dXu
dt

R
k u = -----0- .
X0

In real data, we dont have the instantaneous excretion

, but the average excretion rate,

X u
---------- ,
t

over a much larger interval. What

that means to our calculations is that over the interval of data collection, the total
amount of drug collected divided by the total time interval is the average rate. In
the beginning of the interval the rate was faster than at the end of the interval. So
the average rate must have occurred in the middle of the interval. Thus equation 412 which is the instantaneous rate can be rewritten to
X u
K1 t mid
K1 t mid
---------- = k u X0 e
= R0 e
t
TABLE 4-5 Enalapril

(EQ 4-13)

Urinary Rate Data


X u
---------t

Interval (hr)

t(mid)

Enalapril in
urine X u ,(mg)

0-1

0.5

0.41

0.41

1-2

1.5

0.24

0.24

2-3

2.5

missed sample

3-4

3.5

0.08

0.08

4-6

0.08

0.04

You should be able to transform a data set containing amount of drug in the urine vs. time interX

val into Average Rate, ---------u- , vs.

t ,(t mid the time of the midpoint of the interval), on semilog

yielding a straight line with a slope of

K1 and an intercept of k u X 0 . as shown below.

-1
0

Urinary Excretion Rate (mg/hr)

10

R0 = 0.53 mg/hr

-2
-1

10

1.3 hr

half life
-2

10

T (Mid)

You should be able to determine k u extrapolate the line to t = 0 . The value of Rate (at

t = 0 ), R0, = k r X0 = 0.53 ( mg hr ) which when divided by X 0 .is kr.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-49

I.V. Bolus Dosing

R
1
0.53mg/hr
- = 0.106hr
Thus, -----0- = -----------------------X0

5mg

You should be able to determine k m . K = k u + k m


You should be able to calculate percent metabolized or excreted from a data set.

The rate equation is superior clinically because the ARE method requires collection of all of the urine which is usually only possible when you have a catheterized
patient while the Rate Method does not. (People dont urinate on command, and
your data could be in the toilet, literally.)
An additional advantage of the rate equations is that the

AUC

has the units of

mass, which gives the total amount of drug excreted into the urine directly. Thus:
AUC

R
mg/hr = 1 mg
= -------0 = 0.53
-------------------------1
K1
0
0.53 hr

AN INTERESTING OBSERVATION: If you look at the LaPlace Transform of the


rate equation for any terminal compartment, you would see that the resulting equation is that of the previous compartment times the rate constant through which the
drug entered the terminal compartment. Thus, the rate of drug showing up in the
urine (terminal compartment) is:
dX u
K1 t
K1 t
--------- = k u X 0 e
= R0 e
dt

where ku is the rate constant through which the drug entered the urine and
K1 t
dX
------- = X 0 e
dt

is the equation of the previous compartment.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-50

I.V. Bolus Dosing

4.2 Metabolite
4.2.1

PLASMA
Remember, the LaPlace Transform of the metabolite data yielded
( km Xo )
( km Xo )
K 2t
K 1t
K 1t
K2t
X m = ------------------------ (e
e
) or Xm = ------------------------ (e
e
) depending
( K1 K2 )
( K2 K1 )
on which rate constant that we arbitrarily assigned to be K1, the summation of all
the ways that the drug is removed from the body and K2, the summation of all the
ways that the metabolite is removed from the body. When we begin to manipulate
the data, we know that we have a curve with two different exponents in it. (If they
were the same, the equation would be different.) We dont know which is bigger,
K1 or K2, but we can rewrite the equation to simply reflect Klarge and Ksmall,
knowing that one is K1 and the other is K2 but not which is which. If we, then,
devided both sides of the equation by Vdm, the volume of distribution of the
metabolite, we would get :

km

X0 ( Ksmall t ) ( K l arg e t )
- e
e
C pm = ------------------------------------- --------
K l arg e K small V dm
Utilization:
Curve Stripping

(EQ 4-14)

You should be able to plot a data set of plasma concentration of metabolite vs. time on semi-log
paper yielding a bi-exponential curve.

Kt

0 as t . And e
K l arg e t

k l arg e t

K small t

time, t, e
. In fact
e
time, t, the equation becomes :

0 faster than e
K l arg e t

k small t

0 . So, at some long

is small enough to be ignored. Thus at long

km
X0 ( Ksmall t )
C pm = ---------------------------------- --------(e
)
K
K
V
l arg e

small

(EQ 4-15)

dm

So that the plot of the terminal portion of the graph would yield a straight line with a slope of
-Ksmall and an intercept of I =

km
X0
--------------------------------- --------K
V
K
l arg e

small

dm

You should be able to obtain the slope of the terminal portion of the curve, the negative of
which would be the smaller of the two rate constants, K small , (either the summation of all the
ways that the drug is eliminated,
eliminated,

K1 , or the summation of all the ways that the metabolite is

K2 ).

Subtracting the two previous equations yields

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-51

I.V. Bolus Dosing

km
X 0 ( K big t )
C pm C pm = ---------------------------------- --------(e
)
K l arg e K small Vdm

(EQ 4-16)

which is a straight line on semi-log paper with a slope of -kbig and an intercept of
km
X0
--------I = -----------------------------------. Note: we can get the larger of the two rate constants from this
K
V
K
l arg e

small

dm

method.
TABLE 4-6

Drug

Metabolite

(1)

(2)

(3)

(4)

Time (hr)

Cp
(mcg/L)

Cpm1 (mcg/L)

Cpm

(5)

Cpm

Cpm

181.2

181.2

0.5

24.7

175

150.3

44.4

168.9

124.5

139

71.8

157.5

85.7

65.6

96.5

136.9

40.4

31.1

100

119

19

14.6

94.7

12

76.5

24

34

In the above data Cp vs. Time is the plasma profile of the drug from Table 4-1 on page 2 and
Cpm1 vs. Time is the plasma profile of the metabolite. A plot of Cp vs. Time yielded a straight
line with a slope,(-K1) of -0.375 hr-1,

0.693 = 0.375 hr 1 and and intercept of 295


K1 = ---------------------1
1.85 hr

mic/L,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-52

I.V. Bolus Dosing

Figure 4-1 on page 3 (column 2 vs. 1 in Table 4-6 on page 52)

10 3

Concentration (mic/L)

Cpo = 295 mic/L

100

Concentration (ng/mL)

10 2

50

1.85 hr
10 1
0

Time (hours)

Time (hr)

while a plot of Cpm1 vs. Time( Figure 4-3 on page 53) yields a biexponential plot with a terminal slope of 0.07 hr-1 , k small = 0.693
------------- and extrapolating the terminal line back to time = 0
10 hr

yields 181 mic/L.


FIGURE 4-3.

Nifedipine Metabolite (column 3 vs. 1 in Table 4-6 on page 52)

L)

Nifedipine IV bolus - Metabolite

Concentration (mic/L)

103

mic
Cpm0 = 181 --------L

80
102

40
10 hr
101
0

12

16

20

24

Time (hours)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-53

I.V. Bolus Dosing

You should be able to feather (curve strip) the other rate constant out of the data by plotting the
difference between the extrapolated (to t = 0 ) terminal line (column 4 vs. 1 in Table 4-6 on
page 52) and the observed data (at early times) (column 3 vs. 1 in Table 4-6 on page 52) yielding a straight line with the slope of the line equal to the negative of the other (larger) rate constant (column 5 vs. 1 in Table 4-6 on page 52).
First you would fill in the Cpm column (column 4 in Table 4-6 on page 52) by computing Cpm
for various values of time i.e Cpm = Cpm0 e

ks m a l l t

where k small is the terminal slope of the

graph. Then Cpm Cpm (column 5 in Table 4-6 on page 52) would be column 4 - column 3.
Then a plot of Cpm Cpm vs. time (column 5 vs. 1 in Table 4-6 on page 52) is shown below.
FIGURE 4-4.

Curve strip of Nifedipine Metabolite data

10

Intercept
Column 5

100
102

1.85 hr

50

Half life
1

10

Time (hr)

In this case, the slope of the stripped line line is -0.375 hr-1 and the intercept is 0.181.2 mic/L.
The slope of -0.375 hr-1 should not be surprising as the plot of the data in Figure 4-3 on page 53
resulted in a terminal slope of -.07 hr-1 . Since the data set yielded a bi-exponential plot, separating out the exponents could only yield K1 (0.375 hr-1) or K2 as determined by our Laplace
Transform information. Thus, the terminal slope could be either -K1 or -K2. Since it was obviously not -K1, it had to be -K2. Thus the other rate constant obtained by stripping has to be K1.
You can determine which slope is which rate constant if you have any data regarding intact drug
(i e. either plasma or urine time profiles of intact drug) as the slope of any of those profiles is
always

K1 .

You should be able to determine V dm if you have any urine data regarding intact drug (i.e.
urine time profiles of intact drug) as the intercept of those profiles allow for the solution of k m .
Thus the intercept, I, of the extrapolated line of equation 4-14 could be rearranged to contain
only one unknown variable, V dm

1
mic
0.375hr 25mg 1000
---------------------km X0
mg
= ----------------------------------------------- = -------------------------------------------------------------------------- = 170 L .
1
( K l arg e K small ) I
mic
( 0.375 0.07 ) hr 181.2--------L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-54

I.V. Bolus Dosing

Utilization:
MRT Calculations

You should be able to determine the rate constants using MRT calculations.
In a caternary chain, each compartment contributes its MRT to the overall MRT of the drug,
thus:
Flow Chart 2-4

IV Bolus

K1

MRT(IV) = 1/K1
Suppose the drug were given by IV bolus. Then the drug would have to be metabolized and the
metabolite eliminated. Since the MRTs are additive, the overall MRT of the metabolite would
be made up of the MRTs of the two processes, thus:
Flow Chart 2-5

Metabolite
km

kmu

Xm

MRT(met) = MRT(elim)+MRT(IV)
MRT(met) =

1/K2

+ 1/K1

Thus, using the data from Table 4-3 on page 5 the MRT(IV)Trap is
AUMC
1986.1
AUMC
2100
MRT = ------------------ = ---------------- = 2.42 hr or about MRT = ------------------ = ------------ = 2.67 hr using calculus.
AUC
819.9
AUC
787

And using the data from columns 1 and 3 from Table 4-6 on page 52 the MRT(met) using calcuAUMC- = 36000
lus is MRT = ------------------------------- = 17 hr.
AUC

2116

MRT(elim) = MRT(met) - MRT(IV) = 17 hr - 2.67 hr = 14.33 hr = 1/K2. Thus K2 = 0.07 hr-1.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-55

I.V. Bolus Dosing

4.2.2

URINE
Valid equations:
dX mu
k mu k m X0
K
t
K
t
------------- = --------------------------------------- { e small e l arg e }
dt
( K l arg e K small )

Utilization:

(EQ 4-17)

as in the previous urinary rate equation, clinically we work with the average rate
over a definite interval which results in rewriting equation 4-17 as:
Xmu
k mu k m X0
K
t
K
t
------------= --------------------------------------- { e small mid e l arg e mid }
t
( K l arg e K small )

(EQ 4-18)

You should be able to plot a data set of rate of metabolite excreted vs. time (mid) on semi-log
paper yielding a bi-exponential curve.

You should be able to obtain the slope of the terminal portion of the curve, the negative of
which would be the smaller of the two rate constants (either

K1 or K2 ).

You should be able to feather (curve strip) the other rate constant out of the data by plotting the
difference between the extrapolated (to t = 0 ) terminal line and the observed data (at early
times) yielding a straight line with the slope of the line equal to the negative of the other (larger)
rate constant (either

K1 or K2 ).

You should be able to utilize MRT calculations to obtain K1 and K2 .


You should be able to determine which slope is which rate constant if you have any data regarding intact drug (i.e. either plasma or urine time profiles of intact drug) as the slope of any of
those profiles is always

K1 .

By this time, it should be apparent that data which fits the same shape curve
(mono-exponential, bi-exponential, etc.) are treated the same way. When the
curves are evaluated, the slopes and intercepts are obtained in the same manner.
The only difference is what those slopes and intercepts represent. These representations come from the equations which come from the LaPlace Transforms which
come from our picture of the pharmacokinetic description of the drug. Please
refer back to the section on graphical analysis in the Chapter 1, Math review for a
interpretation of slopes and intercepts of the various graphs.
Temporarily, please refer to exam section 1, chapter 14 for problems for this section (until problems can be generated) as well as additional problems for the previous sections.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

4-56

CHAPTER 5

I.V. Infusion

Author: Michael Makoid and John Cobby


Reviewer: Phillip Vuchetich

OBJECTIVES
1.

Given patient drug concentration and/or amount vs. time profiles, the student will
calculate (III) the relevant pharmacokinetic parameters available ( V d , K, k m , k r ,
AUC ,

Clearance, MRT) from IV infusion data.

2.

I.V. Infusion dosing for parent compounds

3.

Plasma concentration vs. time profile analysis

4.

Rate vs. time profile analysis

5.

Professional communication of IV Infusion information

6.

Computer aided instruction and simulation

7.

Metabolite (active vs. inactive)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-1

I.V. Infusion

5.1 Parent compound


5.1.1

PLASMA

Valid equations:
Kt
Q
C p = -------------( 1 e ) or
K Vd

(EQ 5-1)

Kt
Dose
C p = --------------------------------------( 1 e )
K Vd T infusion

(EQ 5-2)

at any time during the infusion

Q
( C p )ss = -------------K Vd

(EQ 5-3)

at steady state (t is long)

C p = C p( term ) e

Kt

(EQ 5-4)

after termination of infusion

Where C p is the plasma concentration


Dose is the infusion rate shown in equation 5-1 and equation 5-2.
Q = ------------------T infusion
Kt infusion
Q
) is the plasma concentration when the
C p( term ) = -------------- ( 1 e
K Vd
infusion is stopped.

Rewriting equation 5-4 to an equation which may be used by a computer results in:
Q - ( e K T e K T )
Cp = ----------KV
where

T = ( T T i v )

and

T = 0

(EQ 5-5)

for ( T > T iv )

for ( T < T iv )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-2

I.V. Infusion

Using The Scientist@s Unit function makes the change in T straight forward. In
The Scientist@, Unit(+) = 1 and Unit(-) = 0, so defining T = ( T T iv ) UNIT ( T Ti v )
meets these needs.
This equation is utilized in The Scientist@s companion product PKAnalyst@ also
by MicroMath. Since the route of administration is an infusion and we would
know how much we gave (Dose), how fast we gave it (Q), and over how long the
infusion lasted (Tiv), the only other variables in the equation are K and Vd. PKAnalyst asks for Tiv and yields DoverV ( Dose
------------- ) and K as parameters resulting from
Vd
non-linear regression analysis. Dividing Dose by Dose
------------- yields Vd.
Vd

Utilization:

You should be able to determine the infusion rate necessary to obtain a desired
plasma concentration. Rearranging equation 5-3 results in:
K V d ( C p )ss = Q

(EQ 5-6)

You should be able to determine how long it would take to get to a desired plasma
concentration. Using equation 5-1 and equation 5-3, it looks like it will take forever to get exactly to steady state because in order for
Q = -------------Q ( 1 e Kt ) , e Kt 0 which occurs when t = . So,
( C p )ss = -------------K Vd
K Vd
how close is close enough? If ( C p ) = 0.95 ( C p )ss , thats good enough in most
peoples estimation. So in order to find out how long it will take we use equation
5-1, setting ( C p ) = 0.95 ( C p )ss and solve for time. Thus:
Q ( 1 e Kt ) which results in
( C p ) = 0.95 ( C p )ss = -------------K Vd
0.95 = ( 1 e

Kt

0.95 1 = e

Kt

ln ( 0.05 ) = Kt
2.996 = Kt
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-3

I.V. Infusion

---------------2.996 = t
K
2.996
-------------t 1--- = 4.32t 1--- = t ,
0.693 2
2

(EQ 5-7)

or about 4.32 half lives to get to 95% of steady state. Generalizing, then, the number of half-lives it takes to get to steady-state is equal to the logarithm of the
inverse of how close is close (in this case, 5% or 0.05 = 20) devided by the logarithm of two.
Changing infusion rates:

Occasionally, it is necessary to change infusion rates to stabilize the patient. If a


patient were started on an infusion rate, Q1, and then at some subsequent time,
T>T*, the infusion rate was changed to Q2, the equation for the concentration after
the change would be:
( K T )
( k ( T T ) )
Q1
Q2 - ( 1 e ( k ( T T ) ) )
)e
+ ----------Cp = ------------ ( 1 e
KV
KV

(EQ 5-8)

Assuming equilibrium was reached at infusion rate Q1, we could simplify equation
5-8 by setting T = 0 at the time of the rate change (thus we would be interested in
the time after the change) resulting in:
k T
k T
Q1
Q2
Cp = -----------e
+ ------------ ( 1 e
)
KV
KV

(EQ 5-9)

Under these conditions, it would be useful to determine the time to reach the new
equilibrium. As before, within 5% is close enough. Thus if we are coming down
Q2- and if we were
(lowering the Cp, i.e. Q2 < Q1), we would want Cp = 1.05 ----------KV
Q2 . Taking
going up (raising the Cp, i.e. Q2 > Q1), we would want Cp = 0.95 ----------KV
the first condition we find:
k T
Q2
Q1
Q2 - ( 1 e k T )
+ ----------Cp = 1.05 ------------ = ------------ e
KV
KV
KV

(EQ 5-10)

Rearranging and solving for T results in:


Q2
ln 0.05
---------------------Q1 Q2
T = ---------------------------------K

(EQ 5-11)

Similarly, under the second condition, we would find:


Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-4

I.V. Infusion

0.05 Q2
ln ------------------------Q1 Q2
T = ------------------------------------K

(EQ 5-12)

Combining equation 5-11 and equation 5-12 and rearranging results in:
Q1 Q2- 20 ln ----------------------Q1 Q2- 20
ln ----------------------

Q2
Q2
T = ---------------------------------------------- = ---------------------------------------------- t 1 2
K
0.693

(EQ 5-13)

Thus it is the absolute value of the difference of the two rates and the elimination
rate constant which determine the length of time needed to establish a new equilibrium. Under the conditions of Q1 = 0 , that is no previous infusion, and the difference is maximal equation 5-13 simplifies to equation 5-7. Under the conditions
of Q1 = Q2 , the equation is undifined and has no utility (as well as makes no
sense, because the equation was designed to be used when there was a change in
rate.) However, lim T = 0 , thus no change results in zero time to get to the new
Q2 Q1

equilibium. Similar to equation 5-7 as before, the generalization for the number of
half-lives it takes to obtain the new steady-state is the logarithm of (the fractional
difference of the rates (or the steady-state concentrations) times the inverse of how
close is close) devided by the logarithm of two.
As pharmacokinetic equations are additive, you should be able to determine a
loading dose (by I.V. bolus, for example) and a maintenance dose (infusion rate)
for a patient to extablish an equilibrium. If, for example, you want to give a loading dose followed by an IV infusion, the generalization for the number of halflives it takes to obtain the new steady-state is the logarithm of (the fractional difference of the concentrations, Cp0 and Cpss, times the inverse of how close is
close) devided by the logarithm of two.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-5

I.V. Infusion

Discussion:

Example: Using
population average
pharmacokinetic
parameters to make
professional judgements.

IV infusion is a controlled way to get drug into your patient. Using patient population average pharmacokinetic parameters (K, Vd) available in the drug monographs, you are able to make a professional judgement about:
1.

the plasma concentration that you would like to achieve (from therapeutic range) and the time in
which you would like to get there.

2.

the infusion rate necessary to get to the target concentration, and

3.

the time necessary to to get there.

As an example, theophylline is a bronchodilator used in asthma with a therapeutic


range of 10 to 20 mg/L, a volume of distribution of 0.45 (0.3 - 0.7) L/kg and a half
life of about 8 (6 - 13) hours for a non-smoking adult. Your patient weighs 200
pounds and meets these these criteria. The physician decides to maintain him at 15
mg/L. What do you do?
Using population average parameters for K and Vd, equation 5-6 results in:
L- ------------kg - 200 lb 15 mg
0.693
= 53.2mg
- 0.45 ----------- .
----------- ------8 hr
kg 2.2 lb
L
hr
For an eight hour IV infusion, you would need
53.2 mg
------- 8 hr = 425 mg of theophylline.
hr
IV Theophylline comes as aminophylline which is theophylline compound containing 85% theophylline and 15% ethylenediamine. So in order to get 425 mg of
theophylline we have to give
100 mg aminophylline
425 mg theophylline ------------------------------------------------------ = 500 mg aminophylline . So we
85 mg theophylline
prepare our IV infusion using Aminophylline U.S.P. for injection (500 mg aminophylline in 20 mL) by placing the contents of the ampule in 1000 mL of D5W and
calculate the drip rate using an adult IV administration set which regulates the drip
to 10 gtts/mL. Thus the drip rate is:
1020
ml- ---------------hr - 10
gtts- 21 -------gtts- -------------7 gtts--------------------------------8 hr
60 min
ml
min 20 sec

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-6

I.V. Infusion

How long to get to steady


state?

After setting up the infusion, the doctor asks, How long to steady state?
Using equation 5-7, our patient who has an eight hour half life, will take about
4.32 8 hr = 34.6 hr to get to 95% of steady state. The patient doesnt want
relief in a day and a half. He needs to breathe NOW. What would you suggest?

Infusion takes too long.


How do we get relief
now? IV Bolus stat.

It might be possible to give him an IV Bolus dose stat which would get him to
Dose( C p )ss right away. This is done by converting
( C p )ss = -----------to
Vd
V d ( C p ) ss = Dose .
L- ------------kg - 200 lb 15 mg
0.45 ----------- = 613.6 mg Theophylline

kg 2.2 lb
L
Converting to aminophylline yields:
mg aminophylline 725 mg aminophylline . Thus,
613.6 mg Theophylline 100
-----------------------------------------------------85 mg theophylline
if we gave a 725 mg IV bolus dose of aminophylline followed by a concomitant
IV infusion of 500 mg aminophylline over 8 hours, our patient should get to
steady state right away and stay there.

Some protocols require


starting with faster
infusion, then changing
to a slower one to get to
steady state faster.

Sometimes the physician might want to just increase the infusion rate (say double
it for a short time, 2Q) to get to the target concentration faster and then just back
the infusion down. If that is the protocol, the question becomes, How long do
you run the infusion in at the faster rate? Thus:
Kt
Kt
Q
2Q
( C p )ss = -------------- = -------------- ( 1 e ) which yields 1 = 2 ( 1 e ) and so
K Vd
K Vd
Kt
Kt
1
1
--- = 1 e
. Thus --- 1 = e . Taking the ln of both sides ln ( 0.5 ) = Kt
2
2

ln
( 0.5 ) = 0.693
------------------------------ t 1--- = t 1--- = t or that it will take one half-life to get to the target
K
0.693 2
2
plasma concentration (which is the Cpss obtained by the infusion rate of 1Q) if you
run the infusion at a faster rate, 2Q. So for your patient, you might suggest an
infusion of 1000 mg over 8 hours (2Q for one half life) to get to steady state
quickly and then back off to 500 mg over 8 hours for the second 8 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-7

I.V. Infusion

Clearance: New
pharmacokinetic
parameter

Clearance ( Cl = K Vd ) is a pharmacokinetic parameter which relates the fraction


of the volume of distribution which is cleared of the drug per unit time. The volume of distribution is a mathematical construct which relates two knowns, the
Dose of the drug and the resultant Concentration. In linear kinetics, the Dose is
proportional to the Concentration. C D . The units of concentration are
Mass - while the units of dose are Mass. So the units of the proportionality con------------------Volume
stant must be volume in order for the equation to balance. Thus, the volume of distribution is a hypothetical volume and not necessarily a real volume or
physiological space. Consequently, clearance is the hypothetical volume of fluid
from which the drug is irreversibly removed per unit time. So equation 5-3 can be
rewritten:
Q( C p )ss = ----Cl

How do we calculate
Clearance from IV infusion data?

(EQ 5-14)

and equation 5-14 can be rewritten to:


Q Cl = -------------( C p )ss

(EQ 5-15)

Thus, assuming steady state, the clearance can be calculated by dividing the infusion rate by the resultant steady state plasma concentration.
How do we separate K
and Vd out of Clearance?

Graphing equation 5-4 which relates the decline in plasma concentration after cessation of the infusion, the resultant slope of the line yields - K, the elimination rate
constant. Dividing the elimination rate constant, - K, obtained by equation 5-4 into
the clearance obtained by equation 5-15 results in the other necessary pharmacokinetic parameter, Vd.

How can we utilize the


rate of change of
plasma concentration to
determine the pharmacokinetic parameters, K
and Vd?

From our original model


d
X = Q (K X )
dt

(EQ 5-16)

X- . Thus , V C p = X . Rewriting equation 5-16 yields:


and Cp = ----d
Vd

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-8

I.V. Infusion

dCp
Q- K C and rearranging and incorporating equation 5-1 yields
= ----p
dt
Vd
dCp
Q- K -------------Q ( 1 e Kt ) which can be simplified to
= ----
dt
Vd
K Vd
dCp
Q- ----Q- + ----Q- e Kt or
= ----dt
V d V d Vd
dC p
Q- e Kt
= ----dt
Vd

(EQ 5-17)

dCp
Cp
vs. t ( actually,
----------- vs. tmid exactly like we did in urinary
dt
t
rate graphs) of the ascending portion of the plasma profile would result in a
Q- .
straight line with a slope of -K and an intercept of ----Vd
Thus a plot of

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-9

I.V. Infusion

5.2 Problems
Equations needed for solving the problems:
1.

k from the slope of the terminal portion of the graph of Cp vs. T

2.

0.693
t 1 2 = -----------k

3.

Volume of distribution from Cp = -------------- ( 1 e

4.

Clearance Cl = K V d

5.

You wish to maintain a plasma concentration of Cpss.

Q
K Vd

a.

Kt

Calculate the infusion rate necessary to maintain


Cpss. Q = Cp ss K V d

b.

Suggest a loading dose which would give you Cpss immediately.

Dose loading = Cp ss V d
c.

How long will it take to reach steady state?

T 95 = 4.32 T 1 2
d.

Find the plasma concentration if the infusion is discontinued at time = Tdc hours.

Q ( 1 e ( K Tdc ) ) .
Cp dc = -------------K Vd
e.

Find the plasma concentration Tpost hours after infusion is discontinued at time = Tdc hours.

Cp post = Cp dc e

( K T post )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-10

I.V. Infusion

Acyclovir

(Problem 5 - 1)

Problem Submitted By: Maya Leicht

AHFS 08:18.00 Antivirals

Problem Reviewed By: Vicki Long

GPI: 1200001000 Antivirals

Laskin, O., "Clinical pharmacokinetics of acyclovir", Clinical Pharmacokinetics (1983), p. 187 - 201.

Acyclovir (225.21 g/Mole) is an antiviral drug used in the treatment of herpes simplex, varicella zoster, and in suppressive therapy. In this study, patients were given varying doses of acyclovir over one hour by infusion. Acyclovir distributes uniformly into the plasma and tissues such that the plasma concentration is representative of tissue concentration.
Acyclovir is 30% metabolized and 70% renally excreted. The following data was obtained from an intravenous infusion dose of 2.5 mg/kg over one hour where the patient weighed 70 kg.
PROBLEM TABLE 5 - 1. Acyclovir

Plasma concentration
Time (hours)
0

0.25

0.5

12

0.75

17

20

10

umol
-
-----------L

From this data determine the following:


1.

2.

t1 2

3.

Volume of distribution

4.

Clearance

5.

You wish to maintain a plasma concentration of 25 umol L .


a.

Calculate the infusion rate necessary to maintain a plasma concentration of 25 umol L

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 5 hours.

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 5 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-11

I.V. Infusion

Acyclovir on page 11

Concentration

102

101

100
0

Time
1.

k = 0.751 hr-1 (from slope of graph).

2.

t 1 2 = 0.923 hr (from slope of graph).

3.

Volume of distribution = 26.2 L

4.

Clearance = 19.67 l/hr

5.

You wish to maintain a plasma concentration of 25 umol L .


a.

Calculate the infusion rate necessary to maintain a plasma concentration of 25 umol L = 111 mg/hr

b.

Suggest a loading dose for the patient which would give you Cpss immediately. 148 mg

c.

How long will it take to reach steady state? 4 hr

d.

Find the plasma concentration if the infusion is discontinued at time = 5 hours. = 25 umol L

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 5 hours. = 5.6 umol L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-12

I.V. Infusion

Aminophylline

(Problem 5 - 2)

Problem Submitted By: Maya Leicht

AHFS 12:12.00 Sympathomimetics

Problem Reviewed By: Vicki Long

GPI: 4430001000 Xanthine Sympathomimetic

Gilman, T., et al., "Estimation of theophylline clearance during intravenous aminophylline infusions", Journal of Pharmaceutical
Sciences (May 1985), p. 508 - 514.

Aminophylline is used in the treatment of bronchospasm. In this study, aminophylline was given by intravenous infusion to patients with a mean weight of 75.7 kg. The doses given were chosen to maintain a between 10 -20 mg/L based
on desirable body weight. The doses were given at a rate of 0.5 mg/kg/hour (Theophylline) for 84 hr. The following
set of data was collected.
PROBLEM TABLE 5 - 2. Aminophylline

Plasma concentration
Time (hours)
0

mg

------L

0.

12

24

11

30

11.6

36

12.0

48

12.4

54

12.5

66

12.6

72

12.8

84

12.8

88

92

6.4

96

4.6

100

3.2

From this data determine the following:


1.

2.

t1 2

3.

Volume of distribution

4.

Clearance

5.

6.

You wish to maintain a plasma concentration of 15 mg/L in your patient.

a.

Calculate the infusion rate necessary to maintain a plasma concentration of 15 mg/L.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 5 hours.

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 5 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-13

I.V. Infusion

Aminophylline on page 13

CONCENTRATION

102

101

10

20

40

60

80

100

Time
1.

k = 0.085 hr-1

2.

t 1 2 = 8.15 hr

3.

Vd = 35.3 L

4.

Cl = 3 L/hr

5a.

Q = 45 mg/hr

5b.

D L = 530 mg

5c.

5d.

C p = 5.2 mg/L

5e.

C p = 4.4 mg/L

ss
= 35 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-14

I.V. Infusion

Carmustine

(Problem 5 - 3)

Problem Submitted By: Maya Leicht

AHFS 10:00.00 Antineoplastics

Problem Reviewed By: Vicki Long

GPI: 2110201000 Antineoplastics, Nitrosoureas

Henner, W., et al., "Pharmacokinetics and immediate effects of high-dose carmustine in man", Cancer Treatment Reports vol.70
(1986), p. 877 - 880.

Carmustine (BCNU) is an antineoplastic agent with a molecular weight of 214.04 g.


2

In this study a 70 kg, 1.8 M2 patient was given 600 mg m by intravenous infusion over 2 hours. The following data
was obtained.
PROBLEM TABLE 5 - 3. Carmustine

Plasma concentration
Time (minutes)
15

.3

30

.5

60

.7

90

.75

120

.8

135

.5

142.5

.4

150

.3

mg
------L

From this data determine the following:


1.

2.

t1 2

3.

Vd

Cl

5.

A patient with a BSA of1.8 M is to be given BCNU by IV infusion. You wish to maintain a plasma

concentration of 2 uM . Determine the following:


a.

Calculate the infusion rate necessary to maintain a plasma concentration of 2 uM .

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 10 minutes.

e.

Find the plasma concentration 1 hour after infusion is discontinued at time = 10 minutes.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-15

I.V. Infusion

Carmustine on page 15

CONCENTRATION

10

-1

10

50

100

150

TIME

1.

k = 0.031 min-1

2.

t 1 2 = 22 min

3.

Vd = 198 L/M2

Cl = 6.15 L/M2/hr

5.

A patient with a BSA of1.8 M is to be given BCNU by IV infusion. You wish to maintain a plasma

concentration of 2 uM . Determine the following:


a.

Calculate the infusion rate necessary to maintain a plasma concentration of 2 uM .

214g- -----------------mg - -----------198L 1.8M 2 0.031min 1 = 4.73mg


Q = Cp ss V d K = 2mole
------------------- -------------------------------- 285mg
----------------L
mole 1000g M2
min
hr
b.

Suggest

loading

dose

for

the

patient

which

would

give

you

Cpss

immediately.

214g- -----------------mg - -----------198L 1.8M 2 = 150mg


Dose = Cp ss Vd = 2mole
------------------- --------------L
mole 1000g M 2
c.

How long will it take to reach steady state? 4.32 * T 1/2 = 97 min.

d.

Find the plasma concentration if the infusion is discontinued at time = 10 min. = 0.1197 mg/L

e.

Find the plasma concentration 1 hour after infusion is discontinued at time = 10 min. = 0.017 mg/L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-16

I.V. Infusion

Cefotaxime

(Problem 5 - 4)

Problem Submitted By: Maya Leicht

AHFS 08:12.06 Cephalosporins

Problem Reviewed By: Vicki Long

GPI: 0230007510 Cephalosporins - 3rd Generation

Kearns, G., Young, R., and Jacobs, R., "Cefotaxime dosage in infants and children--pharmacokinetic and clinical rationale for an
extended dosage interval", Clinical Pharmacokinetics (1992), p. 284 - 297.

Cefotaxime is a third generation cephalosporin which is widely used as an antimicrobial in neonates, infants, and children. In this study, infants and children were given a 50 mg/kg dose of cefotaxime intravenously over 0.25 hour. The
following data was collected:
PROBLEM TABLE 5 - 4. Cefotaxime

Plasma concentration
Time (hours)
0.00

0.05

35

0.10

70

0.20

140

0.35

155

0.60

130

0.85

110

1.20

80

1.30

75

2.00

45

2.40

35

3.40

15

4.50

6.50

1.7

mg

------L

From this data, assuming that the patient weighs 30 kg, determine the following:
1.

2.

t1 2

3.

Vd

4.

Cl

5.

You wish to maintain a plasma concentration of 80 mg/L. Determine the following:


a.

Calculate the infusion rate necessary to maintain a plasma concentration of 80mg/L

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 0.25 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-17

I.V. Infusion

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 0.25 hours.

Cefotaxime on page 17

CONCENTRATION

103

102

101

100
0

TIME

1.

k = 0.733 hr-1

2.

t 1 2 = 0.945 hr

3.

Vd = 0.276 L/kg

4.

Cl = 0.202 L/kg/hr

4a.

Q = 16.2 mg/kg/hr

4b.

D L = 22.1 mg/kg

4c.

4d.

C p = 13.35 mg/L

4e.

C p = 3.09 mg/L

ss
= 4.1 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-18

I.V. Infusion

Ganciclovir

(Problem 5 - 5)

Problem Submitted By: Maya Leicht

AHFS 08:18.00 Antivirals

Problem Reviewed By: Vicki Long

GPI: 1200002010 Antivirals

Trang, J., et al., "Linear single-dose pharmacokinetics of ganciclovir in newborns with congenital cytomegalovirus infections",
Clinical Pharmacology and Therapeutics (1993), p. 15 - 21.

Ganciclovir is used against the human herpes viruses, cytomegalovirus retinitis, and cytomegalovirus infections of the
gastrointestinal tract. In this study, twenty-seven newborns with cytomegalovirus disease were given 4 mg/kg of ganciclovir intravenously over one hour. Blood samples were taken and the data obtained is summarized in the following
table:
PROBLEM TABLE 5 - 5. Ganciclovir

Plasma concentration --------

ug
mL

Time (hours)
0.5

3.10

1.5

4.50

2.0

3.80

3.0

2.90

4.0

2.30

6.0

1.50

8.0

0.88

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

4.

A patient is to be given ganciclovir by IV infusion to an infant weighing 6.1 kg. You wish
to maintain a plasma concentration of 5.5 mcg/mL. Determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 5.5mcg/mL.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 1 hour.

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 1 hour.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-19

I.V. Infusion

Ganciclovir on page 19

CONCENTRATION

10 1

10 0

10-1
0

Time

1.

k = 0.255 hr-1

2.

t 1 2 = 2.72 hr

3.

Vd = 0.687 L/kg

4.

Cl = 0.175 L/kg/hr

5a.

mg - -----------------1000ml 0.687L
Q = Cp ss V d K = 5.5g
-------------- ------------------ 6.1kg 0.255
------------- = 5.9mg
-------------- ---------------ml 1000g
L
kg
hr
hr

5b.

mg - -----------------1000ml 0.687L
D L = Cp ss Vd = 5.5g
-------------- ---------------------------------- 6.1kg = 23mg
kg

ml 1000g
L

5c.

ss
= 4.32 t1 2 = 11.75hr
95%
5.9mg
-------------- Kt
K 1hr
Q
hr
= -------------- ( 1 e ) = ----------------------------------------------------- (1 e
) = 1.24mg
-----------------K Vd
0.255
L
------------- 0.687L
----------------- 6.1kg
hr
kg

5d.

C p term

5e.

C p = C p term e

K 2hr

= 1.24mg
------------------ 0.6 = 0.74mg
-----------------L
L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-20

I.V. Infusion

Gentamicin

(Problem 5 - 6)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0700002010 Aminoglycosodes

Kaojarern, S., et al., "Dosing regimen of gentamicin during intermittent peritoneal dialysis", Journal of Clinical Pharmacology
(1989), p. 140 - 143.

Gentamicin is an aminoglycoside antibiotic which is frequently used in the treatment of gram-negative bacilli infections. Since it has a low therapeutic index, it is important to determine proper dosage regimens. In this study, patients
on peritoneal dialysis received a 30 minute intravenous infusion of 80 mg gentamicin in 100 mL of 5% dextrose in
water. The following data was collected:
PROBLEM TABLE 5 - 6. Gentamicin

Plasma concentration
Time (hours)
0.50

5.68

1.50

5.15

3.70

4.80

7.35

3.99

11.30

3.35

24.00

2.02

ug
------ mL-

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given gentamicin by IV infusion. You wish to maintain a plasma concentration


of 5.2 ug mL . Determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 5.2 ug mL

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 0.5 hours.

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 0.5 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-21

I.V. Infusion

Gentamicin on page 21

CONCENTRATION

101

100
0

10

15

20

25

Time

1.

k = 0.0431 hr-1

2.

t 1 2 = 16.1 hr

3.

V d = 14.5 L

4.

Cl =0.625 L/hr

5a.

Q = 3.25 mg/hr

5b.

D L = 75 mg

5c.

5d.

C p = 0.11 mg/L

5e.

C p = 0.10 mg/L

ss
= 69.6 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-22

I.V. Infusion

Human Monoclonal Anti-lipid A antibody (HA-1A)


Problem Submitted By: Maya Leicht

AHFS 24:06.00 Antilepemics

Problem Reviewed By: Vicki Long

GPI: 3900000000 Antihyperlipidemic

(Problem 5 - 7)

Fisher, C., et al., "Initial evaluation of human monoclonal anti-lipid A antibody (HA-1A) in patients with sepsis syndrome", Critical Care Medicine (1990), Vol.18, No. 12, p. 1311 - 1315.

HA-1A is an immunoglobulin antibody. In this study, patients received a 250 mg intravenous infusion of HA-1A over
15 minutes. Serum levels were measured before and after infusion and the following data was collected:
PROBLEM TABLE 5 - 7. Human

Monoclonal Anti-lipid A antibody (HA-1A)

Plasma concentration
Time (hours)
0.00

0.75

80

1.00

75

2.00

74

5.00

65

15.00

50

25.00

40

48.00

21

72.00

10

ug-
------ mL

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5. A patient is to be given HA-1A by IV infusion. You wish to maintain a plasma concentration of


Determine the following:

100 g/mL.

a.

Calculate the infusion rate necessary to maintain a plasma concentration of 100 ug mL .

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 1 hour.

e.

Find the plasma concentration 3 hours after infusion is discontinued at time = 1 hour.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-23

I.V. Infusion

Human Monoclonal Anti-lipid A antibody (HA-1A) on page 23

CONCENTRATION

102

101
0

20

40

60

80

Time
1.

k = 0.0282 hr-1

2.

t 1 2 = 24.4 hr

3.

V d = 3.2 L

4.

Cl = 0.09 L/hr

5a.

Q = 9 mg/hr

5b.

D L = 320 mg

5c.

5d.

C p = 2.78 mg/L

5e.

C p = 2.56 mg/L

ss
= 105 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-24

I.V. Infusion

Ifosfamide

(Problem 5 - 8)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Lewis, L., "The pharmacokinetics of ifosfamide given as short and long intravenous infusions in cancer patients", British Journal
of Clinical Pharmacology Vol. 31 (1991), p. 77 - 82.

Ifosfamide is an agent which has shown some pharmacological response in the treatment of cancer. In this study, a 5

gm

dose of ifosfamide was infused over 30 minutes. The median BSA for the subjects was 1.8 m . The

following data was obtained:


PROBLEM TABLE 5 - 8. Ifosfamide

Plasma concentration
Time (hours)
0

0.0

0.5

285.0

260.0

220.0

160.0

112.0

80.0

10

60.0

24

ug
------ mL-

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.
A patient is to be given ifosfamide by IV infusion. The patient has a BSA 1.8 M2. You wish to maintain a
plasma concentration of 336 g/mL. Determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 336 ug mL

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 20 min.

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 20min.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-25

I.V. Infusion

Ifosfamide on page 25

CONCENTRATION

103

102

101

100
0

10

15

20

25

Time
1.

k = 0.1716 hr-1

2.

t 1 2 = 4.04 hr

3.

V d = 16.6 L/M2

4.

Cl = 2.85 L/hr/M2

5a.

Q = 1.725 g/hr

5b.

D L = 10 g

5c.

5d.

C p = 18.7 mg/L

5e.

C p = 13.25 mg/L

ss
= 17.5
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-26

I.V. Infusion

Isosorbide 5-mononitrate
Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

(Problem 5 - 9)

Major, R., et al., "Isosorbide 5-mononitrate kinetics" (1983), p. 653- 660.

Isosorbide 5-mononitrate (5-ISMN) is a metabolite of isosorbide dinitrate. In this study, the kinetics of isosorbide 5mononitrate were looked at in 12 healthy patients after an intravenous infusion of 20 mg at 8 mg/hour for 2.5 hours.
This drug follows one-compartment, open model kinetics. The following data was collected:
PROBLEM TABLE 5 - 9. Isosorbide

5-mononitrate

Time (hours)

Plasma concentration (ng/mL)

0.25

40

0.50

91

0.75

141

1.00

181

1.50

239

2.00

305

2.50

351

3.00

335

3.50

303

4.50

257

5.50

216

7.50

162

9.50

117

11.50

77

14.50

47

18.50

24

26.50

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given 5-ISMN by IV infusion. You wish to maintain a plasma concentration


of 300 ng/mL. If the volume of distribution of 5-ISMN is 44.5, determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 300 ng/mL.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 1 hour.

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 1 hour.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-27

I.V. Infusion

Isosorbide 5-mononitrate on page 27

CONCENTRATION

103

102

101

100
0

10

15

20

25

30

Time
1.

k = 0.168 hr-1

2.

t 1 2 = 4.125 hr

3.

V d = 44.6 L

4.

Cl = 7.5 L/hr

5a.

Q = 2.25 mg/hr

5b.

D L = 13.4 mg

5c.

5d.

C p = 46.4 ng/mL

5e.

C p = 33.2 ng/mL

ss
= 17.8 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-28

I.V. Infusion

Moclobemide

(Problem 5 - 10)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Schoerlin, M., et al., "Disposition kinetics of moclobemide a new MAO-A inhibitor, in subjects with impaired renal function", Journal of Clinical Pharmacology Vol 30 (1990), p. 272 - 284.

Moclobemide is reversibly inhibits the A-isozyme of the monoamine oxidase enzyme system. In this study, twelve
patients received a 96.7 mg dose as an intravenous infusion over 20 minutes. Blood samples were obtained during the
infusion and after the infusion was ended and the following data was obtained:
PROBLEM TABLE 5 - 10. Moclobemide

Time (hours)

Plasma concentration (mg/L)

0.0

0.000

0.2

0.6

0.4

0.7

0.85

0.9

0.750

1.2

0.70

1.6

0.60

1.9

0.50

2.4

0.40

3.4

0.25

4.5

0.15

5.5

0.10

6.4

0.070

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given moclobemide by IV infusion. You wish to maintain a plasma concentration


of 1mg/L. Determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 1mg/L.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 15 min.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-29

I.V. Infusion

e.

Find the plasma concentration 3 hours after infusion is discontinued at time = 15 mins.

Moclobemide on page 29

CONCENTRATION

10 0

10-1

10-2
0

1.

k = 0.44 hr-1

2.

t 1 2 = 1.6 hr.

3.

V d = 90.4 L

4.

Cl = 39.8 L/hr

5a.

Q = 40 mg/hr

5b.

D L = 90 mg

5c.

5d.

C p = 0.1 mg/L

5e.

C p = 0.028 mg/L

Time

ss
= 6.8 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-30

I.V. Infusion

Obidoxime

(Problem 5 - 11)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Bentur, Y., et al., "Pharmacokinetics of obidoxime in organophosphate poisoning associated with renal failure", Clinical Toxicology (1993), Vol. 31, p. 315 - 322.

Obidoxime is an agent which is used as an antidote in organophosphate poisoning. In this study, the pharmacokinetics
of obidoxime were studied in a 20 year old patient who attempted to commit suicide by ingesting Tamaron (60% methamidophos, an organophosphate, in ethylene glycol monethyl ether). She was given 4 mg/kg Obidoxime by intravenous infusion over 10 minutes and the following data was collected:
PROBLEM TABLE 5 - 11. Obidoxime

Time (minutes)

Plasma concentration

10

18

15

17

30

16

45

15

60

14

90

12

120

11

150

9.3

180

240

6.1

300

4.6

g mL

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given obidoxime by IV infusion. The patient has a body weight of 60 kg.
You wish to maintain a plasma concentration of 10 g/mL. Determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 10 ug mL .

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 30 minutes.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-31

I.V. Infusion

e.

Find the plasma concentration 1 hour after infusion is discontinued at time = 30 minutes.

Obidoxime on page 31

CONCENTRATION

102

101

100
0

50

100

150

200

250

300

Time
1.

k = 0.00463 min-1

2.

t 1 2 = 150 min

3.

V d = 0.22L/kg

4.

Cl = 1 mL/min

5a.

Q = 0.61 mg/min

5b.

D L = 132 mg

5c.

5d.

C p = 1.3 mg/L

5e.

C p = 0.98 mg/L

ss
= 10.8 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-32

I.V. Infusion

Perindoprilat

(Problem 5 - 12)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Macfadyen, R., Lees, K., and Reid, J., "Studies with low dose intravenous diacid ACE inhibitor (perindoprilat) infusions in normotensive male volunteers", Journal of Pharmaceutical Sciences (1991), p. 115 - 121.

Perindoprilat and other ACE inhibitors are used in the management of hypertension and chronic congestive heart failure. In this study, a 1 mg dose was infused over a one hour period. The following data was collected:
PROBLEM TABLE 5 - 12. Perindoprilat

Time (minutes)

Plasma concentration

4.0

10

9.0

20

16.0

30

24.0

40

30.0

50

36.0

60

42.0

65

40.0

70

38.0

80

35.0

90

32.0

100

29.0

110

27.0

120

24.0

ng mL

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given perindoprilat by IV infusion. You wish to maintain a plasma concentration


of 30 ng/ml. Determine the following:
a.

Calculate the infusion rate necessary to maintain a plasma concentration of 30 ng/mL.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 5 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-33

I.V. Infusion

e.

Find the plasma concentration 2 hours after infusion is discontinued at time = 5 hours.

Perindoprilat on page 33

CONCENTRATION

102

101

100
0

20

40

60

80

100

120

Time
1.

k = 0.0087 min-1

2.

t 1 2 = 79.6 min

3.

V d = 18.9 L

4.

Cl =164 mL/min

5a.

Q = 5 g/min

5b.

D L = 0.57 mg

5c.

5d.

C p = 27.8 ng/mL

5e.

C p = 9.8 ng/mL

ss
= 5.73 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-34

I.V. Infusion

Sulfonamides

(Problem 5 - 13)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Boddy, A., Edwards, P., and Rowland, M., "Binding of sulfonamides to carbonic anhydrase: influence on distribution within blood
and on pharmacokinetics", Pharmaceutical Research (1989), p. 203- 209

This study looks at the affinity of sulfonamides for carbonic anhydrase. Doses of 8 micromoles/kg were administered
via the jugular vein cannula in approximately 0.5 mL of PEG 400 over 5 minutes at a constant rate. Samples were collected during the infusion period and for 30 minutes afterward. The following set of data was collected:
PROBLEM TABLE 5 - 13.

Sulfonamides

Time (minutes)

Plasma concentration

2.0

17.0

4.0

31.0

5.0

37.0

7.5

32.0

9.0

28.0

12.0

22.5

15.0

18.0

18.0

14.0

23.0

11.0

30.0

6.5

35.0

4.5

( M )

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A 70-kg patient is to be given a sulfonamide by IV infusion. You wish to maintain a plasma


concentration of 30 M. Determine the following:
a.

Calculate the infusion rate which would be necessary to maintain the plasma concentration of 30 M.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 4 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-35

I.V. Infusion

e.

Find the plasma concentration 30 minutes after stopping infusion at time = 4 hours.

Sulfonamides on page 35

CONCENTRATION

102

101

100
0

10

15

20

25

30

35

Time
1.

k = 0.0705 min-1

2.

t 1 2 = 9.8 min

3.

V d = 0.18 L/kg

4.

Cl = 12.7 mL/min/kg

5a.

Q = 26.9 mole/min

5b.

D L = 380 mole

5c.

5d.

C p = 30 mole/L

5e.

C p = 3.6 mole/L

ss
= 42 min
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-36

I.V. Infusion

Terodiline

(Problem 5 - 14)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Hallen, B. ,et al., "Bioavailability and disposition of terodiline in man", Journal of Pharmaceutical Sciences (1994), p. 1241 1246.

Terodiline is an agent which works as an anticholinergic and a calcium antagonist. It is used to treat incontinence. It
is metabolized into p-Hydroxyterodiline, which is further metabolized to 3,4-dihydroxyterodiline. The parent drug and
all of its metabolites are excreted into the urine as well as the feces. A patient is given 12.5 mg of Terodiline by IV
infusion at a rate of 1 mL/ minute for 5 minutes. The following data is collected:
PROBLEM TABLE 5 - 14.

Terodiline
Time (hours)

Plasma concentration

25.000

31

50.000

23

75.000

15

100.000

12

125.000

150.000

175.000

200.000

225.000

g L

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given terodiline by IV infusion. You wish to maintain a plasma concentration


of 40 mcg/L. Determine the following:
a.

Calculate the infusion rate necessary to maintain the plasma concentration of40 mcg/L.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 5 hours.

e.

Find the plasma concentration 2 hours after stopping infusion if the infusion ended at
time = 5 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-37

I.V. Infusion

Terodiline on page 37

CONCENTRATION

102

101

100
0

50

100

150

200

250

Time
1.

k = 0.0136 hr-1

2.

t 1 2 = 50.9 hr

3.

V d = 283 L

4.

Cl =3.85 L/hr

5a.

Q = 0.154 mg / hr

5b.

D L = 11.32 mg

5c.

5d.

C p = 2.63 g/L

5e.

C p = 2.56 g/L

ss
= 220 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-38

I.V. Infusion

Tinidazole

(Problem 5 - 15)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Robson, R., Bailey, R., and Sharman, J., "Tinidazole pharmacokinetics in severe renal failure", Clinical Pharmacokinetics (1984),
p. 88 - 94.

Tinidazole is an antimicrobial similar to metronidazole which is used in the treatment of trichomoniasis, giardiasis,
amoebiasis, and anaerobic infections. This study focuses on the pharmacokinetics of tinidazole in patients suffering
from severe renal failure. Twelve patients received 800 mg of tinidazole dissolved in 400 mL of dextrose monohydrate
solution as an intravenous infusion at a rate of 60 mg/min. Blood samples were taken and the following data was
obtained:
PROBLEM TABLE 5 - 15.

Tinidazole
Time (hours)

Plasma concentration (mg/L)

14.9

13.1

11.2

12

8.9

24

5.1

48

2.1

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given tinidazole by IV infusion. Determine the following:


a.

Calculate the infusion rate necessary to maintain the plasma concentration of 25 mg/L.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 1 hour.

e.

Find the plasma concentration 2 hours after stopping infusion if the infusion was stopped
at time = 1 hour.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-39

I.V. Infusion

Tinidazole on page 39

CONCENTRATION

102

101

100
0

10

1.

k = 0.04136 hr-1

2.

t 1 2 = 16.75 hr

3.

V d = 54.7 L

4.

Cl = 2.26 L/hr

5a.

Q = 56.6 mg/hr

5b.

D L = 1.37 g

5c.

5d.

C p = 1 mg/L

5e.

C p = 0.93 mg/L

20

Time

30

40

50

ss
= 72.4 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-40

I.V. Infusion

Tobramycin

(Problem 5 - 16)

Problem Submitted By: Maya Leicht

AHFS 00:00.00

Problem Reviewed By: Vicki Long

GPI: 0000000000

Cooney, G., et al., "Absolute bioavailability and absorption characteristics of aerosolized tobramycin in adults with cystic fibrosis", Journal of Clinical Pharmacology Vol. 34, (1994), p. 255- 259.

Most persons with cystic fibrosis (CF) become colonized with Pseudomonas aeruginosa in their bronchial secretions
within their second decade of life. These patients require frequent treatment with potent anti-pseudomonal antibiotics
such as Tobramycin. In this study, an intravenous infusion of 2.5 mg/kg tobramycin was given over 35 minutes. The
following data was collected:
PROBLEM TABLE 5 - 16.

Tobramycin

Plasma concentration
Time (minutes)
35

8.00

60

6.00

90

4.50

150

2.50

270

0.75

mg

------L

From this data determine the following:


1.

2.

t1 2

3.

Vd

4.

Cl

5.

A patient is to be given tobramycin by IV infusion. The patient has a body weight of 70 kg. You wish
to maintain a plasma concentration of 10 mg/L. Determine the following:
a.

Calculate the infusion rate necessary to maintain the plasma concentration of 10 mg/mL.

b.

Suggest a loading dose for the patient which would give you Cpss immediately.

c.

How long will it take to reach steady state?

d.

Find the plasma concentration if the infusion is discontinued at time = 30 minutes.

e.

Find the plasma concentration 1 hour after stopping infusion if the infusion wasstopped at
time = 30 minutes.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-41

I.V. Infusion

Tobramycin on page 41

CONCENTRATION

101

100

10-1
0

50

100

150

200

250

300

Time
1.

k = 0.01 min-1

2.

t 1 2 = 69.3 min

3.

V d = 0.269 L/kg

4.

Cl = 2.7 mL/min

5a.

Q = 0.027mg/kg/min = 1.62 mg/kg/hr

5b.

D L = 2.7 mg/kg

5c.

5d.

C p = 2.6 mg/L

5e.

C p = 1.43 mg/L

ss
= 300 min = 5 hr
95%

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

5-42

CHAPTER 6

Biopharmaceutical Factors

Author: Michael Makoid


Reviewer: Phillip Vuchetich

OBJECTIVES
After successfully completing this chapter, the student shall understand:
1.

Physiology and machanisms of absorbtion

2.

Effects of diffusion, cardiac output / blood perfusion, physical properties of the


drug and body on distribution

3.

Biotransformation, first pass effect, and clearance

4.

Renal, biliary, mammary, salivary, other forms of excretion.

5.

identify the effects of physiological changes with age, sex, and disease on the
absorption, distribution, metabolism, and excretion of a drug.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

6-1

CHAPTER 7

Oral Dosing

Author: Michael Makoid and John Cobby


Reviewer: Phillip Vuchetich

OBJECTIVES
After successfully completing this chapter, the student shall be able to
1.

Given patient drug concentration and/or amount vs. Time profiles, the student will
calculate (III) the relevant pharmacokinetic parameters ( V d , K, k m , k r , k a , AUC ,
Clearance, MRT, MAT) available from oral data.

2.

Given patient drug concentration and/or amount vs. Time profiles, the student will
calculate (III) the K from the terminal portion of the curve.

3.

Given patient drug concentration and/or amount vs. Time profiles, the student will
calculate (III) the k a from either the curve stripping Moment techniques.

4.

Given patient drug concentration and/or amount vs. Time profiles, the student will
calculate (III) the Absolute Bioavailability from comparing IV and oral (or
some other process which involves absorption) data.

5.

Given patient drug concentration and/or amount vs. Time profiles, the student will
calculate (III) the Comparative Bioavailability from comparing the generic to the
inovator product.

6.

Given patient drug concentration and/or amount vs. Time profiles, the student will
qualitatively evaluate (IV) bioequivalence as determined by rate of absorption
(peak time) and extent of absorption (Area Under the Curve - AUC, and ( Cp ) max ).

7.

Given patient drug concentration and/or amount vs. Time profiles, the student will
evaluate (IV) bioequivalence data.

8.

Given patient drug concentration and/or amount vs. Time profiles, the student will
lucidly discuss (IV) bioequivalence and recommend (V) to another competant
professional if s/he believes products to be equivalent.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-1

Oral Dosing

9.

The student shall be able to properly use vocabulary relative to bioequivalence.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-2

Oral Dosing

7.1 Oral dosing


7.1.1

VALID EQUATIONS: ( ORAL DOSING, PLASMA)

where

CB

ka
k t
Kt
C p = fD
------ -------------- (e e a )
Vd k a K

(EQ 5-18)

AUC ( oral ) Dose ( oral )


f = -------------------------------------------------------------AUC ( iv ) Dose ( iv )

(EQ 5-19)

AUC ( generic ) Dose ( generic ) CB = ------------------------------------------------------------------------------------AUC ( inovator ) Dose ( inovator )

(EQ 5-20)

ln ( k a K )
t p = ----------------------( ka K )

(EQ 5-21)

X
-----a- = K AUC ( C p + K AUC t )
v

(EQ 5-22)

= the comparative bioavailability

f = the absolute bioavailabilty; the fraction of dose which ultimately reaches systemic circulation (which is made up of the fraction of the dose which is absorbed
times the fraction which gets past the liver (first pass effect))
ka

7.1.2

= absorption rate constant.

UTILIZATION

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-3

Oral Dosing

Ampicillin

(Problem 5 - 17)

The following information is available for ampicillin: 90% is excreted unchanged and a 250 mg IV bolus dose yields an
AUC of 11 mic/mL*hr. The following blood level profile has been reported for two brands of ampicillin which were
given as 500 mg oral capsules.

TABLE 4-7

Time (hr)

g
MEAN SERUM LEVEL -------mL
LEDERLE

BRISTOL

0.5

0.37

0.38

1.0

1.97

1.91

1.5

2.83

2.49

2.0

3.15

3.11

3.0

2.73

2.79

4.0

1.86

1.95

6.0

0.43

0.49

Find the following:.


a.

k for both products.

b.

k a for both products.

c.

k u for both products.

d.

AUC for both products.

e.

f for both products.

f.

t max for both products.

g.

Cl

h.

Vd

Cp 0 for a 250 mg IV dose.

j.

Cp max

k.

Are these two products bioequivalent? Why or why not?

l.

What infusion rate would be necessary to maintain a serum

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-4

Oral Dosing

plasma concentration of 2mcg/mL

The data was plotted as above with the best fit line drawn. From the graph the following parameters were derived:
TABLE 4-8 Comparison

of Ampicillin
Lederle

Bristol

K ( hr )

0.688

0.635

Ratio (L/B)

K a ( hr )

0.858

0.831

T max ( hr )

1.74

1.8

0.97

( C p ) max ( g mL )

2.9

1.03

AUC (trapaziodal)

11.4

11.6

0.98

2) In a clinical study (DiSanto & DeSante, JPS 64:100,1975) prednisone was


administered to 22 adult healthy volunteres (average weight 64.5 kg) either as one
50 mg tablet (product A) or as ten 5 mg tablets (product B). The following data
was observed:
Time (hours)

Concentration (mic/100ml plasma)


A

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-5

Oral Dosing

0.5

40.8

57.3

70.0

77.1

79.5

82.3

80.7

69.4

68.6

60.6

49.4

48.0

35.0

33.7

12

15.3

17.4

24

2.1

3.0

Find ka's for both products.


Calculate peak time and Cp max and AUC for both products.
Can you conclude that these products are bioequivalent ?
(Reasons should include discussion of rate and extent of absorption)

Answer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-6

Oral Dosing

Product A
Ka (hr^-1)

1.19

Tmax (hr)

Product B

Ratio (A/B)

1.8
2

1.52

1.31

Cmax (mcg/100mL)83.2

82.8

1.00

AUC (trapazoidal)676.52

688.81

0.976

Can you conclude that these products are bioequivalent ?


No, Time to peak (Tmax) is outside guidelines.
3) Wilkenstein et al.(Gastroenterology 74:360,1978) tested 12 normal healthy
volunteers in a four way crossover design of four dosage forms containing 300 mg
of cimetadine. The following data was obtained:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-7

Oral Dosing

A.U.C. (mic/ml x hr)------

5.2

5.4

% recovered in urine intact77.177.147.149.0


Peak serum conc.(mic/ml)------ 1.53 1.44
Onset (hr)

Duration (hr)

4.5

Time to peak (hr)0

0.34

0.65

4.0

4.2

4.4

1.0

2.0

A = IV bolus B = IM inj. C = Oral Liq. D = Oral Tab.

The plasma concentration - time profile for product A is as follows:

time(hrs)

(ug/ml)

time(hrs)

(ug/ml)

1.79

0.45

1.36

12

0.08

0.78

a} Using linear regression, find K & Cp0.


b} What is the absolute bioavailability (f) of the liquid.
c} How does that correlate with % recovered intact in the urine?
d} Would you consider the oral forms bioequivalent?
Why/Why not?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-8

Oral Dosing

f} What infusion rate would you suggest to maintain a plasma concentration of


0.75 mic/ml ?
g} How long would it take that infusion rate to attain a therapeutic plasma concentration of 0.5 mic/ml ?
Answer:

IV Bolus Parameters:
Cp max2.4 mic/mL
AUC 8.5
K

0.283 hr^-1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-9

Oral Dosing

a} Using linear regression, find K & Cp0. (graph)

b} What is the absolute bioavailability (f) of the liquid.


5/2/8.5 = 0.61

c} How does that correlate with % recovered intact in the urine?


Very well. Only 61% (f) of liquid gets in and you would expect only 77% of that to
show up in the urine because only 77% of the IV dose shows up in the urine
(.61*.77=.47).

d} Would you consider the oral forms bioequivalent? (No)


Why/Why not? Ratio of peak times ouside guidelines.

e} What infusion rate would you suggest to maintain a plasma concentration of


0.75 mic/ml ?
Q = Cpss * K * V = 0.75 mg/L * 0.283 hr^-1 * 125 L = 26.54 mg/hr

f} How long would it take that infusion to attain a therapeutic plasma concentration of 0.5 mic/ml ?
Cp = Q/(K*V)(1-exp(-K*T) = 0.5 = 26.54/(0.283*125)*(1-exp(-0.283*T)) --> 3.9
hr
4) LYSERGIC ACID DIETHYLAMIDE (LSD) was given to human volunteers at
the dose of 150 mic orally. (Impregnated blotter dosage form.) The following data
was obtained:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-10

Oral Dosing

Time

Cp (ng/ml)

Time
2.0

Cp (ng/ml)

0.25

1.75

4.6

0.5

2.9

3.0

4.1

0.75

3.7

4.0

3.3

1.0

4.2

6.0

2.1

1.5

4.6

8.0

1.4

a) Find ka

b) An IV dose of 100 mic resulted in an AUC of 20.4 ng/ml*hr. Find f.

c) The volunteers ability to concentrate as measured by their ability to do standard


tasks was also monitored. (100% control means no drug interference.) The following data was obtained:
Cp (ng/ml)

% Control

Cp (ng/ml)

5.5

33

1.5

65

4.1

40

1.1

80

2.9

52

% Control

If 100 mic dose were given by IV bolus, how long would it be before the volunteer
would regain 80% of his control?
Answwer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-11

Oral Dosing

Evaluation of the graph of Concentration vs. time yields:

Cpmax

4.63 ng/mL

T max

1.7 hr

AUC (trap)30.07
K

0.225 hr^-1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-12

Oral Dosing

m (-K)

-0.225 hr^-1

Ka

1.22 hr^-1

f (AUCoral/Doseoral)/(AUCiv/Doseiv) = .98

Evaluation of the graph of response vs ln(concentration) yields:


dR/dln(c) = 27.86

Multiplying dR/dln(c) * dln(c)/dt (m of the previous graph) yields dR/dt = 27.86 *


-0.225 = 6.26%/hr

100 mic dose IV yields Cp0 of (Cp0 =AUC * K = 20.4 * 0.225) 4.59ng/mL.

The response of a 100 mic dose is (R = 27.86*ln(4.59)+19.9) 62.3%

Response = Response at t=0 - dR/dt * t


20% = 62.3% - 6.26%/hr * t hours
T

= 6.76 hours

5. The following data was collected from a double blind cross over study between
500 mg dose of cloxacillin made by Bristol (Tegopen@) and a generic product
which you might want to put in your store.
Time

(Conc. mic/ml)

Time

TEGOPEN GENERIC

(Conc. mic/ml)
TEGOPEN GENERIC

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-13

Oral Dosing

0.25

.41

0.1

1.5

6.93

7.75

0.5

8.56

6.39

4.95

5.16

0.75

11.97

11.44

11.28

11.42

9.57

9.64

1.25

3
4

2.19
1.48

2.29
1.30

Calculate the comparative bioavailability.


Would you consider these products bioequivalent? Why/Why not?
Answer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-14

Oral Dosing

Evaluation of the above graphs yields:


Tegopen

Cpmax (mic/mL)10.8

GenericRatio (G/T)

9.94

0.92

T max (hr)

0.74

0.89

1.20

AUC (trap)

21.7

21.06

0.97

K (hr^-1)

0.72

0.8

ka (hr^-1)

4.3

2.69

Actual evaluation of ka and peak time is dificult because of the pucity of data at
early time points however all relavent parameters meet guidlines.
7. The F.D.A. reported the following data submitted to be consideration regarding
the equivalence of Mylan Pharmaceuticals' Tetracycline with that of Lederle and
an intervenous bolus dose. (Dose 250 mg).
Time(hrs) Conc.(mcg/ml) Time(hrs)
Lederle Mylan I.V.

Conc.(mcg/ml)

Lederle Mylan I.V.

0.5

0.55 0.20 5.2

2.70 2.60 2.9

1.80 1.35 4.8

2.20 1.80 2.1

1.5

2.11 1.75 4.4

1.35 1.25 1.26

2.35 2.10 4.0

12

0.83 0.74 0.76

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-15

Oral Dosing

2.65 2.25 3.4

15

0.50 0.45 0.46

Would you consider Mylan to be bioequivalent to the Lederle product ?


Calculate the absolute bioavailability of Lederle Tetracycline.(.77)
f) Calculate the volume of distribution of tetracycline. (44.3 L)
g) Tetracycline has a pKa of 9.7. Tetracyclines tend to localize in the dentin and
enamel of developing teeth causing hypoplasia and permanent discoloration of
teeth. Would you recomend tetracyline for a 110 pound lactating mother ?
Support your argument with the dose of the child. (Child's weight 11 lbs. and he
eats 2 oz of milk every 2 hours. Mom's average plasma concentration is maintained at 3 mic/ml by taking 250 qid. pH of the milk is 6.1, pH of blood is 7.4)
Answer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-16

Oral Dosing

Pharmacokinetic parameters:
Lederle
Cpmax (mic/mL)2.75 2.42
Tmax (hr)

3.04

AUC

Mylan

IV

5.65
3.08

26.4

0
23.3

k (hr^-1)

0.165

0.161

Ka (hr^-1)

0.684

0.729

31.4
0.167

Ratio of bioequivalence parameters (Cpmax, Tmax and AUC) are all within guidelines. So, the would be considered bioequivalent.

Absolute bioavailability f (= (AUCoral/DOSEoral)/(AUCiv/DOSEiv) = (26.4/


250)/(31.4/250) is 0.84.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-17

Oral Dosing

Volume of Distribution (Dose/Cp0 = 250 mg/ 5.65 mg/mL) is 44.2 L

The ratio of milk to blood is about 200.

r(m/b) = (10^(pKa-pH) + 1)milk / (10^(pKa-pH) + 1)blood


= (10^(9.7-6.1)+1)/(10^(7.4-6.1)+1) = 10^3.6/10^1.3 = 10^2.3 = 200

Dose the kid gets is mom's plasma concentration * Ratio(M/b) * volume of milk /
day = 3 mic/mL * 200 * 60cc * 12 feedings = 432 mg.day
Mom gets 1000 mg/day

Ratio of dose on a mg/kg basis (kid/mom) = (432/5)/1000/50) = 4.32 - Kid's getting more than mom.
Fifty miligrams of ketameperidine was given by IV bolus. The following urinary
profile was obtained for the only metabolite N-methyl-ketameperidine:

Collection period (hr)

Mean urinary excretion rate (mg/hr)

0.0 - 0.5

2.26

0.5 - 1.5

5.83

1.5 - 2.5

5.43

2.5 - 3.5

4.60

3.5 - 5.0

2.36

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-18

Oral Dosing

5.0 - 7.0

1.47

7.0 -10.0

0.96

10.0 -18.0

0.44

Calculate K, km and ku.


What Percent of ketameperidine was metabolized?

Answer:
With only one data point in the early time points, the larger rate constant is in question. The terminal slope is assumed to be K. The AUC will yield the amount of
ketameperidine which was metabolized (dXmu/dt * t = Xmu).

K (hours^-1) 0.216
AUC (mg)30.3

30.3 mg showed up as metabolite = 60.6% of 50 mg dose.

km = 60.6% * K = 0.131 hours^-1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-19

Oral Dosing

kr = K - km = 0.085 hours^-1
Aminophylline consists of THEOPHYLLINE (85% W/W) & Ethylene diamine
(15% W/W)
THEOPHYLLINE is the active compound measured in blood.
THEOPHYLLINE has a volume of distribution of 0.45 l/kg.
THEOPHYLLINE is 10% excreted unchanged and 90% metabolized to inactive
metabolites.
THEOPHYLLINE has a therapeutic range between 20 and 10 mg/l.
AUC FROM 0 to infinity for THEOPHYLLINE (given as 400 mg AMINPHYLLINE) is 120 mg/l x hr.
The average plasma concentration of THEOPHYLLINE given as 400 mg of AMINOPHYLLINE is as follows:
time

conc. time

conc.

(hrs)

(mg/L) (hrs)

(mg/L)

0.5

7.24

4.0

8.06

1.0

9.56

6.0

6.89

2.0

10.00 8.0

5.57

3.0

8.84

4.53

10.0

Find f, K, ka, Vd,total body clearance.


Find the infusion rate necessary to maintain a plasma concentration of 15 mg/l.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-20

Oral Dosing

Answer:
AUC (mg/L)*hr117.8
K (hr^-1)

0.096

ka (hr^-1)

2.11

f = (AUCoral/DOSEoral)/(AUCiv/DOSEiv) =
= (117.8 / 400 )/(120 / 400 ) = 0.98

Vd
AUC * K = Cp0iv
120 * 0.096 = 11.52 mg/L

Vd = Dose/Cp0 = (400mg*0.85)/11.52 = 29.5 L

TBC = K * Vd = 0.096/hr * 26.5L = 2.83 L/hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-21

Oral Dosing

Infusion rate = Q = Cpss * TBC = 15 mg/L * 2.83 L/hr = 42.45 mg/hr Theophylline = 42.45/.85 = 50 mg/hr Aminophylline

Abbott labs has provided the following data conserning their ORETIC tablets
(hydrochlorthiazide tablets U.S.P.) Dose given was 50 mg.

time

conc. time

conc.

(hrs)

(mg/L) (hrs)

(mg/L)

0.5

0.05

3.0

0.31

1.0

0.21

4.5

0.23

1.5

0.27

6.0

0.18

2.0

0.31

8.0

0.12

a Find K, ka, Cmax,

Answer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-22

Oral Dosing

The data is plotted both without (first figure) and with (second figure) a lag-time
which is associated with the release of the drug from the delivery system. Note
that the addition of the lag-time improves the fit.

The parameters obtained from each fit are:

WithoutWith

Cpmax (mg/L)0.22

0.31

Tmax (hr)

2.28

3.45

AUC (mg/L*hr)2.2

2.26

K (hr^-1)

0.216

0.201

ka (hr^-1)

0.380

1.10

t lag (hr)

0.0

0.393

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-23

Oral Dosing

It takes the tablet about 20 minutes to release the drug!

Wilkenstein et al.(Gastroenterology 74:360,1978) tested 12 normal healthy volunteers in a four way crossover design of four dosage forms containing 300 mg of
cimetadine. The following data was obtained:
A
B
C
D
AUC(mic/ml x hr)

---

recovered in urine intact77.177.1

---

5.2

54.9

55.8

Peak serum conc.(mic/ml)--- --Onset (hr)

Duration (hr)

4.5

Time to peak (hr)

1.53

1.44

4.6

5.4

0.34

0.65

4.2

4.4

1.0

2.0

A = IV Bolus B=IM injection C = Oral liquid D= Oral tablet

The plasma concentration vs. time profile for product A is as follows:


time (hrs)

conc.(ug/ml)

1.79

1.36

0.78

0.45

12

0.08

a} find K, Cp0.
Both can be found from the graph. K = .283/hr Cp0 = 2.36 mic/ml

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-24

Oral Dosing

b} What is the absolute bioavailability (f) of the liquid.


5.2/8.5 = 0.61

c} How does that correlate with % recovered intact in the urine?


Very well. Only 61% (f) of liquid gets in and you would expect only 77% of that
to show up in the urine
because only 77% of the IV dose shows up in the urine
(0.61 * .77 = .47).

d} How can you explain the variation in % recovered intact in the urine?

e} Would you consider the oral forms bioequivalent ? Why/Why not?


No. The ratio of peak times is outside the guidelines.

f} What infusion rate would you suggest to maintain a plasma concentration of


0.75 mic/ml?
Q = Cpss * K * V = 0.75 mg/L * 0.283/hr * 125L = 26.54 mg/hr

g} How long would it take that infusion rate to attain a therapeutic plasma concentration of 0.5 mic/ml ?
Cp = Q/(K * V)(1-exp(-K*T) = 0.5 = 26.54/(0.283 *125)*(1-exp(-0.283 * T)) > 3.9 hr
Roxane labs of Columbus, Ohio offers the following data for your review of their
Quinidine Sulfate tablets (Dose 200 mg). It is compared against the reference
standard by Ely Lilly and company at the same dose.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-25

Oral Dosing

Time (hours)Concentration (mcg/ml)


Roxane

Lilly

.42

.58

.73

.77

.71

.74

.61

.66

.45

.52

.32

.34

12

.20

.22

a) Calculate the comparative bioavailability.


b) Would you consider Roxane Quinidine Sulfate to be bioequivalent to the Lilly
product ?

Answers

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-26

Oral Dosing

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-27

Oral Dosing

Roxane labsEli Lilly


w/o

w/o

Rw/o(R/L)Rw(R/L)

Cpmax (mcg/mL)0.650.740.740.76 0.88

0.97

AUC (mcg/mL*hr)6.086.236.756.84 0.90

0.91

Tmax (hr)

2.69

2.05

2.33

2.10

T lag (hr)

0.0

0.70

0.0

0.36

1.15

0.98

Yes. Ratios are within guidelines.


Shand et al. offers the following data for propranolol :

Answers:

Time Concentration (ng/ml)


(hours) 10 mg I.V. 80 mg oral
0.5

--

50

--

77

1.5

--

100

29

100

24

90

18

78

15

59

11

45

32

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-28

Oral Dosing

a) find ka
b) Calculate the absolute bioavailability of propranolol.
c) Calculate TBC

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-29

Oral Dosing

IV data

Oral Data
w/o

AUC (ng/mL*hr)201.3562.8 540


Cpmax (ng/mL) 47.7 97.8

99.7

Tmax (hr)

2.0

K (hr^-1)

0.239

0.324 0.421

ka (hr^-1)

---

0.715 0.548

2.1

T lag

0.0

0.02

Absolute bioabailability = (AUCoral/DOSEoral)/(AUCiv/DOSEiv)


= (562.8/80)
(540 / 80)

/(201.3/10) = 0.35 or using lag time data


/(210.3/10) = 0.335

TBC = Dose / AUC = 10,000 mic/ 201.3 mic/L*hr

= 50 L/hr or

0.35*80,000mic /562.8 mic/L*hr = 50 L/hr


Niazi et al. offers the following data for meperadine :
Meperidine : is 95% metabolized
has an absolute bioavailability of 0.4
has a hepatic plasma extraction ratio of 0.6
has a volume of distribution of 100 L.
has a half life of 3.5 hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-30

Oral Dosing

a) Calculate TBC
TBC = K * V = (0.198/hr)(100L) = 19.8 L/hr

b) Calculate the intrinsic hepatic plasma clearance of meperidine.


19.8 L/hr * .95 = 18.8 L/hr

c) Calculate the effect on total body clearance in a patient with viral hepititis (FI =
0.3).
Clh*/Clh = (.3)(1)/1 + .6(.3 - 1) = .3/.58 = .517
(.517)(18.8) = 9.72
TBC = 1 + 9.72 = 10.72

d) Calculate the effect on total body clearance in a patient with stenosis (FR = 0.3).
Clr*/Clr = (1)(.3)/.3 + .6(1 - .3) = .3/.72 = .417
TBC = 18.8 + .417 = 19.22

e) Comment on which patient might need modification in therapy and why.


The patient with viral hepatitis would need modification in therapy. Because
of the decrease in TBC, we
can see that the drug is staying the body much
longer than normal, therefore the dosage regimen should
be decreased.
Chlorthalidone is used to treat high blood pressure. The following information is
offered regarding a generic
and a brand name chlorthalidone 50 mg tablet:
Time Conc. (mcg/ml)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-31

Oral Dosing

(hours)

Hygroton@Generic

.5

0.14

0.15

0.51

0.64

1.23

1.67

1.94

2.48

2.20

2.91

2.64

3.49

2.86

3.52

12

3.43

3.82

24

3.22

3.38

48

2.45

2.74

72

1.53

1.91

96

1.20

1.40

120

0.76

0.77

Pharmacokinetic parameters
Cpmax (mg) 3.73

4.62

Time to peak (hr) 13.810.8


AUC (0 to Inf)293

336

Xu inf (mg)18.3

22.1

Ka (hr^-1)0.168

0.253

Ke (hr^-1)0.019

0.019

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-32

Oral Dosing

Average mean83.1

84.5

blood presure

a) Calculate the comparative bioavailability.


(336/50mg)/(293/50mg) = 1.15
b) Would you consider the generic product to be bioequivalent to the USV
(Hygroton@) product? Prepare a short statement that you would tell a patient
regarding why you would or would not make a generic substitution for this drug.
No. The maximum concentration the generic is too much greater than that of
the brand name product.
They are not considered to be bioequivalent.

R(G/H)
Cpmax (mg) 1.23

outside

Time to peak (hr)0.78outside


AUC (0 to inf)115

ok

Buspirone is a new anxiolytic agent that has been found to be effective for the
treatment of generalized anxiety disorder at a mean dose of approximately 20 mg/
day orally in divided doses. Buspirone is metabolized almost entirely. Less than
0.1% is found intact in the urine. The following data has been presented by Gammans (Am J Med:80(supp 3b),41-51;1986):
Time (hours)Concentration (ng/ml)
(hours)

1 mg I.V.20 mg oral

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-33

Oral Dosing

0.25

--

1.07

0.50

4.33

1.76

1.0

3.75

2.45

2.80

2.51

2.10

2.05

1.57

1.60

0.8

0.91

a) find ka
b) Find Oral Peak Time and Oral Cmax.
c) Calculate the absolute bioavailability of buspirone.

answer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-34

Oral Dosing

IV

Oral

Cpmax (ng/mL) 5.0 2.6


AUC (0 to inf)17.4

13.9

Tmax (hr)

1.5

K (hr^-1)

0.290 0.289

ka (hr^-1)

1.3

Absolute bioavailability, f, = (AUCoral/DOSEoral)/(AUCiv/DOSEiv)


= ( 13.9 / 20)

/( 17.4/ 1

= 0.04
Valproate is a carboxylic acid anticonvulsant. Its activity may be related, at least
in part, to increase concentrations of the neurotransmitter inhibitor gamma aminobutyric acid in the brain. It is used alone or in combination with other anticonvulsants. in the prophylactic management of petit mal. It appears to be almost
entirely cleared by liver function with negligible amounts excreted into the urine
unchanged. It comes as soft gelatin capsules of 250 mg and enteric coated tablets
250 and 500 mg as well as oral syrup of 250 mg / 5 cc. Two different formulations of Valproate (250 mg) were prepared by Abbott and compared. The data is as
follows:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-35

Oral Dosing

Time(Hr.)Formulation BFormulation A
0.5

3.4

AUC = 287 mg/L * hr

1.0

6.0

Ka = 0.7 hr^-1

1.5

7.9

Ke = 0.065 hr^-1

2.0

9.3

2.5

10.3

3.0

10.9

4.0

11.6

6.0

11.4

8.0

10.5

12.0

8.3

18.0

5.7

24.0

3.8

1) find ka for formulation B.


2) Five hundred mg of valproate was administered by IV bolus. The AUC for that
route was 574 mg/L * hr. Calculate f for formulation A. Calculate Cp0 for the IV
dose.
3) Find Peak Time and Cmax for formulation A.
4) Calculate the comparative bioavailability of formulation B.
5) Would you consider formulation B to be bioequivalent to Formulation A ? Prepare a short statement in which you would substantiate that stand that you might

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-36

Oral Dosing

need to respond to another health professional who asked you to stock that formulation for his patients.
6) Calculate the Total Body Clearance (TBC) of valproate.

Answers:
Formulation B

R(A/B)

AUC = 243.3 mg/L * hr 1.18


Cpmax = 11.7 mg/L1.12
Tp max = 4.70 hr0.79
ka

= 0.493 hr^-1

= 0.0655

Tmax(A) = ln(ka/K)/(ka-K)= 3.75 hr


cpmax =
(ka/(ka-k))*(fX0/Vd)*(exp(-k*tmax)-exp(-ka*tmax)
13.3 mg/L

Absolute bioavailability, f,=(AUCoral/DOSEoral)/(AUCiv/DOSEiv)


= (287/250)/(574/500)
= 1.0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-37

Oral Dosing

Comparative bioavailability =(AUCb/DOSEb)/(AUCa/DOSEa) = 243.3/287 =


0.85
TBC = Dose / AUC = 500 mg / 574 mg /L * hr
The following data was made available by Lederle Labs regarding its generic
Procainamide HCl. (Dose 250mg).
Procainamide is a base (pka =9.1). As the hydrochloride salt it is 87% Procainamide.

Time (hrs)Conc.(mcg/ml) Procainamide Base


Lederle

Squibb

0.33

0.68

0.26

0.5

0.82

0.67

0.66

1.17

0.93

1.23

1.12

1.45

1.33

1.31

1.19

1.35

1.39

1.12

1.18

0.93

0.96

0.95

0.74

0.74

0.77

0.51

0.51

0.51

0.32

0.30

0.33

12

0.11

0.09

0.14

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

I.V.

7-38

Oral Dosing

a) find ka of the Squibb product


b) ka of the Lederle product.
c) Calculate the comparative bioavailability.
d) Would you consider Lederle to be bioequivalent to the Squibb product ?
e) Calculate the absolute bioavailability of Lederle Procainamide.
f) Calculate the volume of distribution of procainamide.
g) Would you recommend your patient breast feed her newborn? Prepare a short
consult for her physician. Support your argument with the dose of the child.
(Child's weight 11 lbs. and he eats 2 oz of milk every 2 hours. Mom's average
plasma concentration is maintained at 4 mic/ml from a 1 g dose ever 6 hours. pH of
the milk is 6.3, pH of blood is 7.4)
Procainamide is cleared about 60% by liver and 40% by kidney function. 20 % of
cardiac output (70 ml/min/kg) goes to liver, 25% goes to the kidney. Mom's
weight is 130 lb. Assuming her plasma vs time profile to be similar to the Lederle
product (i.e. pharmacokinetic parameters obtained from this information can be
used):
h) Calculate Total body clearance
i) Calculate the intrinsic hepatic plasma clearance of procainamide.
j) Calculate the effect on her total body clearance if she were to contract viral hepatitis which effect liver function (FI = 0.4). Prepare a short consult for her physician as to whether you would recommend a change in therapy. d) Calculate the
effect on her total body clearance stenosis of the liver (FR = 0.4). Prepare a short
consult for her physician as to whether you would recommend a change in therapy.
Answers:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-39

Oral Dosing

IV

LederleSquibb R(L/S)

AUC (0 to inf)8.577.46.8

1.09

Cpmax

1.8

1.28

1.25

1.02

Tmax

1.43

1.45

0.99

0.212 0.247 0.256

ka

---

1.51

1.93

t lag

0.24

Absolute bioavailability, f,=(AUCoral/DOSEoral)/(AUCiv/DOSEiv)


= 7.4

/ 8.57

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-40

Oral Dosing

= 0.86

Vd = Dose/Cp0 = 0.87*250mg/1.8mg/L= 120.8 L

Ratio of milk to blood = (10^(9.1-6.3)+1)/(10^(9.1-7.4)+1)= 12.4


Kid's dose = 4 mic/mL * 12.4 * 60 mL/feeding * 12 feedings/day * 1 mg/1000 mic
= 36 mg/day
Ratio of kid's daily dose/# to Mother's daily dose/# = (36mg/11#)/(1000mg*4/
130#) = 0.42. The kid gets about half of the mother's dose!

Nifedipine (Procardia @) is a calcium channel blocker which specifically inhibits


potential-dependent channels not receptor-operated channels, preventing calcium
influx of cardiac and vascular smooth muscle (coronary, cerebral). Calcium
channel blockers reduce myocardial contractility and A-V node conduction by
reducing the slow inward calcium current. They are indicated in angina, cardiac
dysrhythmias, and hypertension among others. Nifedipine appears to be metabolized entirely into an inactive metabolite, an acid and subsequently further metabolized to a lactone. Both the acid and the lactone are excreted into the urine and
the feces.

Echizen and Eichelbaum (Clin Pkin 1986; 11:425-49) and Kleinbloesem et al (Clin
Pcol Therap 1986; 40: 21-8) Reviewed the pharmacokinetics of Nifedipine. While
the drug is not routinely given by IV bolus and does not strictly conform to a one
compartment model, lets treat the data as if those problems can be ignored. The
following data is offered for evaluation:
25mg IV

10 mg oral tablet
Formula AFormula B

Time Cp

Cp

Cp

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-41

Oral Dosing

(hr.)

(mic/l)

(mic/l)

0.5

29.3

33.1

42.1

43.7

1.5

45.7

43.7

44.4

39.8

36.2

25.5

(mic/l)

139

3
4

65.6

27

20.7

31.1

13.6

9.9

14.6

6.5

4.7

1.5

1.0

12

a} Find ka's of the two products.


b} Calculate peak time and Cp max for both products.
d} Can you conclude that these products are bioequivalent ? (you must support you
argument)
e) Calculate the absolute bioavailability of product A.
f} What infusion rate would you suggest to maintain a plasma concentration of 30
mic/L ?
Answers:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-42

Oral Dosing

IV

R(A/B)

Cpmax (mic/L)294.25 45.7 44.01.04


Tmax (hr) 0

1.57

1.18 1.33

AUC(0 to inf)785219.7182.7 1.20


ka (hr^-1)---

1.0

1.6

K (hr^-1) 0.375 0.374 0.375

No,Tmax is outside the guidelines.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-43

Oral Dosing

Absolute bioavailability, f,=(AUCoral/DOSEoral)/(AUCiv/DOSEiv)


=(219.7/10)/(785/25)
=0.7

Q = Cpss * K * V = 0.955 mg/hr


Tetracycline HCl has a pKa of 9.7. Tetracyclines tend to localize in the dentin and
enamel of developing teeth causing hypoplasia and permanent discoloration of
teeth. Would you recommend tetracycline for a lactating mother ? Support your
argument with the dose of the child. (Child's weight 11 lbs. and he eats 2 oz of
milk every 2 hours. Mom's average plasma concentration is maintained at 4 mic/
ml she is taking 250 mg T.I.D. ( Milk pH = 6.1, Blood pH = 7.4)

Tm/Tb = 109.7 - 6.1/109.7 - 7.4 = 20/1


The concentrarion of tetracycline in the mother's milk is 80 mic/ml
The child takes in 720 ml of milk per day
80 mic/ml * 720 ml = 57600 mic = 57.6 mg
57.6mg/5kg = 11.52mg/kg = dose that the child is getting from the mother's
milk.

I would not recomend tetracycline for a lactating mother. The dose that a nursing child gets from the milk
too high.

Oxazepam (acid, pKa 11.5) is an anxyolytic sedative with the usual adult dose 10
mg 3 times daily. If the circulating plasma concentration of oxazapam were 20

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-44

Oral Dosing

mic/ml for nursing 120 lb mother, would her 9 lb infant be getting a comparable
mg per kg daily dose if he consumes 2 oz of his mothers milk every 2 hours. Prepare a short consult for her physician in which you might (or might not) recommend the patient stop breast feeding while she is on this medication. Include
appropriate calculations.

Om/Ob = 1011.5 - 6.1/1011.5 - 7.4 = 20/1


The concentration of the mother's milk would then be 400 mic/ml
400 mic/ml * 720 ml = 288000 mic given to baby = 288mg
288mg / 4.1kg = 70 mg/kg = dose/kg given to baby
This dose is much greater then that given to the mother. The mother should
discontinue breast feeding
while taking Oxazepam.
Bioequivalence studies are sometimes done within the same company to check if
the tablets of the same drug, but different strengths (with the strength normalized)
could be considered equivalent (i.e. could two 5 mg tablets be considered equal to
one 10 mg tablet). While not strictly kosher (products are not pharmaceutical
equivalents because of different strengths), it is done. Here is the results of such a
study in which Zomax 100 and 200 mg tablets were compared. (Yes, I know that
Zomax was removed from the market after a short life of only 6 months.)

Zomax 100 mg tablet 200 mg tablet 50 mg IV bolus


Time Conc AUC

Conc AUC

Conc.

(hr)

(mg/L) (0->t)

(mg/L) (0->t)

(mg/L)

0.25

1.41

0.18

4.03

0.50

0.5

1.98

0.60

5.13

1.65

0.75

2.15

1.12

5.18

2.94

2.12

1.65

4.89

4.20

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

1.14

7-45

Oral Dosing

1.56

3.49

3.37

8.33

0.764

1.05

4.80

2.26

11.14

0.512

0.707 5.67

1.51

13.03

0.343

0.318 6.70

0.68

15.22

0.154

0.143 7.16

0.306 16.20

0.069

1) What is the elimination rate constant for zomax (hr) ?


A) 0.2 B) 0.3 *C) 0.4 D) 0.5 E) 0.6

2) What is the volume of distribution of zomax given by IV bolus (L) ?


A) 43.85 B) 33.3 *C) 29.4 D) 25.9 E) 0.034

AUC = D/(Vd * K)
Vd = D/(AUC * K)
= 50mg/(4.25 * 0.4)
= 29.4 L

3) What is the volume of distribution of zomax given by 100 mg oral tablet ?


A) 43.85 *B) 33.3 C) 29.4 D) 25.9 E) 0.034

Vd = 100mg/(7.48 * 0.4)
= 33.4 L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-46

Oral Dosing

4) What is the AUC(0->infinity_ for the IV bolus dose ?


A) 2.68 B) 2.85 C) 3.55 D) 4.08 *E) 4.25

5) What is the AUC(0->infinity) for the 100 mg tablet ?


A) 7.16 *B) 7.5 C) 16.20 D) 17

E) 37.38

6) What is the absolute bioavailability of the 100 mg tablet ?


A) 0.84 *B) 0.88 C) 1

D) 1.14 E) 1.19

(7.48/100)/(4.25/50) = 0.88

7) What is the AUC(0->infinity) for the 200 mg tablet ?


A) 7.16 B) 7.5 C) 16.20 *D) 17

E) 73.98

8) What is the absolute bioavailability of the 200 mg tablet ?


A) 0.84 B) 0.88 *C) 1

D) 1.14 E) 1.19

(16.9/200)/(4.25/50) = 1

9) What is K * AUC (0->infinity) for the 100 mg tablet (mic/ml) ?


A) 2.9 *B) 3.0 C) 6.5 D) 6.8 E) 14.95
7.48 * 0.4 = 2.99

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-47

Oral Dosing

10) What is the absorption rate constant for the 100 mg tablet ?
A) 1.7 B) 2.2 *C) 2.6 D) 3.2 E) 3.7

11) What is the intercept of the extrapolated line for the 200 mg tablet ?
A) 3.5 B) 4.1 C) 5.6 D) 6.1 *E) 7.6

12) What is the absorption rate constant for the 200 mg tablet ?
A) 1.7 B) 2.2 C) 2.6 D) 3.2 *E) 4.01

13) What is the Tmax for the 100 mg tablet ?


A) 0.5

B) 0.67 C) 0.75 *D) 0.85 E) 0.95

14) What is the Tmax for the 200 mg tablet ?


A) 0.5

*B) 0.67 C) 0.75 D) 0.85 E) 0.95

15) Would you consider these two tablets bioequivalent (given normalization for
dose) (consider all ratios to be the 100 mg / 200 mg parameter normalized as to
dose where applicable)?
A) Yes
B) No, because the ratio of the ka's is 0.70
C) No, because the ratio of the AUCs is 0.44

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-48

Oral Dosing

D) No, because the ratio of the Cmaxs is 0.41


*E) No, because the ratio of the Tmaxs is 1.27

16) What infusion rate would you recommend to maintain an average plasma concentration of 1 mic/ml ?
A) 17.5 B) 13.3 *C) 11.8 D) 10.4 E) 9.0

Vd = D/Cp0 = 50mg/1.7mg/L = 29.4


Q = Cpss * K * V = 1mg/L * 0.4/hr * 29.4L = 11.8

17) What would be the concentration (mg/L) 2 hrs after discontinuing the infusion assuming you reached steady state ?
A) 0.67 B) 0.55 *C) 0.45 D) 0.37 E) 0.30

Cpss = Cp0 * e-Kt


= 1mg/L * e(-0.4 * 2)
= 0.44
A 110 pound mother breast feeds her 11 pound infant while on morphine sulfate
(base, pKa = 9.85). Mother's average circulating plasma levels are 0.5 ug/ml following a 10 mg IV dose q4h. (pH Milk = 6.1, pH blood = 7.4)

18) What is the Ratio of morphine concentration in the milk as compared to the
blood ?
A) 0.05 B) 0.5

C) 1

D) 2

*E) 20

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-49

Oral Dosing

Mm/Mb = 10(9.85 - 6.1)/10(9.85 - 7.4)


= 20

19) How much (mg) morphine is contained in 120 cc of breast milk (the child consumes 2 ounces every 2 hours)
*A) 1.2 B) 0.12 C) 0.06 D) 0.03 E) 0.003

Mother's blood conc. is 0.5mic/ml therefore her milk conc. is 10 mic/ml.


10mg/L * 0.12L = 1.2 mg

20) In your professional judgment, will the child's dose cause a problem ?
A) No, morphine does not concentrate in the milk and thus the milk is ok to drink.
B) No, the dose is too small. The ratio of the child's dose to the mother's dose is
0.12.
C) Yes, even though the dose is small, we don't want any drug to get to the child.
*D) Yes, the dose is comparable to the mother's dose. The ratio of the child's to the
mother's dose is 1.2.
E) Not my job. I only give what the doctor orders.
Answers:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-50

Oral Dosing

IV

Tablet Tablet

50 mg 100 mg200 mgR(100/200)


AUC(0 to inf)4.257.4816.9

0.89

Cpmax

1.7

2.15

5.23

0.82

Tmax

0.82

0.64

1.28

0.4

0.4

-.4

ka

---

2.76

4.01

Tmax ratio is ouside guidelines.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-51

Oral Dosing

Answers are rounded off. When you pick a foil, use that number in subsequent calculations when needed.

Rifampin (unionized free base pKa 7.9) is a drug used to treat TB. The following
data was collected following a 600 mg oral tablet from the inovator (Treatment A),
and a 600 mg oral tablet from a generic (treatment B), and a 400 mg IV dose
(Treatment C).

Concentration (mic/mL)AUC(0->t)
TreatmentA

0.5

5.3

4.8

10.3

8.6

1.5

10.2

9.8

9.4

9.8

2.5

8.9

9.2

7.5

8.4

4.7

5.9

6.7

3.7

3.6

4.1

2.2

2.2

2.5

1.3

10

1.3

1.5

0.8

12

0.8

0.92

0.5

Time (hours)
1.2
7.8

4.55
9.15

6.1

14.05
18.8

AUC(0->inf)53.957.7

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-52

Oral Dosing

(mic/mL*hr)
Lag time (min)18.610.5
Cp max10.6

9.9

Ka (hr^-1)2.66

1) What is the Cp0 for C (mg/L)?

a) 0

b) 7.8 *c) 10

d) 12

e) 15

Cp0 = AUC * K
= 39.8 * 0.25
= 9.95

2) What is the volume of distribution of Rifampin (L)?


d) 33.3 e) 26.7

a) 60

b) 51.3 *c) 40

Vd = D/Cp0
= 400mg/(9.95mg/L)
= 40.2 L

3) What is the half life for rifampin (hr)?


1.5

*a) 2.8

b) 2.3

c) 2.0

d) 1.75 e)

t1/2 = .693/0.25
= 2.77

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-53

Oral Dosing

4) What is the elimination rate constant for rifampin (hr^-1)? *a) 0.25 b) 0.3 c)
0.35 d) 0.4 e) 0.45

5) Calculate the AUC (0->1hr) for C (mic/mL*hr).


8.9 e) 17.8

a) 1.95 *b) 3.9 c) 7.8 b)

6) Calculate the AUC (12hr->inf.) for C (mic/mL*hr).


d) 1.25 e) 1.11

*a) 2

b) 1.67 c) 1.43

0.5/0.25 = 2
7) Calculate the AUC (0->inf) for C (mic/mL*hr).
40 e) 60

a) 16

b) 26.85 c) 35

8) Calculate the absolute bioavailability for the generic product.


0.95 c) 1 d) 1.05 e) 1.43

*d)

a) 0.70 *b)

(57.7/600)/(39.8/400) = 0.966
9) Calulate the comparative bioavailability for the generic product.
0.95 c) 1 *d) 1.05

a) 0.70 b)

e) 1.43

(57.7/600)/(53.9/600) = 1.07

10) Using Wagner-Nelson method, calculate the Ka for the generic product (hr^1). a) 0.45 b) 1 c) 1.55 d) 2 e) 2.45

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-54

Oral Dosing

11) Calculate the peak time for the generic product (min).
91 e) 105

a) 37 b) 67 c) 86 d)

tp = [ln(Ka/K)]/(Ka - K)
= [ln(1.37/0.25)]/(1.37 - 0.25)
= 1.52 hr = 91 min
12) Calculate the peak time for the brand name product (min).
86 d) 95 e) 105

a) 37 *b) 59 c)

tp = [ln(2.66/0.25)]/(2.66 - 0.25)
= 0.98 hr = 59 min
13) Are the two products bioequivalent?
a) yes, all federal requirements are met.
*b) no, the ratio of the peak times are out side federal requirements.
c) no, the ratio of the lag times are out side federal requirements.
d) no, the ratio of the Kas are out side federal requirements.
e) no, the ratio of the comparative bioavailabilities are out side federal requirements.

14) What is the ratio of the concentration of milk (pH 6.1) to blood (pH 7.4)? a)
0.05 b) 0.05 c) 1 d) 15.4 *e) 20

Rm/Rm = 10(7.9 - 6.1)/10(7.9 - 7.4)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-55

Oral Dosing

= 20

15) The average plasma concentration for the mother (110#) is 2.5 mg/L from a
600 mg once a day dosing regimen. If the baby (11#) drinks 780 mL of milk a day
(2 - 2.5 ounces every 2 hours), what is his daily dose (mg)?
a) 0.1 b) 0.13 c) 2

d) 30

*e) 39

Mother's blood average blood conc. is 2.5 mg/L therefore her milk conc. is 50
mg/L.
If the baby drinks 780 ml of milk he/she will get 39 mg of the drug.
16) Would you recommend mom stop breast feeding? (What % of the mom's daily
dose (mg/kg) is the baby's daily dose (mg/kg)?)
a) No, the child's dose is less than 1% of the mother's dose on a mg/kg/day basis.
b) No, the child's dose is about 5% of the mother's dose on a mg/kg/day basis.
c) Maybe, the child's dose is about 10% of the mother's dose on a mg/kg/day basis.
*d) Yes, the child's dose is about 50% of the mother's dose on a mg/kg/day basis.
e) Yes, the child's dose is about the same as the mother's dose on a mg/kg/day
basis.
17) While Rifampin is not administered by IV infusion, what would be the infusion rate necessary to maintain an average plasma concentration of 2.5 mg/L (mg/
hr)? *a) 25 b) 50 c) 100 d) 150 e) 200

Vd = D/Cp0
= 400/10.02
= 39.9 L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-56

Oral Dosing

Q = Cpss * K * Vd
= 2.5 * 0.25 * 39.9
= 25 mg/hr
18) While Rifampin is not administered by IV bolus, what would be the loading
dose necessary to obtain a plasma concentration of 2.5 mg/L (mg)? a) 25 b) 50
*c) 100 d) 150 e) 200

Loading Dose = Cpss * Vd


= 2.5 * 39.9
= 100mg
19) While Rifampin is not administered by IV infusion, what would be the infusion rate necessary to obtain a plasma concentration of 2.5 mg/L in about 2.5 to 3
hours (mg/hr)? a) 25 *b) 50 c) 100 d) 150 e) 200

Cp = [Q/(K * Vd)] * (1 - e-kt)


Q = (Cp * K * Vd)/(1 - e-kt)
= (2.5mg/L * 0.25 * 39.9L)/[1 - e(-0.25 * 2.75)]
= 50 mg/hr
20) Rifampin is a semisynthetic derivative of rifamycin B, an antibiotic derived
from Streptomyces mediterranei. The minimum inhibitory concentration for N.
menengitidis is 0.1 - 1 mic/mL. It is distributed well into bodily fluids. About 30%
shows up in the urine as free drug and active metabolite while 60% shows up in the
feces as metabolite. The secretary is hounding me to finish the exam, so the
answer to 20 is a. Also, rifampin is 85% protien bound at physiological concentrations. *a) 25 b) 50 c) 100 d) 150 e) 200
Answers:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-57

Oral Dosing

IV

Tmax

1.27

1.7

0.25

0.25

0.25

ka

2.66

1.37

---

AUC

53.9

57.7

39.8

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-58

Oral Dosing

Cpmax

10.6

9.9

10.02

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #2

(1) Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
a}Cpss b}feathering c}Wagner-Nelson method d}clearance e}f

(2) For each of the following pairs of variables (ordinate against abscissa), draw
a graph illustrating the qualitative profile of their relationship. Where appropriate, indicate the nature of important slopes, intercepts, and values. Unless your
specifically indicate on your plot that semi-log paper is being considered (write "SL"), it will be assumed that rectilinear paper is being considered. Graphs are for a
drug given by oral route where applicable.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-59

Oral Dosing

a} total amount of drug collected minus the amount collected at the time in the
urine vs time
b} Plasma concentration of a drug given by oral route vs time
c} Plasma concentration of metabolite of a drug given by IV bolus vs time
d} Steady state plasma concentration vs infusion rate
e} Steaty state plasma consentration vs clearance

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #3

SECTION I

(1) Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:)
a}Cpss b}f c}Absolute Bioavailability d}Comparative Bioavailability

(2) Compare and contrast:a}Wagner-Nelson and feathering methods


ment of plasma and urine data using Wagner-Nelson

b} Treat-

(3) For each of the following pairs of variables (ordinate against abscissa), draw
a graph illustrating the qualitative profile of their relationship. Where appropriate, indicate the nature of important slopes, intercepts, and values. Unless your
specifically indicate on your plot that semi-log paper is being considered (write "SL"), it will be assumed that rectilinear paper is being considered. Graphs are for a
drug given by oral route where applicable.
dXu/dt vs t for a drug given orally.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-60

Oral Dosing

dXmu/dt vs t for a drug given by IV bolus.


Steady state plasma concentration vs infusion rate
Steady state plasma concentration vs elimination rate constant

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #4

SECTION I

1. Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
a) clearance b)f c) absolute bioavailability d) comparative bioavailability e)
AUC

2. By means of an annotated phase diagram explain how freeze-dried pharmaceutical injectables are made.

3. For each of the following pairs of variables (ordinate against abscissa) draw a
graph illustrating the qualitative profile of their relationship. Where appropriate,
indicate the nature of important slopes, intercepts, and values. Unless you specifically indicate on your plot that semi-log paper is being considered (write "SL"), it
will be assumed that rectilinear paper is being considered.
Pharmacological Response vs time
Peak time vs ka for oral dose
Fractional change in total body clearance vs. renal clearance

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-61

Oral Dosing

AUC vs ka
AUC vs ke

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #5

SECTION I
1. Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
a) first pass effect b) f c) Intrinsic clearance
d) comparative bioavailability e) Extraction ratio

2. By means of an annotated phase diagram explain how a metastable polymorph


can be formed and how these polymorphs might effect the bioavailability of the
drug.

3. For each of the following pairs of variables (ordinate against abscissa) draw a
graph illustrating the qualitative profile of their relationship. Where appropriate,
indicate the nature of important slopes, intercepts, and values. Unless your specifically indicate on your plot that semi-log paper is being considered (write "S-L"),
it will be assumed that rectilinear paper is being considered
fractional change in total body clearance vs plasma flow for drugs having a large
extraction ratio.
Peak time vs ka for oral dose
Fractional change in total body clearance vs. hepatic clearance.
AUC vs ka

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-62

Oral Dosing

AUC vs clearance

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #6

SECTION I
1. Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
Henderson-Hasselbach relationship
Therapeutic alternatives
Therapeutic equivalents
comparative bioavailability
Extraction ratio

Briefly discuss generic substitution by the pharmacist. Include such topics as when
it might be admissable and the liabilities involved.

3. For each of the following pairs of variables (ordinate against abscissa) draw a
graph illustrating the qualitative profile of their relationship. Where appropriate,
indicate the nature of important slopes, intercepts, and values. Unless you specifically indicate on your plot that semi-log paper is being considered (write "S-L"),
it will be assumed that rectilinear paper is being considered

a)
fractional change in total body clearance vs fractional change in plasma
flow for drugs having a small extraction ratio.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-63

Oral Dosing

b)

Peak time vs dose for oral dose

c)
Fractional change in total body clearance vs. fractional change in hepatic
clearance for drugs having a large extration ratio.
d)

Ratio of milk to blood for basic drugs vs pKa.

e)

Ratio of milk to blood for acidic drugs vs pKa.

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #7

SECTION I
1. Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
a) Bioequivalance
b) Intrinsic Clearance
c) first pass effect
d) Henderson - Hasselbach equation
e) f

2.Compare and contrast absolute and relative bioavailability.


3.For each of the following pairs of variables (ordinate against abscissa) draw a
graph illustrating the qualitative profile of their relationship. Where appropriate,
indicate the nature of important slopes, intercepts, and values. Unless you specifically indicate on your plot that semi-log paper is being considered (write "S-L"),
it will be assumed that rectilinear paper is being considered

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-64

Oral Dosing

a)

Cpmax vs f for a drug given orally.

b)

Cpmax vs dose for a drug given orally.

c)

Cpmax vs Vd for a drug given orally.

d)

TBC vs Fi(H) for a drug with a high extraction ratio in the liver.

e)

TBC vs Fr(H) for a drug with a high extraction ratio in the liver

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM #9


SECTION I
1. Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
a) Henderson-Hasselbach relationship
b) Therapeutic alternatives
c) Therapeutic equivalents
d) Comparative bioavailability
e) Extraction ratio

2. Briefly discuss generic substitution by the pharmacist. Include such topics as


when it might be admissible and the liabilities involved.

3. For each of the following pairs of variables (ordinate against abscissa) draw a
graph illustrating the qualitative profile of their relationship. Where appropriate,
indicate the nature of important slopes, intercepts, and values. Unless you specifically indicate on your plot that semi-log paper is being considered (write "SL"), it
will be assumed that rectilinear paper is being considered
a)
fractional change in total body clearance vs fractional change in plasma
flow for drugs having a small extraction ratio.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-65

Oral Dosing

b)

Peak time vs dose for oral dose.

c)
Fractional change in total body clearance vs. fractional change in intrinsic
hepatic clearance for drugs having a large extraction ratio.
d)

Ratio of blood to milk concentrations for basic drugs vs pKa.

e)

Ratio of blood to milk concentrations for acidic drugs vs pKa.

PHARMACOKINETICS SECOND HOUR PRACTICE EXAM # 10


SECTION I
1. Succinctly define, stating rigorously the meaning of any symbols used and the
dimensions of measurement:
a) Henderson-Hasselbach relationship
b) Therapeutic alternatives
c) Therapeutic equivalents
d) Comparative bioavailability
e) Absolute bioavailability
f) Bioequivalents
2. Compare and Contrast: Feathering and Wagner-Nelson method.

3. For each of the following pairs of variables (ordinate against abscissa), draw a
graph illustrating the qualitative profile of their relationship. Where appropriate,
indicate the nature of important slopes, intercepts, and values. Unless you indicate
on your plot that semi-log paper is being considered (write SL), it will be assumed
that rectilinear paper is being considered. Graphs are for a drug given by an oral
delivery system where applicable.
a)

Cpss vs. K

b)

Cp vs. ka

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-66

Oral Dosing

c)

Ratio of Milk to Blood for acidic drugs vs. pKa

Pharmacokinetics practice exam #11


Pharmaceutical alternatives may have different:
I. therapeutic moieties
II. dosage forms or strengths
III. salt or ester forms of the same therapeutic moiety

2) Pharmaceutical equivalents must have the same:


I. active ingredients and strength
II. dosage form and route of administration
III. rate and extent of absorption

3) Bioequivalent drug products must have the same:


I. active ingredients and strength
II. dosage form and route of administration
III. rate and extent of absorption

4) Therapeutic equivalents are:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-67

Oral Dosing

I. pharmaceutical alternatives
II. pharmaceutical equivalents
III. bioequivalents

5) The Wagner-Nelson method


I. uses curve stripping or feathering techniques
II. can be used to find ku and km
III. uses AUC calculations

6) The Federal guidelines for for bioequivalence require that the following pharmacokinetic parameters be within + 20 % of
the innovator's product:
I. AUC, Peak time, Cpmax
II. Ka, Ke
III. Vd

7) The steady state plasma concentration of a drug given by intravenous infusion is


dependent on:
I. length of time of of infusion
II. volume of distribution
III. elimination rate constant, K.

8) The peak time of a drug given by the oral route is dependent on:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-68

Oral Dosing

I. the absorption rate constant


II. the metabolism rate constant
III. the excretion rate constant

9) The slope of the terminal portion of the graph of the metabolite of a drug which
(the drug, not the metabolite) was given
by intravenous bolus injection could be:
I. - the elimination rate constant of the metabolite
II. - the elimination rate constant of the drug
III. - the absorption rate constant of the metabolite

10) Comparative bioavailability includes calculations of the ratio(s) of the following pharmacokinetic parameters of two
oral products (generic / Innovator) normalized for dose :
I. AUC (0 to Inf)
II. Peak time
III. Cpmax

Pharmacokinetics practice second hour exam #12

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-69

Oral Dosing

Where the are only three foils (possible quesses), please use K type system:
(NOTE: if foils are equivalent, all must be selected)
A) I ONLY
B) III ONLY
C) I AND II ONLY
D) II AND III ONLY
E) I, II, AND III

1) Steady state plasma concentration obtained by continuous infusion is inversely


proportional to:
I Infusion rate
II elimination rate constant
III volume of distribution

2) Steady state plasma concentration obtained by continuous infusion is directly


proportional to:
I Infusion rate
II time
III volume of distribution

3) When calculating the AUC for an oral product using the trapezoidal rule, concentrations necessary to calculate the
first trapezoid are:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-70

Oral Dosing

I the intercept of the extrapolated line of the plasma vs. time profile.
II the concentration at time zero, Cp0
III the concentration at the first time point

4) When calculating the AUC for an IV product using the trapezoidal rule, concentrations necessary to calculate the
first trapezoid are:
I the intercept of the extrapolated line of the plasma vs. time profile.
II the concentration at time zero, Cp0
III the concentration at the first time point

5) Absolute bioavailability is a calculation which


I must be between .80 and 1.20
II compares an oral product to an IV bolus dose.
III is the ratio of the normalized AUCs of the products tested.

6) Comparative bioavailability is a calculation which


I is the ratio of the normalized AUCs of the products tested.
II must be between .80 and 1.20
III compares an oral product to an IV bolus dose.
7) When plotting the Wagner-Nelson function vs. time, a plot which proceeds horizontally for a measurable time and then
declines:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-71

Oral Dosing

I is because of poor data in the early part of the data set.


II only the declining portion should be used to calculate ka.
III is an indication of a delay in release of the drug from the delivery system, a lag
time.

8) When considering ion trapping, comparing a drug which forms sulfate salts
distributing between mother's milk and
blood, the ratio of total drug in milk to total drug in blood (Rm/b) can be
I greater than one.
II one.
III less than one.

9) When considering ion trapping, comparing a drug which forms sodium salts
distributing between mother's milk and
blood, the ratio of total drug in milk to total drug in blood (Rm/b) can be
I greater than one.
II one.
III less than one.

10) When using dry starch as a tablet disintegrating agent,


I tablet hardness is directly proportional to starch content.
II starch acts by allowing the water to wick into the tablet.
III a threshold minimum amount of starch is necessary before any disintegration
action is apparent.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-72

Oral Dosing

Cyclosporine A

(Problem 8 - 6)

Quigano, R., et al., "Effect of atropine of gastrointestinal motility and the bioavailability of cyclosporine A in rats", Drug Metabolism and Disposition, Vol. 21, No. 1, (1993), p. 141 - 143.

In this study rats with an average weight of 300 g were given either an IV bolus dose of cyclosporine A (CyA)
or an oral dose of CyA. Subsequently, doses of atropine were given; however, the data below is that which was gathered prior to atropine administration. A summary of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Bioequivalence

Dose (mg/kg)
ug- hr
AUC ------ mL

ug- hr 2
AUMC ------ mL

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug-
Cp at 1 hour ------ mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

1.

MRT iv

2.

k e , the elimination rate constant

3.

t1 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-73

Oral Dosing

4.

Cp 0

5.

Vd

6.

The plasma concentration ( Cp ) of cyclosporine A at 1 hour after the iv dose was given.

7.

AUC 0 1 hour for the iv dose

8.

f , the absolute bioavailability of oral cyclosporine A.

MRT oral .

10.

MAT oral

11.

k a , the apparent absorption rate constant.

12.

t peak for the oral dose.

13.

Cp max , the maximum concentration of the oral dosage form given as a single dose.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-74

Oral Dosing

Fosinopril

(Problem 8 - 7)

Gehr, T., et al., "The pharmacokinetics and pharmacodynamics of fosinopril in haemodialysis patients", European Journal of Clinical Pharmacology, Vol. 45, No. 5, (1993), p. 431 - 436.

Fosinopril (MW 562.6) is a new Angiotension Converting Enzyme (ACE) Inhibitor used in the treatment of
hypertension. Following oral administration, fosinopril is rapidly and almost completely hydrolyzed to its pharmacologically active metabolite, fosinoprilate (MW 435.2). About 50% of the drug is excreted unchanged through the kidneys. In this study, patients received either 7.5 mg of fosinoprilat administered intravenously or 10 mg of fosinopril
administered orally. A summary of the some of data obtained from this experiment is given below.

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Bioequivalence

Dose (mg/kg)
ug- hr
AUC ------ mL

ug- hr 2
AUMC ------ mL

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug-
Cp0 ------ mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

From the preceding data, please calculate the following:


1.

MRT iv

2.

k e , the elimination rate constant

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-75

Oral Dosing

3.

t1 2

4.

Cp 0

5.

Vd

6.

The plasma concentration ( Cp ) of fosinopril at 1 hour after the iv dose was given.

7.

AUC 0 1 hour for the iv dose

8.

f , the absolute bioavailability of oral fosinopril.

MRT oral .

10.

MAT oral

11.

k a , the apparent absorption rate constant.

12.

t peak for the oral dose.

13.

Cp max , the maximum concentration of the oral dosage form given as a single dose.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-76

Oral Dosing

Verapamil

(Problem 8 - 8)

Rutledge, D., Pieper, J., and Mirvis, D., "Effects of chronic phenobarbital on verapamil disposition in humans", The Journal of
Pharmacology and Experimental Therapeutics, Vol. 246, No. 1, (1988), p. 7 - 13.

This study focused on the effects of phenobarbital, a hepatic-enzyme inducer, on verapamil. Seven healthy
male volunteers with an average weight of 78.8 kg participated in the study. The patients received either an single oral
verapamil dose of 80 mg or a single intravenous verapamil dose of 0.15 mg/kg over 3 minutes. A summary of the some
of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Bioequivalence

Dose (mg/kg)
ug- hr
AUC ------ mL

ug- hr 2
AUMC ------ mL

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug-
Cp at 1 hour ------ mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

1.

MRT iv

2.

k e , the elimination rate constant

3.

t1 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-77

Oral Dosing

4.

Cp 0

5.

Vd

6.

The plasma concentration ( Cp ) of verapamil at 1 hour after the iv dose was given.

7.

AUC 0 1 hour for the iv dose

8.

f , the absolute bioavailability of oral verapamil.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-78

Oral Dosing

Zidovudine

(Problem 8 - 9)

Trang, J., et al., "Zidovudine bioavailability and linear pharmacokinetics in female B6C3F1 mice", Drug Metabolism and Disposition Vol, 21 (1993), p.189 - 193.

Zidovudine (AZT) is a potent inhibitor of HIV-1 during viral replication. It has been approved for the treatment of AIDS. In this study a 30 mg/kg dose of AZT was given to mice either iv or orally. :A summary of the some
of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Bioequivalence

Dose (mg/kg)
ug- hr
AUC ------ mL

ug- hr 2
AUMC ------ mL

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug-
Cp at 1 hour ------ mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

1.

MRT iv

2.

k e , the elimination rate constant

3.

t1 2

4.

Cp 0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-79

Oral Dosing

5.

Vd

6.

The plasma concentration ( Cp ) of zidovudine at 1 hour after the iv dose was given.

7.

AUC 0 1 hour for the iv dose

8.

f , the absolute bioavailability of oral zidovudine.

MRT oral .

10.

MAT oral

11.

k a , the apparent absorption rate constant.

12.

t peak for the oral dose.

13.

Cp max , the maximum concentration of the oral dosage form given as a single dose.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

7-80

CHAPTER 8

Bioavailability, Bioequivalence,
and Drug Selection

Author: Rasma Chereson


Reviewer: Umesh Banakar

OBJECTIVES
1.

Given sufficient data to compare an oral product with another oral product or an
IV product, the student will estimate (III) the bioavailability (compare AUCs) and
judge (VI) professional acceptance of the product with regard to bioequivalence
(evaluate (VI) AUC, T p and ( Cp ) max ).

2.

The student will write (V) a professional consult using the above calculations.

3.

The student will be able to calculate (III) the absolute bioavailability of drug products.

4.

The student will be able to discuss (II) the various factors affecting bioavailability.

5.

The student will be able to discuss (II) the various methods of assessing bioavailablity.

6.

The student will be able to discuss (II) In Vivo / In Vitro Correlations.

7.

The student will be able to enumerate (II) FDA requirements regarding bioequivalence.

8.

The student shall be able to utilize (III) the FDA Orange Book to make drug
product selections.

9.

The student shall be able to discuss (II) and utilize (III) reasonalble guidelines
regarding drug product selections.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-1

Bioavailability, Bioequivalence, and Drug Selection

8.1 Bioavailability, Bioequivalence and Drug Product Selection


Bioavailability and bioequivalence of drug products, and drug product selection
have emerged as critical issues in pharmacy and medicine during the last three
decades. Concern about lowering health care costs has resulted in a tremendous
increase in the use of generic drug products; currently about one half of all prescriptions written are for drugs that can be substituted with a generic product (1).
Over 80% of the approximately 10,000 prescription drugs available in 1990 were
available from more than one source (2). With the increasing availability and use
of generic drug products, health care professionals are confronted with an
ever-larger array of multisource products from which they must select those that
are therapeutically equivalent.
This phenomenal growth of the generic pharmaceutical industry and the abundance of multisource products have prompted some questions among many health
professionals and scientists regarding the therapeutic equivalency of these products, particularly those in certain critical therapeutic categories such as anticonvulsants and cardiovasculars (1, 3-5). Inherent in the currently accepted guidelines
for product substitution is the assumption that a generic drug considered to be
bioequivalent to a brand-name drug will elicit the same clinical effect. As straightforward as this statement regarding bioequivalence appears to be, it has generated
a great deal of controversy among scientists and professionals in the health care
field. Numerous papers in the literature indicate that there is concern that the current standards for approval of generic drugs may not always ensure therapeutic
equivalence (6-18).
The availability of different formulations of the same drug substance given at the
same strength and in the same dosage form poses a special challenge to health care
professionals, making these issues very relevant to pharmacists in all practice settings. Since pharmacists play an important role in product-selection decisions,
they must have an understanding of the principles and concepts of bioavailability
and bioequivalence.

8.1.1

RELATIVE AND ABSOLUTE BIOAVAILABILITY


Bioavailability is a pharmacokinetic term that describes the rate and extent to
which the active drug ingredient is absorbed from a drug product and becomes
available at the site of drug action. Since pharmacologic response is generally
related to the concentration of drug at the receptor site, the availability of a drug
from a dosage form is a critical element of a drug product's clinical efficacy. However, drug concentrations usually cannot be readily measured directly at the site of

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-2

Bioavailability, Bioequivalence, and Drug Selection

action. Therefore, most bioavailability studies involve the determination of drug


concentration in the blood or urine. This is based on the premise that the drug at
the site of action is in equilibrium with drug in the blood. It is therefore possible to
obtain an indirect measure of drug response by monitoring drug levels in the blood
or urine. Thus, bioavailability is concerned with how quickly and how much of a
drug appears in the blood after a specific dose is administered. The bioavailability
of a drug product often determines the therapeutic efficacy of that product since it
affects the onset, intensity and duration of therapeutic response of the drug. In
most cases one is concerned with the extent of absorption of drug, (that is, the fraction of the dose that actually reaches the bloodstream) since this represents the
"effective dose" of a drug. This is generally less than the amount of drug actually
administered in the dosage form. In come cases, notably those where acute conditions are being treated, one is also concerned with the rate of absorption of a drug,
since rapid onset of pharmacologic action is desired. Conversely, these are
instances where a slower rate of absorption is desired, either to avoid adverse
effects or to produce a prolonged duration of action.
"Absolute" bioavailability, F, is the fraction of an administered dose which actually
reaches the systemic circulation, and ranges from F = 0 (no drug absorption) to F =
1 (complete drug absorption). Since the total amount of drug reaching the systemic circulation is directly proportional to the area under the plasma drug concentration as a function of time curve (AUC), F is determined by comparing the
respective AUCs of the test product and the same dose of drug administered intravenously. The intravenous route is the reference standard since the dose is, by definition, completely available.
AUC ev
F = ---------------AUC iv

(EQ 8-1)

(where AUCEV and AUCIV are, respectively, the area under the plasma concentration-time curve following the extravascular and intravenous administration of a
given dose of drug. Knowledge of F is needed to determine an appropriate oral
dose of a drug relative to an IV dose.
"Relative" or Comparative bioavailability refers to the availability of a drug
product as compared to another dosage form or product of the same drug given in
the same dose. These measurements determine the effects of formulation differences on drug absorption. The relative bioavailability of product A compared to
product B, both products containing the same dose of the same drug, is obtained by
comparing their respective AUCs.
AUC
RelativeBioavailabilty = ---------------A
AUC B
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 8-2)

8-3

Bioavailability, Bioequivalence, and Drug Selection

where drug product B is the reference standard. When the bioavailability of a


generic product is considered, it is usually the relative bioavailability that is
referred to. A more general form of the equation results from considering the possibility of different doses,
AUC Generic
----------------------------Dose Generic
ComparativeBioavailability = ----------------------------AUC Brand
------------------------Dose Brand

(EQ 8-3)

The difference between absolute and relative bioavailability is illustrated by the


following hypothetical example. Assume that an intravenous injection (Product
A) and two oral dosage forms (Product B and Product C), all containing the same
dose of the same drug, are given to a group of subjects in a crossover study. Furthermore, suppose each product gave the values for AUC indicated in Table 8-1 on
page 4.
TABLE 8-1. Data

for Absolute and Relative Bioavailability

Drug Product

Area Under the Curve (mcg/ml) x hr

A Intravenous injection

100

B Oral dosage form, brand or reference standard

50

C Oral dosage form, generic Product

40

The F for Product B and Product C is 50% (F = 0.5) and 40% (F = 0.4), respectively. However, when the two oral products are compared, the relative bioavailability of Product C as compared to Product B is 80%.

8.1.2

FACTORS INFLUENCING BIOAVAILABILITY


Before the therapeutic effect of an orally administered drug can be realized, the
drug must be absorbed. The systemic absorption of an orally administered drug in
a solid dosage form is comprised of three distinct steps:
1.

disintegration of the drug product

2.

dissolution of the drug in the fluids at the absorption site

3.

transfer of drug molecule across the membrane lining the gastrointestinal tract into the systemic
circulation.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-4

Bioavailability, Bioequivalence, and Drug Selection

Any factor that affects any of these three steps can alter the drug's bioavailability
and thereby its therapeutic effect. While there are more than three dozen of these
factors that have been identified (19-38), the more significant ones are summarized
here.
The various factors that can influence the bioavailability of a drug can be broadly
classified as dosage form-related or patient-related. Some of these factors are
listed in Table 8-2 on page 5 and Table 8-3 on page 5, respectively.
TABLE 8-2 Bioavailability

Factors related to the dosage form

Physicochemical properties of the drug

Formulation and manufacturing variables

Particle size

Amount of disintegrant

Crystalline structure

Amount of lubricant

Degree of hydration of crystal

Special coatings

Salt or ester form

Nature of diluent
Compression force

TABLE 8-3 Bioavailability

Factors Related to the patient

Physiologic factors

Interactions with other substances

Variations in absorption power along GI tract

Food

Variations in pH of GI fluids

Fluid volume

Gastric emptying rate

Other drugs

Intestinal motility
Perfusion of GI tract
Presystemic and first-pass metabolism
Age, sex, weight
Disease states

The physical and chemical characteristics of a drug as well as its formulation are
of prime importance in bioavailability because they can affect not only the absorption characteristics of the drug but also its stability. Since a drug must be dissolved
to be absorbed, its rate of dissolution from a given product must influence its rate
of absorption. This is particularly the case for sparingly soluble drugs. All the factors listed in Table 8-2 on page 5 can alter the dissolution rate of the drug, its bioavailability, and ultimately, its therapeutic performance.
One of the more important factors that affects the dissolution rate of slowly dissolving substances is the surface area of the dissolving solid (39). Peak blood levels occurred much faster with the smaller particles than the larger ones, primarily
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-5

Bioavailability, Bioequivalence, and Drug Selection

as a result of their faster dissolution rate. Particle size can also have a significant
effect on AUC(40). Serum levels of phenytoin after administration of equal doses
containing micronized (formulation G) and conventional (formulation F) drug
were measured. Based on the AUC, almost twice as much phenytoin was
absorbed after the micronized preparation (40).
There are numerous reports of the effects of formulation and processing variables
on the dissolution of active ingredients from drug products; an apparently inert
ingredient may affect drug absorption. For example, magnesium stearate, a lubricant, commonly used in tablet and capsule formulations, is water-insoluble and
water-repellent. Its hydrophobic nature tends to retard drug dissolution by preventing contact between the solid drug and the aqueous GI fluids. Thus, increasing the amount of magnesium stearate in the formulation results in a slower
dissolution rate of the drug, and decreased bioavailability(34) .
The nature of the dosage form itself may have an effect on drug absorption characteristics. The major pharmaceutical dosage forms for oral use are listed in Table 84 on page 6 in order of decreasing bioavailability of their active ingredients. The
decreasing bioavailability is related to the number of steps involved in the absorption process following administration. The greater the number of steps a product
must undergo before the final absorption step, the slower is the availability and the
greater is the potential for bioavailability differences to occur. Thus, solutions
(elixirs, syrups, or simple solutions) generally result in faster and more complete
absorption of drug, since a dissolution step is not required. Enteric-coated tablets,
on the other hand, do not even begin to release the drug until the tablets empty
from the stomach, resulting in poor and erratic bioavailability.
TABLE 8-4 Bioavailability

Fastest availability

and oral Dosage Forms

Solutions
Suspensions
Capsules
Tablets
Coated tablets

Slowest availability

Controlled-release formulations

Bioavailability studies with pentobarbital from various dosage forms show the
absorption rate of pentobarbital after administration in various oral dosage forms
decreased in the following order: aqueous solution > aqueous suspension of the
free acid > capsule of the sodium salt > tablet of the free acid (41).
In addition to the dosage form-related factors identified above, bioavailability may
also be affected by a variety of physiologic and clinical factors related to the
patient (Table 8-3 on page 5). Considerable inter-subject differences in the bioBasic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-6

Bioavailability, Bioequivalence, and Drug Selection

availability of some drugs have been observed. These can often be attributed to
individual variations in such factors as GI motility, disease state and concomitantly-administered food or drugs.
One example of the myriad of physiologic factors that can affect the bioavailability
of an orally-administered drug is a patient's gastric emptying rate. Since the proximal small intestine is the optimum site for drug absorption, a change in the stomach emptying rate is likely to alter the rate, and possibly the extent, of drug
absorption. Any factor that slows the gastric emptying rate may thus prolong the
onset time for drug action and reduce the therapeutic efficacy of drugs that are primarily absorbed from the small intestine. In addition, a delay in gastric emptying
could result in extensive decomposition and reduced bioavailability of drugs that
are unstable in the acidic media of the stomach (e.g. penicillins and erythromycin).
Differences in stomach emptying among individuals have been implicated as a
major cause of variations in the bioavailability of some drugs, particularly those
with acid-resistant enteric coatings. In a study (42), after the administration of 1.5
g acetaminophen to 14 patients, the maximum plasma concentration ranged from
7.4 to 37 mcg/ml, and the time to reach the maximum concentration ranged from
30 to 180 minutes. Both these parameters of bioavailability were linearly related
to the gastric emptying half-life found in each patient.
There are numerous factors that affect gastric emptying rate (Table 8-5 on page 8)
(43). Factors such as a patient's emotional state, certain drugs, type of food
ingested and even a patient's posture can alter the time course and extent of drug
absorption.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-7

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-5

Factors influencing Gastric Emptying Rate

INFLUENCE ON GASTRIC
EMPTYING RATE

FACTOR
Increased viscosity of stomach contents

decreased

Body position
lying on left side

decreased

Emotional state
stress

increased or decreased

depression

decreased

anxiety

increased

Activity, exercise

decreased

Type of meal
fatty acids, fats

decreased

carbohydrates

decreased

amino acids

decreased

pH of stomach contents
decreased

decreased

increased

increased

Disease states
gastric ulcers

decreased

Crohn's disease

decreased

hypothyroidism

decreased

hyperthyroidism

increased

Drugs
atropine

decreased

propantheline

decreased

narcotic analgesics

decreased

amitriptyline

decreased

metoclopramide

increased

Since drugs are generally administered to patients who are ill, it is important to
consider the effects of the disease process on the bioavailability of the drug. Disease states, particularly those involving the GI tract, such as celiac disease, Crohn's
disease, achlorhydria, and hypermotility syndromes can certainly alter the absorption of a drug (32). In addition, some diseases concerning the cardiovascular system and the liver may also alter circulating drug levels after oral dosing.
Drugs are frequently taken with food, and patients often use mealtimes to remind
them to take their medications. However, food can have a significant effect on the
bioavailability of drugs. The influence of food on drug absorption has been recog-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-8

Bioavailability, Bioequivalence, and Drug Selection

nized for some time, and several reviews have been published on the influence of
food on drug bioavailability (30-32, 36, 44). Food may influence drug absorption
indirectly, through physiological changes in the GI tract produced by the food,
and/or directly, through physical or chemical interactions between the drug molecules and food components. When food is ingested, stomach emptying is delayed,
gastric secretions are increased, stomach pH is altered, and splanchnic blood flow
may increase. These may all affect bioavailability of drugs. Food may also interact directly with drugs, either chemically (e.g. chelation) or physically, by adsorbing the drug or acting as a barrier to absorption. In general, gastrointestinal
absorption of drugs is favored by an empty stomach, but the nature of drug-food
interactions is complex and unpredictable; drug absorption may be reduced,
delayed, enhanced or unaffected by the presence of food. Table 8-6 on page 9
summarizes some of the studies that have indicated the effect of food on the bioavailability of a variety of drugs.
TABLE 8-6

Effect of Food on Drug Absorption

Reduced Absorption

Delayed Absorption

Increased Absorption

Ampicillin

Acetaminophen

Chlorothiazide

Aspirin

Aspirin

Diazepam

Atenolol

Cephalosporins (most)

Griseofulvin

Captopril

Diclofenac

Hydralazine

Erythromycin

Digoxin

Labetalol

Ethanol

Furosemide

Metoprolol

Hydrochlorothiazide

Nitrofurantoin

Nitrofurantoin

Penicillins

Sulfadiazine

Propranolol

Tetracyclines (most)

Sulfisoxazole

Riboflavin

Source: Ref. 32

The effect of food and type of diet on the bioavailability of erythromycin is shown
in a study by Welling (45). The absorption of the antibiotic is significantly
reduced when it is administered with food compared with its absorption under fasting conditions. This reduced absorption is primarily a result of degradation of the
acid-labile erythromycin due to prolonged retention in the stomach.
Delayed absorption due to food has been demonstrated in the case of cephradine in
a study by Mischler (46). Similar results have been observed with other oral cephalosporins.
Some drugs demonstrate enhanced bioavailability in the presence of food. This
has been attributed to a variety of factors, including improved compound solubility
and more time for dissolution because of delayed gastric emptying. In the case of
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-9

Bioavailability, Bioequivalence, and Drug Selection

highly metabolized agents, such as propranolol and metoprolol, the enhanced


availability may be due to increased splanchnic blood flow causing reduced
first-pass clearance. The circulating levels of these drugs dosed under fasting and
non-fasting conditions have been presented in a study by Melander (47).
The volume of fluid with which an orally administered dose is taken can also affect
a drug's bioavailability. Drug administration with a larger fluid volume will generally improve its dissolution characteristics and may also result in more rapid stomach emptying. Thus, more efficient and more reliable drug absorption can be
expected when an oral dosage form is administered with a larger volume of fluid.
(45) .
Interactions between drugs can have a significant effect on the bioavailability of
one or both drugs. Such interactions may be direct, as in chelation of tetracycline
by polyvalent metal ions in antacids or the adsorption of digoxin by
cholestyramine resin, or indirect, as with the increased rate of acetaminophen
absorption due to the increased gastric emptying rate produced by metoclopramide. Most of the reported drug-drug interactions have resulted in a reduction in
the rate and/or extent of drug absorption, the most frequent causes being complexing of a drug with other substances, reduced GI motility and alterations in drug
ionization (24, 30, 32, 48, 49). Table 8-7 on page 10 summarizes the major mechanisms of GI drug interactions affecting bioavailability.
TABLE 8-7

Drug interactions affecting absorption

1. Change in gastric or intestinal pH


2. Change in gastrointestinal motility
3. Change in gastrointestinal perfusion
4. Interference with mucosal function (drug-induced malabsorption syndromes)
5. Chelation
6. Exchange resin binding
7. Aadsorption
8. Solution in poorly absorbable liquid

Source: Ref. 23

An example of a direct interaction between drugs affecting bioavailability is the


interaction between iron and tetracycline. This is a well-documented and clinically significant interaction which can result in a dramatic reduction in serum concentration of tetracycline (50).
The above potential sources of alteration in a drug's bioavailability must be kept in
mind when attempting to evaluate the relative performance of drug products on the
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-10

Bioavailability, Bioequivalence, and Drug Selection

basis of studies performed with healthy human volunteers. These studies are generally performed under tightly-controlled fasting conditions in the absence of other
drugs. In practice, however, drugs are seldom taken under such ideal conditions,
and the factors leading to changes in drug absorption must be taken into consideration.

8.1.3

METHODS OF ASSESSING BIOAVAILABILITY


Bioavailability testing is a means of predicting the clinical efficacy of a drug; the
estimation of the bioavailability of a drug in a given dosage form is direct evidence
of the efficiency with which a dosage form performs its intended therapeutic function.
The bioavailability of a drug substance formulated into a pharmaceutical product
is fundamental to the goals of dosage form design and essential for the clinical efficacy of the medication. Thus, bioavailability testing, which measures the rate and
extent of drug absorption, is a way to obtain evidence of the therapeutic utility of a
drug product. Bioavailability determinations are performed by drug manufacturers
to ensure that a given drug product will get the therapeutic agent to its site of
action in an adequate concentration. Bioavailability studies are also carried out to
compare the availability of a drug substance from different dosage forms or from
the same dosage form produced by different manufacturers.

In-vivo methods

One method for assessing the bioavailability of a drug product is through the demonstration of a clinically significant effect. However, such clinical studies are
complex, expensive, time-consuming and require a sensitive and quantitative measure of the desired response. Further, response is often quite variable, requiring a
large test population. Practical considerations, therefore, preclude the use of this
method except in initial stages of development while proving the efficacy of a new
chemical entity.
Quantification of pharmacologic effect is another possible way to assess a drug's
bioavailability. This method is based on the assumption that a given intensity of
response is associated with a particular drug concentration at the site of action;
e.g., variation of miotic response intensity can be directly related to the oral dose
of chlorpromazine. However, monitoring of pharmacologic data is often difficult,
precision and reproducibility are difficult to establish, and there are only a limited
number of pharmacologic effects (e.g. heart rate, body temperature, blood sugar
levels) that are applicable to this method.
Because of these limitations, alternative methods have been developed to predict
the therapeutic potential of a drug. The current method to assess the clinical performance of a drug involves measurement of the drug concentrations in the blood

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-11

Bioavailability, Bioequivalence, and Drug Selection

or urine. In such studies a single dose of the drug product is administered to a


panel of normal, healthy adult (18- to 35-year old) subjects. Blood and/or urine
samples are collected over a period of time following administration and are analyzed for drug content. Based on the blood concentration as a function of time
and/or urinary excretion profile, inferences are drawn regarding the rate and extent
of absorption of the drug. These studies are relatively easy to conduct and require
a limited number of subjects.
Blood level studies-

Blood level studies are the most common type of human bioavailability studies,
and are based on the assumption that there is a direct relationship between the concentration of drug in blood or plasma and the concentration of drug at the site of
action. By monitoring the concentration in the blood, it is thus possible to obtain
an indirect measure of drug response. Following the administration of a single
dose of a medication, blood samples are drawn at specific time intervals and analyzed for drug content. A profile is constructed showing the concentration of drug
in blood at the specific times the samples were taken . The key parameters to note
are:
1.

AUC

, The area under the plasma concentration-time curve, The AUC is proportional to the

total amount of drug reaching the systemic circulation, and thus characterizes the extent of
absorption.
2.

Cmax , The maximum drug concentration. The maximum concentration of drug in the plasma
is a function of both the rate and extent of absorption. Cmax will increase with an increase in
the dose, as well as with an increase in the absorption rate.

3.

Tmax , The time at which the Cmax occurs. The Tmax reflects the rate of drug absorption, and
decreases as the absorption rate increases.

Bioavailability (the rate and extent of drug absorption) is generally assessed by the
determination of these three parameters.
Since the AUC is representative of, and proportional to, the total amount of drug
absorbed into the circulation, it is used to quantitate the extent of drug absorption.
The calculation of AUC has been discussed in Chapter 4. A variety of pharmacokinetic methods have been suggested for the calculation of absorption rates (51-56).
For clinical purposes, it is generally sufficient to determine Cmax and Tmax. If all
other factors are constant, such as the extent of absorption and rate of elimination,
then Cmax is proportional to the rate of absorption and Tmax is inversely proportional to the absorption rate. Thus, the faster the absorption of a drug the higher
the maximum concentration will be and the less time it will take to reach the maximum concentration.
Urinary Excretion Data -

An alternative bioavailability study measures the cumulative amount of unchanged


drug excreted in the urine. These studies involve collection of urine samples and
the determination of the total quantity of drug excreted in the urine as a function of

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-12

Bioavailability, Bioequivalence, and Drug Selection

time. These studies are based on the premise that urinary excretion of the
unchanged drug is directly proportional to the plasma concentration of total drug.
Thus, the total quantity of drug excreted in the urine is a reflection of the quantity
of drug absorbed from the gastrointestinal tract. Consider the following example:
two products, A and B, each containing 100 mg of the same drug are administered
orally. A total of 80 mg of drug is recovered in the urine from Product A, but only
40 mg is recovered from Product B. This indicates that twice as much drug was
absorbed from Product A as from Product B. (The fact that neither product
resulted in excretion of the entire dose might be due to the existence of other routes
of elimination, e.g. metabolism).
This technique of studying bioavailability is most useful for those drugs that are
not extensively metabolized prior to urinary elimination. As a rule-of-thumb,
determination of bioavailability using urinary excretion data should be conducted
only if at least 20% of a dose is excreted unchanged in the urine after an IV dose
(56). Other conditions which must be met for this method to give valid results
include:
1.

the fraction of drug entering the bloodstream and being excreted intact by the kidneys must
remain constant.

2.

collection of the urine has to continue until all the drug has been completely excreted (five times
the half-life 1).

Urinary excretion data are primarily useful for assessing extent of drug absorption,
although the time course for the cumulative amount of drug excreted in the urine
can also be used to estimate the rate of absorption. In practice, these estimates are
subject to a high degree of variability, and are less reliable than those obtained
from plasma concentration-time profiles (57). Thus, urinary excretion of drug is
not recommended as a substitute for blood concentration data; rather, these studies
should be used in conjunction with blood level data for confirmatory purposes.
Single-dose versus
Multiple-Dose-

Most bioavailability evaluations are made on the basis of single-dose administration. The argument has been made that single doses are not representative of the
actual clinical situation, since in most instances, patients require repeated administration of a drug. When a drug is administered repeatedly at fixed intervals, with
the dosing frequency less than five half-lives, drug will accumulate in the body and
eventually reach a plateau, or a steady-state
At steady-state, the amount of drug eliminated from the body during one dosing
interval is equal to the available dose (rate in = rate out); therefore, the area under
the curve during a dosing interval at steady-state is equal to the total area under the
curve obtained when a single dose is administered. This AUC can therefore be

1.

Half life is defined as the length of time required to lose 50% of the drug in the body, assuming first order elimination.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-13

Bioavailability, Bioequivalence, and Drug Selection

used to assess the extent of absorption of the drug, as well as its absolute and relative bioavailability.
Multiple-dose administration has several advantages over single-dose bioavailability studies, as well as some limitations. These are summarized in Table 8-8 on
page 14 (54, 59).
TABLE 8-8 Multiple

dose vs. single dose studies in bioavailability studies

Advantages:
Eliminates the need to extrapolate the plasma concentration profiles to obtain the total AUC
after a single dose
Eliminates the need for a long wash-out period between doses
More closely reflects the actual clinical use of the drug
Allows blood levels to be measured at the same concentrations encountered therapeutically
Because blood levels tend to be higher than in the single-dose method, quantitative determination is easier and more reliable
Saturable pharmacokinetics, if present, can be more readily detected at steady-state
Limitations:
Requires more time to complete
More difficult and costly to conduct (requiring prolonged monitoring of subjects
Greater problems with compliance control
Greater exposure of subjects to the test drug, increasing the potential for adverse reactions

When a drug obeys linear, first-order kinetics, it is possible to estimate the results
that would be obtained during multiple dosing from single-dose studies. Projection is easily made with regard to the extent of absorption, using the AUC following a single dose. Results from bioequivalence studies indicate that conclusions on
the extent of absorption as assessed by the AUC can be made equally well on the
basis of a single or multiple dose study (60). Assessing the rate of absorption during multiple-dosing from single-dose studies has presented a greater problem.
Although a number of single-dose characteristics have been suggested as indicators of rate of absorption during multiple dosing (e.g. percent peak-trough fluctuation and percent peak-trough swing), results of bioequivalence studies indicate that
only the plateau time (the time during which the concentration exceeds 75% of the
maximum concentration, t 75% Cmax) and the residual concentration at the end of
the dose interval produce consistent results in assessing the rate of absorption in
single- and multiple-dose studies (54, 61).
In the case of drugs exhibiting nonlinear kinetics, establishing a linear relationship
between single- and multiple-dose bioavailability data has proven to be a difficult
task. Thus, it has been recommended that for drugs with either saturable elimination or a nonlinear first-pass effect, steady-state studies be carried out to assess
their bioavailability (62).
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-14

Bioavailability, Bioequivalence, and Drug Selection

8.1.4

STUDY DESIGN
Bioavailability studies involve the administration of the test dosage form to a panel
of subjects, after which blood and/or urine samples are collected and analyzed for
drug content. Based on the concentration profile of the drug, a judgement is made
regarding the rate and extent of absorption of the drug. Normally, the study is conducted in a group of healthy, male subjects who are of normal height and weight,
and range in age from 18 to 35 years (6). Questions have been raised regarding the
extent to which such a population reflects the performance of a given drug product
in a actual patient population. At first glance, it would seem that bioavailability
should be determined in patients actually suffering from the disease for which the
drug is intended, or in patients representative of the age and sex of subjects who
would be using the drug. However, there are several very good reasons for using
healthy volunteers rather than patients. In bioavailability studies, it is assumed
that there are no physiologic changes in the subjects during the course of the study.
If actual patients were used, this would not be a valid assumption, due to possible
changes in the disease state. Another potential problem with using patients is that
many patients take more than one drug. This could result in a drug-drug interaction which could influence the bioavailability of the test drug. In addition, diet and
fluid volume intake, both of which can influence a drug's bioavailability are more
difficult to control in a patient population than in a panel of healthy test subjects.
In general, it is more difficult with patients to have a standardized set of conditions
which are necessary for a dependable bioavailability study. However, it must be
recognized that factors that may affect a drug's performance in a patient population
may not be detected in a group of healthy subjects. Thus, it is best to conduct a
separate study in patients to determine if the disease, for which the drug is intended
to be used, alters the bioavailability of the drug.
Other important considerations in the methodology of a bioavailability study are
sample size, period of trial, and sampling. For statistical purposes, twelve subjects
are considered to be a minimum sample size. Otherwise there will not be enough
data to draw valid conclusions (63). The bioavailability testing period should be of
a sufficient length of time to ensure that drug absorption has been completed. This
length of time is at least three times the half-life of the drug; generally a period of
four to five times the half-life is used (63, 64). Blood samples should be taken
with sufficient frequency to permit an accurate determination of tmax, Cmax and
AUC.

8.1.5

IN-VITRO DISSOLUTION AND BIOAVAILABILITY


Pharmaceutical scientists have for many years been attempting to establish a correlation between some physicochemical property of a dosage form and the biological

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-15

Bioavailability, Bioequivalence, and Drug Selection

availability of the drug from that dosage form. The term commonly used to
describe this relationship is "in-vitro/in-vivo correlation" (65). Specifically, it is
felt that if such a correlation could be established, it would be possible to use
in-vitro data to predict a drug's in-vivo bioavailability. This would drastically
reduce, or in some cases, completely eliminate the need for bioavailability tests.
The desirability for this becomes clear when one considers the cost and time
involved in bioavailability studies as well as the safety issues involved in administering drugs to healthy subjects or patients. It would certainly be preferable to be
able to substitute a quick, inexpensive in-vitro test for in-vivo bioavailability studies. This would be possible if in-vitro tests could reliably and accurately predict
drug absorption and reflect the in-vivo performance of a drug in humans.
Disintegration Tests-

The early attempts to establish an indicator of drug bioavailability focused on disintegration as the most pertinent in-vitro parameter. The first official disintegration test appeared in the United States Pharmacopeia (USP) in 1950. However,
while it is true that a solid dosage form must disintegrate before significant dissolution and absorption can occur, meeting the disintegration test requirement only
insures that the dosage form (tablet) will break up into sufficiently small particles
in a specified length of time. It does not ensure that the rate of solution of the drug
is adequate to produce suitable blood levels of the active ingredient. Therefore,
while the test for tablet disintegration is very useful for quality control purposes in
manufacturing, it is a poor index of bioavailability.

Dissolution Tests-

Since a drug must go into solution before it can be absorbed, and since the rate at
which a drug dissolves from a dosage form often determines its rate and/or extent
of absorption, attention has been directed at the dissolution rate. It is currently
considered to be the most sensitive in-vitro parameter most likely to correlate with
bioavailability.

Official dissolution tests -

There are two official USP dissolution methods: Apparatus 1, (basket method),
and Apparatus 2 (paddle method). For details of these dissolution tests, the reader
is recommended to consult USPXXII/NFXVII (66).
Dissolution tests are an extremely valuable tool in ensuring the quality of a drug
product. Generally, product-to-product variations are due to formulation factors,
such as particle size differences, excessive amounts of lubricant and coatings.
These factors are reactive to dissolution testing. Thus, dissolution tests are very
effective in discriminating between and within batches of drug product(s). The
dissolution test, in addition, can exclude definitively any unacceptable product.

Limitations of
dissolution tests-

There are, however, problems with in-vitro dissolution testing which should be
noted - problems which make correlation with in- vivo availability difficult. The
first is related to instrument variance and the absence of a standard method. The
tests described in the USP are but a few of the large number of dissolution methods
proposed to predict bioavailability. Since the dissolution rate of a dosage form is
dependent on the methodology used in the dissolution test, changes in the appara-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-16

Bioavailability, Bioequivalence, and Drug Selection

tus, dissolution medium, etc., can dramatically modify the results. Table 8-9 on
page 17 lists some of the factors related to the dissolution testing device that can
affect the dissolution rate of the drug.
TABLE 8-9

Device factors affecting dissolution

1. Degree of agitation
2. Size and shape of container
3. Composition of dissolution medium
pH
ionic strength
viscosity
surface tension
4. Temperature of dissolution medium
5. Volume of dissolution medium
6. Evaporation
7. Hydrodynamics (flow pattern)
Source: Ref. 67

Another significant problem is related to the difference between the in-vitro and
in-vivo environments in which dissolution occurs. In-vitro studies are generally
carried out under controlled conditions in one, or perhaps two, standardized solvents. The in-vivo environment (the gastrointestinal tract), on the other hand, is a
continuously changing, complex environment. There are many variables which
can affect the dissolution rate of a drug in the gastrointestinal tract, including pH,
enzyme secretions, surface tension, motility, presence of other substances and
absorption surfaces (68). Thus, drugs frequently dissolve in the body at rates quite
different from those observed in an in-vitro test situation. Most of the official dissolution tests tend to be acceleration dissolution tests which bear limited or no
relationship with in-vivo dissolution.
Adding to the complexity of correlating dissolution with in-vivo absorption are
factors such as drug-drug interactions, age, food effects, health, genetic background, biorhythm and physical activity (32, 69). All these factors may have an
effect on the rate and extent of absorption of a drug. Thus, the in-vivo environment is far more complex, variable, and unpredictable than any in-vitro test environment, making in-vitro / in-vivo correlations very difficult. A simple dissolution
test in a standardized vehicle cannot reflect the in vivo absorption of a drug across a
population (70).
Parameters used-

Proper selection of the in-vitro and in-vivo parameters to be correlated is critical


in achieving a meaningful correlation. The in-vitro parameter should be selected
that has the greatest effect on the absorption characteristics of the drug (71). There
are several approaches to establishing a correlation between the dissolution of a

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-17

Bioavailability, Bioequivalence, and Drug Selection

drug in in- vitro and the bioavailability of a drug in-vivo. The in-vitro - in-vivo
correlative methods used most often are of the single-point type where the dissolution rate (expressed as the percent of drug dissolved in a given time, or the time
required for a given percent of the drug to dissolve) is correlated to a certain
parameter of the bioavailability. Examples of in-vivo parameters used include
Cmax, AUC, time to reach half-maximal plasma concentration, the average
plasma concentration after 0.5 or 1 hour, maximum urinary excretion rate, and
cumulative percent excreted in urine after a given time (71- 78). According to
Wagner, the best in-vitro variable to use is the time for 50 percent of the drug to
dissolve, and the best variable from in-vivo data to use is the time for 50 percent of
the drug to be absorbed (79).
Ideally, one would hope to find a linear relationship between some measurement of
the dissolution test and some measurement based on bioavailability studies.
Unfortunately, most attempts to accomplish this objective have failed.

8.1.6

IN-VITRO / IN-VIVO CORRELATION STUDIESThere have been many attempts to establish in-vitro / in-vivo correlations for a
large variety of drugs. Some of these studies have been summarized by Welling,
Banakar, and Abdou (71, 80-82).
While there are many published examples of satisfactory correlations between
absorption parameters and in-vitro dissolution tests, most studies have resulted in
poor, or moderate, in-vitro - in-vivo correlations, often involving agreement with
only one of the critical bioavailability parameters. Moreover, the positive correlations that have been found generally apply only to the specific formulation studied.
There have been instances where the dissolution rates or various formulations of
the same drug have been significantly different, yet little or no difference was
observed in their bioavailability parameters (83-85). There have also been cases
where a drug has failed to meet compendia dissolution standards but has demonstrated adequate bioavailability (86). Welling states: "To the writer's knowledge,
there have been no studies that have accurately correlated in- vitro and in-vivo data
to the point that the use of upper and lower limits for in-vitro dissolution parameters can be confidently used to predict in-vivo behavior and, therefore, to replace
in-vivo testing" (71).
Even if an in-vitro test could be designed that would accurately reflect the dissolution process in the gastrointestinal tract, dissolution is only one of many factors
that affect a drug's bioavailability. For example, saturable presystemic metabolism
may affect the extent of drug absorption, but this would not be predicted by an

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-18

Bioavailability, Bioequivalence, and Drug Selection

in-vitro test. Dissolution studies also would not predict poor bioavailability due to
instability in gastric fluid or complexation with another drug or food component.
Thus, the ultimate evaluation a drug product's performance under the conditions
expected in clinical therapy must be an in-vivo test; a dissolution test is unlikely to
entirely replace bioavailability testing (70, 87, 88). In-vitro methods are important
in the development and optimization of dosage forms while in-vivo tests are essential in obtaining information on the behavior of medication in living organisms.
One cannot be substituted for the other (69).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-19

Bioavailability, Bioequivalence, and Drug Selection

8.2 Bioequivalence
Definitions

With the phenomenal increase in the availability of generic drugs in recent years,
the issues of bioavailability and bioequivalence have received increasing attention.
In order for a drug product to be interchangeable with the pioneer (innovator or
brand name) product, it must be both pharmaceutically equivalent and bioequivalent to it. According to the FDA, "pharmaceutical equivalents" are drug products
that contain identical active ingredients and are identical in strength or concentration, dosage form, and route of administration (89). However, pharmaceutical
equivalents do not necessarily contain the same inactive ingredients; various manufacturers' dosage forms may differ in color, flavor, shape, and excipients. The
terms "pharmaceutical equivalents" and "chemical equivalents" are often used
interchangeably.
"Bioequivalence" is a comparison of the bioavailability of two or more drug products. Thus, two products or formulations containing the same active ingredient are
bioequivalent if their rates and extents of absorption are the same. When a new
formulation of an existing drug is developed, its bioavailability is generally evaluated relative to the standard formulation of the originator. Indeed, a bioequivalence trial against the standard formulation is the key feature of an Abbreviated
New Drug Application (ANDA) submitted to the Food and Drug Administration
by a manufacturer who wishes to produce a generic drug. For a generic drug to be
considered bioequivalent to a pioneer product, there must be no statistical differences (as specified in the accepted criteria) between their plasma concentration-time profiles. Because two products rarely exhibit absolutely identical
profiles, some degree of difference must be considered acceptable, as will be discussed later.
Since the concentration of a drug in blood is used as an assessment of its clinical
performance, inherent in the demonstration that two preparations containing
equivalent amounts of the same drug produce similar concentrations of the drug
entity in blood is the assumption that they will elicit equivalent drug responses.
Thus, two products that are deemed to be bioequivalent are also assumed to be
therapeutically equivalent, and therefore interchangeable. This principle is fundamental to the concept of bioequivalence and is the basic premise on which it is
founded.
In general, the FDA considers two products to be "therapeutic equivalents" if they
each meet the following criteria (90):
1.

they are pharmaceutical equivalents,

2.

they are bioequivalent (demonstrated either by a bioavailability measurement or an in vitro standard),

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-20

Bioavailability, Bioequivalence, and Drug Selection

Background

3.

they are in compliance with compendial standards for strength, quality, purity and identity,

4.

they are adequately labelled, and

5.

they have been manufactured in compliance with Good Manufacturing Practices as established
by the FDA.

The first intimations of bioequivalence problems with multi-source drug products


were given by early investigations of the availability of vitamins, aspirin, tetracycline, and tolbutamide (91-97). In 1974, after an extensive review of the bioavailability of drugs, Koch-Weser concluded that " . . . among drugs thus far tested
bioinequivalence of different drug products has been far more common than
bioequivalence" (98). Of particular note were the studies involving digoxin; the
findings of these investigations sparked the discussion about bioequivalence
assessment that still continues today. Significant differences were seen in the bioavailability of digoxin not only between products supplied by different companies,
but also between lots obtained from the same manufacturer (99). Because of the
narrow therapeutic range for this drug, and because the drug is utilized in the treatment of cardiac patients, these findings generated a great deal of concern.
Similar reports of bioinequivalence and therapeutic inequivalence appeared for
other drugs as well, including phenytoin, phenylbutazone, chloramphenicol, tolbutamide and thyroid (6). The clinical significance of these reported differences in
bioavailability relates to the therapeutic index of the drug, the dose of the drug and
the nature of the disease. In 1973 the Ad Hoc Committee on Drug Product Selection of the American Pharmaceutical Association published a list of drugs with a
potential for therapeutic inequivalence based on reported evidence of bioinequivalence (100). The drugs fall in three categories: "high," "moderate," or "low risk"
based on the clinical implications (Table 8-10 on page 22).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-21

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-10 Drugs

with various risk potential for inequivalence

High Risk Potential

Moderate Risk Potential

Low or Negligible Risk


Potential

aminophylline

amphetamines

acetaminophen

aspirin (when used in high dose


levels)

(sustained-release)

codeine

ampicillin

ferrous sulfate

bishydroxycoumarin

chloramphenicol

hydrochlorothiazide

digoxin

chlorpromazine

ephedrine

dipheylhydantoin (phenytoin)

digitoxin

isoniazid

para-aminosalicylic acid

erythromycin

meprobamate

prednisolone

griseofulvin

penicillin VK

prednisone

oxytetracycline

sulfisoxazole

quinidine

penicillin G (buffered)

warfarin

pentobarbital
phenylbutazone
phenacetin
potassium chloride (solid dosage
forms)
salicylamide
secobarbital
sulfadiazine
tetracycline
tolbutamide

The concern about the bioinequivalence of some drugs led to the establishment in
1974 of the Drug Bioequivalence Study Panel of the Office of Technology Assessment (OTA). The objective was to ensure that drug products of the same physical
and chemical composition would produce similar therapeutic effects. Among the
11 recommendations of the Panel was the conclusion that not all chemical equivalents were interchangeable, but the goal of interchangeability was achievable for
most oral drug products (101). The Report recommended that a system should be
organized as rapidly as possible to generate an official list of interchangeable drug
products. The OTA Report, as well as the growing awareness within the scientific
and regulatory communities of bioavailability problems with marketed drug products, focused the attention of the FDA on bioequivalence and bioavailability problems and issues.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-22

Bioavailability, Bioequivalence, and Drug Selection

8.2.1

BIOEQUIVALENCE REGULATIONS
In 1977, the FDA implemented a series of bioavailability and bioequivalence regulations which formed the basis of subsequent discussion, if not controversy, of
therapeutic equivalency of drug products (102). The regulations are divided into
two separate regulations; Subpart B - Procedures for Determining the Bioavailability of Drug Products and Subpart C - Bioequivalence Requirements. While
Table 11 summarizes the key provisions of the bioavailability regulations, those
for bioequivalence requirements are summarized in Table 8-11 on page 23.
TABLE 8-11 Key

provisions for bioavailabilty regulations

1. Defines bioavailability in terms of both the rate and extent of drug absorption.
2. Describes procedures for determining the bioavailability of drug products.
3. Sets forth requirements for submission of in vivo bioavailability data.
4. Sets forth criteria for waiver of human in vivo bioavailability studies.
5. Provides general guidelines for the conduct of in vivo bioavailability studies.
6. Imposes a requirement for filing an Investigational New Drug Application.
Source: Ref. 103

Criteria for establishing


a bioequivalence
requirement -

The 1977 Bioequivalence regulations set forth the following criteria and evidence
supporting the establishment of a bioequivalence requirement for a given drug
product:
1.

Evidence from well-controlled clinical trials or controlled observations in patients that such
products do not give comparable therapeutic effects.

2.

Evidence from well-controlled bioequivalence studies that such products are not bioequivalent
drug products.

3.

Evidence that the drug products exhibit a narrow therapeutic ratio, (e.g., there is less than a
two-fold difference in the median lethal dose (LD50) and median effective dose (ED50) value
or have less than a two-fold difference in the minimum toxic concentration and minimum effective concentrations in the blood), and safe and effective use of the drug product requires careful
dosage titration and patient monitoring.

4.

Competent medical determination that a lack of bioequivalence would have a serious adverse
effect in the treatment or prevention of a serious disease or condition.

5.

Physicochemical evidence of any of the following:


a.

The active drug ingredient has a low solubility in water--e.g., less than 5 mg/ml.

The dissolution rate of one or more such products is slow--e.g., less than 50 percent in
thirty minutes when tested with a general method specified by an official compendium or the
FDA.
b.

The particle size and/or surface area of the active drug ingredient is critical in determining
bioavailability.
c.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-23

Bioavailability, Bioequivalence, and Drug Selection

Polymorphs, solvates, complexes, and such, exist that could contribute to poor dissolution
and may affect absorption.
d.
e.

There is a high excipient/active drug ratio present in the drug product--e.g., greater than 5

to 1.
The presence of specific inactive ingredients (e.g. hydrophilic or hydrophobic excipients)
that either may be required for absorption of the active drug or may interfere with such absorption.
f.

6.

Pharmacokinetic evidence of any of the following:


The drug is absorbed in large part in a particular segment of the gastrointestinal tract or is
absorbed from a localized site.
a.

Poor absorption of the drug, even when it is administered as a solution--e.g., less than 50
percent compared to an intravenous dose.
b.
c.

The drug undergoes first-pass metabolism in the intestinal wall or liver.

The drug is rapidly metabolized or excreted, requiring rapid dissolution and absorption for
effectiveness.
d.

The drug is unstable in specific portions of the gastrointestinal tract, requiring special
coatings and formulations--e.g., enteric coatings, buffers, film coatings--to ensure adequate
absorption.
e.

The drug follows nonlinear kinetics in or near the therapeutic range, and the rate and
extent of absorption are both important to bioequivalence.
f.

Types of Bioequivalence
Requirements

In the event that a drug meets one or more of the above six criteria, a bioequivalence requirement is established. The requirement could be either an in-vivo or an
in-vitro investigation, as specified by the FDA. The types of bioequivalence
requirements include the following:
1.

An in-vivo test in humans.

2.

An in-vivo test in animals that has been correlated with human in- vivo data.

3.

An in-vivo test in animals that has not been correlated with human in- vivo data.

4.

An in-vitro bioequivalence standard, i.e., an in-vitro test that has been correlated with human
in-vivo bioavailability data.

5.

A currently available in-vitro test (usually a dissolution rate test) that has not been correlated
with human in-vivo bioavailability data.

The regulations state that in-vivo testing in humans would generally be required if
there is well-documented evidence that pharmaceutical equivalents intended to be
used interchangeably meet one of the first three criteria used to establish a
bioequivalence requirement:
1.

The drug products do not give comparable therapeutic effects.

2.

The drug products are not bioequivalent.

3.

The drug products exhibit a narrow therapeutic ratio (as described above), and safe and effective use of the product requires careful dosage titration and patient monitoring.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-24

Bioavailability, Bioequivalence, and Drug Selection

Criteria for waiver of


evidence of in-vivo
bioavailability -

Although a human in-vivo test is considered to be preferable to other approaches


for the most accurate determination of bioequivalence, there is a provision in the
1977 regulations for waiver of an in-vivo bioequivalence study under certain circumstances. For some drug products, the in-vivo bioavailability of the drug may
be self-evident or unimportant to the achievement of the product's intended purposes. The FDA will waive the requirement for submission of in-vivo evidence of
bioavailability or bioequivalence if the drug product meets one of the following
criteria:
1.

The drug product is a solution intended solely for intravenous administration, and contains the
active drug ingredient in the same solvent and concentration as an intravenous solution that is
the subject of an approved full New Drug Application (NDA).

2.

The drug product is a topically applied preparation intended for local therapeutic effect.

3.

The drug product is an oral dosage form that is not intended to be absorbed, e.g., an antacid.

4.

The drug product is administered by inhalation and contains the active drug ingredient in the
same dosage form as a drug product that is the subject of an approved full NDA.

5.

The drug product is an oral solution, elixir, syrup, tincture or other similar soluble form, that
contains an active drug ingredient in the same concentration as a drug product that is the subject
of an approved full NDA and contains no inactive ingredient that is known to significantly
affect absorption of the active drug ingredient.

6.

The drug product is a solid oral dosage form (other than enteric-coated or controlled-release)
that has been determined to be effective for at least one indication in a Drug Efficacy Study
Implementation (DESI) notice and is not included in the FDA list of drugs for which in vivo
bioequivalence testing is required.

7.

The drug product is a parenteral drug product that is determined to be effective for at least one
indication in a DESI notice and shown to be identical in both active and inactive ingredients formulation, with a drug product that is currently approved in an NDA. (Excluded from the waiver
provision are parenteral suspensions and sodium phenytoin powder for injection.)

According to the regulations, the bioavailability of certain drug products may be


demonstrated by evidence obtained in-vitro in lieu of in-vivo data. Thus, the FDA
also permits waiver of the in-vivo requirements if a drug product meets one of the
following criteria:
1.

The drug product is one for which only an in-vitro bioequivalence requirement has been
approved by the FDA.

2.

The drug product is in the same dosage form, but in a different strength, and is proportionally
similar in its active and inactive ingredients to another drug product made by the same manufacturer and the following conditions are met:
a.

the bioavailability of this other product has been demonstrated

b.

both drug products meet an appropriate in-vitro test approved by the FDA

the applicant submits evidence showing that both drug products are proportionally similar
in their active an inactive ingredients.
c.
3.

The drug product is shown to meet an in-vitro test that assures bioavailability, i.e., an in-vitro
test that has been correlated with in-vivo data.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-25

Bioavailability, Bioequivalence, and Drug Selection

4.

5.

The drug product is a reformulated product that is identical, except for color, flavor, or preservative, to another drug product made by the same manufacturer, and both of the following conditions are met:
a.

the bioavailability of the other product has been demonstrated.

b.

both drug products meet an appropriate in vitro test approved by the FDA.

The drug product contains the same active ingredient and is in the same strength and dosage
form as a drug product that is the subject of an approved full NDA or Abbreviated New Drug
Application (ANDA) and both drug products meet an appropriate in-vitro test that has been
approved by the FDA.

Although the above list of criteria for waiver of an in-vivo bioavailability study is
quite lengthy, currently virtually all new tablet or capsule formulations from which
measurable amounts of drug or metabolites are absorbed into the systemic circulation require a human bioequivalence study for approval (104).
TABLE 8-12 Key

Provisions for bioequivalence requirements

1. Defines procedures for establishing a bioequivalence requirement.


2. Sets forth criteria to establish a bioequivalence requirement.
3. Describes types of bioequivalence requirements.
4. Sets forth requirement for in-vitro batch testing and certification.
5. Describes requirements for marketing a drug product subject to a bioequivalence requirement.
6. Sets forth requirements for in-vivo testing of a drug product not meeting an in-vitro bioequivalence standard.
Source: Ref. 103

8.2.2

STUDY DESIGN
A single-dose bioequivalency study is generally performed in normal, healthy,
adult volunteers. The subject population should be selected carefully, so that product formulations, and not intersubject variations, will be the only significant determinants of bioequivalence (105). A minimum of 12 subjects is recommended,
although 18 to 24 subjects are used to increase the data base for statistical analysis.
The test and the reference products are usually administered to the subjects in the
fasting state (overnight fast for at least 10 hours, plus 2 to 4 hours after administration of the dose), unless some other approach is more appropriate for valid scientific reasons. These subjects should not take any other medication for one week
prior to the study or during the study. The bioavailability is determined by the collection of either blood samples or urine samples over a period of time and measurement of the concentration of drug present in the samples.
Generally, a crossover study design is used. Using this method, both the test and
the reference products are compared in each subject, so that inter-subject variables,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-26

Bioavailability, Bioequivalence, and Drug Selection

such as age, weight, differences in metabolism, etc., are minimized. Each subject
thus acts as his own control. Also, with this design, subjects' daily variations are
distributed equally among all dosage forms or drug products being tested.
The subjects are randomly selected for each group and the sequence of drug
administration is randomly assigned. The administration of each product is followed by a sufficiently long period of time to ensure complete elimination of the
drug (washout period) before the next administration. The washout period should
be a minimum of 10 half-lives of the administered drug (106). A waiting period of
one week between administration is usually an adequate washout period of most
drugs.
With a drug requiring a washout period of one week, a typical randomized twoway crossover bioequivalency study is shown in Table 8-13 on page 27.
TABLE 8-13 Two

way cross over design

Treatment
Groupa

Week 1

I
II
a

A
B

Week 2
B
A

10 subjects per group

Assuming that the in-vivo performances of the two formulations are to be compared by examining their blood level profiles, one must be certain that an adequate
number of blood samples are taken. Blood samples should be drawn with sufficient frequency to provide an accurate characterization of the drug concentration-time profile from which tmax, Cmax and AUC can be determined. Typically,
a total of 10 to 15 sampling times might be required (107). Moreover, all samples
should be taken at the same time for both the test and the reference product to permit proper statistical analysis.
Additional features which contribute to good study design include:
1.

All drug samples obtained for the test and reference preparations should be analyzed by the
same method.

2.

Identical test conditions must be used for the two groups of subjects. For example, the types of
foods, fluid intake, physical activity, and posture should all be rigidly controlled in the study.

3.

The physical characteristics of the subjects (such as age, height, weight, and health) should be
standardized.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-27

Bioavailability, Bioequivalence, and Drug Selection

Several important questions have been raised specifically regarding the design of
the bioequivalence tests. One of these deals with the selection of the appropriate
reference standard, since this is a critical component of a protocol (6, 108). Normally, the reference product is that available from the innovator company holding
the New Drug Application. However, in cases where there may be some question
as to the bioavailability of such a product, the study may utilize a solution of the
drug instead of or in addition to the marketed product. The use of a solution can,
of course, result in some difficulty in interpretation of the data: a solid dosage
form, when compared to a solution, will usually exhibit a lower Cmax and a longer
tmax. The clinical significance of these differences may be difficult to assess.
In some instances, the FDA must designate a specific product as the reference
standard from among two or more possible products; e.g., Proventil tablets, 4 mg
(Schering), not Ventolin tablets 4 mg (Allen and Hanburys), is the reference
product in bioequivalence studies of albuterol sulfate conventional tablets (108).
Advantages of Multipledose vs. single dose
studies:

Another important question is whether the bioequivalence trial should compare


single doses of the formulations or if it should compare "steady-state" conditions
reached after multiple dosing. It would seem that multiple dosing would be the
logical choice for drugs intended for long-term use since this would give a more
realistic comparison in view of the way in which the drug is normally administered. Other advantages of conducting a multiple-dose study over a single-dose
study include (54, 59):
1.

Multiple-dosing eliminates the long washout periods required between single-dose administrations. The switch-over from one formulation to the other can take place in steady state.

2.

Single-dose studies may pose problems of sufficiently long sampling periods in order to get reliable estimates of terminal half-life, which is needed for correct calculation of the total AUC.

3.

Multiple-dose studies yield higher concentrations of drug in the blood, making accurate measurement easier. In addition, since drug concentrations need to be measured only over a single
dosing interval at steady state, the need to measure lower concentrations during a disposition
phase is avoided.

4.

Multiple-dosing studies can be conducted in patients, rather than healthy volunteers, allowing
the use of higher doses.

5.

Usually, smaller intersubject variability is observed in steady-state studies, which may permit
the use of fewer subjects.

6.

Nonlinear pharmacokinetics, if present, can be more readily detected at steady-state following


multiple-dosing.

Thus, for some drug products, multiple-dose bioequivalence studies are appropriate and should be performed. In fact, according to one of the conclusions of the
Bio- International '92 conference on the bioequivalence of highly variable drugs, a
multiple-dose study is required in the case of compounds exhibiting nonlinear
pharmacokinetics (110). The circumstances under which a multiple-dose study
may be required are summarized in the regulations (109):

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-28

Bioavailability, Bioequivalence, and Drug Selection

1.

When there is a difference in the rate of absorption but not in the extent of absorption.

2.

When there is excessive variability in bioavailability from subject to subject.

3.

When the concentration of the active moiety in the blood resulting from a single dose is too low
for accurate determination.

4.

When the drug product is a controlled-release dosage form.

On the other hand, multiple-dose bioequivalence studies are undesirable in some


respects. Healthy subjects should not be dosed with any drug for an extended
period of time (59). Multiple-dose studies are also generally more difficult to
carry out, especially with regard to ensuring subject compliance with dosing and
dietary restrictions. Therefore, most bioequivalence studies are conducted as single-dose studies. Multiple-dose studies should be performed only when a single-dose study is not a reliable indicator of bioavailability (111).

8.2.3

ASSESSMENT OF BIOEQUIVALENCE
In order for different formulations of the same drug substance to be considered
bioequivalent, they must be equivalent with respect to the rate and extent of drug
absorption. Thus, the two predominant issues involved in the assessment of
bioequivalence are: the pharmacokinetic parameters that best characterize the rate
and extent of absorption and, the most appropriate method of statistical analysis of
the data.

Pharmacokinetic criteria

With regard to the choice of the appropriate pharmacokinetic characteristics,


Westlake suggests comparisons of the formulations should be made with respect to
only those parameter(s) of the blood level profile that possess some meaningful
relation to the therapeutic effect of the drug (107). Since the AUC is directly proportional to the amount of drug absorbed, this pharmacokinetic parameter is most
commonly used to characterize the extent of absorption, both in single- and multiple- dose studies.
The choice of an appropriate pharmacokinetic characteristic for the rate of absorption is still being discussed with considerable controversy (112, 113). Although a
broad array of methods exists for calculating absorption rates (e.g. moment analysis, deconvolution procedures and curve-fitting), the most commonly used parameters are peak concentration (Cmax) and time to peak concentration (tmax).
Although these parameters have been observed to have significant variances and
may be difficult to determine accurately, they remain the parameters generally
requested as rate characteristic by most regulatory authorities for immediate-release products (112).

Statistical criteria

After a bioequivalence study is conducted and the appropriate parameters are


determined, the pharmacokinetic data must be examined according to a set of predetermined criteria to confirm or refute the bioequivalency of the test and refer-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-29

Bioavailability, Bioequivalence, and Drug Selection

ence formulations. That is, one must determine whether the test and reference
products differ within a predefined level of statistical significance. Since the statistical outcome of a bioequivalence study is the primary basis of the decision for
or against therapeutic equivalence of two products, it is critically important that the
experimental data be analyzed by an appropriate statistical test.
In the early 1970s, bioequivalence was usually determined only on the basis of
mean data. Mean AUC and Cmax values for the generic product had to be within
+20% of those of the reference (innovator) product (108). Although the 20% value
was somewhat arbitrary, it was felt that for most drugs, a 20% change in the dose
would not result in significant differences in the clinical response to drugs (114).
A relatively common misconception is that current regulatory standards still allow
this difference of 20% in the means of the pharmacokinetic variables (Cmax and
AUC) of the test and reference formulations. The FDA's statistical criteria for
approval of generic drugs now requires the application of confidence limits to the
mean data, using an analysis known as the two one-sided tests procedure (115).
This change came about as a result of the conclusion of the FDA Bioequivalence
Task Force in 1986 that the use of a 90% confidence interval based on the two
one-sided t-tests approach was the best available method for evaluating bioequivalence (111).
Westlake was the first to suggest the use of confidence intervals as a means of testing for bioequivalence (116). Recognizing that no two products will result in identical blood-level profiles, and that there will be differences in mean values between
products, Westlake pointed out that the critical issue was to determine how large
those differences could be before doubts as to therapeutic equivalence arose (107,
117). A test formulation was considered to be bioequivalent to a reference formuCp max test
AUC test
lation if 0.8 < ------------------< 1.2 and 0.8 < -------------------< 1.2 . (119). By this proceAUC ref
Cp max ref
dure, if test and reference products were not bioequivalent (i.e. means differed by
more than 20%), there was a 5% chance of concluding that they are bioequivalent.
The current FDA guidelines are that two formulations whose rate and extent of
absorption differ by -20%/+25% or less are generally considered bioequivalent
(90). In order to verify that the -20%/+25% rule is satisfied, the two one-sided statistical tests are carried out: one test verifies that the bioavailability of the test
product is not too low and the other to show that it is not too high. The current
practice is to carry out the two one-sided tests at the 0.05 level of significance.
Computationally, the two one-sided tests are carried out by computing a 90% confidence interval. For approval of an ANDA, a generic manufacturer must show
that the 90% confidence interval for the ratio of the mean response (usually AUC

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-30

Bioavailability, Bioequivalence, and Drug Selection

and Cmax) of its product to that of the innovator is within the limits of 0.8 to 1.25.
Since these tests are carried out at the 0.05 level of significance, there is no more
than a 5% chance that they will be approved as equivalent if they differ by as much
or more than is allowed by the equivalence criteria (-20%/+25%).
Since this test requires that the 90% confidence interval of the difference between
the means be within a range of -20%/+25%, it is more stringent than simply requiring the comparison of the test and reference products' AUC and Cmax to be within
the 80 to 125% range. If the mean response of the generic product in the study
population is near 20% below or 25% above the innovator mean, one or both of the
confidence limits will fall outside the acceptable range and the product will fail the
bioequivalence test. Thus, the confidence interval requirement ensures that the
difference in mean values for AUC and Cmax will actually be less than -20%/
+25%. It should be pointed out that the standards vary among drugs and drug
classes. For example, antipsychotic agents may fall within a 30% variation and
antiarrhythmic agents may be allowed a 25% variation (122).
The actual differences between brand and generic products observed in bioequivalence studies have been reported to be small. The FDA has stated that for
post-1962 drugs approved over a two-year period under the Waxman-Hatch bill
(1984), the mean bioavailability difference between the generic and pioneer products has been about 3.5% (120). In addition, 80% of the generic drugs approved
by the FDA between 1984 and 1986 differed from the innovator products by an
observed difference of only +5%. Such differences are small when compared to
other variables of drug therapy and would not be expected to produce clinically
observable differences in patient response.

8.2.4

CONTROVERSIES AND CONCERNS IN BIOEQUIVALENCE


The design, performance and evaluation of bioequivalence studies have received a
great deal of attention over the past decade from academia, the pharmaceutical
industry and regulatory agencies. A number of concerns and questions have been
raised about the conduct of bioequivalence studies as well as the guidelines and
criteria used to determine bioequivalence (112). Many of these concerns were triggered by the passage of the Drug Price Competition and Patent Term Restoration
Act (The Waxman-Hatch Amendments) by Congress in 1984. This Act provided
for an expedited approval by the FDA of generic drugs, thereby expanding the
potential generic market for prescription generic drugs (121). Shortly after the
passage of this Act, numerous published reports appeared in the scientific literature questioning the FDA's ability to ensure that generic drugs were equivalent to
the brand name drugs they were copying. Most of the concerns of the scientific
community centered around adequate standards for evaluation of bioequivalence

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-31

Bioavailability, Bioequivalence, and Drug Selection

and correlation between bioequivalence and therapeutic equivalence. Some of the


issues and concerns that were raised are summarized in Table 8-14 on page 32 (8,
13).
TABLE 8-14 Issues

and Concerns regarding bioequivalence

Correct analysis of drugs in biological fluids


Appropriate choice of pharmacokinetic parameters to assess bioequivalence
Generalizing results obtained in healthy volunteers to patients
Problems involved in extrapolating from single-dose studies to steady-state
Importance of evaluating active metabolites
Inadequate statistical criteria to evaluate bioequivalency
Bioequivalence does not always ensure therapeutic equivalence
Lack of clear guidelines for evaluation of bioequivalence

At the center of the controversy were the methods and criteria used by the FDA to
determine bioequivalence. Assessment of bioequivalence was done on the basis of
mean data: mean AUC and Cmax values for the generic product had to be within
+20% of those of the innovator product for approval. A statistical test was
employed to assess the power of the test to detect a 20% mean difference in treatments. For drugs that could not meet the statistical criteria because of inherent
variability, another rule was used, the so-called "75/75" rule: that in at least 75%
of the subjects, the test formulation must fall within the range of 75% to 125% of
the reference standard to be considered equivalent (122). It was felt by many that
these rules permitted too much variability in the bioavailability of test drugs and
could result in therapeutic failure or increased risk of side effects (4, 15, 123).
Statistically, the power approach and the 75/75 rule were shown to have poor performance characteristics and bioequivalence evaluation based on these methods
was discontinued by the FDA in 1986. In their place, the Agency currently
employs the two one-sided tests procedure, as previously discussed.
Although the decision of bioequivalence is now made in a more statistically valid
way and the associated concerns have diminished somewhat, some important
questions and controversies in bioequivalence remain. These are primarily centered around study design, the criteria used to establish or refute equivalence, and
the assumption that products that are bioequivalent are therapeutically equivalent.
One criticism of bioequivalence testing is that it is almost always done in a panel
of young, healthy male volunteers rather than in the target population for which the
drug is intended. Clearly, the performance of a drug product in a 20-year-old male
will not be the same as in an 85-year-old woman. Serious concerns have been
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-32

Bioavailability, Bioequivalence, and Drug Selection

raised that different results would be observed in elderly patients, in women, in


patients with diseases of the gastrointestinal tract, and in patients with diminished
renal or hepatic function. However, although factors such as age and disease state
might affect the actual observed concentrations of drug, the products being compared should be affected in a similar fashion, and one can still be compared to the
other. If two products show an equivalent level in healthy volunteers, their levels
should be elevated to the same extent in patients with impaired hepatic function.
Thus, they can still be compared to each other. Healthy male volunteers are generally used in bioequivalence studies to assure a homogeneous study population and
to permit focus on formulation factors that might affect bioavailability. In addition, healthy subjects are more likely to remain stable during the study. The condition of actual patients might change due to the disease resulting in greater
variability in the data. The FDA does recognize the possibility that some conditions could cause two products that are bioequivalent in healthy subjects to be bioinequivalent in certain patients and is prepared to modify its guidelines if
necessary.
A study design-related area of concern is average versus individual bioavailability.
Current procedures assess equivalence in terms of average bioavailabilities, and do
not address within-subject equivalence. In recent years, there has been increased
interest expressed in the variability of response, particularly variability within an
individual. This has given rise to the most recent controversy in bioequivalence
assessment, namely whether average bioequivalence is adequate to allow interchangeability of drugs in an individual (112). Anderson and Hauck believe that a
different, more stringent, notion of bioequivalence, referred to as individual
bioequivalence, is needed to provide assurance that an individual patient can be
switched from one formulation to another (124).
The second major area of controversy has focused on the criteria used to determine
bioequivalence. Implicit in the FDA guidelines is the assumption that a -20%/
+25% change in mean serum concentration of drugs can be safely tolerated. However, there is little documentation demonstrating whether 20% variation in bioavailabilities does or does not affect the safety and efficacy of drugs. There are
certain critical therapeutic categories (Table 8-15 on page 34) in which minor fluctuations in blood levels may have a substantial impact on therapeutic outcome or
toxicity (125, 126). In view of this, some scientists believe that the FDA should be
more stringent, requiring the mean values for AUC to be within 10% rather than
20%/25%. The Bioequivalence Task Force, in its 1988 report, concluded that for
certain drugs or drug classes, there is clinical evidence that may indicate a need for
tighter limits than the then-generally applied +20% rule (111). The Task Force
recommended that the Agency consider using as an "additional nonstatistical criterion" a mean difference in AUC of +10%; however, this additional criterion would
not be essential to ensuring drug bioequivalence.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-33

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-15

Critical Therapeutic Catagories of Drugs

Category

Example

Cardiovascular drugs

digoxin

Anticonvulsants

phenytoin

Bronchodilating agents

theophylline

Oral anticoagulants

warfarin

In general, the choice of the appropriate bioequivalence range should be done on


clinical grounds; for a drug with a narrow therapeutic range, more stringent limits
should be considered. On the other hand, the current requirements for Cmax for
some drugs may be too stringent, considering the difficulty in accurately estimating this value. For example, it has been suggested that the acceptable bioequivalence range for Cmax for fast-releasing nifedipine formulations should be 70% to
130%, rather than the usual 80% to 125%. In light of this, many, including the
Pharmaceutical Research and Manufacturers of America (formerly the Pharmaceutical Manufacturers Association [PMA]), feel that the FDA should repudiate its
-20%/+25% rule and develop drug-by- drug bioequivalence criteria (127).
A third source of controversy in bioequivalence is the very foundation on which
the whole concept of bioequivalence is based: the central assumption is that if two
products are shown to be bioequivalent by currently accepted standards, then they
are also therapeutically equivalent, and thus interchangeable. A number of critics
have challenged this "bioequivalence = therapeutic equivalence" equation, pointing out that this relationship has not been conclusively established for most drugs
(9, 13, 16, 128). These terms are, in fact, not interchangeable; bioequivalence
means that two products have basically superimposable blood level curves (within
specified limits) while therapeutic equivalence means the products produce similar
effects. There may be situations where two products have similar blood concentrations, yet if the drug has a narrow therapeutic range, they may have significantly
different therapeutic effects. On the other hand, there may be products which have
widely varying blood level profiles, but exhibit very little difference in their clinical effect. This might be the case for drugs with a wide therapeutic range. In addition, the therapeutic efficacy of some drugs is not necessarily related to their blood
levels, e.g., some psychoactive drugs, where the end point of drug effects is psychological and behavioral response (129).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-34

Bioavailability, Bioequivalence, and Drug Selection

Williams suggests several ways that the integrity of a bioequivalence study as a


prediction of therapeutic equivalence could be assessed (104). One way involves
the performance of specific clinical studies to confirm that products shown to be
bioequivalent in healthy subjects would be bioequivalent in the patient population
as well. A second way suggested is through post-marketing surveillance of therapeutic response produced by different formulations of the same drug under actual
conditions of use. A third method is based on anecdotal reports. Williams points
out that none of these methods have been systematically employed to confirm current bioequivalence methodology.
Thus, a number of problems remain in the bioequivalence process which should be
addressed. FDA scientists themselves have readily acknowledged the existence of
shortcomings in the bioequivalence testing program. However, a great deal of
progress has been made in this area in the last twenty years. The improved design
of the studies, the interpretation of the data, the increased scientific rigor of the
acceptance criteria, as well as the more rigorous auditing and inspection program
have made bioequivalence data an appropriate and valid means of approving
generic drug products.

8.2.5

GENERIC DRUGS AND PRODUCT SELECTION


Generic drug utilization has increased dramatically in the last 20 years. In 1975,
approximately 9% of all prescription drugs dispensed were generic versions (130).
This percentage rose to 20% in 1984, and 40% in 1991. It has been variously estimated that the generic share of all new prescriptions will be 46% to 65% in 1995
(131-133).
This rise of generics has not gone altogether smoothly, however; the popularity of
generic drugs took a sharp downturn in 1989 when scandal rocked the generic drug
industry. This involved illegal and unethical acts by some generic drug companies
-- payoffs to FDA employees and fraudulent drug-approval test -- aimed at getting
drugs approved ahead of other firms (134-138).
Although these events did shake the confidence of pharmacists, physicians and the
public in the quality of generic drugs and cast a shadow over generics generally,
these concerns were relatively short-lived. Numerous surveys conducted one to
two years after the scandal unfolded indicated that confidence in generic drugs had
been regained and that the generic industry was in better shape with pharmacists
than it had been before the scandal occurred (139-146). Given the seriousness of
the events, the speed with which generics came back was impressive. This was
due in part to the FDA's reaction to the scandal: a multilevel reorganization of its
generic drug operations and a comprehensive inspection of the leading manufac-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-35

Bioavailability, Bioequivalence, and Drug Selection

turers of generic drugs (134, 140, 147, 148). It was felt that this stringent FDA
review of generics proved the overall integrity of the companies that emerged with
a clean bill of health. After a sharp drop in the use of generic drugs in 1989, they
began to rise nearly as quickly as they fell, and by mid-1990, sales of generics
were approaching their previous record high (141).
This trend in generic drug utilization is expected to continue its upward spiral, with
newly generic drugs coming to market at an increasing rate. There are several factors that have contributed to this period of considerable growth in the generic drug
industry. One major factor was the passage of the Drug Price Competition and
Patent Term Restoration Act (Waxman-Hatch Act) in 1984. This act, by eliminating the requirement for clinical safety and efficacy testing for generics of drugs
introduced after 1962, greatly expedited the entry of generic drugs into the marketplace. The purpose of this act was to facilitate generic competition and thereby
reduce health care costs. This act significantly expanded the number of drugs eligible to be manufactured as generics. Another factor fueling the surge of generic
products is the abundance of brand name drugs whose patents began expiring in
1986. Between 1991 and 1994, patents expired on brand-name drugs whose combined annual sales totaled $10 billion (141). These include Procardia, Ceclor,
Tagamet, Cardizem, Feldene, Naprosyn, and Xanax. All told, more than
100 drugs worth upwards of $25 billion in sales will have come off patent by the
year 2000 (149). Table 8-16 on page 37 lists some recent and impending patent
expirations (150, 151). As a result of these patent expirations on popular drugs,
there has been an explosion of new generic drug applications.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-36

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-16 Recent

and pending patent expirations

Brand Name
Procardia
Tenormin
Ceclor
Cardizem
Feldene
Naprosyn
Xanax
Tagamet
Seldane
Micronase
Capoten
Zantac
Trental
Noroxin

Generic Name
Nifedipine
Atenolol
Cefaclor
Diltiazem
Piroxicam
Naproxen
Alprazolam
Cimetidine
Terfenadine
Glyburide
Captopril
Ranitidine
Pentoxifylline
Norfloxacin

Patent Expiration Date*


1991
1991
1992
1992
1992
1993
1993
1994
1994
1994
1995
1995
1997
1998

*Extentions may be granted


Perhaps the major factor promoting generic drug utilization is the increased attention to containing health-care costs. Pushed by a drive for lower-cost medication
by federal and state governments, private insurers, corporate benefit managers,
regulatory agencies and consumer groups, generic drug usage is at a peak. Additional impetus could come from health care reform, wherein generic drugs are
viewed as a key to controlling pharmaceutical costs. Managed care programs are
expected to cover more than 70% of all outpatient prescriptions by the end of the
decade, with an accompanying greater demand for generic products (152). Thus
the demand for generic drugs will continue to rise, in a climate that favors health
care reform, lower- cost medications and broad-based prescription benefits (153).
With the increasing availability of generic drugs, pharmacists are called upon more
and more often to select a patient's drug product from a myriad of multisource
products. The pharmacist's role in product selection has increased dramatically in
the past decade and the proper selection of multisource drug products has become
a major professional responsibility of pharmacists. Although most pharmacists do
not, realistically, evaluate the bioequivalence of two products from blood level
data, professional judgement does need to be exercised; and this requires an understanding and application of the biopharmaceutical principles discussed.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-37

Bioavailability, Bioequivalence, and Drug Selection

8.2.6

THE ORANGE BOOK


One of the factors that led to the widespread repeal of the state anti-substitution
laws in the 1970's was an effort by the states to contain drug costs and the establishment of maximum allowable costs (MACs) for reimbursement of drugs under
Medicaid. By allowing the pharmacist to select the manufacturer of a drug, the
less- expensive generic version could be dispensed. However, before the pharmacist could knowledgeably select a generic drug, he had to know which generics
were bioequivalent to the innovator product and thus, interchangeable. (There was
substantial evidence at this time that not all pharmaceutically equivalent products
were bioequivalent). To answer this need, the states began preparing either positive or negative formularies, often turning to the FDA for assistance in this undertaking.
In response to the many requests for assistance from the states in developing their
formularies, the FDA Commissioner notified state officials of FDA's intent to provide a list of all prescription drug products that have been approved as being safe
and effective, along with therapeutic equivalence determinations for multisource
prescription products. This list, entitled Approved Drug Products with Therapeutic Equivalence Evaluations, more commonly known as "The Orange Book" was
first published in 1980 and is now in its 14th edition. It is published annually and
updated monthly. The Orange book is generally considered to be the most reliable
guide for determining which drug products are therapeutically equivalent.
The Prescription Drug Products List contains:
1.

all the drug products approved by the FDA as being safe and effective under the Federal Food,
Drug and Cosmetic Act, and

2.

2.the therapeutic equivalence evaluations for all approved multisource prescription drug products (those pharmaceutical equivalents available from more than one manufacturer).

Currently, multisource products comprise almost 80% of the approximately 10,000


drugs on the Prescription Drug Product List. The therapeutic evaluation for these
products have been prepared to serve as information and advice to state health
agencies, pharmacists and prescribers to promote knowledgeable drug product
selection and to foster containment of health costs.

8.2.7

THERAPEUTIC EQUIVALENCE
Drug products are considered to be therapeutic equivalents if they are pharmaceutical equivalents and if they can be expected to have the same clinical effect when
administered to patients as specified in the labeling (90). In general, the FDA

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-38

Bioavailability, Bioequivalence, and Drug Selection

evaluates as therapeutically equivalent those drug products that satisfy the following general criteria:
1.

They are approved as safe and effective.

2.

They are pharmaceutical equivalents; i.e. they


contain identical amounts of the same active ingredient in the same dosage form and route
of administration, and
a.

b.

meet compendial and other applicable standards for quality, purity, strength and identity.

3.

They are bioequivalent. Bioequivalence may be established by either an in-vivo or in-vitro test,
depending on the drug. If the drug presents a known or potential bioequivalence problem then
an appropriate standard must be met which demonstrates a comparable rate and extent of
absorption.

4.

They are adequately labeled.

5.

They are manufactured in compliance with Current Good Manufacturing Practice regulations.

The FDA believes that drug products meeting the above criteria are therapeutically
equivalent and can be substituted with the full expectation that the substituted
product will produce the same therapeutic effect as the prescribed product.

8.2.8

THERAPEUTIC EQUIVALENCE EVALUATION CODESThe FDA uses a two-letter coding system for multisource products. The first letter
in the code allows users to determine whether a particular product has been evaluated therapeutically equivalent to other pharmaceutically equivalent products. The
second letter in the code provides additional information about the basis of FDA's
evaluation. The various categories are summarized in Table 8-17 on page 40.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-39

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-17 Therapuetic

equivalency codes

"A" Drug Products

"B" Drug Products

Drug products the FDA considers to be therapeutically equivalent; i.e. drug

Drug products the FDA does not consider to be therapeutically equivalent; i.e.

products for which:

drug products for which actual or potential bioequivalence problems have not

1.

2.

There are no actual or potential bioequivalence problems. These are

been resolved by adequate evidence of bioequivalence. Often the problem is

designated as:

with specific dosage forms rather than with the active ingredient. These products

AA

Products in conventional dosage forms

are classified as "B" for one of three reasons:

AN

Solutions and powders for


aerosolization

AO

Injectable oil solutions

AP

Injectable aqueous solutions

AT

Topical products

1.

2.

The active ingredients or dosage forms have documented or potential


bioequivalence problems, and no adequate studies demonstrating
bioequivalence have been submitted.
The quality standards are inadequate or the FDA has insufficient
basis to determine therapeutic equivalence.

3.

Actual or potential bioequivalence problems have been resolved via


adequate in vivo and/or in vitro tests. These are designated as AB.

The drug product is under regulatory review.


These products are designated as:
BC

Controlled-release tablets, capsules and injectables

BD

Active ingredients and dosage forms with documented


bioequivalence problems

BE

Delayed-release oral dosage forms (e.g. enteric-coated


products)

BN

Products in aerosol-nebulizer drug delivery systems

BP

Active ingredients and dosage forms with potential


bioequivalence problems

BR

Suppositories or enemas that deliver drugs for systemic


absorption

BS

Products having drug standard deficiencies

BT

Topical products with bioequivalence issues

BX

Insufficient data to determine therapeutic equivalence

B*

Drug products requiring further FDA investigation and


review to determine equivalence

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-40

Bioavailability, Bioequivalence, and Drug Selection

There are two basic categories into which multisource drugs have been placed, "A"
or "B". Drug products rated "A" are products that the FDA considers to be therapeutically equivalent to the pharmaceutically equivalent original product. These
fall into one of two classes:
1.

There are no known or suspected bioequivalence problems.

2.

Actual or potential bioequivalence problems have been resolved with adequate in vivo and/or in
vitro evidence supporting bioequivalence.

Category "B" consists of drug products that the FDA does not at this time consider
to be therapeutically equivalent to the pharmaceutically equivalent reference product. Certain types of products are rated B by virtue of their specialized dosage
forms. For example, controlled-release dosage forms are rated BC, unless
bioequivalence data have been submitted as evidence of equivalence. In this case,
the product would be coded AB.
The fact that a product is in the "B" category does not mean it should not be dispensed; it simply means that a B rated product should not be substituted for a pharmaceutically equivalent product. For example, glyburide is marketed as
Micronase and DiaBeta by two different manufacturers. Both these products
are clinically effective, but because bioequivalence between the two has not been
studied, they are B rated and are not interchangeable.
To avoid possible significant variations among generic drugs as a result of comparison to different reference drugs, the FDA began designating a single reference
listed drug against which all generic versions must be shown to be bioequivalent.
The reference listed drug is identified by the symbol "+" in the Prescription Drug
Product List. This symbol was used for the first time in the 1993 edition of the
Orange Book.
Limitations and
exclusions-

Although the Orange Book is a very valuable reference for pharmacists performing drug product selection, it has certain limitations, which must be recognized. It
was not intended to serve as a single comprehensive reference on all multisource
drugs. Many prescription drug products are not listed in the Orange Book, making
evaluation of their therapeutic equivalence difficult, if not impossible. Exclusion
of a drug from the Orange Book means that the FDA has not evaluated its safety,
efficacy and quality. Table 18 lists the classes of products excluded from the
Orange Book. Because the equivalence of these excluded products is unknown,
interchanging of these products should be avoided.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-41

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-18 Drug

Products excluded from the Orange Book

1.

Drugs marketed before the passage of the Federal Food, Drug, and Cosmetic Act of 1938. These are not
included because the FDA has not reviewed these drugs for safety and efficacy and does not have the necessary
information to make therapeutic equivalence evaluations.
Examples: digoxin, morphine, codeine, thyroid, levothyroxine, phenobarbital and nitroglycerin

2.

Drugs for which the FDA has no NDA or ANDA on file.


Examples: Anusol-HC, Naldecon (and their generic counterparts)

3.

Drugs still undergoing Drug Efficacy Study Implementation (DESI) review. These are drugs that were
marketed between 1938 and 1962 on the basis of safety, but not efficacy. Although most of these drugs have
been reviewed and are listed in the Orange Book, there are still a number of these pre-1962 drugs which have
not yet been classified as "effective" under the DESI program, and are not listed.
Examples: nitroglycerin controlled-release capsules, pentaerythritol tetranitrate, isocarboxazid,
hydrocortisone-iodochlorhydroxyquin cream
In addition, nitroglycerin transdermal patches are still undergoing efficacy studies, and are not listed in the
Orange Book.

Another limitation of the Orange Book that all pharmacists should be aware of is
that the drug listings contain the names of only the companies that actually hold an
approved NDA or ANDA; they may not be the same as the actual manufacturer or
distributor. It is fairly common practice for a drug to be manufactured pursuant to
an NDA or an ANDA but distributed under license agreement by another company. In this instance, the distributor would not be listed in the Orange Book.
Since pharmacists are, understandably, generally unaware of the name of the actual
holder of the NDA or ANDA, it is often difficult for them to determine the therapeutic equivalence of a particular multisource product if it is not listed in the
Orange Book. For example, there are over thirty manufacturers and distributors
marketing approved, therapeutically equivalent versions of furosemide 40 mg tablets (154). However, only twelve of these companies are actually listed in the
Orange book, since these are the actual holders of an NDA or ANDA. Therefore,
the pharmacist would have to verify the therapeutic equivalence evaluation of the
non-listed products by obtaining the information from the manufacturer, packager,
or supplier.
Legal status and
pharmacists'
responsibility-

The Orange Book per se has no legal status. The FDA stresses that it is a source of
information and advice on drug product selection, but it does not mandate the drug
products which may be dispensed nor the products that should be avoided. Thus,
the Orange Book does not carry the weight of regulation or law, and the FDA
assumes no liability for drug products selected on the basis of its equivalence evaluation.
The Orange Book points out that "FDA evaluation of therapeutic equivalence in no
way relieves practitioners of their professional responsibilities in prescribing and
dispensing such products with due care." There are circumstances where pharma-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-42

Bioavailability, Bioequivalence, and Drug Selection

cists will have to exercise professional care and sound judgement in selecting a
drug product for a particular patient. Although two products may be rated as being
therapeutically equivalent in the Orange Book, they may not be equally suitable
for a particular patient. Drugs that share the "A" code may still vary in ways that
could affect patient acceptance. They may differ in shape, color, taste, scoring,
configuration, packaging, preservatives, expiration time, and in some instances,
labeling. If products with such differences are substituted for each other, there is
potential for patient confusion or decreased patient acceptance. For example, a
patient may be sensitive to an inert ingredient in one product that another product
does not contain. Or, a patient may become confused if the color or shape of a
product varies from that to which he has become accustomed. A patient may reject
the administration of a substituted product because of differences in taste or
appearance. When such characteristics of a specific product are important in the
treatment of a particular patient, the pharmacist should select a product with these
considerations in mind as well as bioequivalence.
Despite its limitations and shortcomings, the Orange Book is a very useful guide
for rational product selection. Pharmacists can utilize the information presented
there, in combination with sound professional judgment, to make decisions on
behalf of their patients regarding the choice of the most appropriate drug product.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-43

Bioavailability, Bioequivalence, and Drug Selection

8.3 Drug Product Selection


Multisource drug product selection has become a very important component of
contemporary pharmacy practice. The National Prescription Audit (NPA) has, for
some years now, been chronicling the heightened role played by pharmacists in the
selection of which brand (or generic version) of a multiple-source drug will be dispensed to the patient. From 1983 through 1993, the pharmacist's role in selecting
brand or generic products for dispensing has increased dramatically, as shown in
Table 19. In the first half of 1993, pharmacists controlled 41% of dispensing decisions, as compared to 16% in 1983. It is evident that the substitution trend is
strong and is continuing to gain ground. This expansion of pharmacy's province in
brand choice decisions is the result of several factors: economic pressures for
lower prescription costs, repeal of anti- substitution laws and increased acceptance
of generics by patients, physicians and pharmacists. Perhaps the most significant
factor in escalating the overall level of pharmacists' brand choice decisions has
been the expiration of the patents of high- volume pioneer brands, as previously
discussed. This has resulted in significant expansion in the potential for pharmacist choice.
TABLE 8-19 Pharmacists

Brand Selection

Year

Percent of all new prescriptions involving


pharmacists brand choicea

1983
1984
1985
1986
1987
1988
1989
1990
1991
1992
Jan.-June 1993

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

16
18
20
23
25
27
30
32
34
38
41

8-44

Bioavailability, Bioequivalence, and Drug Selection

8.3.1

CONSIDERATIONS IN SELECTING A MANUFACTURER


The selection of a pharmaceutical manufacturer of a multisource product has
become an important professional responsibility for pharmacists. This responsibility has become an especially critical part of a pharmacists role in light of the
increasing number of generic products available and in light of some of the problems that have occurred in the generic drug industry (the "generic drug scandal" of
1989). The pharmacist is entrusted by the public to select manufacturers that offer
the best quality at the best price.

So how does the pharmacist select the manufacturer of a multisource


drug product? What
factors should be considered?

Thoughtful selection of a multisource drug product is not an easy task, and


requires a consideration of not only the drug product itself, but also the manufacturer, and in some cases, the patient. Several options are open to the pharmacist
performing drug product selection: to select a product solely on the basis of economics, to select a product on the basis of the reputation of the manufacturer, or to
make a decision based on product bioequivalence and quality and on the basis of
the product's conformity with official compendial standards and with those established by the FDA. The first option, while offering a financial advantage, does not
provide assurance of therapeutic efficacy. The second option, although subjective,
is easily applied and does offer a degree of security to the pharmacist. The third
option is the most challenging to the pharmacists, requiring the application of principles of biopharmaceutics and pharmacokinetics in arriving at a decision. Ideally,
the pharmacist should take into consideration all the above options when selecting
a drug product for a patient.
When pharmacists were asked which factors are most important to them in selecting a manufacturer of a generic product, the primary criteria indicated were the
reputation and quality of the company (159-162). Bioequivalence to the
brand-name product was also ranked as being an important factor in product selection. However, the most frequently used sources for assessing bioequivalence
were manufacturer reputations (based previous experience) and product literature
provided by the distributing company. Company-sponsored material must be carefully evaluated. Unfortunately, promotional literature does not generally contain
sufficient data to permit rational analysis of whether or not products are bioequivalent (163). Also, relying on personal methods of information gathering for assessing bioequivalence is not very reliable. Interestingly, only 23% of pharmacists
reported using the Orange Book in assessing bioequivalence (161). Selection of
drug products should be based on sound scientific and clinical grounds. Developments in the science of pharmacokinetics and the related area of bioavailability
have given pharmacists the tools necessary to make sound choices among multisource products. In response to the profession's need for information and advice
on how to select appropriate drug products from multiple sources, the American
Pharmaceutical Association formed a Bioequivalency Working Group to establish
guidelines for product selection (Table 8-20 on page 47) (164). This Group made

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-45

Bioavailability, Bioequivalence, and Drug Selection

recommendations of factors that pharmacists should consider when selecting drug


products to be dispensed to their patients. If pharmacists consider the factors indicated as part of the professional judgement process when making drug product
selections, it is likely that the best interest of the patients will be served.
The appropriate selection of a generic drug product involves much more than just
cost considerations or reliance on state and federal laws and regulations. It
requires a knowledge of the drug entity and its physical and chemical properties,
the condition to be treated, and its significance, and the history and attitude of the
manufacturer. One of the criteria often used to evaluate a manufacturer's record is
the number and type of recalls of that company's products. Product selection may
also require taking into consideration the patient, the disease, previous drug therapy, and duration of therapy before a decision is made. Gagnon presented a
step-by-step analysis procedure that pharmacists can use in evaluating multisource
suppliers of a pharmaceutical product (Table 8-21 on page 49) (165). Using this
procedure, each manufacturer is rated in each area listed, thus enabling the pharmacist to make the most rational choice.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-46

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-20 Guidelines

for product selection

DISPENSING DECISIONS
? State Rules and Regulations. Pharmacists should be cognizant of legal requirements that address the issue of drug product selection. Many states have positive or negative
formularies to provide guidance in drug product selection.
? Bioequivalency Information/Orange Book Ratings. Only products with proven bioequivalency should be selected to be dispensed in lieu of the innovator product.
Products that are listed in the FDA's Approved Drug Products and Therapeutic Equivalence Evaluations (the Orange Book) as "A" rated should be selected when such products
are available. For pre-1938 drugs, the selection should be based on data obtained from the literature, because bioequivalency testing is not required by the FDA for these
drug products.
? Dosage Form. The type of dosage form should be considered whenever one drug product is selected from among multisource drug products. This is especially true with
extended or delayed release medications.
? Previous Drug Use. Two questions should be considered regarding previous drug product usage. First, is the prescribed drug a continuation of already successful therapy?
If it is, the impact of any change in source of the medication should be considered. The pharmacist should also know which product the patient was using previously, including
any medications in the hospital if the patient was recently discharged. Second, was the original product dispensed a generic product? If so, preference should be given to
continuing to dispense the same generic product from the same source.
? Patient Status. The pharmacist should consider how well controlled the patient is and how susceptible that patient might be to small changes in drug absorption. If a
patient has labile control or has experienced great difficulty in achieving control, the pharmacist should continue therapy with a product from a single source throughout therapy.
? Diseases. The seriousness of the disease and its potential impact on the patient may influence the pharmacist's willingness to change products.
? Drug Class or Category. Drugs with narrow therapeutic ranges and with known clinically significant bioavailability problems should be substituted with care and/or after
discussion with the prescriber.
? Cost. The cost of the product , while an important consideration, should be a secondary consideration in selecting among products judged by the pharmacist to be
bioequivalent.
? Patient Opinion. An informed patient, cooperating with a physician and pharmacist in his or her drug therapy, is an important element in ensuring the best possible
therapeutic outcomes. The pharmacist should take into account the patient's need when selecting from multisource drug products and inform the patient of any potential
consequences associated with alternate product selections.

PURCHASE DECISIONS
? Current State Laws and Regulations. Some states have positive or negative formulary systems that place regulatory restrictions on the products considered therapeutically
equivalent. The state formulary may not always be in agreement with classifications listed in the FDA's Orange Book. Therefore, pharmacists should be familiar with both.
? Bioequivalency Information/Orange Book. Products shown to be bioequivalent through reference to the Orange Book or other reliable source of bioequivalency
information are preferred. Purchase decisions for drugs marketed prior to 1938 should be based on data obtained from the literature or the manufacturer, because bioequivalency
testing may not be required by the FDA for these drug products.
? Drug Category. Greater attention should be given to purchasing strategies for drug products used for serious or life-threatening diseases and in situations where therapeutic
activity of the product is confined to a narrow range of biologic fluid concentration.
? Availability. A continuous supply from the same manufacturer is essential even in the event that the distributor has changed to ensure that refills of prescriptions will
contain the same product as originally dispensed. However, in those instances when the manufacturer of a generic drug product has to be changed, care should be exercised
to ensure that the new drug product is equivalent to the formerly stocked drug product.
? Supplier's Reputation. The reputation of the manufacturer in terms of its ability to adhere to good manufacturing practices (GMP) that ensure that each dosage form is
manufactured correctly and in a consistent manner is an important consideration. When purchasing a product from a distributor rather than directly from the manufacturer,
the procedure used by that supplier in selecting manufacturers for multisource products is also an important consideration. Establishment Inspection Reports and recall
reports are available from FDA through a Freedom of Information (FOI) request. These are valuable tools in this decision.
? Cost.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-47

Bioavailability, Bioequivalence, and Drug Selection

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-48

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-21 Evaluation

of Multi-source Suppliers

Factors and Cues


Product Information
Size(s) available
Dosage form(s) available
Bioequivalence data results using Orange Book
Existence of identification codes on solid dosage forms
Average number of months between product receipt and expiration date
Results of cost-effectiveness information from manufacturer
Complete product literature provided from manufacturer
Strength(s) available
State/federal formulary rules, e.g., MAC limits
Economics
Price(s)
Deals and other discounts
Terms of sale
Clear and equitable pricing policy
Large sizes available at discount prices
Product Quality
NDA/ANDA on file at FDA
Pharmaceutical elegance of products, e.g., broken tablets, powder in bottles
Less than 3 year FDA on-site inspection
Results of on-site FDA inspection
Company willing to allow pharmacist to inspect plant
Results of quality control analysis
Company willing to supply samples for testing
Product acceptance by physicians
Product acceptance by patients
Service Quality
Returns policy
Rapid resolution of complaints
Company product availability record
Liability protection policy
Terms of unconditional guarantee
Company commitment to education of practitioners
Availability of company representative
Existence of 24-hour emergency customer service telephone number
Product availability through wholesalers
Ease of placing orders
Company customer information center, including an 800 number
Company Reputation
Number of recalls in last 3 years
Severity of recalls in last 3 years
Who initiated recalls (FDA or company)
Company has a recall strategy
Other regulatory actions against company
Company has wide product line
FDA quality assurance profile
Company has crisis communication strategy

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-49

Bioavailability, Bioequivalence, and Drug Selection

Pharmacists have the responsibility of correctly selecting and dispensing multisource products that will have the greatest likelihood of achieving a positive therapeutic outcome in a cost-effective manner. The more information pharmacists
have about a product and its manufacturer, the more likely they will be to make the
most appropriate choice. Price cannot be the single factor in selecting a product. It
is also clear, as Joseph Oddis stated, "Rational drug product selection entails far
more than simply consulting the FDA's Orange Book or looking at the price catalogue" (166).

8.3.2

SPECIAL CASES
While in most situations selection of drug products that are therapeutically equivalent can be done without undue complications, there are some circumstances
where problems could occur. Depending on the drug, its formulation, the disease
being treated, and the condition of the patient, generic substitution may not be
advisable. Some of these special situations require extra attention and handling by
the pharmacist.
There are a number of drugs that could present problems when interchanged.
Drugs that are poorly water soluble may have inherent problems with rate and
extent of dissolution, resulting in poor or variable bioavailability. Drugs that are
potent and thus present in very low amounts in a dosage form could present problems due to formulation factors. Some dosage forms may have inherent bioavailability problems, such as controlled-release products. And drugs which are
considered "critical" also need special consideration. "Critical" drugs have been
defined as drugs with a narrow therapeutic range, where a change in plasma concentration might result in adverse clinical outcome; drugs that are considered primarily for control of a disease rather than for alleviation of temporary symptoms;
and drugs that have inherent or historical bioavailability or bioequivalence problems (8, 19). Seven classes of drugs have been identified that have demonstrated
bioequivalence problems or, because of the nature of the product, have the potential for creating therapeutic problems if product interchange is permitted (Table 822 on page 51) (167-168).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-50

Bioavailability, Bioequivalence, and Drug Selection

TABLE 8-22 Catagories

of drugs with demonstrated bioequivalence problems


Digitalis glycosides
- digoxin
Warfarin anticoagulants
Theophylline products
Thyroid preparations (including levothyroxine)
Conjugated and esterified estrogens
Antiarrhythmic agents
- quinidine salts
- procainamide
Anticonvulsants
- phenytoin
- carbamazepine
- primidone

There have been numerous reports of drugs implicated in therapeutic problems due
to bioinequivalence difficulties. In addition to those in the categories given in
Table 8-22 on page 51, these include furosemide, propranolol, diazepam, prednisone, nitrofurantoin, and amitriptyline (20, 126, 167, 169-180). Although the
documentation implicating these drugs in therapeutic failures due to bioavailability problems is primarily anecdotal in nature (and thus disregarded by the FDA),
the performance of these products should still be closely observed and monitored,
and care should be taken when selecting drugs from these categories.
In addition to "critical" drugs, critical patients and critical diseases have also been
identified when special care should be taken in performing product selection (8,
166). Critical patients are the very old and the very young, those suffering from
multiple diseases who are managed with multiple drugs, and those who live alone,
making observation of adverse drug effects unlikely. Critical diseases are generally chronic in nature and difficult to stabilize, where drug-disease interactions can
present major problems (e.g. congestive heart failure, asthma, diabetes, cardiac
disorders, and psychoses). In all the above special "critical" circumstances, there
is a high risk of therapeutic problems, and product selection requires extra attention and precautions. In fact, product substitution and interchange in these cases is
generally discouraged. Once a product (brand or generic) has been selected for a
course of therapy, the pharmacist should not change to a different product if it can
be avoided. If interchange is performed, it should be done only with the utmost
care, and the patient should be monitored for any adverse outcomes.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-51

Bioavailability, Bioequivalence, and Drug Selection

Pharmacist's
professional
responsibility-

Drug product selection has been and continues to be a primary and challenging professional responsibility of pharmacists. It is one where the pharmacist
must exercise professional care and sound judgement to make decisions on behalf
of the patient to maximize safety and efficacy, while minimizing cost. Pharmacists
have a professional obligation to patients to take whatever steps are necessary to
assure themselves that the medicines they are dispensing are safe and effective.
Although some of this activity is currently constrained by bureaucratic and regulatory restrictions that often discourage, or entirely prevent, individual professional
evaluation and initiative, with a greater appreciation and understanding of the scientific, clinical, and regulatory issues that form the basis of the process, pharmacists can make decisions that result in better patient care. Pharmacists must take
steps to ensure the quality and integrity of the drug products dispensed to their
patients. To accomplish this, pharmacists must look to pharmaceutical manufacturers to supply them with a quality product they can trust. Thus, the manufacturer
of a multisource product must be carefully selected to ensure that the products they
supply are of proper quality. If necessary, pharmacists should conduct independent
research into the reputation and integrity of the manufacturer, or, if products are
purchased through a buying group, should make sure that established policies and
guidelines are in place to review multisource products. When considering purchasing drug products, the pharmacist should request the manufacturer to provide
certain documentation and information, and should then evaluate this information
(see Table 23).
TABLE 8-23

Considerations when evaluating a Multi-Source vendor

1.Willingness to supply requested information


2.Bioavailability and bioequivalence data
3.Dissolution testing results
4.FDA bioequivalence rating
5.The actual manufacturer of the product, if not the supplier
6.FDA inspection reports
7.History of the manufacturer's recall record
8.Willingness of the manufacturer to permit on-site visitations
9.Evaluate economic considerations such as price, shipping, terms, discounts, insurance, return policies, and packagng.

And finally, pharmacists can counsel the patients on the importance of


using the same drug product throughout a course of therapy, even though they
might go to a different pharmacy. To further emphasize this, it has been suggested
that the initial prescription and subsequent refills of a drug product considered
questionable for interchange should contain auxiliary labeling that stresses the
importance of continuing to use that product (167).
Drug product selection is an important professional responsibility, but it is
not an easy task. It requires the pharmacist to use his/her current knowledge, and
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-52

Bioavailability, Bioequivalence, and Drug Selection

all the currently available information in order to arrive at and render a decision
regarding the most appropriate product to use for a specific patient.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-53

Bioavailability, Bioequivalence, and Drug Selection

8.4 Summary
With the dramatic increase in the availability and utilization of generic drug products in recent years, pharmacists are being faced with an ever-increasing array of
multisource products. Appropriate selection of a product from the plethora of
products on the market is not always an easy task; the quality of the drug product
must be considered, as well as the cost. The principles of biopharmaceutics indicate that the formulation and method of manufacture of a drug product can have a
marked effect on the bioavailability of the active ingredient. Thus, generic equivalents may not necessarily be therapeutically equivalent. Guidelines and criteria
have been established by the FDA to help judge whether one product can be substituted for another with assurance of equivalent therapeutic effect.
For pharmacists to provide informed product selection, it is essential that
they be knowledgeable about, and familiar with, these guidelines and criteria. This
requires an understanding of bioavailability, bioequivalence, and how they are
determined. The pharmacist can serve a major role in ensuring that only high quality products are dispensed, and in this way help reduce health care costs without
compromising quality of care.
Acknowledgment

The author gratefully acknowledges the assistance of Umesh V. Banakar in the


preparation of this manuscript.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-54

Bioavailability, Bioequivalence, and Drug Selection

8.4.1

QUESTIONS
1.

The term bioavailability refers to the


a.

dissolution of a drug in the gastrointestinal tract.

b.

amount of drug destroyed in the liver by first-pass metabolism.

c.

distribution of drug to the body tissues over time.

relationship between the physical and chemical properties of a drug and its systemic
absorption.
d.
e.

2.

measurement of the rate and amount of drug that reaches the systemic circulation.

The bioavailability of various drug products can be evaluated by comparing their plasma concentration-time curves. The three most important parameters of comparison that can be
obtained directly from the curves are
a.

biologic half-life (t1/2), absorption rate constant, area under the curve (AUC).

b.

time of peak concentration (tmax), absorption rate constant, elimination rate constant.

c.

maximum drug concentration (Cmax), time of peak concentration (tmax), duration of action.

d.

area under the curve (AUC), time of peak concentration (tmax), maximum drug concentration

(Cmax).
e.

3.

rate of elimination, area under the curve (AUC), rate of absorption.

Two products are bioequivalent if they


a.

contain the same amount of the same active ingredient.

b.

have equal areas under the curve after the administration of the same dose.

c.

have the same value for Cmax after administration of the same dose.

have equivalent rates and extents of absorption of the drug after administration of equal
doses.
d.
e.

4.

5.

are pharmaceutically equivalent.

If an oral capsule formulation of drug A produces a plasma concentration- time curve having
the same area under the curve (AUC) as that produced by an equivalent dose of drug A given
intravenously, it can generally be concluded that:
a.

there is no advantage to the IV route.

b.

the absolute bioavailability of the capsule formulation is equal to 1.

c.

the capsule formulation is essentially completely absorbed.

d.

the drug is very rapidly absorbed.

e.

b and c are correct.

5.Which of the following is NOT a criterion for therapeutic equivalence of two products,
according to the FDA?
a.

They must be pharmaceutical equivalents.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-55

Bioavailability, Bioequivalence, and Drug Selection

6.

7.

8.

b.

All ingredients - active and inactive - must be the same.

c.

They have been manufactured in compliance with Good Manufacturing Practices.

d.

They are bioequivalent.

e.

They are approved as safe and effective by the FDA.

A test oral formulation has the same area under the plasma concentration- time curve as the reference formulation. This means that the two formulations
a.

are bioequivalent by definition.

b.

deliver the same total amount of drug to the body but are not necessarily bioequivalent.

c.

are bioequivalent if they both meet USP dissolution standards.

d.

deliver the same total amount of drug to the body and are, therefore, bioequivalent.

e.

have the same rate of absorption.

In-vitro dissolution rate studies on drug products are useful in bioavailability evaluations only if
they can be correlated with
a.

in-vivo bioavailability studies in humans.

b.

the chemical stability of the drug.

c.

USP disintegration requirements.

d.

in-vivo studies in at least three species of animals.

e.

the therapeutic response observed in patients.

Which of the following statements regarding bioequivalence is TRUE?


If the mean AUC and Cmax values for a generic product are within + 20% of those of the reference product, the two products are bioequivalent.
a.

b.

If we can be 90% certain that the mean values of AUC and Cmax for two products are within

80% to 125% of each other, then the two products are considered bioequivalent.

Bioequivalence studies are generally conducted in a panel of patients consisting of the target population for which the drug is intended.
c.

Bioequivalence studies are generally conducted as multiple-dose studies utilizing the


cross-over design.
d.

If two products are shown to be bioequivalent, we can always say with certainty that they
will be therapeutically equivalent.
e.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-56

Bioavailability, Bioequivalence, and Drug Selection

9.

9.Which of the following statements about the FDA Orange Book is TRUE?
Drugs that are excluded from the Orange Book are not safe and effective and should not be
dispensed.
a.

b.

It contains therapeutic equivalence evaluations for all the drugs approved by the FDA.

c.

Products placed in the "B" category should not be dispensed.

The Orange Book is an official compendium, and pharmacists can legally only dispense
those products listed as bioequivalent.
d.

The drug listings contain the names of only the companies that actually hold an approved
NDA or ANDA for a drug.
e.

10.

8.4.2

10.Growth in the utilization of generic drug products can be attributed to


a.

passage of the 1984 Waxman-Hatch Act.

b.

expiration of patents of many popular brand products.

c.

pressures to reduce health care costs.

d.

the growth of managed health care organizations.

e.

all of the above.

ANSWERS TO QUESTIONS
1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-57

Bioavailability, Bioequivalence, and Drug Selection

8.5 Bioavailibility Equations


The following set of equations were used to solve the bioavailability problem set.
The problem sets for the first two drugs have been done for you. The others are
done exactly the same way. The answers follow the problems.

1.

AUMC iv
as discussed in chapter 4.
MRT iv = --------------------AUC iv

2.

1
k = --------------MRT iv

3.

ln 2
t 1 2 = -------k

4.

Cp 0iv = AUC k

5.

Dose iv
Vd = ---------------Cp 0iv

6.

Cp iv = Cp 0 e

7.

AUC oral Dose iv


f = -------------------- ----------------Dose oral AUC iv

8.

AUMC po
- as discussed in chapter 4
MRT po = ---------------------AUC po

9.

MAT po = MRT po MRT iv

10.

1
k a = -----------MAT

11.

kt

k
ln ----a-
k
t p = ---------------ka k

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-58

Bioavailability, Bioequivalence, and Drug Selection

12.

ka
kt
k t
Cp max = fD
------ ------------ (e p e a p )
V ka k

14.

( AUC generic ) ( Dose generic )


Relative Bioavailability (R.B. or C.B.) = --------------------------------------------------------------------( AUC Brand ) ( Dose Brand )

15.

Bioequivalent: Yes if all three:


0.80 < CB < 1.25
t p generic
0.80 < -------------< 1.25
tp brand
C p max g eneric
0.80 < ----------------------- < 1.25
C p max b rand

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-59

Bioavailability, Bioequivalence, and Drug Selection

8.6 Problems

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-60

Bioavailability, Bioequivalence, and Drug Selection

Caffeine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 1)

AHFS 00:00.00
GPI: 0000000000

Aramaki, S., et al., "Pharmacokinetics of caffeine and its metabolites in horses after intravenous, intramuscular, or oral administration", Chem Pharm Bull, Vol. 30, No. 11, (1991), p. 2999 - 3002.

This study deals with the pharmacokinetics of caffeine. Caffeine doses of 2.5 mg/kg were administered both intravenously and orally to horses with an average weight of about 500 kg. A summary of the some of data obtained from this
experiment is given below. Fill in the empty cells.
TABLE 8-24 Caffeine

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg/kg)

2.5

2.5

2.5

2.5

ug
AUC -------- hr
mL

63.1

60.7

60

57

2
ug
AUMC -------- hr
mL

1442

1556.8

1600

1723

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug-
Cpmax ------ mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-61

Bioavailability, Bioequivalence, and Drug Selection

Cefetamet Pivoxil
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 2)

AHFS 00:00.00
GPI: 0000000000

Ducharme, M., et. al., "Bioavailability of syrup and tablet formulations of cefetamet pivoxil", Antimicrobial Agents and Chemotherapy, Vol. 37, No. 12, (1993), p. 2706 - 2709.

Cefetamet pivoxil is a prodrug of cefetamet. This study compares the bioavailability of cefetamet pivoxil in tablet
form versus syrup form. A summary of the some of data obtained from this experiment is given below. Fill in the
approprate cells.
.
Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

250

500

500

500

ug
AUC -------- hr
mL

30.64

53.68

50

47

2
ug
AUMC -------- hr
mL

101.66

191.64

205.6

225.3

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug-
Cpmax ------ mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-62

Bioavailability, Bioequivalence, and Drug Selection

Cefixime

(Problem 8 - 3)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Faulkner, R. ,et al., "Absolute bioavailability of cefixime in man", Journal of Clinical Pharmacology, Vol. 28 (1988), p. 700 - 706.

Cefixime is a broad-spectrum cephalosporin which is active against a variety of gram positive and gram negative bacteria. In this study, sixteen subjects each received a 200 mg intravenous dose and then a 200 mg capsule with a
washout period between the administration of each dosage form. A summary of the some of data obtained from this
experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Capsule

Generic Capsule

Dose (mg)

200

200

200

ug
AUC -------- hr
mL

47

23.6

20.2

2
ug
AUMC -------- hr
mL

183.3

162.8

187.5

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug-
Cp at 1 hour ------ mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-63

Bioavailability, Bioequivalence, and Drug Selection

Ceftibuten
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 4)

AHFS 00:00.00
GPI: 0000000000

"The pharmacokinetics of ceftibuten in humans"

Ceftibuten is a new oral cephalosporin with potent activity against enterobacteriaceae and certain gram positive organisms. In this study two groups received either a 400 mg oral dosage form of ceftibuten or a 200 mg iv bolus
dose of ceftibuten. A summary of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

200

400

400

400

ug
AUC -------- hr
mL

75.2

65.9

64.2

64

2
ug
AUMC -------- hr
mL

211.2

213.4

220

208

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-64

Bioavailability, Bioequivalence, and Drug Selection

Cimetidine

(Problem 8 - 5)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Sandborn, W., et al., "Pharmacokinetics and pharmacodynamics of oral and intravenous cimetidine in seriously ill patients", Journal of Clinical Pharmacology, Vol. 30, (1990), p. 568 - 571.

Cimetidine is a histamine receptor antagonist which is used in the treatment of gastric and duodenal ulcer disease. In this study, patients received 300 mg of cimetidine as an iv bolus on the first day and data was collected. On
the second day, the patients received 300 mg orally and data was collected. A summary of the some of data obtained
from this experiment is given below.

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

300

300

300

ug
AUC -------- hr
mL

3.81

2.48

2.50

2
ug
AUMC -------- hr
mL

5.33

11.73

10.73

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-65

Bioavailability, Bioequivalence, and Drug Selection

Diurnal Variability in Theophylline Bioavailability


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 6)

AHFS 00:00.00
GPI: 0000000000

Bauer, L., Gibaldi, M., and Vestal, R., "Influence of pharmacokinetic diurnal variation on bioavailability estimates", Clinical Pharmacokinetics, vol. 9, (1984), p. 184 - 187.

This article discusses the effects of diurnal variation on the bioavailability and clearance of theophylline. In
this study patients received a 500 mg dose every 12 hours either orally or by iv bolus. A summary of the some of data
obtained from this experiment for the time period between midnight and noon is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

500

500

500

500

ug
AUC -------- hr
mL

160.25

144.58

140

144

2
ug
AUMC -------- hr
mL

1821

1662

1785

1700

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-66

Bioavailability, Bioequivalence, and Drug Selection

cis-5-Fluoro-1-[2-Hydroxymethyl-1,3-Oxathiolan-5-yl] Cytosine
(FTC)
(Problem 8 - 7)
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Frick, L. , et al., "Pharmacokinetics, oral bioavailability, and metabolic disposition in rats of (-)-cis-5-Fluoro-1-[2-Hydroxymethyl-1,3-Oxathiolan-5-yl] Cytosine, a nucleoside analog active against human immunodeficiency virus and hepatitis B virus", Antimicrobial Agents and Chemotherapy, Vol. 37, No. 11, (1993), p. 2285 - 2292.

FTC is a 2',3'-didoexynucleoside analog that may be useful against HIV and HBV. In this study, rats with an
average weight of 270 g were given either iv or oral doses of 100 mg/kg. A summary of the some of data obtained
from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg/kg)

100

100

100

ug
AUC -------- hr
mL

265

168

175

2
ug
AUMC -------- hr
mL

19514

12600

13125

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug-
Cpmax ------ mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-67

Bioavailability, Bioequivalence, and Drug Selection

Hydromorphone
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 8)

AHFS 00:00.00
GPI: 0000000000

Vallner, J., et al., "Pharmacokinetics and bioavailability of hydromorphone following intravenous and oral administration to
human subjects", Journal of Clinical Pharmacology, Vol. 21, (1981), p. 152 - 156.

Hydromorphone hydrochloride is an analog of morphine which has about seven times the effect of morphine
when given intravenously. In this study, volunteers were given a 2 mg intravenous dose and a 4 mg oral dose of hydromorphone on separate days. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

ug
AUC ------ hr
L

83

87.2

96

2
ug
AUMC ------ hr
L

289.4

401

432

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 ------
L
Vd (L)
ug
Cp at 1 hour ------
L
f
------
Cpmax ug
L
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-68

Bioavailability, Bioequivalence, and Drug Selection

Isosorbide Dinitrate
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 9)

AHFS 00:00.00
GPI: 0000000000

Straehl, P. and Galeazzi, R., "Isosorbide dinitrate bioavailability , kinetics, and metabolism", Clinical Pharmacology and Therapeutics, Vol. 38m (1985), p. 140 - 149.

Isosorbide dinitrate is used in the treatment of angina pectoris, vasospastic angina, and congestive heart failure.
In this study volunteers received a 5 mg intravenous dose given over 5 minutes and a 10 mg tablet. The different dosage forms were separated by a washout period. A summary of the some of data obtained from this experiment is given
below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg/kg)

10

10

AUC ug
------ hr
L

370.3

158

165

2
AUMC ug
------ hr
L

487

310

305

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 ------
L
Vd (L)
------
Cp at 1 hour ug
L
f
ug
Cpmax ------
L
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-69

Bioavailability, Bioequivalence, and Drug Selection

Ketanserin

(Problem 8 - 10)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Kurowski, M., "Bioavailability and pharmacokinetics of ketanserin in elderly subjects", Journal of Clinical Pharmacology, Vol. 28,
(1988), p. 700 - 706.

Ketanserin is a 5-hydroxytryptamine S2-antagonist. This study focuses on the kinetics of Ketanserin in the
elderly. Subjects were given either a 10 mg intravenous dose or a 40 mg oral tablet. A summary of the some of data
obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

10

40

40

ng
AUC -------- hr
mL

247

520

400

2
ng
AUMC -------- hr
mL

3991

8922

8922

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ng
Cp0 --------
mL
Vd (L)
ng
Cp at 1 hour --------
mL
f
ng
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-70

Bioavailability, Bioequivalence, and Drug Selection

Methotrexate

(Problem 8 - 11)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Seideman, P., et al., " The pharmacokinetics of methotrexate and its 7-hydroxy metabolite in patients with rheumatoid arthritis",
British Journal of Clinical Pharmacology, 35 (1993), p. 409 - 412.

The drug Methotrexate is a folic acid which has been shown to inhibit dihydrofolate reductase. The importance of this drug at present is mostly seen in the area of oncology, but lately it has been used for rheumatoid arthritis.
Methotrexate has a molecular weight of 454.4. In this study, the drug was administered both by IV bolus and orally as
a 15 mg dose. The following data was obtained:
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

15

15

15

AUC nmole
---------------- hr
L

2752

2708

2700

2
AUMC nmole
---------------- hr
L

15887

18400

18500

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
nmole
Cp0 ----------------
L
Vd (L)
----------------
Cp at 1 hour nmole
L
f
nmole
Cpmax ----------------
L
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-71

Bioavailability, Bioequivalence, and Drug Selection

Moclobemide

(Problem 8 - 12)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Schoerlin, M. et al., "Disposition kinetics of moclobemide, a new MAO-A inhibitor, in subjects with impaired renal function", Journal of Clinical Pharmacology, Vol. 30 (1991), p. 272 - 284.

Moclobemide is an antidepressant agent that reversibly inhibits the A-isozyme of the monoamine oxidase
enzyme system. In this study, single IV and oral doses were administered to a patient. A summary of the some of data
obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

150

100

100

ug
AUC -------- hr
mL

2.58

1.70

1.52

2
ug
AUMC -------- hr
mL

6.35

5.91

5.90

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-72

Bioavailability, Bioequivalence, and Drug Selection

Nalbuphine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 13)

AHFS 00:00.00
GPI: 0000000000

Nalbuphine hydrochloride is an agonist-antagonist opiod which is used for its analgesic actions. In this study,
volunteers were given single doses of four different nalbuphine forms. The data below focuses on a 10 mg iv dose and
a 45 mg dose of an oral solution. A summary of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

10

45

40

40

ng
AUC -------- hr
mL

86.9

70.3

62.5

60

2
ng
AUMC -------- hr
mL

288

306

280

270

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ng
Cp0 --------
mL
Vd (L)
ng
Cp at 1 hour --------
mL
f
ng
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-73

Bioavailability, Bioequivalence, and Drug Selection

Nefazodone
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 14)

AHFS 00:00.00
GPI: 0000000000

Shukla, U. et al., "Pharmacokinetics, absolute bioavailability, and disposition of nefazodone in the dog", Drug Metabolism and
Disposition, Vol. 21, No. 3, (1993), p. 502 - 507.

Nefazodone was given to four healthy, adult, male beagles with an average weight of 11.0 kg. Each dog was
given a 10 mg/kg dose as a either a intravenous injection or as an oral solution or tablet. A summary of the some of
data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg/kg)

10

10

10

10

ng
AUC -------- hr
mL

6023

829

800

700

2
ng
AUMC -------- hr
mL

29283

4875

4800

4500

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ng
Cp0 --------
mL
Vd (L)
ng
Cp at 1 hour --------
mL
f
ng
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-74

Bioavailability, Bioequivalence, and Drug Selection

Ondansetron

(Problem 8 - 15)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Colthup, P., et al., "Determination of ondansetron in plasma and its pharmacokinetics in the young and elderly", Journal of Pharmaceutical Sciences, Vol. 80, No. 9(1991), p. 868 - 871.

Ondansetron is a 5-hydroxyltryptamine compound which is useful in treating the nausea and vomiting which is
caused by the use of chemotherapy and radiation in the cancer patients. In order to determine the absolute bioavailability of oral Ondansetron, doses of 8 mg were given to two groups. One group received an oral dose and the other group
received an intravenous dose. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

ng
AUC -------- hr
mL

246.5

139

145

2
ng
AUMC -------- hr
mL

1138

795

870

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ng
Cp0 --------
mL
Vd (L)
ng
Cp at 1 hour --------
mL
f
ng
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-75

Bioavailability, Bioequivalence, and Drug Selection

Omeprazole

(Problem 8 - 16)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Anderson, T., et al, "Pharmacokinetics of various single intravenous and oral doses of omeprazole", Eur Journal of Clinical Pharmacology, 39, (1990), p. 195 - 197.

Omeprazole (mw: 345.42) is an agent which inhibits gastric acid secretion from the parietal cell. It is useful in
treating such problems as ulcers and gastroesophageal reflux disease. One group received an iv bolus dose and the
other group received an oral dose. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Capsule

Generic Capsule

Dose (mg)

20

40

40

mole
AUC ---------------- hr
L

3.2

3.5

3.0

2
mole
AUMC ---------------- hr
L

3.2

5.25

4.5

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
mole
Cp0 ----------------
L
Vd (L)
mole
Cp at 1 hour ----------------

f
mole
Cpmax ---------------
L

Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-76

Bioavailability, Bioequivalence, and Drug Selection

Paroxetine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 17)

AHFS 00:00.00
GPI: 0000000000

Lund, J., et al., "Paroxetine: pharmacokinetics and cardiovascular effects after oral and intravenous single doses in man", Journal
of Pharmacology and Toxicology, Vol. 51, (1982), p. 351 - 357.

Paroxetine kinetics and cardiovascular effects were studied in male subjects after single oral doses of 45 mg
and slow intravenous infusion of 23 - 28 mg. A summary of the some of data obtained from this experiment is given
below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

28

45

45

ng
AUC -------- hr
mL

467

750

675

2
ng
AUMC -------- hr
mL

6671

11250

10463

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ng
Cp0 --------
mL
Vd (L)
ng
Cp at 1 hour --------
mL
f
ng-
Cpmax ------ mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-77

Bioavailability, Bioequivalence, and Drug Selection

Ranitidine

(Problem 8 - 18)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Garg, D., et al., "Pharmacokinetics of ranitidine in patients with renal failure", Journal of Clinical Pharmacology, Vol. 26 (1986),
p. 286 - 291.

Ranitidine is an agent used in the treatment of peptic ulceration. In this study, ten patients with renal failure
received either a 50 mg intravenous bolus dose or a 150 mg tablet. A summary of the some of data obtained from this
experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

50

150

150

ng
AUC -------- hr
mL

5159

6422

6753

2
ng
AUMC -------- hr
mL

53415

78752

84413

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ng
Cp0 --------
mL
Vd (L)
ng
Cp at 1 hour --------
mL
f
ng
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-78

Bioavailability, Bioequivalence, and Drug Selection

Sulpiride
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 8 - 19)

AHFS 00:00.00
GPI: 0000000000

Bressolle, F., Bres, J., and Faure-Jeantis, A., "Absolute bioavailability , rate of absorption, and dose proportionality of sulpiride in
humans", Journal of Pharmaceutical Sciences ,Vol. 81, No. 1 (1992), p. 26 - 32.

Sulpiride is a substituted benzamine antipsychotic. In this study, the drug was administered to two groups.
The first group received a 200 mg oral dose and the second group received a 100 mg intravenous infusion. A summary
of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

100

200

200

200

ug
AUC -------- hr
mL

8.27

8.79

8.6

8.0

2
ug
AUMC -------- hr
mL

79.1

87.3

91.1

84.5

Bioequivalence

MRT (hr)
MAT (hr)
ke (hr-1)
ka (hr-1)
ug
Cp0 --------
mL
Vd (L)
ug
Cp at 1 hour --------
mL
f
ug
Cpmax --------
mL
Tmax (hr)
Relative Bioavailability
Generic Equivalent (Yes / No)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-79

Bioavailability, Bioequivalence, and Drug Selection

8.7 Solutions
8.7.1

CAFFEINE ON PAGE 61

Aramaki, S., et al., "Pharmacokinetics of caffeine and its metabolites in horses after intravenous, intramuscular, or oral administration", Chem Pharm Bull, Vol. 30, No. 11, (1991), p. 2999 - 3002.

This study deals with the pharmacokinetics of caffeine. Caffeine doses of 2.5 mg/kg were administered both intravenously and orally to horses with an average weight of about 500 kg. A summary of the some of data obtained from this
experiment is given below. Fill in the empty cells.
TABLE 8-25 Caffeine

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg/kg)

2.5

2.5

2.5

2.5

ug
AUC -------- hr
mL

63.1

60.7

60

57

2
ug
AUMC -------- hr
mL

1442

1556.8

1600

1723

MRT (hr)

22.9

25.7

26.7

30.2

2.79

3.81

7.36

0.358

0.262

0.136

0.78

0.59

0.31

0.96

0.95

.90

1.98

1.83

1.45

0.79

6.69

8.19

12.1

1.5

MAT (hr)
ke (hr-1)

0.0438

ka (hr-1)
ug
Cp0 --------
mL

2.76

V d (L/kg)

0.91

ug-
Cp at 1 hour ------ mL

2.64

f
ug
Cpmax --------
mL

Bioequivalence

2.76

Tmax (hr)
Relative Bioavailability

0.95

Generic Equivalent (Yes / No)

NO

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-80

Bioavailability, Bioequivalence, and Drug Selection

h
1442ug
--------------mL
---------------------------- = 22.9
2
h
63.1ug
--------------mL

1.

MRT = AUMC
------------------ =
AUC

2.

1 = ------------1 = 0.044 h 1
k = -----------MRT 22.9h

3.

ln 2 = 0.693
t 1 2 = ---------------------------- = 15.75h
1
k
0.044h

4.

1
h
ugCp 0 = AUC k = 63.1 ug
------------ 0.0044h
= 2.76 ------mL
mL

5.

hours

The horses have an average weight of 500 kg.

Dose = 2.5 mg
------- 500kg = 1250mg
kg
ug- = 2.78 mg
Cp 0 = 2.78 ------------mL
L
1250mg- = 449.6L = ------------------------2.5mg kg = 0.91 ----LVd = Dose
------------- = ------------------Cp 0
mg
mg
kg
2.78 ------2.78------L
L
6.

7.

Cp = Cp0 e

kt

ug- ( e 0.044 ( 1 ) ) = 2.64 ------ug= 2.78 ------mL


mL

h
2.5 mg
------60 ug
------------AUC oral Dose iv
mL
kg = 0.95
f = --------------------- ----------------- = -------------------- -----------------------Dose oral AUC iv
h
2.5 mg
------- 63.1ug
------------kg
mL
2

8.

MRT po

ug h 1556.8 --------------mL - = 25.7h


= AUMC
------------------ = -------------------------------AUC
ug
h
60.7------------mL

Where AUMC is that which is given for the oral dose.


Where AUC is that which is given for the oral dose.
9.

MAT po = MRT po MRT iv = 25.7h 22.9h = 2.79h

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-81

Bioavailability, Bioequivalence, and Drug Selection

10.

1 = --------1 - = 0.358hr 1
k a = -----------MAT
2.79
0.358hr
k a
ln -----------------------

1
ln -----

0.044h
k
tp = ---------------- = ------------------------------------------------------ = 6.7hr
1
1
ka k
0.358hr 0.044hr
1

11.

12.

13.

ktp
katp
( 0.044 6.7 )
( 0.358 6.7 )
fD
ka
0.96 1250mg
0.358hr
e
) = ----------------------------------- ------------------------------------------------- ( e
Cp max = ------ -------------- ( e
e
)
1
V ka k
449L
( 0.358 0.044 ) hr

( AUC

) ( Dose

generic
generic
Relative Bioavailability (R.B. or C.B.) = ---------------------------------------------------------------------

( AUC Brand ) ( Dose Brand )

ug-
57 ------ mL

hr 2.5 mg
-------

km
CB = ------------------------------------------------------------- = 0.95
ug- hr 2.5 mg
60 -------------
mL

km
14.

Bioequivalent: Yes if all three = Yes:

0.80 < CB < 1.25 CB = 0.95 = Yes


t p generic
0.80 < -------------< 1.25
tp brand

t p generic
-------------= 12.1hr
---------------- = 1.5 = NO
8.19hr
t pbrand

C p max g eneric
0.80 < ----------------------- < 1.25
C p max b rand

ug1.45 ------C p max g eneric


mL- = 0.79 = NO
------------------------ = -----------------C p max b rand
ug1.83 ------mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-82

Bioavailability, Bioequivalence, and Drug Selection

8.7.2

CEFETAMET PIVOXIL ON PAGE 62

Ducharme, M., et. al., "Bioavailability of syrup and tablet formulations of cefetamet pivoxil", Antimicrobial Agents and Chemotherapy, Vol. 37, No. 12, (1993), p. 2706 - 2709.

Cefetamet pivoxil is a prodrug of cefetamet. This study compares the bioavailability of cefetamet pivoxil in tablet
form versus syrup form. A summary of the some of data obtained from this experiment is given below. Fill in the
approprate cells.
.
Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

250

500

500

500

ug- hr
AUC ------ mL

30.64

53.68

50

47

ug- hr 2
AUMC ------ mL

101.66

191.64

205.6

225.3

MRT (hr)

3.32

3.57

4.11

4,79

0.252

0.794

1.48

3.97

1.26

0.678

12.62

9.03

5.91

0.88

0.82

0.77

13.1

9.6

7.4

0.77

0.70

1.49

2.15

1.44

MAT (hr)
ke

(hr-1)

0.301

ka (hr-1)
ug
Cp0 --------
mL

9.23

Vd (L)

27.1

ug-
Cp at 1 hour ------ mL

6.83

f
ug
Cpmax --------
mL

Bioequivalence

9.23

Tmax (hr)
Relative Bioavailability

0.94

Generic Equivalent (Yes / No)

NO
2

1.

mg h
101.66 ----------------L
MRT = AUMC
------------------ = ---------------------------------= 3.32 hours
2
AUC
mg
h
30.64 ----------------L
Where AUMC is that which is given for the intravenous dose.
Where AUC is that which is given for the intravenous dose.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-83

Bioavailability, Bioequivalence, and Drug Selection

2.

1
1
1
k = -----------= ------------- = 0.301 h
MRT 3.32h

3.

ln 2 = --------------------0.693 = 2.3h
t 1 2 = -------1
k
0.301h

4.

1
h
Cp 0 = AUC k = 30.64 mg
-------------- 0.301h = 9.22 mg
------L
L

5.

6.

7.

250mg
V d = Dose
------------- = ------------------ = 27.06L
Cp 0
9.24mg
------L
Cp = Cp 0 e

kt

0.301 ( 1 )
mg
= 9.24 mg
------- ( e
) = 6.84------L
L

h
53.68mg
-------------AUC oral Dose iv
L
250mg - = 0.876
f = --------------------- ----------------- = ----------------------------- ---------------------------Dose oral AUC iv
500mg
h
30.64 mg
-------------L
2

8.

MRT po

mg h
191.64 ----------------L
= AUMC
------------------ = ---------------------------------= 3.57h
AUC
mg
h
53.68 --------------L

Where AUMC is that which is given for the oral dose.


Where AUC is that which is given for the oral dose.
9.
10.

MAT po = MRT po MRT iv = 3.57h 3.32h = 0.252hr


1 = -----------1 - = 3.97hr 1
k a = -----------MAT
0.252
4.0h

k a
ln --------------------

ln -----
0.301h 1
k
t p = ---------------- = ------------------------------------------- = 0.7h
1
1
ka k
4.0h 0.301h
1

11.

12.

13.

0.301 ( 0.7 )
3.97 ( 0.7 )
ka ( e kt e k a t ) = 0.88
( 500mg )
3.97hr
mg
Cp max = fD
------ -------------------------------------------- ---------------------------------------------- ( e
e
) = 13.1------1
V ka k
27.1L
L
( 3.97 0.301 ) hr

( AUC

) ( Dose

generic
generic
Relative Bioavailability (R.B. or C.B.) = ---------------------------------------------------------------------

( AUC Brand ) ( Dose Brand )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-84

Bioavailability, Bioequivalence, and Drug Selection

ug-
47 ------ mL

hr 500mg
CB = -------------------------------------------------------- = 0.94
ug- hr 500mg
50 ------ mL

14.

Bioequivalent: Yes if all three = Yes:

0.80 < CB < 1.25 CB = 0.94 = Yes


t p generic
0.80 < -------------< 1.25
tp brand

t p generic
-------------= 2.15hr
---------------- = 1.44 = NO
1.49hr
t pbrand

C p max g eneric
0.80 < ----------------------- < 1.25
C p max b rand

ug7.4 ------C p max g eneric


mL
------------------------ = ---------------- = 0.77 = NO
C p max b rand
ug9.6 ------mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-85

Bioavailability, Bioequivalence, and Drug Selection

8.7.3

CEFIXIME ON PAGE 63

Faulkner, R. ,et al., "Absolute bioavailability of cefixime in man", Journal of Clinical Pharmacology, Vol. 28 (1988), p. 700 - 706.

Cefixime is a broad-spectrum cephalosporin which is active against a variety of gram positive and gram negative bacteria. In this study, sixteen subjects each received a 200 mg intravenous dose and then a 200 mg capsule with a
washout period between the administration of each dosage form. A summary of the some of data obtained from this
experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Capsule

Generic Capsule

Dose (mg)

200

200

200

ug
AUC -------- hr
mL

47

23.6

20.2

2
ug
AUMC -------- hr
mL

183.3

162.8

187.5

MRT (hr)

3.9

6.9

9.3

3.0

5.38

0.334

0.186

1.5

0.77

0.50

0.43

2.5

1.6

0.64

3.4

4.6

1.33

MAT (hr)
ke

(hr-1)

0.256

ka (hr-1)
ug
Cp0 --------
mL

12.1

Vd (L)

16.6

ug
Cp at 1 hour --------
mL

9.3

f
ug-
Cpmax ------ mL

Bioequivalence

12.1

Tmax (hr)
Relative Bioavailability

0.86

Generic Equivalent (Yes / No)

NO

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-86

Bioavailability, Bioequivalence, and Drug Selection

8.7.4

CEFTIBUTEN ON PAGE 64

"The pharmacokinetics of ceftibuten in humans"

Ceftibuten is a new oral cephalosporin with potent activity against enterobacteriaceae and certain gram positive organisms. In this study two groups received either a 400 mg oral dosage form of ceftibuten or a 200 mg iv bolus
dose of ceftibuten. A summary of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

200

400

400

400

ug- hr
AUC ------ mL

75.2

65.9

64.2

64

ug- hr 2
AUMC ------ mL

211.2

213.4

220

208

MRT (hr)

2.94

MAT (hr)
ke (hr-1)

3.24

3.43

3.25

0.297

0.485

0.309

3.37

2.06

3.24

16.9

15.3

16.4

0.44

0.42

0.43

17.3

15.3

16.7

1.09

0.76

1.05

0.78

0.74

0.390

ka (hr-1)
ug-
Cp0 ------ mL

25.6

Vd (L)

7.8

ug
Cp at 1 hour --------
mL

18.2

f
ug
Cpmax --------
mL

Bioequivalence

25.6

Tmax (hr)
Relative Bioavailability

Generic Equivalent (Yes / No)

NO

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-87

Bioavailability, Bioequivalence, and Drug Selection

8.7.5

CIMETIDINE ON PAGE 65

Sandborn, W., et al., "Pharmacokinetics and pharmacodynamics of oral and intravenous cimetidine in seriously ill patients", Journal of Clinical Pharmacology, Vol. 30, (1990), p. 568 - 571.

Cimetidine is a histamine receptor antagonist which is used in the treatment of gastric and duodenal ulcer disease. In this study, patients received 300 mg of cimetidine as an iv bolus on the first day and data was collected. On
the second day, the patients received 300 mg orally and data was collected. A summary of the some of data obtained
from this experiment is given below.

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

300

300

300

ug- hr
AUC ------ mL

3.81

2.48

2.50

ug- hr 2
AUMC ------ mL

5.33

11.73

10.73

MRT (hr)

1.40

4.73

4.29

3.33

2.89

0.300

0.346

0.32

0.37

0.65

0.66

0.40

0.44

1.1

2.1

2.0

0.94

MAT (hr)
ke

(hr-1)

0.715

ka (hr-1)
ug-
Cp0 ------ mL

2.72

Vd (L)

110

ug
Cp at 1 hour --------
mL

1.33

f
ug
Cpmax --------
mL

Bioequivalence

2.72

Tmax (hr)
Relative Bioavailability

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-88

Bioavailability, Bioequivalence, and Drug Selection

8.7.6

DIURNAL VARIABILITY IN THEOPHYLLINE BIOAVAILABILITY


ON PAGE 66

Bauer, L., Gibaldi, M., and Vestal, R., "Influence of pharmacokinetic diurnal variation on bioavailability estimates", Clinical Pharmacokinetics, vol. 9, (1984), p. 184 - 187.

This article discusses the effects of diurnal variation on the bioavailability and clearance of theophylline. In
this study patients received a 500 mg dose every 12 hours either orally or by iv bolus. A summary of the some of data
obtained from this experiment for the time period between midnight and noon is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

500

500

500

500

ug- hr
AUC ------ mL

160.25

144.58

140

144

ug- hr 2
AUMC ------ mL

1821

1662

1785

1700

MRT (hr)

11.40

11.50

12.75

11.8

0.13

1.39

0.44

7.58

0.721

2.26

11.8

6.02

10.7

0.90

0.87

0.90

12.1

9.20

11.1

1.21

.059

3.3

1.5

0.45

MAT (hr)
ke

(hr-1)

0.088

ka (hr-1)
ug-
Cp0 ------ mL

14.1

Vd (L)

35.5

ug
Cp at 1 hour --------
mL

12.9

f
ug
Cpmax --------
mL

Bioequivalence

14.1

Tmax (hr)
Relative Bioavailability

1.03

Generic Equivalent (Yes / No)

NO

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-89

Bioavailability, Bioequivalence, and Drug Selection

8.7.7

CIS-5-FLUORO-1-[2-HYDROXYMETHYL-1,3-OXATHIOLAN-5-YL]
CYTOSINE (FTC) ON PAGE 67

Frick, L. , et al., "Pharmacokinetics, oral bioavailability, and metabolic disposition in rats of (-)-cis-5-Fluoro-1-[2-Hydroxymethyl-1,3-Oxathiolan-5-yl] Cytosine, a nucleoside analog active against human immunodeficiency virus and hepatitis B virus", Antimicrobial Agents and Chemotherapy, Vol. 37, No. 11, (1993), p. 2285 - 2292.

FTC is a 2',3'-didoexynucleoside analog that may be useful against HIV and HBV. In this study, rats with an
average weight of 270 g were given either iv or oral doses of 100 mg/kg. A summary of the some of data obtained
from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg/kg)

100

100

100

ug- hr
AUC ------ mL

265

168

175

ug- hr 2
AUMC ------ mL

19514

12600

13125

MRT (hr)

73.6

75

75

1.36

1.36

0.734

0.734

1.18

1.23

0.63

0.66

2.1

2.2

1.04

5.54

5.54

1.0

MAT (hr)
ke

(hr-1)

.0136

ka (hr-1)
ug-
Cp0 ------ mL

3.6

Vd (L/kg)

27.7

ug
Cp at 1 hour --------
mL

3.55

f
ug
Cpmax --------
mL

Bioequivalence

3.6

Tmax (hr)
Relative Bioavailability

1.04

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-90

Bioavailability, Bioequivalence, and Drug Selection

8.7.8

HYDROMORPHONE ON PAGE 68

Vallner, J., et al., "Pharmacokinetics and bioavailability of hydromorphone following intravenous and oral administration to
human subjects", Journal of Clinical Pharmacology, Vol. 21, (1981), p. 152 - 156.

Hydromorphone hydrochloride is an analog of morphine which has about seven times the effect of morphine
when given intravenously. In this study, volunteers were given a 2 mg intravenous dose and a 4 mg oral dose of hydromorphone on separate days. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

83

87.2

96

289.4

401

432

3.49

4.60

4.50

1.11

1.03

0.899

0.987

12.6

14.7

0.53

0.56

14.6

16.6

1.13

1.87

1.77

0.95

ug
AUC ------ hr
L
2
------ hr
AUMC ug
L

MRT (hr)
MAT (hr)
ke

(hr-1)

0.287

ka (hr-1)
ug
Cp0 ------
L

23.8

Vd (L)

84

------
Cp at 1 hour ug
L

17.9

f
ug
Cpmax ------
L

Bioequivalence

23.8

Tmax (hr)
Relative Bioavailability

1.1

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-91

Bioavailability, Bioequivalence, and Drug Selection

8.7.9

ISOSORBIDE DINITRATE ON PAGE 69

Straehl, P. and Galeazzi, R., "Isosorbide dinitrate bioavailability , kinetics, and metabolism", Clinical Pharmacology and Therapeutics, Vol. 38m (1985), p. 140 - 149.

Isosorbide dinitrate is used in the treatment of angina pectoris, vasospastic angina, and congestive heart failure.
In this study volunteers received a 5 mg intravenous dose and a 10 mg tablet. The different dosage forms were separated by a washout period. A summary of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg/kg)

10

10

370.3

158

165

487

310

305

1.32

1.96

1.85

0.65

0.53

1.546

1.875

60.1

66.2

0.21

0.22

60.4

67.8

1.12

0.90

0.81

0.90

ug
AUC ------ hr
L

2
ug
AUMC ------ hr
L

MRT (hr)
MAT (hr)
ke (hr-1)

0.760

ka (hr-1)
------
Cp0 ug
L

282

Vd (L)

17.75

ug
Cp at 1 hour ------
L

132

f
ug
Cpmax ------
L

Bioequivalence

282

Tmax (hr)
Relative Bioavailability

1.04

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-92

Bioavailability, Bioequivalence, and Drug Selection

8.7.10

KETANSERIN ON PAGE 70

Kurowski, M., "Bioavailability and pharmacokinetics of ketanserin in elderly subjects", Journal of Clinical Pharmacology, Vol. 28,
(1988), p. 700 - 706.

Ketanserin is a 5-hydroxytryptamine S2-antagonist. This study focuses on the kinetics of Ketanserin in the
elderly. Subjects were given either a 10 mg intravenous dose or a 40 mg oral tablet. A summary of the some of data
obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

10

40

40

ng- hr
AUC ------ mL

541

112.5

103.9

ng- hr 2
AUMC ------ mL

11700

24900

22900

MRT (hr)

21.6

22.1

22.0

0.5

0.4

2.0

2.5

43.6

42.7

.052

0.48

47.6

44.5

0.94

1.93

1.63

0.84

MAT (hr)
ke (hr-1)

Bioequivalence

0.0402

ka (hr-1)
ng-
Cp0 ------ mL

25.0

Vd (L)

400

ng
Cp at 1 hour --------
mL

23.9

f
ng
Cpmax --------
mL

25.0

Tmax (hr)
Relative Bioavailability

0.92

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-93

Bioavailability, Bioequivalence, and Drug Selection

8.7.11

METHOTREXATE ON PAGE 71

Seideman, P., et al., " The pharmacokinetics of methotrexate and its 7-hydroxy metabolite in patients with rheumatoid arthritis",
British Journal of Clinical Pharmacology, 35 (1993), p. 409 - 412.

The drug Methotrexate is a folic acid which has been shown to inhibit dihydrofolate reductase. The importance of this drug at present is mostly seen in the area of oncology, but lately it has been used for rheumatoid arthritis.
Methotrexate has a molecular weight of 454.4. In this study, the drug was administered both by IV bolus and orally as
a 15 mg dose. The following data was obtained:
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

15

15

15

nmole
AUC ---------------- hr

2752

2708

2700

2
nmole
AUMC ---------------- hr
L

15887

18400

18500

MRT (hr)

5.77

6.79

6.85

1.02

1.08

0.979

0.927

265

256

0.98

0.98

323

318

0.98

2.15

2.23

1.04

MAT (hr)
ke

(hr-1)

0.173

ka (hr-1)
nmole
Cp0 ----------------
L

477

Vd (L)

69.3

nmole
Cp at 1 hour ----------------

401

f
----------------
Cpmax nmole

Bioequivalence

477

Tmax (hr)
Relative Bioavailability

1.0

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-94

Bioavailability, Bioequivalence, and Drug Selection

8.7.12

MOCLOBEMIDE ON PAGE 72

Schoerlin, M. et al., "Disposition kinetics of moclobemide, a new MAO-A inhibitor, in subjects with impaired renal function", Journal of Clinical Pharmacology, Vol. 30 (1991), p. 272 - 284.

Moclobemide is an antidepressant agent that reversibly inhibits the A-isozyme of the monoamine oxidase
enzyme system. In this study, single IV and oral doses were administered to a patient. A summary of the some of data
obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

150

100

100

ug- hr
AUC ------ mL

2.58

1.70

1.52

ug- hr 2
AUMC ------ mL

6.35

5.91

5.90

MRT (hr)

2.46

3.48

3.80

1.02

1.42

0.985

0.704

0.344

0.250

.099

.088

.037

0.29

.079

1.53

1.85

1.21

MAT (hr)
ke (hr-1)

0.406

ka (hr-1)
ug-
Cp0 ------ mL

1.05

Vd (L)

143

ug
Cp at 1 hour --------
mL

0.698

f
ug
Cpmax --------
mL

Bioequivalence

1.05

Tmax (hr)
Relative Bioavailability

0.89

Generic Equivalent (Yes / No)

NO

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-95

Bioavailability, Bioequivalence, and Drug Selection

8.7.13

NALBUPHINE ON PAGE 73

Nalbuphine hydrochloride is an agonist-antagonist opiod which is used for its analgesic actions. In this study,
volunteers were given single doses of four different nalbuphine forms. The data below focuses on a 10 mg iv dose and
a 45 mg dose of an oral solution. A summary of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

10

45

40

40

ng- hr
AUC ------ mL

86.9

70.3

62.5

60

ng- hr 2
AUMC ------ mL

288

306

280

270

MRT (hr)

3.31

4.35

4.48

4.5

1.04

1.17

1.19

0.963

0.858

0.843

11.1

9.2

8.7

0.180

0.180

0.173

12.5

10.7

10.2

0.95

1.76

1.88

1.90

1.01

MAT (hr)
ke (hr-1)

.0301

ka (hr-1)
ng-
Cp0 ------ mL

26.2

Vd (L)

381

ng
Cp at 1 hour --------
mL

19.4

f
ng
Cpmax --------
mL

Bioequivalence

26.2

Tmax (hr)
Relative Bioavailability

0.96

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-96

Bioavailability, Bioequivalence, and Drug Selection

8.7.14

NEFAZODONE ON PAGE 74

Shukla, U. et al., "Pharmacokinetics, absolute bioavailability, and disposition of nefazodone in the dog", Drug Metabolism and
Disposition, Vol. 21, No. 3, (1993), p. 502 - 507.

Nefazodone was given to four healthy, adult, male beagles with an average weight of 11.0 kg. Each dog was
given a 10 mg/kg dose as a either a intravenous injection or as an oral solution or tablet. A summary of the some of
data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg/kg)

10

10

10

10

ng- hr
AUC ------ mL

6023

829

800

700

ng- hr 2
AUMC ------ mL

29283

4875

4800

4500

MRT (hr)

4.86

5.88

6.0

6.43

1.02

1.14

1.57

0.982

0.879

0.638

94.8

85.7

60.7

0.138

0.133

0.116

112.7

105.6

84.0

0.80

2.0

2.16

2.62

1.21

MAT (hr)
ke (hr-1)

0.210

ka (hr-1)
ng-
Cp0 ------ mL

1238

Vd (L)

8.07

ng
Cp at 1 hour --------
mL

1009

f
ng
Cpmax --------
mL

Bioequivalence

1238

Tmax (hr)
Relative Bioavailability

0.88

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-97

Bioavailability, Bioequivalence, and Drug Selection

8.7.15

ONDANSETRON ON PAGE 75

Colthup, P., et al., "Determination of ondansetron in plasma and its pharmacokinetics in the young and elderly", Journal of Pharmaceutical Sciences, Vol. 80, No. 9(1991), p. 868 - 871.

Ondansetron is a 5-hydroxyltryptamine compound which is useful in treating the nausea and vomiting which is
caused by the use of chemotherapy and radiation in the cancer patients. In order to determine the absolute bioavailability of oral Ondansetron, doses of 8 mg were given to two groups. One group received an oral dose and the other group
received an intravenous dose. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

ng- hr
AUC ------ mL

246.5

139

145

ng- hr 2
AUMC ------ mL

1138

795

870

MRT (hr)

4.62

5.72

6.0

1.10

1.38

0.907

0.723

15.9

14.7

0.56

0.59

19.2

18.8

2.1

2.4

MAT (hr)
ke

(hr-1)

0.217

ka (hr-1)
ng-
Cp0 ------ mL

53.4

Vd (L)

150

ng
Cp at 1 hour --------
mL

43

f
ng
Cpmax --------
mL

Bioequivalence

53.4

Tmax (hr)

1.1

Relative Bioavailability

1.04

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-98

Bioavailability, Bioequivalence, and Drug Selection

8.7.16

OMEPRAZOLE ON PAGE 76

Anderson, T., et al, "Pharmacokinetics of various single intravenous and oral doses of omeprazole", Eur Journal of Clinical Pharmacology, 39, (1990), p. 195 - 197.

Omeprazole (mw: 345.42) is an agent which inhibits gastric acid secretion from the parietal cell. It is useful in
treating such problems as ulcers and gastroesophageal reflux disease. One group received an iv bolus dose and the
other group received an oral dose. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Capsule

Generic Capsule

Dose (mg)

20

40

40

mole hr
AUC ---------------

3.2

3.5

3.0

2
mole
hr
AUMC ---------------

3.2

5.25

4.5

MRT (hr)

1.0

1.5

1.5

0.5

0.5

1.63

1.40

0.55

0.47

1.8

1.5

0.86

0.69

0.69

MAT (hr)
ke

(hr-1)

ka (hr-1)
mole
Cp0 ----------------

3.2

Vd (L)

52.4

mole
Cp at 1 hour ---------------
L

1.18

f
mole
Cpmax ----------------

Bioequivalence

3.2

Tmax (hr)
Relative Bioavailability

0.86

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-99

Bioavailability, Bioequivalence, and Drug Selection

8.7.17

PAROXETINE ON PAGE 77

Lund, J., et al., "Paroxetine: pharmacokinetics and cardiovascular effects after oral and intravenous single doses in man", Journal
of Pharmacology and Toxicology, Vol. 51, (1982), p. 351 - 357.

Paroxetine kinetics and cardiovascular effects were studied in male subjects after single oral doses of 45 mg
and slow intravenous infusion of 28 mg. A summary of the some of data obtained from this experiment is given below.

From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

28

45

45

ng
AUC -------- hr
mL

467

750

675

2
ng
AUMC -------- hr
mL

6671

11250

10463

MRT (hr)

14.3

MAT (hr)
ke (hr-1)

15

15.5

0.72

1.22

1.40

.082

37.9

25.5

0.90

44.8

37.6

0.84

2.25

3.27

1.45

0.07

ka (hr-1)
ng
Cp0 --------
mL

32.7

Vd (L)

856

ng-
Cp at 1 hour ------ mL

30.5

f
ng
Cpmax --------
mL

Bioequivalence

32.7

Tmax (hr)
Relative Bioavailability

0.90

Generic Equivalent (Yes / No)

NO

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-100

Bioavailability, Bioequivalence, and Drug Selection

8.7.18

RANITIDINE ON PAGE 78

Garg, D., et al., "Pharmacokinetics of ranitidine in patients with renal failure", Journal of Clinical Pharmacology, Vol. 26 (1986),
p. 286 - 291.

Ranitidine is an agent used in the treatment of peptic ulceration. In this study, ten patients with renal failure
received either a 50 mg intravenous bolus dose or a 150 mg tablet. A summary of the some of data obtained from this
experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Brand Tablet

Generic Tablet

Dose (mg)

50

150

150

ng- hr
AUC ------ mL

5159

6422

6753

ng- hr 2
AUMC ------ mL

53415

78752

84413

MRT (hr)

10.4

12.3

12.5

1.91

2.15

0.524

0.466

240

231

.0415

0.436

423

432

1.02

3.96

4.26

1.07

MAT (hr)
ke (hr-1)

Bioequivalence

0.0966

ka (hr-1)
ng-
Cp0 ------ mL

498

Vd (L)

100

ng
Cp at 1 hour --------
mL

452

f
ng
Cpmax --------
mL

498

Tmax (hr)
Relative Bioavailability

1.05

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-101

Bioavailability, Bioequivalence, and Drug Selection

8.7.19

SULPIRIDE ON PAGE 79

Bressolle, F., Bres, J., and Faure-Jeantis, A., "Absolute bioavailability , rate of absorption, and dose proportionality of sulpiride in
humans", Journal of Pharmaceutical Sciences ,Vol. 81, No. 1 (1992), p. 26 - 32.

Sulpiride is a substituted benzamine antipsychotic. In this study, the drug was administered to two groups.
The first group received a 200 mg oral dose and the second group received a 100 mg intravenous infusion. A summary
of the some of data obtained from this experiment is given below.
From the preceding data, please calculate the following:

Parameter

IV

Oral Solution

Brand Tablet

Generic Tablet

Dose (mg)

100

200

200

200

ug- hr
AUC ------ mL

8.27

8.79

8.6

8.0

ug- hr 2
AUMC ------ mL

79.1

87.3

91.1

84.5

MRT (hr)

9.56

MAT (hr)
ke (hr-1)

9.93

10.6

10.6

0.367

1.02

1.0

2.72

0.972

1.0

0.798

0.526

0.498

0.53

0.52

0.48

0.807

0.687

0.643

0.94

1.24

2.57

2.52

0.98

0.865

ka (hr-1)
ug-
Cp0 ------ mL

0.865

Vd (L)

116

ug
Cp at 1 hour --------
mL

0.779

f
ug
Cpmax --------
mL

Bioequivalence

0.865

Tmax (hr)
Relative Bioavailability

0.93

Generic Equivalent (Yes / No)

YES

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-102

Bioavailability, Bioequivalence, and Drug Selection

8.8 References
1.

Miller S.W., Strom J.G., Drug Product Selection: Implications for the Geriatric Patient, The Consultant Pharmacist, 5(1):30-37, 1990.

2.

The Food and Drug Letter, 365:2, 1990.

3.

Lamy, P., Critical Patients, Critical Drugs, Critical Diseases, Maryland Pharmacist, 61:22-25, 1985.

4.

Colaizzi, J., Lowenthal, D., Critical Therapeutic Categories: A Contraindication to Generic Substitution?, Clin.
Therap., 8:370-379, 1986.

5.

Foster, T.S., Selecting Therapeutically Equivalent Products: Special Cases, Am. Pharm., NS31 (11):49-54,
1991.

6.

Meyer, M., The Therapeutic Equivalence of Drug Products. A Second Look, The University of Tennessee Center for the Health Sciences, Memphis, 1985.

7.

Levy, G., The Therapeutic Implications of Brand Interchange, Am. J. Hosp. Pharm., 17:756-760, 1960.

8.

Lamy, P., Generic Equivalents: Issues and Concerns, J. Clin. Pharmacol., 26:309-316, 1986.

9.

Dettelbach, H.R., A Time to Speak Out on Bioequivalence and Therapeutic Equivalence, J. Clin. Pharmacol.,
26:307-308, 1986.

10.

Schwartz, L., The Debate Over Substitution Policy, Am. J. Med., 79:38-44, 1985.

11.

Berger, B., Drug Product Selection: Are All Drugs Created Equal?, M. M & M, Sep:46-53, 1980.

12.

Lamy, P, What Should We Know about Generics?, Geriatric Medicine Today, 5 (2):25-27, 1986.

13.

Horwitz, N., Generic Bioequivalence Tests are Flawed, Medical Tribune, 26 (26):1, 1985.

14.

Gottschalk, L.A., Clinical Relevance of the Bioavailability/Bioequivalence Controversy, J. Clin. Psychiatry,


47(9, Suppl):3-5, 1986.

15.

Barone, J.A., Colaizzi, J.L., Critical Evaluation of Thioridazine Bioequivalence, Drug Intell. Clin. Pharm.,
19:847-858, 1985.

16.

Strom, B.L., Generic Drug Substitution Revisited, N. Eng. J. Med., 316: 1456-1462, 1987.

17.

Weaver, L.C., Drug Cost Containment and the Case for Generics, IPU Review, 12:320-324, 1987.

18.

Nuwer, M.R., et al., Generic Substitutions for Antiepileptic Drugs, Neurology, 40:1647-1651, 1990.

19.

Blake, M.I., Drug Product Equivalency, Drug Topics, 132(Oct. 3):84-89, 1988.

20.

Lofholm, P.W., Multisource Product Selection, US Pharmacist, 16:44-45, 1991.

21.

Jochsberger, T., Factors Influencing Drug Product Selection - Part I, Pharmacy Times, 47(1):66-75, 1981.

22.

Jochsberger, T. Factors Influencing Drug Product Selection - Part II, Pharmacy Times, 47(2):68-75, 1981.

23.

Koch-Weser, J. Bioavailability of Drugs, Medical Intelligence, 291:233-237, 1974.

24.

Welling, P.G., Drug Bioavailability and its Clinical Significance, in Progress in Drug Metabolism, Vol. 4,
Bridges, J.W. and Chasseaud, L.F., Eds. John Wiley & Sons Ltd., New York, p. 131-163, 1980.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-103

Bioavailability, Bioequivalence, and Drug Selection

25.

Blanchard, J. and Sawchuk, R.J., Drug Bioavailability: An Overview, in Principles and Perspectives in Drug
Bioavailability, Blanchard, J., Sawchuk, R.J. and Brodie, B.B., ed., Karger, Basel, p. 1-19, 1979.

26.

Edwards, D.J., Bioavailability, Bioequivalence and Therapeutic Equivalence: Concepts and Issues for Pharmacy Students, A. J. Ph. Ed., 54:178-181, 1990.

27.

Gibaldi, M., Biopharmaceutics and Clinical Pharmacokinetics, Fourth Edition, Lea & Febiger, Philadelphia, p.
24-79, 1991.

28.

Banakar, U.V., Issues in Contemporary Drug Delivery, Part II: Biopharmaceutical Considerations, J. Pharm.
Technol., 6:122-131, 1990.

29.

Riley, T.N. and Ravis, W.R., Key Concepts in Drug Bioequivalence, U.S. Pharmacist, 12(2):41-53, 1987.

30.

Welling, P.G., Interactions Affecting Drug Absorption, Clin. Pharmacokinetics, 9:404-434, 1984.

31.

Toothaker, R.D. and Welling, P.G., The Effect of Food on Drug Bioavailability, Ann. Rev. Pharmacol. Toxicol.,
20:173-179, 1980.

32.

Welling, P.G., Pharmacokinetics, Processes and Mathematics, American Chemical Society, Washington D.C.,
35-76, 1986.

33.

Selen, A., Factors Influencing Bioavailability and Bioequivalence, in Pharmaceutical Bioequivalence, Welling, P.G., Tse, F.L.S. and Dighe, S.V., editors, Marcel Dekker, Inc., New York, p. 117-148, 1991.

34.

Shargel, L. and Yu, A.B.C., Applied Biopharmaceutics and Pharmacokinetics, Appleton & Lange, Norwalk,
Connecticut, p. 111-167, 1993.

35.

Cadwallader, D.E., Biopharmaceutics and Drug Interactions, Third Edition, Raven Press, New York, p. 39-86,
1983.

36.

Welling, P.G., Influence of Food and Diet on Gastrointestinal Drug Absorption: A Review, J. Pharmacokin.
Bioph., 5(4):291-334, 1977.

37.

Notari, R.E., Biopharmaceutics and Clinical Pharmacokinetics, Marcel Dekker, New York, p. 160-171, 1987.

38.

Abdou, H.M., Dissolution, Bioavailability and Bioequivalence, Mack Publishing Company, Easton, Pennsylvania, p. 53-105, 1989.

39.

Kakemi, K., Absorption and Excretion of Chloramphenicol, Symposium on Drug Absorption, Metabolism and
Excretion, Paper B-IV, Preprints of Papers, Scientific Section of the American Pharmaceutical Association,
Las Vegas, 1962.

40.

Neuvonen, P.J., Pentikainen, P.J., and Elfing, S.M., Factors Affecting the Bioavailability of Phenytoin, Int. J.
Clin. Pharmacol. Biopharm., 15:84, 1977.

41.

Sjogren, J., Solvell, L., and Karlsson, I., Studies on the Absorption Rates of Barbiturates in Man., Acta Med.
Scand., 178:553, 1965.

42.

Heading, R.C., et al., The Dependence of Paracetamol Absorption on the Rate of Gastric Emptying, Br. J.
Pharmacol., 47:415, 1973.

43.

Mayersohn, M., in Principles and Perspectives in Drug Bioavailability, Blanchard, J., Sawchuk, R.J. and Brodie, B.B., ed., S. Karger, Basel, p. 211, 1979.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-104

Bioavailability, Bioequivalence, and Drug Selection

44.

Gibaldi, M., Drug Interactions, The Annals of Pharmacotherapy, 26:829-834, 1992.

45.

Welling, P.G., Huang, H., Hewitt, P.F., and Lyons, L.L., Bioavailability of Erythromycin Stearate: Influence of
Food and Fluid Volume, J. Pharm. Sci., 67:764-766, 1978.

46.

Mischler, T.W., Sugerman, A.A., Willard, D.A., Brannick, L.J., and Neiss, E.S., Influence of Probenecid and
Food on the Bioavailability of Cephradine in Normal Subjects, J. Clin. Pharmacol. 14:604-611, 1974.

47.

Melander, A., Danielson, K., Schersten, B., Wahlin, E., Clin. Pharmacol. Ther., 22:108-112, 1977.

48.

Hartshorn, E.A., and Tatro, D.S., Principles of Drug Interactions, in Drug Interaction Facts, Olin, B.R., ed.,
Facts and Comparisons, St. Louis, Missouri, 1993.

49.

Hansten, P.D. and Horn, J.R., Drug Interactions and Updates, Lea & Febiger, Malvern, Pennsylvania, 1-27,
1990.

50.

Neuvonen, P.J. and Turakka, H., Inhibitory Effect of Various Iron Salts on the Absorption of Tetracycline in
Man, Eur. J. Clin. Pharmacol., 7:357-360, 1974.

51.

McGilveray, I., Consensus Report on Issues in the Evaluation of Bioavailability, Pharm.Res., 8:136-138, 1991.

52.

Endrenyi, L., Fritsch, S. and Yan, W., Cmax/AUC is a Clearer Measure than Cmax for Absorption Rates in Investigation of
Bioequivalence, Int. J. Clin. Pharmacol. Therap. Toxicol., 29:394-399, 1991.

53.

Aarons, L., Assessment of Rate of Absorption in Bioequivalence Studies, J. Pharm. Sci., 76:853-855, 1987.

54.

Steinijans, V.W., Sauter, R., Jonkman, J.H.G., Schulz, H.U., Stricker, H., and Blume, H., Bioequivalence Studies: Single vs. Multiple Dose, Int. J. Clin. Pharmacol. Therap. Toxicol., 27:261-266, 1989.

55.

Gibaldi, M., Biopharmaceutics and Clinical Pharmacokinetics, Fourth Edition, Lea & Febiger, Malvern, Pennsylvania, p. 20, 151-153, 1990.

56.

Abdou, H.M., Dissolution, Bioavailability, and Bioequivalence, Mack Publishing Company, Easton, Pennsylvania, p. 405-411, 1989.

57.

Notari, R.E., Biopharmaceutics and Clinical Pharmacokinetics, Marcel DekkerInc., New York, New York, p.
189, 1987.

58.

Wartak, J., Clinical Pharmacokinetics, Praeger, New York, New York, p. 154, 1983.

59.

Tse, F.L.S., Robinson, W.T. and Choc, M.G. Study Design for the Assessment of Bioavailability and
Bioequivalence in Pharmaceutical Bioequivalence, Welling, A.G., Tse, F.L.S. and Dighe, S.V., ed., Marcel
Dekker Inc., New York, New York, p. 17-34, 1991.

60.

Junginger, H., Studies on Bioavailability and Bioequivalence APV Guideline, Drugs Made in Germany,
30:161-166, 1987.

61.

Schulz, H.U. and Steinijans, V.W., Striving for Standards in Bioequivalence Assessment: a Review, Int. J. Clin.
Pharmacol. Ther. Toxicol., 29, 293-298, 1991.

62.

Jackson, A.J., Prediction of Steady State Bioequivalence Relationships Using Single Dose Data II - Nonlinear
Kinetics, Bioph. Drug Disp., 10:489-503, 1989.

63.

Ueda, C.T., Essentials of Bioavailability and Bioequivalence, Concepts in Clinical Pharmacology.


Upjohn Company, p. 11, 1979.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

The

8-105

Bioavailability, Bioequivalence, and Drug Selection

64.

Guidelines on the design of a single-dose in-vivo bioavailability study, Bioavailability and Bioequivalence
Requirements, 21 CFR 320: 26, 1991.

65.

In-vitro/In-vivo Correlation for Extended-Release Oral Dosage Forms, Stimuli to the Revision Process, The
United States Pharmacopeial Convention, Inc., p. 4160, 1988.

66.

USP XXII/NF XVII, United States Pharmacopeial Convention, Inc., Rockville, Maryland, p. 1578-1579, 1990.

67.

Banakar, U.V., Factors that Influence Dissolution Testing, in Pharmaceutical Dissolution Testing, Marcel Dekker, Inc., New York, New York, p. 135, 1992.

68.

Welling, P.G., In-Vitro Methods to Determine Bioavailability: In Vitro-In-Vivo Correlations, in Pharmaceutical


Bioequivalence, Welling, P.G., Tse, F.L.S. and Dighe, S.V., ed., Marcel Dekker, Inc., New York, New York, p.
224, 1991.

69.

Httenrauch, R. and Speiser, P., In Vitro-In Vivo Correlation: An Unrealistic Problem, Pharm. Res., 2:97-107,
1985.

70.

PMA's Joint Committee on Bioavailability, The Role of Dissolution Testing in Drug Quality, Bioavailability,
and Bioequivalence Testing, Pharm. Technol., 9:62-66, 1985.

71.

Welling, P.G., In Vitro Methods to Determine Bioavailability: In Vitro-In Vivo Correlations, in Pharmaceutical
Bioequivalence, Welling, P.G., Tse, F.L.S., and Dighe, S.V., ed., Marcel Dekker, Inc., New York, New York, p.
225- 232, 1991.

72.

Sullivan, T.J., Sakmar, E. and Wagner, J.G., Comparative Bioavailability: A New Type of In Vitro-In Vivo Correlation Exemplified by Prednisone, J. Pharmacokin. Biopharm., 4:173-181, 1976.

73.

Yau, M.K.T. and Meyer, M.C., In Vivo-In Vitro Correlations With a Commercial Dissolution Simulator I: Methenamine, Nitrofurantoin, and Chlorothiazide, J. Pharm. Sci., 70:1017-1023, 1981.

74.

Milsap, R.L., Ayres, J.W., Mackichan, J.J., and Wagner, J.G., Comparison of Two Dissolution Apparatuses
with Correlations of In Vitro-In Vivo Data for Prednisone and Prednisolone Tablets, Biopharm. Drug Disp.,
1:3-17, 1979.

75.

Levy, G., Leonards, J. R. and Procknal, J.A., Development of In Vitro Dissolution Tests Which Correlate Quantitatively with Dissolution Rate- Limited Drug Absorption in Man., J. Pharm. Sci., 54:1719-1722, 1965.

76.

Yau, M.K.T. and Meyer, M.C., In Vivo-In Vitro Correlations with a Commercial Dissolution Simulator II:
Papaverine, Phenytoin, and Sulfisoxazole, J. Pharm. Sci., 72:681-686, 1983.

77.

Shah, V.P., Prasad, V.K., Alston, T., Cabana, B.E., Gural, R.P. and Meyer, M.C., Phenytoin I: In Vitro-In Vivo
Correlation for 100 mg. Phenytoin Sodium Capsules, J. Pharm. Sci., 72:306-308, 1983.

78.

Shah, A.C. and Dakkuri, A., Correlation of In Vitro Rate of Dissolution with In Vivo Bioavailability: An Overview, Pharm. Technol., Sep:67-97, 1982.

79.

Wagner, J.G., Rate of Dissolution In Vitro and In Vivo Part VI: Correlation of In Vivo with In Vitro Data - Theoretical and Practical Considerations, Drug Intell. Clin. Pharm., 4:160-163, 1970.

80.

Banakar, U. and Block, L. Beyond Bioavailability, Pharm. Technol., 7:107, 1983.

81.

Abdou, H.M., Dissolution, Bioavailability and Bioequivalence, Mack Publishing Company, Easton, Pennsylvania, p. 508-511, 1989.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-106

Bioavailability, Bioequivalence, and Drug Selection

82.

Banakar, U.V., Pharmaceutical Dissolution Testing, Marcel Dekker, Inc., New York, New York, p. 358-382,
1992.

83.

Hartley, R., Aleksandrowicz, V., Bowmer, C.J., Cawood, A., and Forsythe, W.I., Dissolution and Relative Bioavailability of Two Carbamazepine Preparations for Children with Epilepsy, J. Pharm. Pharmacol.,
43:117-119, 1991.

84.

Mattok, G., McGilveray, I. and Mainville, C., Acetaminophen III: Dissolution Studies of Commercial Tablets
of Acetaminophen and Comparison with In Vivo Absorption Parameters, J. Pharm. Sci., 60:561, 1971.

85.

Stoiropoulus, J., Deutch, T. and Plakogiannis, F., Comparative Bioavailability of Three Commercial Acetaminophen Tablets, J. Pharm. Sci., 70:422, 1981.

86.

Ylitalo, P. and Lundell, G., Bioavailability of Digoxin Tablets in Relation to their Dissolution In Vitro, J.
Pharm. Sci., 7:1264-1266, 1975.

87.

FDA Bioequivalence Task Force Report Conclusions, FDC Reports, February 15, p. 16, 1988.

88.

Kottke, M.K. and Rhodes, C.T., Limitations of Presently Available In Vitro Release Data for the Prediction of
In Vivo Performance, Drug Dev. Ind. Pharm., 17:1157-1176, 1991.

89.

Definitions, Bioavailability and Bioequivalence Requirements, 21 CFR, 320.1, 1991.

90.

Approved Drug Products With Therapeutic Equivalence Evaluations, 14th edition, U.S. Department of Health
and Human Services, Public Health Service, Food and Drug Administration, Center for Drug Evaluation and
Research, Rockville, MD., 1994.

91.

Melnick, D., Hochberg, M. and Oser, B.L., Physiological Availability of the Vitamins I, The Human Bioassay
Technique, J. Nutr., 30:67-79, 1945.

92.

Morrison, A.B., Chapman, D.G. and Campbell, J.A., Futher Studies on the Relation Between In Vitro Disintegration Time of Tablets and the Urinary Excretion Rates of Riboflavin, J. Am. Pharm. Assoc. Sci. Ed.,
48:634-637, 1959.

93.

Morrison, A.B. and Campbell, J.A., The Relationship Between Physiological Availability of Salicylates and
Riboflavin and In Vitro Disintegration Time of Enteric Coated Tablets, J. Am. Pharm. Assoc. Sci. Ed.,
49:473-478, 1960.

94.

Middleton, E.J., Davies, J.M., and Morrison, A.B., Relationship Between Rate of Dissolution, Disintegration
Time, and Physiological Availability of Riboflaving in Sugar-coated Tablets, J. Pharm. Sci., 53:1378-1380,
1964.

95.

Levy, G., Comparison of Dissolution and Absorption Rates of Different Commercial Aspirin Tablets, J.
Pharm. Sci, 50:388-392, 1961.

96.

Levy, G., Effect of Dosage Form Properties on Therapeutic Efficacy of Tolbutamide Tablets, Can. Med. Assoc.
J, 90:978-979, 1964.

97.

MacDonald, H., Pisano, F. and Burger, J., Physiologic Availability of Various Tetracyclines, Clin. Med.,
Dec:30-33, 1969.

98.

Koch-Weser, J., Bioavailability of Drugs, Med. Intell., 291:503-506, 1974.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-107

Bioavailability, Bioequivalence, and Drug Selection

99.

Lindenbaum, J., Mellow, M.H., Blackstone, M.O. and Butler, V.P., Variation in Biologic Availability of
Digoxin from Four Preparations, New Engl. J. Med., 285:1344-1347, 1971.

100.

Report of the Ad Hoc Committee on Drug Product Selection of the Academy of General Practice of Pharmacy
and the Academy of Pharmaceutical Sciences, J. Am. Pharm. Assoc., NS13(6):278-280, 1973.

101.

Office of Technology Assessment, Drug Bioequivalence Study Panel, Drug Bioequivalence, U.S. Government
Printing Office, Washington, D.C., 1974.

102.

Bioavailability and Bioequivalence Requirements, Federal Register, 42:1624- 1653, 1977.

103.

Cabana, B.E., Bioavailability/Bioequivalence, Food Drug Cosm. Law J., 32:512-526, 1977.

104.

Williams, R.L., Bioequivalence and Therapeutic Equivalence, in Pharmaceutical Bioequivalence, Welling,


P.G,. Tse, F.L.S., and Dighe, S.V., eds., Marcel Dekker, Inc., New York, p. 1-14, 1991.

105.

Henderson, J.D., Dighe, S.V., Williams, R.L., Subject Selection and Management in Bioequivalence Studies,
Clin. Research Reg. Affairs, 9:71- 87, 1992.

106.

Cadwallader, D.E., Biopharmaceutics and Drug Interactions, Third Edition, Raven Press, New York, P.
87-106, 1983.

107.

Westlake, W.J., Design and Statistical Evaluation of Bioequivalence Studies in Man., in Principles and Perspectives in Drug Bioavailability, Blanchard, J., Sawchuk, R.J. and Brodie, B.B., ed., Karger, Basel, p.
192-210, 1979.

108.

Dighe, S.V. and Adams, W.P., Bioequivalence: A United States Regulatory Perspective, in Pharmaceutical
Bioequivalence, Welling, P.G., Tse, F.L.S., and Dighe, S.V., eds., Marcel Dekker, Inc., New York, p. 347-380,
1991.

109.

Guidelines on the Design of a Multiple-Dose In-Vivo Bioavailability Study, in Bioavailability and Bioequivalence Requirements, 21 CFR 320.27.

110.

Bio-International '92, Conference on Bioavailability, Bioequivalence and Pharmacokinetics Studies, Pharm.


Res., 10:1806-1811, 1993.

111.

FDA Bioequivalence Task Force Report Conclusions, F-D-C Reports, p. 15, Feb. 15, 1988.

112.

Steinijans, V.W., Hauschke, D., and Jonkman, J.H.G., Contoversies in Bioequivalence Studies, Clin. Pharmacokinet., 22:247-253, 1992.

113.

Consensus Report on "Issues in the Evaluation of Bioavailability", Pharm. Res., 8:136-138, 1991.

114.

Meyer, M.C., Scientific Basis of Bioavailability and Bioequivalence Testing, Am. Pharm., NS31(8):47-52,
1991.

115.

Schuirmann, D.J., A Comparison of the Two One-Sided Tests Procedure and the Power Approach for Assessing the Equivalence of Average Bioavailability, J. Pharmacokinet. Biopharm., 15:657-680, 1987.

116.

Westlake, W.J., Use of Confidence Intervals in Analysis of Comparative Bioavailability Trials, J. Pharm. Sci.,
61:1340-1341, 1972.

117.

Westlake, W.J., Bioavailability and Bioequivalence of Pharmaceutical Formulations, in Biopharmaceutical


Statistics for Drug Development, Peace, K.E., ed., Marcel Dekker, Inc., New York, p. 329-352, 1988.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-108

Bioavailability, Bioequivalence, and Drug Selection

118.

Metzler, C.M., Statistical Criteria, in Pharmaceutical Bioequivalence, Welling, P.G., Tse, F.L.S., and Dighe,
S.V., eds., Marcel Dekker, Inc., New York, p. 35-67, 1991.

119.

Rescigno, A., Bioequivalence, Pharm. Res., 9:925-928, 1992.

120.

Additional Generics on the Market, FDA Drug Bulletin, Nov:14-15, 1986.

121.

Mattison, N., Pharmaceutical Innovation and Generic Drug Competition in the U.S.A.: Effects of the Drug
Price Competition and Patent Term Restoration Act of 1984, Pharmaceut. Med., 1:177-185, 1986.

122.

Madan, P.L., Bioavailability and Bioequivalence, The Underlying Concepts, U.S. Pharm., 17 (Nov. Hosp.
Ed.):H10-H30, 1992.

123.

Haynes, J.D., FDA 75/75 Rule: A Response, J. Pharm. Sci., 72:99-100, 1983.

124.

Anderson, S. and Hauck, W.W., Consideration of Individual Bioequivalence, J. Pharmacokinet. Biopharm.,


18:259-273, 1990.

125.

Carter, M.G. and Sanderson, J.H., Generic Prescribing and Clinical Bioinequivalence, Pharm. J., Nov. 26:683,
1988.

126.

Somberg, J. and Sonnenblick, E., Perspective: The Bioequivalence of Generic Drugs, Cardiovascular Rev.
Rep., 6:1010-1015, 1985.

127.

Gore, M.J., Cost, Safety & Efficacy: Defining the Pharmacist's Role in Drug Product Selection, Consultant
Pharm., 6:771-789, 1991.

128.

Somberg, J.C., Bioequivalence or Therapeutic Equivalence, J. Clin. Pharmacol., 26:1, 1986.

129.

Garrett, E.R., Weinstein, C.B.A., (Commentaries), Integr. Psychiatry, 3:89S- 96S, 1985.

130.

Covington, T.R., Generic Drug Utilization: Overview and Guidelines for Prudent Use, Clin. Research Reg.
Affairs, 9:103-126, 1992.

131.

Shah, H.K., Generics Capture New Prescription Markets, Perspectives in Pharmacy Economics, 4:3, 1992.

132.

Glaser, M., On the Move Again, Drug Topics Supplement, 6S-12S, 1993.

133.

Generic Survey, Amer. Druggist, 208(3):36-41, 1993.

134.

Major Events During the Generic Drug Investigations, Am. Pharm., NS30(7):38-39, 1990.

135.

Drugs: Still Safe?, Consumer Reports, May:310-313. 1990.

136.

Conlan, M.F., More Charges Coming in Generic Industry Probe, Drug Topics, 135(2):56-57, 1991.

137.

Yorke, J., FDA Ensures Equivalence of Generic Drugs, FDA Consumer, Sept:11-15, 1992.

138.

Thompson, L.R., After the Scandals: New Generic Counseling, Am. Pharm., NS30(7):31-33, 1990.

139.

Conlan, M.F., After the Storm, Drug Topics, 135(12):40-44, 1991.

140.

Martin, S., Generic Drug Scandals Raise Questions About Safety, Am. Pharm., NS29(10):23-24, 1989.

141.

Spalding, B.J., The Generic Industry: One Year Later, Am. Drug., 202(3):14- 16, 1990.

142.

Loudin, A., Fallout From the Generic Drug Scandal, Pharm. Update, 2(5):1-9, 1991.

143.

Pal, S. and D'Angelo, A.C., More Patient Questions, More Brand Name Rxs, U.S. Pharm., 15(3):66-70, 1990.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-109

Bioavailability, Bioequivalence, and Drug Selection

144.

Consumer Confidence in Generic Drug Products Down in Wake of Industry Scandal, But Satisfaction with
Products Remains High, Survey Reveals, A.J.H.P., 47:468, 1990.

145.

Pal, S. and D'Angelo, A.C., FDA Reputation Suffers, Pharmacists Ask for More Product Information, U.S.
Pharm., 15(4):20-25, 1990.

146.

Gross, L.H., Can Pharmacists, Physicians, and the Public Trust Generics? Am. Druggist, 200(3):25-28, 1989.

147.

Most Generics are Safe, But Approval Process Remains in 'Disrepair', Am. Pharm., NS30(1):14, 1990.

148.

Conlan, M.F., Getting it Back on Track, Drug Topics, 134(3):42-47, 1990.

149.

Cardinale, V., Generics: Where Now? Drug Topics (Supplement):3S, 1993.

150.

Conlan, M.F., Future is Sunny for Generics as Popular Rxs Come Off Patent, Drug Topics, 134 (20):14-19,
1990.

151.

Starr, C., Which Drugs Are Going Off Patent or Losing Exclusivity?, Drug Topics (Supplement):14S-15S,
1993.

152.

Summers, K.H., Counseling Patients Who Take Generic Drug Products, Drug Topics (Supplement):45S-54S,
1993.

153.

Laskoski, G., Generics: Too Good For Their Own Good, Am. Druggist, 208(3):30-35, 1993.

154.

Shafermeyer, K.W., Schondelmeyer, S.W., and Wilson, G.T., The FDA Orange Book: Expectations Versus
Realities, J. Pharm. and Law, 1:13-26, 1992.

155.

Chappell, S.C., Pharmacists Now Control 27% of Dispensing Decisions Involving New Rxs, Pharm. Times,
54(10):55-62, 1988.

156.

Simonsen, L., Rx Dispensing Trends: Pharmacists Make More Selection Decisions, Pharm. Times,
57(10):53-59, 1991.

157.

Simonsen, L., Rx Brand Choice: Pharmacists Decide for 35% of All New Rxs, Pharm. Times, 58(10):92-98,
1992.

158.

Simonsen, L., Generic Prescribing and RPh Substitution Continue to Climb, Pharm. Times, 59(10):29-30,
1993.

159.

D'Angelo, A.C., How To Win Pharmacists and Influence Purchasing, U.S. Pharm., 16(6):35-36, 1991.

160.

Charupatanapong, N. and Rascati, K.L., Pharmacists' Satisfaction With Drug Product Selection Legislation,
Am. Pharm., NS28(10):27-32, 1988.

161.

Smith, M., Monk, M. and Banahan, B., Factors Influencing Substitution Practices, Am. Druggist,
203(5):88-96, 1991.

162.

Segal, R., Wantz, D.L. and Brusadin, R.A., Pharmacists' Decision Making in the Selection of Generic Pharmaceuticals, J. Pharm. Market. Mngment., 4(1):75-91, 1989.

163.

McCormack, J.P., Levine, M. and Miller, P., Bioequivalence: Just the Facts Please, CPJ-RPC, Sep:404-407,
1990.

164.

Guidelines for Pharmacists Performing Product Selection, Pharm. Today, Feb. 2:4-5, 1990.

165.

Gagnon, J.P., Key Factors and Cues for Evaluating Pharmaceutical Manufacturers, Pharm. Times, 56(4):45-48,
1990.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-110

Bioavailability, Bioequivalence, and Drug Selection

166.

Feinberg, J.L., A Pharmacist's Survival Guide to the Generic Drug Scandal, Consult. Pharm., 5(1):15-23,
1990.

167.

Foster, T.S., Selecting Therapeutically Equivalent Products: Special Cases, Am. Pharm., NS31(11):49-54,
1991.

168.

Ross, M.B., Status of Generic Substitution: Problematic Drug Classes Reviewed, Hosp. Formul., 24:441-449,
1989.

169.

Sanderson, J.H. and Lewis, J.A., Differences in Side-Effect Incidence in Patients on Proprietary and Generic
Propranolol, The Lancet, Apr. 26:967- 968, 1986.

170.

Ansbacher, R., Conjugated Estrogens: Do Not Substitute, Am. Pharm., NS30(7):27-28, 1990.

171.

Locniskar, A., Greenblatt, D.J., Harmatz, J.S., and Shader, R.I., Bioinequivalence of a Generic Brand of Diazepam, Biopharm. Drug Disp., 10:597-605, 1989.

172.

Rentmeester, T.W., Doelman, J.C., and Hulsman, J.A.R.J., Carbamazepine: Merkpreparaat en Generiek,
Pharm. Weekbl., 125(43):1108-1110, 1990.

173.

Welty, T.E., Pickering, P.R., Hale, B.C. and Arazi, R., Loss of Seizure Control With Generic Substitution of
Carbamazepine, Annals Pharmacotherapy, 26:775-776, 1992.

174.

Grubb, B.P., Recurrence of Ventricular Tachycardia After Conversion From Proprietary to Generic Procainamide, Am. J. Cardiol., 63:1532-1533, 1989.

175.

Cunha, B.A., Nitrofurantoin - Bioavailability and Therapeutic Equivalence, Adv. Therapy, 5(3):54-63, 1988.

176.

Nuwer, M.R., Correspondence, Neurology, 41:1165, 1991.

177.

Check, W.A., Caution Urged in Prescribing of Generic Antiarrhythmic Drugs, Consult. Pharm.,
5(11):718-721, 1990.

178.

Fincham, J.E., Therapeutic Failure with Generic Hydrochlorothiazide- Triamterene in an Elderly Female: A
Case Report, J. Ger. Drug Therapy, 5(4):85-89, 1991.

179.

Fincham, J.E., Therapeutic Failure with Generic Clonidine in An Elderly Female: A Case Report, J. Ger. Drug
Therapy, 3(1):83-87, 1988.

180.

Ansbacher, R., Bioequivalence of Conjugated Estrogen Products, Clin. Pharmacokinet., 24(4):271-274, 1993.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

8-111

CHAPTER 9

Clearance

OBJECTIVES
1.

Given patient information regarding organ function, the student will calculate (III)
changes in clearance and other pharmacokinetic parameters inherent in compromised patients.

2.

Determine the total clearance based on Dose and AUC.

3.

Determine clearance of an organ based on dose, AUC, and fraction of drug eliminated by the organ

4.

Determine change in clearance due to functional changes in an organ.

5.

Determine change in clearance due to change in blood flow through an organ.

6.

Prepare a professional consult (V) and justify (VI) modifications in drug therapy
based on clearance of a drug.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-1

Clearance

9.1 Equations
Rate of Elimination
Cl = --------------------------------------------------Serum Concentration

(EQ 9-1)

D os
Cl tot = f--------------AUC

(EQ 9-2)

Cl r = Cl tot ( Fraction of drug that is renally eliminated )

(EQ 9-3)

Cl H = Cl tot ( Fraction of drug that is hepatically eliminated )

(EQ 9-4)

L
min
L
Q r = 0.0191 -------------------- renal blood perfusion 70kg 60 --------- 80 -----blood
min kg
hr
hr

(EQ 9-5)

L
min
L
Q H = 0.0238 -------------------- hepatic blood perfusion 70kg 60 --------- 100 -----blood
min kg
hr
hr

(EQ 9-6)

Er = ( Cl r ) Q r

(EQ 9-7)

E H = ( Cl H ) Q H

(EQ 9-8)

Q Cl
f u Cl int = ---------------Q Cl

(EQ 9-9)

Cl int

Q Cl--------------Q
Cl
= ---------------fu

(EQ 9-10)

f u Cl int
F i = ------------------------f u Cl int

(EQ 9-11)

FR = Q
------Q

(EQ 9-12)

Fi FR
F Cl = ----------------------------------------F R + Er ( Fi FR )

(EQ 9-13)

H + Cl r
Cl tot
k V = Cl
FCl tot = ------------- = -------------------------------------------Cl tot
k V
Cl H + Cl r

(EQ 9-14)

0.80 FCl t o t 1.20

(EQ 9-15)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-2

Clearance

9.2 Definitions and Terms


Clearance:
The hypothetical volume of a fluid from which a substance is
totally and irreversibly removed per unit time.
Dimensions:

L T

Examples of fluids: blood, serum, plasma, bile, gut contents, CSF.


Systemic or Total Body Clearance: Removal process is elimination
(excretion and metabolism). Fluid is usually plasma or serum (rarely blood).
( Cl ) , ( Cl tot )

Renal Clearance: Removal process is urinary excretion of


unchanged drug. Fluid is usually plasma or serum (rarely blood).
( Cl r )

Metabolic Clearance: Removal process is metabolism. Fluid is usually blood (rarely plasma or serum).
( Cl m )

( Cl H )

Hepatic Clearance: This is

Cl m

when the liver is the metabolic

organ.
Creatinine Clearance: This is Cl r applied to endogenous creatinine.
It is used to monitor renal function, and thus is a valuable parameter for calculating
dosage regimens in elderly patients or those suffering from renal dysfunction. Creatinine t 1 2 = 231min
( Cl cr )

Value for normal males: 117 20 ml/min


Value for normal females: 108 20 ml/min
Cl inulin

Inulin Clearance: This is

Cl r

for inulin, and yields the glomerular

filtration rate.
Value for normal males: 124.5 9.7 ml/min
Value for normal females: 108.8 13.5 ml/min

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-3

Clearance

9.3 Measurement of Creatinine Clearance


The mass of endogenous creatinine excreted into the urine collected over a given
time interval ( t ) is determined. The mean serum creatinine concentration ( Cs ) cr
over that interval is calculated from sample determinations; this should be the concentration halfway through the interval. In practice, t = 24hr and, as ( C s ) cr is relatively constant, the serum sample is taken at any convenient time.
Let a be a volume of serum having a creatinine concentration of ( C s ) cr . The mass
of creatinine in the serum will be a ( Cs ) cr . If this creatinine is totally and irreversibly removed from the serum to the urine in the time interval, t , then
a ( C s ) cr = Xu

(EQ 9-16)

Xu
a = -------------( C s ) cr

(EQ 9-17)

Thus,

The volume of serum from which this creatinine is removed in unit time is
this is the definition of clearance. Hence,
( Xu t )
a = -------------------------T
Cl cr = ----t
( C s ) cr

a t ;

(EQ 9-18)

Siersbaek-Neilson et al. report a value of 11.1 g ml for ( Cs ) cr in 149 males (aged


20-99). The value of ( X u t ) T decreased with age from 16.53 g min per Kg body
weight (age 20-29) to 6.53

min

per Kg body weight (age 90-99). For a 25 year

old 70Kg male, equation 9-18 yields

ml
Cl cr = 104.2 --------min

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-4

Clearance

9.4 Model Correlations


Although intrinsically model independent, clearance can also be related to compartmental models.

9.4.1

RENAL CLEARANCE
The plasma renal clearance of a drug may be measured analogously to creatinine
clearance:
( Xu t )
Cl r = --------------------------TCp

(EQ 9-19)

The practical versions of and states:


( Xu t )

= k u X = k u VC p

(EQ 9-20)

Comparing equation 9-19 and equation 9-20,


Cl r = k u V

(EQ 9-21)

This relates clearance to model parameters. What is the slope of a plot of ( Xu t ) T


against

Cp ?

Note that if Cl r > 117 20ml min (males), it may indicate active secretion of the drug
into the kidney tubules. If Cl r < 108 20ml min (females), it may indicate reabsorption of the drug from the kidney tubules.

9.4.2

SYSTEMIC CLEARANCE AND METABOLIC CLEARANCE

How could you measure


Cl ? and Cl m

By analogy,
0.693V
Cl tot = KV = ----------------t1 2

and

K = ku + km

, so

Cl tot = Cl r + Cl m

(EQ 9-22)

Consequently, fractional changes in clearance,


Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-5

Clearance

F Cl

tot

Cl h + Cl r
K V
= ------------------------ = ---------------KV
Cl h + Cl r

(EQ 9-23)

where X is new or altered variable. Hepatic function and renal function are not a
priori connected, although some physiological functional changes might result in
similar changes in clearance of both organs. We can see from equation 9-12 that
changes in total body clearance can result in changes in either K, V, or both. The
consequences of that will be discussed in the section on dosage regimens.

9.4.3

USE IN PHARMACOKINETIC EQUATIONS


Systemic Clearance (Cl) can be used in many equations where the drug is removed
by elimination (renal excretion and metabolism). If renal excretion is the only
removal process, use Cl r l if metabolism, use Cl m . Some examples:
Intravenous infusion:

( Cp )

ss

Q= ----Cl

Oral and Intravenous Bolus:

f( Xa)
fD
fD
Cl = ---------------0 = --------------- = -----------

AUC
C p dt C p dt
0

This equation becomes a means of calculating Cl from plasma data.


Dosage Regimen: ( Cp ) ss

fD = ----------- Cl

These examples are all model-independent expressions, which are very useful in
calculating dosage regimens. The importance of clearance terms rests on their ability to account for variations in both t 1 2 and / or V simultaneously, as both these
parameters can change in disease states and with age.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-6

Clearance

9.5 Physiological Factors Affecting Clearance


9.5.1

INTRINSIC CLEARANCE ( Cl int )


Intuitively, it may be recognized that two factors will affect the clearance of a
drug:
1. The rate at which blood is presented to the eliminating organ.
2. The intrinsic ability of the eliminating organ to clear the drug.
Mathematically, a hyperbolic equation has been derived to illustrate the relative
effect of these factors. (Note: this is one model of clearance. There are several others which also illustrate the effect of these factors.)

Liver Drug Metabolism

QH f u ( Cl H ) int
Cl H = -----------------------------------------Q H + f u ( Cl H ) int

(EQ 9-24)

Where Q H is the rate of blood flow through the liver (assumed 23.8 ml/min/Kg
body weight in normal adult),
fu is the fraction unbound of the drug, and
( Cl H )

int

is the intrinsic hepatic clearance of the drug.

If there were no physiological limits to the rate of blood flow( Q H ), hence equation 9-24 becomes
Cl H = ( Cl H ) int

(EQ 9-25)

This equation provides a definition for intrinsic clearance, namely the clearance of
a drug were there to be no physiological limits on the rate of blood flow through
the clearing organ.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-7

Clearance

Kidney drug excretion

By analogy for excretion of unchanged drug by the kidney:


Q r f u ( Cl r ) int
Cl r = --------------------------------------Q r + fu ( Cl r ) int

(EQ 9-26)

Where Q r is the rate of blood flow through the kidney (assumed 19.1 ml/min per
Kg body weight in normal adults), and
( Cl r )

int

is the intrinsic renal clearance of the drug.

Note that the value of Cl cr (assumed 1.75 ml.min per Kg body weight in normal
adults) is about 9% of Q r .

9.5.2

EXTRACTION RATIO (E)


This is defined as the ratio of the clearance of a drug compared to the rate of
blood flow through the clearing organ. As such, it indicates what fraction of the
drug in the blood is cleared (extracted) on each passage through the clearing organ.
Note: when using clearance to calculate extraction ratio, blood flow must be used.

Drug metabolism by the


liver

Cl
E H = --------HQH
Where

EH

(EQ 9-27)

is the steady-state hepatic extraction ratio.

By comparison with equation 9-24,


f u ( Cl H ) int
E H = -----------------------------------------Q H + f u ( Cl H ) int

(EQ 9-28)

Thus, the range of values of E H is from zero, when ( Cl H ) int = 0 , to one, when
Q H = 0 or ( Cl H ) int Q H . For example, propanolol has E H = 0.75 , yielding
17.9ml
Cl H = ----------------------------------------------------min Kg body weight

and

( Cl H )

int

71.4ml
= ----------------------------------------------------min Kg body weight

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

in normal adult males.

9-8

Clearance

Kidney excretion of
unchanged drug

where

Er

Cl
Er = --------r
Qr

(EQ 9-29)

f u ( Cl r ) int
Er = --------------------------------------Q r + f u ( Cl r ) int

(EQ 9-30)

is the steady-state renal extraction ratio.

Thus the range of values is from zero, when


( Cl r )

and

int

Qr

( Cl r )

. For example, digoxin has

int

1.89ml
= ----------------------------------------------------min Kg body weight

( Cl r )

E r = 0.09

int

= 0

, to one, when

, yielding

Qr = 0

or

1.72ml
Cl r = ---------------------------------------------------min Kg body weight

in normal adult males. In this case, note that

Cl r Cl c r

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-9

Clearance

9.6 Hepatic Function and Clearance


9.6.1

ALTERATIONS IN HEPATIC BLOOD FLOW


For a given drug, equation 9-24 predicts that alterations in the hepatic blood perfusion rate will cause a change in drug clearance, assuming the intrinsic hepatic
clearance is unaltered. A general equation may be derived relating the ratio of
hepatic clearances at two blood perfusion rates to the fractional change in perfusion rate and the extraction ratio of the drug.
FR
Cl
-----------H- = ---------------------------------------Cl H
F R + EH ( 1 F R )
where
Cl H

Cl H

(EQ 9-31)

denotes normal hepatic clearance,

denotes altered hepatic clearance

Q H
F R = ---------QH

is the new flow rate over the old flow rate, the fractional change in blood

perfusion rate, and


EH

is the hepatic extraction ratio under normal conditions.

The equation predicts that, for any given decrease in blood perfusion rate, drugs
having a large normal extraction ratio will experience a proportionally greater
reduction in clearance than drugs having a small normal extraction ratio.
Liver blood flow can be reduced by congestive heart failure, for example. The
intrinsic hepatic clearance can be represented by the inherent activity of the
enzymes responsible for drug metabolism.

9.6.2

ALTERATIONS IN HEPATIC INTRINSIC CLEARANCE


For any given drug, equation 9-24 predicts that alterations in the intrinsic hepatic
clearance will cause a change in drug clearance, assuming the blood flow rate is
unchanged. A general equation may be derived relating the ratio of hepatic clearance at two intrinsic hepatic clearances to the fractional change in intrinsic hepatic
and the extraction ratio of the drug.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-10

Clearance

Fi
Cl
-----------H- = ----------------------------------1 + EH ( F i 1 )
Cl H
where

f u ( Cl int )
F i = ------------------------------f u ( Cl int )

(EQ 9-32)

is the fractional change in fraction unbound times the frac-

tional change in intrinsic hepatic clearance.


The equation predicts that, for any given decrease in intrinsic hepatic clearance,
drugs having a small normal extraction ratio will experience a proportionately
greater reduction in clearance than drugs having a large normal extraction ratio.
The intrinsic hepatic clearance of a drug can be reduced by cirrhosis or increased
by enzyme inducers, such as phenobarbitol.

9.6.3

TABULATED OR GRAPHICAL ALTERATIONS


A table or graph of clearance changes when the hepatic blood flow (but not the
intrinsic hepatic clearance) is altered shows that drugs having a low extraction
ratio ( EH = 0.1 ) need little adjustment in dosage. Even if the hepatic blood flow
were halved ( FR = 0.5 ) , the hepatic clearance is still 91% of its normal value. Conversely, dosage adjustment is necessary for drugs having a high extraction ratio
and predominantly eliminated by hepatic metabolism (e.g., propanolol).
A table or graph of clearance changes when the intrinsic hepatic clearance (but not
the hepatic blood flow) is altered shows that drugs having a high extraction ratio
( E H = 0.9 ) need little adjustment in dosage. Even if the intrinsic hepatic clearance
were halved ( Fi = 0.5 ) , the hepatic clearance is still 91% of its normal value. Conversely, dosage adjustment is necessary for drugs having a low extraction ratio and
predominately eliminated by hepatic metabolism (e.g., phenylbutazone).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-11

Clearance

9.7 Renal Function and Clearance


Approximately 25% of cardiac output goes to the kidneys or approximately 735
ml/min of plasma is presented to the kidneys of a 70 kg man (19.1 mL/min/kg x 70
kg). Approximately 125 ml/min (1.8 mL/min/kg) of that goes to the glomeruli for
filtration (Glomerular Filtration Rate, GFR). Unbound drug is filtered into the
proximal renal tubule at this point. The remaining plasma (as blood) is shunted
around the tubule in the arterioles adjascent to the proximal tubule where drug
may be actively secreted from the arteriol into the proximal tubule or actively
reabsorbed in the opposite direction. As the blood flows down the vessels adjascent to the loop of Henle, the drug may be also passively reabsorbed into the blood
vessel as the water in the urine is being reabsorbed and the urine is being concentrated.
This leads to some interesting possibilities:
1.

Cl r = f u GFR . It is likely that the drug is filtered only, in this case, . It is also possible that

secretion and reabsortion balance and cancel each orther out but are still occurring. The actual
clearance of the drug may be low as the drug may be bound to plasma protiens or red blood
cells.
11. Cl r > f u GFR .

Net active secretion is infered in this case. These active mechanisms are non-

specific and consequently, drugs actively secreted compete with each other. Secretion, if it
occurs, occurs on the unbound drug and thus is also effected by changes in free fraction. In
cases where secretion is very rapid and as a consequence, virtually all of the drug is removed by
the single pass through the kidney (Er ~1), the disssociation of the drug from the protien or out
of the red blood cells is not a hinderance. Some reabsorption may occur but it is less than secretion.
12. Cl r < f u GFR .

Net active reabsorbtion is infered in this case. Active reabsorption occurs for

many exogenous compounds, including glucose and vitamins. For many compounds, reabsorption is passive, occurring only as a consequence of the concentration gradient produced as water
is removed from the urine as is proceeds down the renal tubule. Since the membrane is lipoidal
in nature, polar compounds, ionized acids and ionized bases are less likely to be reabsorbed.
Thus changing the pH of the urine would result in changing the reabsorption characteristics of
weakly acidic or basic drugs.

For low molecular weight drugs (<2,000 dalton) , filtration always occurs. Active
secretion, active reabsorption and passive reabsorption may occur.
It has been found that renal blood flow is little affected by changes in blood flow
elsewhere. However, in chronic renal dysfunction there are two effects which
exhibit a parallel decline. One is a decrease in glomerular filtration rate (GFR), as
measured by Cl cr , and the other is the net secretion of drugs into the kidney
tubules. Note that p-amino hippurate (PAH) clearance measures the sum of both
effects.
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-12

Clearance

For any given drug, equation 9-26 predicts that alterations in both the renal blood
perfusion rate (as manifest by the GFR) and the intrinsic renal clearance will cause
a change in drug clearance. A general equation may be derived relating the ratio of
renal clearances at two different blood flow rates and two different intrinsic renal
clearances.
Cl
-----------r = F i
Cl r

(EQ 9-33)

where F i is the fractional change in drug unbound times the fractional change in
intrinsic renal clearance.
In this case,
f u
F i = ------F
fu
R
where

FR

(EQ 9-34)

is the fractional change in blood flow rate (or GFR).

Thus, equation 9-33 shows that the renal clearance of a drug is reduced by a constant fraction, independent of the renal extraction ratio ( E r ) . This fractional
decrease can be estimated by changes in creatinine clearance:
Cl cr
FR = -----------Cl cr
where

Cl cr

(EQ 9-35)

is the altered creatinine clearance

Substituting, equation 9-33 through equation 9-35, we get:


Cl r
Cl cr
----------- = -----------Cl r
Cl cr

(EQ 9-36)

This equation shows why, in cases of chronic renal dysfunction, a change in the
measured creatinine clearance indicated a likely change in drug renal clearance.
Hence, dosage adjustments are made on this basis, particularly for drugs predominantly eliminated by renal filtration (e.g., gentamicin, digoxin).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-13

Clearance

9.8 General Equations for Changes in Clearance


For each clearing organ,
FI FR
Cl
F Cl = ------ = --------------------------------------Cl
F R + E ( F I FR )

(EQ 9-37)

When a drug has a high extraction ratio, E 1 , then equation 9-26 becomes
F Cl F R

(EQ 9-38)

and when a drug has a low extraction ratio, E 0 , then equation 9-26 becomes
F Cl F I

(EQ 9-39)

Thus, the clearance of drugs with a high extraction ratio are more effected by physiological changes in flow of blood to the clearing organ, while drugs with a low
extraction ratio are more effected by physiological changes in the function of the
organ.

9.8.1

PLASMA/BLOOD RATIO
Calculation of Extraction Ratio requires measurement in whole blood by definition. Since most clinical measurements are done in plasma, knowledge of the
plasma/blood ratio is necessary. Blood in made up of plasma and red blood cells
(RBCs). Thus the amount of drug in the blood is made up of the amount of drug in
the plasma and the amount of drug in the RBCs.
C b Vb = C p Vp + C rbc V rbc
where

(EQ 9-40)

C x V x = AMOUNT x

and

b = blood, p = plasma, rbc = red blood cell

If we define the ratio of the concentration of the drug in the RBCs to the concentration of the free drug in plasma as

C rbc
= -------------fu C p

(EQ 9-41)

and
V rbc = H V b

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 9-42)

9-14

Clearance

Vp = ( 1 H) Vb

(EQ 9-43)

Pluging equation 9-41 thru equation 9-43 into equation 9-40 results in:
Cb Vb = ( 1 H ) V b Cp + f u H Vb Cp

(EQ 9-44)

Rearranging and simplifying results in:


Cb
------ = 1 + H ( fu 1 )
Cp

H 1 + ( Cb C p )
= ----------------------------------------fu H

(EQ 9-45)

(EQ 9-46)

Thus equation 9-46 determines the affinity of the drug for the RBCs. Drugs with a
high affinity for the RBCs should result in a smaller volume of distribution.
For drugs that are primarily filtered by the glomeruli, the renal extraction ratio is:
GFR f u C p
Rate of filtration - = ------------------------------E rf = ----------------------------------------------Rate of presentation
Qr Cb

(EQ 9-47)

Putting equation 9-45 into equation 9-47 results in:


GFR f u
E rf = ----------------------------------------------------------Q r ( 1 + H ( fu 1 ) )

(EQ 9-48)

Thus:
1.

Cl
Qr

If the the ratio of --------r to calculated Erf from equation 9-48 is one, it is likely that the drug is
filtered only.

13.

Cl

If the the ratio of --------r to calculated Erf from equation 9-48 is greater than one, active secretion
Qr

is infered in this case.


14.

Cl

If the the ratio of --------r to calculated Erf from equation 9-48 is less than one, active reabsorbQr

tion is infered in this case.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-15

Clearance

9.8.2

HALF LIFE AND ELIMINATION RATE CONSTANT IN RELATIONSHIP


TO CLEARANCE
The elimination rate constant is related to the volume of distribution and the total
body clearance by equation 9-22 above which when rewritten yields:
Rate of Elimination
( Mass ) ( Time )
Cl
( Volume ) ( Time )
K = ------------------------------------------------ = ---------------------------------------- = ------ = ---------------------------------------------Amount in the body
Mass
V
Volume

(EQ 9-49)

Cl pw
Cl
K = Cl
------ = -------b- = -------------u
V
Vb
Vpw u

(EQ 9-50)

where b = blood and pwu = unbound plasma water. Clearance of drug from blood
(Clb) is useful in considering drug extraction in the eliminating organs. Volume
and clearance terms based on unbound drug concentration are particularly useful
in therapeutics, because only the unbound drug is thought to cause the therapeutic
action.

9.8.3

EFFECTS OF ALTERATIONS IN PROTEIN BINDING ON CLEARANCE


Protein binding of drugs may be altered in disease states and by interferance binding by other drugs on the protein. These changes in binding effect Fi , the fractional change in intrinsic clearance in both renal and hepatic clearances even
though the actual intrinsic clearance, the indicator of organ function, may be uneffected as shown in equation 9-11:
f u Cl int
F i = ------------------------f u Cl int
where Cl int = Cl int . Thus, a change in protein binding will cause a proportional
change in Fi of both clearances. There is more discussion in the chapter on protein
binding.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-16

Clearance

9.9 Problems
For each of the problems, do questions A through R now and S through W after
completing chapter 10. In doing questions S through W, please try to obtain a
plasma concentration of free drug within 120 % of normal
Cp

ss
min free

Cp

ss
max free

and 80 % of

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-17

Clearance

Acebutolol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 1)

AHFS 00:00.00
GPI: 0000000000

Piquette-Miller, M., et. al., Effect of aging on the pharmacokinetics of acebutolol enantiomers, Journal of Clinical Pharmacology, Vol. 32, (1992), p. 148 - 156. Kukes, VG; Gneushev ET; Mamedov TS; Gneusheva IA; Acebutolol and diacetolol: thier binding to plasma and erythrocytes and secretion with saliva. Farmakol-Toksikol. 1991 Jan-Feb; 54(1)

Acebutolol is a beta-adrenergic blocking agent which is often used in the treatment of hypertension.(Use Qr = 72 L/hr;
PROBLEM TABLE 9 - 1. Acebutolol

Patient Condition

Normal

Normal

Dose(mg)

200

400

0.4

fu

0.867

1.93

Vd (L)

k (hr-1)

T 1/2 (hr)

5.5

%Clr

40

%Clnr

60

AUC (mg/L*hr)

3.97

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

Cl h

Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

12

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Qh = 90 L/hr)
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-18

Clearance

TABLE 9 - 1. Answers for Acebutolol

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

200

400 BID

400 BID

400 BID

400 BID

200 QID

FRR

0.5

FIR

0.5

0.5

FRH

0.5

FIH

0.5

0.4

fu

0.867

1.93

Vd (L)

160

k (hr-1)

0.126

0.101

0.102

0.12

0.091

T 1/2 (hr)

5.5

6.87

6.77

5.89

7.64

%Clr

40

25

26.1

42.9

55.5

%Clnr

60

75

73.9

57.1

44.6

AUC (mg/L*hr)

3.97

9.93

9.78

8.51

5.5

Cl tot (L/hr)

20.2

16.1

16.4

18.8

14.5

Cl h (L/hr)

12.1

12.1

12.1

10.7

6.5

Cl r (L/hr)

8.1

4.03

4.27

8.1

8.1

Eh

0.126

Er

0.112

0.8

0.81

0.93

0.72

12

12

12

12

2.18

1.75

1.77

2.04

0.785

g
------ mL

1.11

1.24

1.23

1.15

1.03

g
------ mL

0.57

0.72

0.71

0.61

0.80

ss g
-------min free mL

0.25

0.37

0.36

0.28

0.6

P
Q

Cl h
Cl r

int

int

(L/hr)

16

(L/hr)

10.5

FCL

(hr)
N

U
Cp

ss
max free

V
Cp
W
Cp

ss
avg free

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-19

Clearance

Bisoprolol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 2)

AHFS 00:00.00
GPI: 0000000000

Kirch, W., et. al., Pharmacokinetics of bisoprolol during repeated oral administration to healthy volunteers and patients with kidney or liver disease, Clinical Pharmacokinetics, Vol. 13, (1987), p. 110 - 117.

Bisoprolol (comes as 5 and 10 mg tablets) is a - selective adrenergic antagonist. It is used in the treatment of hypertension and angina pectoris.(Use Qr = 72 L/hr; Qh = 90 L/hr)
PROBLEM TABLE 9 - 2. Bisoprolol

Patient Condition

Normal

Normal

Dose(mg)

10

10 TID

0.7

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

10

%Clr

50

%Clnr

50

AUC (mg/L*hr)

0.661

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp
W
Cp

ss g
-------avg free mL
ss

g
------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-20

Clearance

TABLE 9 - 2. Answers for Bisoprolol

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

10

10

10

10

10

10

FRR

0.5

FIR

0.5

0.5

FRH

0.5

FIH

0.5

0.9

fu

0.7

Vd (L)

152.8

k (hr-1)

0.0693

0.052

0.0526

0.067

0.0525

T 1/2 (hr)

10

13.3

13.2

10.3

13.2

%Clr

50

33.3

34

51.3

66

%Clnr

50

66.7

66

48.7

34

AUC (mg/L*hr)

0.661

0.85

0.87

0.69

0.87

Cl tot (L/hr)

10.6

7.94

8.0

10.3

8.0

Cl h (L/hr)

5.3

5.3

5.3

5.0

2.7

Cl r (L/hr)

5.3

2.65

2.8

5.3

5.3

Eh

0.055

Er

0.074

0.75

0.76

0.97

0.76

12

12

12

P
Q

Cl h
Cl r

int

int

(L/hr)

5.6

(L/hr)

5.7

FCL

(hr)
N

U
Cp

0.8

0.9

0.9

0.78

0.91

0.108

0.099

0.098

0.11

0.098

g
------ mL

0.083

0.073

0.073

0.085

0.073

g
------ mL

0.062

0.052

0.052

0.064

0.052

ss g
-------max free mL

V
Cp
W
Cp

ss
avg free
ss
min free

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-21

Clearance

Cefonicid
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 3)

AHFS 00:00.00
GPI: 0000000000

Fillastre, J., et. al., Pharmacokinetics of cefonicid in uraemic patients, Journal of Antimicrobial Chemotherapy, Vol. 18, (1986),
p. 203 - 211.

Cefonicid is a beta-lactamase resistant cephalosporin which is useful in treating many infections caused by Gram-positive and Gram-negative organisms. Cefonicid is 80% renally excreted.(Use Qr = 72 L/hr; Qh = 90 L/hr)
PROBLEM TABLE 9 - 3. Cefonicid

Patient Condition

Normal

Normal

Dose(mg)

1000

1000 TID

fu

0.06

Vd (L)

k (hr-1)

T 1/2 (hr)

5.3

%Clr

80

%Clnr

20

AUC (mg/L*hr)

654

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp
W
Cp

ss g
-------avg free mL
ss

g
------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-22

Clearance

TABLE 9 - 3. Answers for Cefonicid

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

1000

1000

1000

1000

1000

1000

FRR

0.5

FIR

0.5

0.5

FRH

0.5

FIH

0.5

fu

0.06

Vd (L)

11.7

k (hr-1)

0.131

0.078

0.079

0.131

0.118

T 1/2 (hr)

5.3

8.8

8.8

5.3

5.9

%Clr

80

66.7

66.9

80

88.9

%Clnr

20

33.3

33.1

20

11.1

AUC (mg/L*hr)

654

1090

1083

654

727

Cl tot (L/hr)

1.53

0.917

0.93

1.5

1.4

Cl h (L/hr)

0.3

0.31

0.31

0.3

0.15

Cl r (L/hr)

1.22

0.612

0.62

1.22

1.22

Eh

0.0032

Er

0.017

0.6

0.6

0.9

12

12

1.51

1.4

1.4

1.51

1.36

g
------ mL

7.9

8.4

8.4

7.9

8.4

g
------ mL

4.9

5.5

5.4

4.9

5.4

ss g
-------min free mL

2.8

3.3

3.3

2.8

3.3

P
Q

Cl h
Cl r

(L/hr)

5.11

(L/hr)

20.7

int

int

FCL

(hr)
N

U
Cp

ss
max free

V
Cp
W
Cp

ss
avg free

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-23

Clearance

Cefpirome
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 4)

AHFS 00:00.00
GPI: 0000000000

Lameire, N., et. al., Single-dose pharmacokinetics of cefpirome in patients with renal impairment, Clinical Pharmacology and
Therapeutics, Vol. 52, (1992), p. 24 - 30.

Cefpirome is a third-generation, broad-spectrum cephalosporin which is useful against many cephalosporin-resistant


organisms.
PROBLEM TABLE 9 - 4.

Cefpirome

Patient Condition

Normal

Normal

Dose(mg)

2000 IV

2000 TID

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

2.6

%Clr

85

%Clnr

15

AUC (mg/L*hr)

342

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp
W
Cp

ss g
-------avg free mL
ss

g
------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-24

Clearance

TABLE 9 - 4. Answers for Cefpirome

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-25

Clearance

Cefprozil
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 5)

AHFS 00:00.00
GPI: 0000000000

Shyu, W., et. al., Pharmacokinetics of cefprozil in healthy subjects and patients with renal impairment, Journal of Clinical Pharmacology, Vol. 31, (1991), p. 362 - 371.

Cefprozil is a broad-spectrum oral cephalosporin.


PROBLEM TABLE 9 - 5.

Patient Condition

Normal

Dose(mg)

1000

0.95

fu

0.7

Vd (L)

k (hr-1)

T 1/2 (hr)

1.2

%Clr

75

%Clnr

25

AUC (mg/L*hr)

58.1

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Cefprozil

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-26

Clearance

TABLE 9 - 5. Answers for Cefprozil

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-27

Clearance

Chloramphenicol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 6)

AHFS 00:00.00
GPI: 0000000000

Ambrose, P., Clinical pharmacokinetics of chloramphenicol and chloramphenicol succinate, Clinical Pharmacokinetics, Vol. 9,
(1984), p. 222 - 238.

Chloramphenicol succinate is a prodrug which is converted in vivo to the active form, chloramphenicol.
PROBLEM TABLE 9 - 6.

Patient Condition

Normal

Dose(mg)

1000

fu

0.4

Vd (L/kg)

k (hr-1)

T 1/2 (hr)

0.6

%Clr

30

%Clnr

70

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

2.8

(L/hr)

int

FCL

Normal

(L/hr)

int

Chloramphenicol

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-28

Clearance

TABLE 9 - 6. Answers for Chloramphenicol

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-29

Clearance

Enalapril
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 7)

AHFS 00:00.00
GPI: 0000000000

Ohnishi, A., et. al., Kinetics and dynamics of enalapril in patients with liver cirrhosis, Clinical Pharmacology and Therapeutics,
Vol. 45, (1989), p. 657 - 665.

Enalapril is an ACE inhibitor which is a prodrug that is metabolized in the liver to the active metabolite, enalaprilat.
PROBLEM TABLE 9 - 7. Enalapril

Patient Condition

Normal

Normal

Dose(mg)

10 mg po

10 BID1

0.65

fu

0.55

Vd (L)

k (hr-1)

T 1/2 (hr)

0.63

%Clr

27

%Clnr

73

AUC (mg/L*hr)

0.123

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-30

Clearance

TABLE 9 - 7. Answers for Enalapril

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-31

Clearance

Enoxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 8)

AHFS 00:00.00
GPI: 0000000000

Somogyi, A., and Bochner, F., The absorption and disposition of enoxacin in healthy subjects, Journal of Clinical Pharmacology, Vol. 28, (1988), p. 707 - 713.

Enoxacin is a fluorinated quinolone which is used to treat infections caused by gram-negative organisms and
Pseudomonoas aeruginosa.
PROBLEM TABLE 9 - 8.

Enoxacin

Patient Condition

Normal

Normal

Dose(mg)

400

200 bid

0.9

fu

0.8

Vd (L)

k (hr-1)

T 1/2 (hr)

7.75

%Clr

60

%Clnr

40

AUC (mg/L*hr)

15.61

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

12

U
Cp

ss

g
------max free mL

V
Cp
W
Cp

ss g
-------avg free mL
ss

g
------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-32

Clearance

TABLE 9 - 8. Answers for Enoxacin

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-33

Clearance

Enprofylline
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 9)

AHFS 00:00.00
GPI: 0000000000

Nadai, M., et. al, Dose-dependent pharmacokinetics of enprofylline and its renal handling in rats, Journal of Pharmaceutical
Sciences, Vol. 80, No. 7, (1991), p. 648 - 651

Enprogylline is a xanthine bronchodilator which is more potent than theophylline.


Enprofylline

PROBLEM TABLE 9 - 9.

Patient Condition

Normal

Normal

Dose(mg/kg)

2.5

2.5 TID

fu

0.4

Vd (L/kg)

k (hr-1)

T 1/2 (hr)

0.36

%Clr

90

%Clnr

10

AUC (mg/L*hr)

3.6

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

TABLE 9 - 9. Answers for Enprofylline

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-34

Clearance

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-35

Clearance

Erythromycin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 10)

AHFS 00:00.00
GPI: 0000000000

Welling and Creig (JPS 67, 1057-9,1978).

Erythromycin is a macrolide antibiotic.(Use Qr = 72 L/hr; Qh = 90 L/hr)


PROBLEM TABLE 9 - 10.

Erythromycin

Patient Condition

Normal

Normal

Dose(mg)

300

300 TID

.8

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

10

%Clnr

90

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

0.06

0.06

0.12

0.06

0.18

57

57

100

57

150

16.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-36

Clearance

TABLE 9 - 10. Answers for

Erythromycin

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

300

300

300

200

300

200

FRR

0.5

FIR

0.5

0.5

FRH

0.5

FIH

0.5

.8

fu

0.06

0.06

0.12

0.06

0.18

Vd (L)

57

57

100

57

150

k (hr-1)

0.255

0.242

0.235

0.227

0.075

T 1/2 (hr)

2.71

2.85

2.92

3.05

9.27

%Clr

10

5.3

3.1

11.2

37

%Clnr

90

94.7

96.9

88.8

63

AUC (mg/L*hr)

14.5

17.4

6.7

18.5

14.2

Cl tot (L/hr)

16.5

13.8

23.7

13.0

11

Cl h (L/hr)

13.1

13.1

23.4

11.5

7.02

Cl r (L/hr)

1.45

0.73

0.73

1.45

4.19

Eh

0.136

Er

0.020

0.95

1.63

0.89

0.77

24

2.94

2.79

2.74

2.63

2.59

g
------ mL

0.29

0.30

0.23

0.30

0.23

g
------ mL

0.12

0.13

0.10

0.14

0.11

0.038

0.042

0.034

0.049

0.038

P
Q

Cl h
Cl r

(L/hr)

252

(L/hr)

24.7

int

int

FCL

(hr)
N

U
Cp

ss
max free

V
Cp
W
Cp

ss
avg free

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-37

Clearance

Fleroxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 11)

AHFS 00:00.00
GPI: 0000000000

Singlas, E., et. al., Disposition of fleroxacind, a new trifluoroquinolone, and its metabolites - pharmacokinetics in renal failure
and influence of haemodialysis, Clinical Pharmacokinetics, Vol. 19, No. 1, (1990), p. 67 - 79.

Fleroxacin is a trifluorinated quinolone with activity against a variety of gram-negative and gram-positive organisms.
Fleroxacin

PROBLEM TABLE 9 - 11.

Patient Condition

Normal

Normal

Dose(mg)

400

200 tid

0.95

fu

0.5

1.45

Vd (L)

k (hr-1)

T 1/2 (hr)

14

%Clr

65

%Clnr

35

AUC (mg/L*hr)

92

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-38

Clearance

TABLE 9 - 11. Answers for Fleroxacin

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-39

Clearance

Fosinopril
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 12)

AHFS 00:00.00
GPI: 0000000000

Hui, K., et. al., Pharmacokinetics of fosinopril in patients with various degrees of renal function, Journal of Clinical Pharmacology and Therapeutics, Vol. 49, No. 4, (1991), p. 457 - 466.

Fosinopril is an angiotensin converting enzyme inhibitor which is a prodrug that is metabolized to active form, fosinoprilat.
PROBLEM TABLE 9 - 12.

Fosinopril

Patient Condition

Normal

Normal

Dose(mg)

7.5 IV

20 po qd

1 (Oral
0.36)

fu

0.01

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

100

AUC (mg/L*hr)

5.1

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

24

U
Cp

ss

g
------max free mL

V
Cp
W
Cp

ss g
-------avg free mL
ss

g
------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-40

Clearance

TABLE 9 - 12. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Fosinopril

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-41

Clearance

Glutathione
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 13)

AHFS 00:00.00
GPI: 0000000000

Mulders, T., et. al., Characterization of glutathione conjugation in humans: stereoselectivity in plasma elimination pharmacokinetics and urinary excretion of (R)- and (S)-2-bromoisovalerylurea in healthy volunteers, Clinical Pharmacology and Therapeutics, Vol. 53, (1993), p. 49 - 58.

This study explored the pharmacokinetics of glutathione.


Glutathione

PROBLEM TABLE 9 - 13.

Patient Condition

Normal

Normal

Dose(mg)

600

500 tid

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

4.4

%Clr

35

%Clnr

65

AUC (mg/L*hr)

276

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-42

Clearance

TABLE 9 - 13. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Glutathione

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-43

Clearance

Guanadrel
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 14)

AHFS 00:00.00
GPI: 0000000000

Halstenson, C., et. al., Disposition of guanadrel in subjects with normal and impaired renal function, Journal of Clinical Pharmacology, Vol. 29, (1989), p. 128 - 132.

Guanadrel is adrenergic blocker used in the treatment of hypertension.


Guanadrel

PROBLEM TABLE 9 - 14.

Patient Condition

Normal

Normal

Dose(mg)

25 mg po

25 BID

fu

0.8

Vd (L)

k (hr-1)

T 1/2 (hr)

3.7

%Clr

40

%Clnr

60

AUC (mg/L*hr)

0.234

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

12

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-44

Clearance

TABLE 9 - 14. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Guanadrel

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-45

Clearance

Monoxidine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 15)

AHFS 00:00.00
GPI: 0000000000

Kirch, W., Hutt, H., and Plnitz, V., The influence of renal function on clinical pharmacokinetics of monoxidine, Clinical Pharmacokinetics, Vol. 15, (1988), p. 245 - 253.

Monoxidine is a centrally acting antihypertensive agent which stimulates 2 -adrenergic receptors.


PROBLEM TABLE 9 - 15.

Monoxidine

Patient Condition

Normal

Normal

Dose(mg)

0.25 IV

0.25 tid

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

2.75

%Clr

95

%Clnr

AUC (mg/L*hr)

0.05

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-46

Clearance

TABLE 9 - 15. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Monoxidine

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-47

Clearance

Nalmefene
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 16)

AHFS 00:00.00
GPI: 0000000000

Dixon, R., et. al., Nalmefence: safety and kinetics after single and multiple oral doses of a new opiod antagonist, Journal of
Clinical Pharmacology, Vol. 27, (1987), p. 233 - 239.

Nalmefene is a pure opiod antagonist which is currently being investigated for use.
Nalmefene

PROBLEM TABLE 9 - 16.

Patient Condition

Normal

Dose(mg)

20 IV

0.6 oral

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

9.8

%Clr

70

%Clnr

30

AUC (mg/L*hr)

0.3

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FRR=0.5

(L/hr)

int

Normal

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-48

Clearance

TABLE 9 - 16. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Nalmefene

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-49

Clearance

Nitrendipine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 17)

AHFS 00:00.00
GPI: 0000000000

Dylewicz, P, et. al, Bioavailability and elimination of nitrendipine in liver disease, European Journal of Clinical Pharmacology,
Vol, 32, (1987), p. 563 - 568.

Nitrendipine is a calcium antagonist


PROBLEM TABLE 9 - 17.

Nitrendipine

Patient Condition

Normal

Normal

Dose(mg)

5 IV

25 po BID

1 (oral 0.2)

fu

0.05

Vd (L)

k (hr-1)

T 1/2 (hr)

11.7

%Clr

0.1

%Clnr

99.9

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

90

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-50

Clearance

TABLE 9 - 17. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Nitrendipine

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-51

Clearance

Ofloxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 18)

AHFS 00:00.00
GPI: 0000000000

Lamerire, N., et. al., Ofloxacin pharmacokinetics in chronic renal failure and dialysis, Clinical Pharmacokinetics, Vol. 21, No.
4, (1995), p. 357 - 371.
PROBLEM TABLE 9 - 18.

Ofloxacin

Patient Condition

Normal

Normal

Dose(mg)

300

300

0.93

fu

0.74

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

97

%Clnr

AUC (mg/L*hr)

28.47

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

12

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-52

Clearance

TABLE 9 - 18. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Ofloxacin

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-53

Clearance

Omeprazole

(Problem 9 - 19)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

(Use Qr = 72 L/hr; Qh = 90 L/hr)


PROBLEM TABLE 9 - 19.

Omeprazole

Patient Condition

Normal

Normal

Dose(mg)

10

10 BID

0.5

fu

0.04

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

100

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FIH=0.5
0.75

0.06

0.08

30

12

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

TABLE 9 - 19. Answers for

FRH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

Omeprazole

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

10

10

10

10

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-54

Clearance

FRR

0.5

FIR

0.5

0.5

FRH

0.5

FIH

1.5

0.5
0.75

0.5

fu

0.04

Vd (L)

30

k (hr-1)

0.347

0.347

0.493

0.31

0.184

T 1/2 (hr)

1.41

2.2

3.8

%Clr

%Clnr

100

AUC (mg/L*hr)

0.48

0.48

0.338

0.53

0.68

Cl tot (L/hr)

10.4

10.4

14.8

9.38

5.5

Cl h (L/hr)

10.4

10.4

14.8

9.38

5.5

Cl r (L/hr)

Eh

0.108

Er

1.42

0.90

0.53

12

P
Q

Cl h
Cl r

(L/hr)

291.5

(L/hr)

int

int

FCL

0.06

(hr)
N

U
Cp

2.83

2.71

3.2

0.0076

0.0076

0.0051

0.0079

0.011

g
------ mL

0.0032

0.0032

0.0025

0.0035

0.0045

g
------ mL

0.00095

0.00095

0.00099

0.0012

0.0012

ss g
-------max free mL

V
Cp
W
Cp

ss
avg free
ss
min free

0.08

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-55

Clearance

Piperacillin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 20)

AHFS 00:00.00
GPI: 0000000000

Johnson, C., et. al., Single-dose pharmacokinetics of piperacillin and tazobactam in patients with renal disease, Clinical Pharmacology and Therapeutics, Vol. 51, (1992), p. 32 - 41.

Piperacillin is a beta-lactam antibiotic.


Piperacillin

PROBLEM TABLE 9 - 20.

Patient Condition

Normal

Normal

Dose(mg)

3000

2000 qid

fu

0.82

Vd (L)

k (hr-1)

T 1/2 (hr)

0.95

%Clr

75

%Clnr

25

AUC (mg/L*hr)

276

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-56

Clearance

TABLE 9 - 20. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Piperacillin

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-57

Clearance

Piroxicam
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 21)

AHFS 00:00.00
GPI: 0000000000

Boudinot, S., Funderburg, E., and Boudinot, F., Effects of age on the pharmacokinetics of piroxicam in rats, Journal of Pharmaceutical Sciences, Vol. 82, No. 3, (1993), p. 254 - 257.

Piroxicam is a nonsteroidal anti-inflammatory drug (NSAID) commonly used in the treatment of arthritis.
Piroxicam

PROBLEM TABLE 9 - 21.

Patient Condition

Normal

Normal

Dose(mg)

70

20 qd

fu

0.007

Vd (L)

k (hr-1)

T 1/2 (hr)

50

%Clr

%Clnr

95

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-58

Clearance

TABLE 9 - 21. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Piroxicam

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-59

Clearance

Quinidine

(Problem 9 - 22)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Quinidine sulfate is used to treat ventricular and supraventricular arrythmias and is available in 200 and 300 mg tablets.
It is known to bind to -acid glycoprotein (AAG), which is an acute phase reactant. AAG rises in trauma, inflamation,
malignancy and stress and falls in hepatic disease, nephrotic syndrome and malneurtrition for example.(Use Qr = 72 L/
hr; Qh = 90 L/hr)
Quinidine

PROBLEM TABLE 9 - 22.

Patient Condition

Normal

Normal

Dose(mg/kg)

10

10 TID

0.7

fu

0.2

0.83

Vd (L/kg)

2.6

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr/kg)

Cl h (L/hr/kg)

0.20

Cl r (L/hr/kg)

0.056

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

0.10

0.25

0.15

0.3

2.0

2.75

2.2

3.0

6.4

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

1
8

(hr)
N

U
Cp

ss
max free

V
Cp
W
Cp

g
------ mL

MTC
1.7

ss g
-------avg free mL
ss
min free

g
------ mL

MEC
0.37

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-60

Clearance

TABLE 9 - 22. Answers for

Quinidine

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

10

10

10

10

10

FRR

0.5

FIR

0.25

0.5

0.75

1.5

FRH

0.5

FIH

0.5

1.25

0.75

0.5

0.7

fu

0.20

0.10

0.25

0.15

0.30

0.83

Vd (L)

2.6

2.0

2.75

2.2

3.0

k (hr-1)

0.098

0.057

0.101

0.087

0.061

T 1/2 (hr)

12.1

6.9

7.9

11.3

%Clr

22

12.3

10

22

46

%Clnr

78

87.7

90

78

54

AUC (mg/L*hr)

27.3

61.3

20.6

36.5

19.0

Cl tot (L/hr)

0.256

0.114

0.28

0.19

0.18

Cl h (L/hr)

0.20

0.10

0.25

0.15

0.1

Cl r (L/hr)

0.056

0.014

0.028

0.04

0.08

Eh

0.0021

Er

0.00078

0.45

1.1

0.75

0.72

1.3

0.66

1.16

1.0

0.71

0.82

0.79

0.95

0.79

0.75

0.57

0.63

0.65

0.56

0.59

0.37

0.5

0.44

0.39

0.46

P
Q

Cl h
Cl r

(L/hr)

1.0

(L/hr)

0.28

int

int

FCL

(hr)
N

U
Cp

ss
max free

V
Cp
W
Cp

ss
avg free
ss
min free

g
------ mL

MTC
1.7

g
------ mL
g
------ mL

MEC
0.3

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-61

Clearance

Tazobactam
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 23)

AHFS 00:00.00
GPI: 0000000000

Johnson, C., et. al., Single-dose pharmacokinetics of piperacillin and tazobactam in patients with renal disease, Clinical Pharmacology and Therapeutics, Vol. 51, (1992), p. 32 - 41.

Tazobactam is an irreversible beta-lactamase inhibitor.


Tazobactam

PROBLEM TABLE 9 - 23.

Patient Condition

Normal

Normal

Dose(mg)

375 IV

375 qid

fu

0.96

Vd (L)

k (hr-1)

T 1/2 (hr)

0.89

%Clr

68

%Clnr

32

AUC (mg/L*hr)

30.3

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

TABLE 9 - 23. Answers for

Tazobactam

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-62

Clearance

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-63

Clearance

Theophylline
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 24)

AHFS 00:00.00
GPI: 0000000000

Wagner, J., Theophylline - pooled Michaelis-Menten parameters and implications, Clinical Pharmacokinetics, Vol. 10, (1985),
p. 432 - 442.
PROBLEM TABLE 9 - 24. Theophylline
Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-64

Clearance

TABLE 9 - 24. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Theophylline

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-65

Clearance

Tolrestat

(Problem 9 - 25)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Troy, S., et. al., The effect of renal disease on tolrestat pharmacokinetics, Clinical Pharmacology and Therapeutics, Vol. 51,
(1992), p. 271 - 277.

Tolrestat is an aldose reductase inhibitor used in the treatment of diabetic neuropathy, diabetic nephropathy, and diabetic retinopathy. (Use Qr = 72 L/hr; Qh = 90 L/hr)
PROBLEM TABLE 9 - 25.

Tolrestat

Patient Condition

Normal

Normal

Dose(mg)

200

200 tid

0.8

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

10.6

%Clr

25

%Clnr

40 bile 35 metab.

AUC (mg/L*hr)

86

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss

g
------max free mL

V
Cp
W
Cp

ss g
-------avg free mL
ss

g
------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-66

Clearance

TABLE 9 - 25. Answers for

Tolrestat

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

200

200 TID

200 TID

200 TID

200 TID

200 BID

FRR

0.5

FIR

0.5

0.5

FRH

0.5

FIH

0.5

0.8

fu

Vd (L)

28.5

k (hr-1)

0.0654

0.057

0.057

0.065

0.041

T 1/2 (hr)

10.6

12.1

12.1

10.7

16.9

%Clr

25

14.3

14.3

25.3

39.8

%Clnr

75

85.7

85.7

74.7

60.2

AUC (mg/L*hr)

86

98.3

98.2

87

137

Cl tot (L/hr)

1.86

1.63

1.63

1.84

1.17

Cl h (L/hr)

1.4

1.4

1.4

1.4

0.7

Cl r (L/hr)

0.46

0.23

0.47

0.47

0.47

Eh

0.0155

Er

0.0065

0.88

0.88

0.99

0.63

12

0.75

0.66

0.66

0.75

0.71

g
------ mL

13.8

15.3

15.3

13.9

14.4

g
------ mL

10.8

12.3

12.3

10.9

11.4

g
------ mL

8.2

9.6

9.7

8.3

8.8

P
Q

Cl h
Cl r

(L/hr)

1.42

(L/hr)

0.47

int

int

FCL

(hr)
N

U
Cp

ss
max free

V
Cp
W
Cp

ss
avg free
ss
min free

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-67

Clearance

Vancomycin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 9 - 26)

AHFS 00:00.00
GPI: 0000000000

Macais, W., Meuller, B., and Scarim, S., Vancomycin pharmacokinetics in acute renal failure; preservation of nonrenal clearance, Clinical Pharmacology and Therapeutics, Vol., 50, (1991), p. 688 - 694.

Vancomycin is a glycopeptide antibiotic used in the treatment of infections caused by Gram-positive organisms.
PROBLEM TABLE 9 - 26.

Vancomycin

Patient Condition

Normal

Normal

Dose(mg)

1000

500 QID

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

70

%Clnr

30

AUC (mg/L*hr)

543

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

0.44

(L/hr)

int

FCL

FIR=0.5

(L/hr)

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-68

Clearance

TABLE 9 - 26. Answers for

Patient Condition
A

Dose(mg)

fu

Vd (L)

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

Cl r (L/hr)

Eh

Er

P
Q

Cl h
Cl r

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

(L/hr)

int

FCL

Normal

(L/hr)

int

Normal

Vancomycin

(hr)
N

U
Cp

ss g
-------max free mL

ss

Cp

g
------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-69

Clearance

Xipamide

(Problem 9 - 27)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Knauf, H, et. al., Xipamide disposition in liver cirrhosis, Clinical Pharmacology and Therapeutics, Vol. 48, No. 6, (1990), p. 328
- 632.

Xipamide is a diuretic that has been used in the treatment of congestive heart failure, hypertension, advanced renal failure, and hepatic edema.(Use Qr = 72 L/hr; Qh = 90 L/hr)
PROBLEM TABLE 9 - 27.

Xipamide

Patient Condition

Normal

Normal

Dose(mg)

40

40 QID

0.8

fu

0.01

Vd (L)

21

k (hr-1)

T 1/2 (hr)

%Clr

%Clnr

AUC (mg/L*hr)

Cl tot (L/hr)

Cl h (L/hr)

1.38

Cl r (L/hr)

0.72

Eh

Er

P
Q

Cl h
Cl r

FRH=0.5

FIH=0.5

0.01

0.02

0.01

0.025

(L/hr)

FCL

FIR=0.5

(L/hr)

int

int

FRR=0.5

(hr)
N

U
Cp

ss g
-------max free mL

V
Cp

ss g
-------avg free mL

Cp

ss g
-------min free mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-70

Clearance

TABLE 9 - 27. Answers for

Xipamide

Patient Condition

Normal

Normal

FRR=0.5

FIR=0.5

FRH=0.5

FIH=0.5

Dose(mg)

40

40 QID

40 QID

20 QID

40 QID

20 QID

FRR

0.5

FIR

0.5

0.5

2.5

FRH

0.5

FIH

0.5

0.8

fu

0.01

0.01

0.02

0.01

0.025

Vd (L)

21

k (hr-1)

0.1

0.083

0.146

0.1

0.118

T 1/2 (hr)

6.9

8.4

4.7

7.0

5.9

%Clr

34

21

12

35

72

%Clnr

66

79

88

65

28

AUC (mg/L*hr)

15.2

18.4

7.8

15.4

6.5

Cl tot (L/hr)

2.1

1.74

3.1

2.1

2.5

Cl h (L/hr)

1.38

1.38

2.71

1.36

0.7

Cl r (L/hr)

0.72

0.36

0.36

0.72

1.8

Eh

0.0144

Er

0.01

0.83

1.46

1.18

P
Q

Cl h
Cl r

(L/hr)

140

(L/hr)

73

int

int

FCL

U
Cp

0.87

0.71

0.847

0.86

0.034

0.039

0.034

0.034

0.038

g
------ mL

0.025

0.031

0.026

0.025

0.027

g
------ mL

0.019

0.024

0.019

0.019

0.019

ss g
-------max free mL

V
Cp
W
Cp

(hr)

ss
avg free
ss
min free

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

9-71

CHAPTER 10

Dosage Regimen (Healthy,


Aged, and Diseased Patients)

Author: Michael Makoid


Reviewer: Phillip Vuchetich

OBJECTIVES
1.

Given population average patient data, the student will devise (V) dosage regimens which will maintain plasma concentrations of drug within the therapeutic
range.

2.

Given specific patient information, the patient will justify (VI) dosage regimen
recommendations.

3.

Given patient information regarding organ function, the student will devise (V)
and justify (VI) dosage regimen recommendations for the compromised patient.

4.

The student will write (V) a professional consult using the above calculations

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-1

Dosage Regimen (Healthy, Aged, and Diseased Patients)

10.1 Therapeutic Drug Monitoring


10.1.1

THERAPEUTIC RANGE
The pharmacokinetics of a drug determine the blood concentration achieved from
a prescribed dosing regimen. During multiple drug dosing, the blood concentration
will reflect the drug concentration at the receptor site; and it is the receptor site
concentration that determines the intensity of the drugs effect. Therefore, in order
to predict a patients response to a drug regimen, both the pharmacokinetics and
pharmacological response characteristics of the drug must be understood.
There exists a fundamental relationship between drug pharmacokinetics and pharmacologic response. The relationship between response and ln-concentration is
sigmoidal. A threshold concentration of drug must be attained befor any response
is ellicited at all. Therapy is accheived when the desired effect is attained because
the required concentration has been reached. That concentration would set the
lower limit of utility of the drug, and is called Effective Concentration (MEC).
Most drugs are not clean, that is exhibit only the desired therapeutic response.
They also exhibit undesired side effects, sometimes called toxic effects at a higher,
hopefully a lot higher, concentration. At some concentration, these toxic side
effects become become intollerable. That concentration, or one below it, would
set the upper limit of utility for the drug and is called the Maximum Therapeutic
Concentration or Minimum Toxic Concentration (MTC). Patient studies have
generated upper (MTC) and lower (MEC) plasma concentration ranges that are
deemed safe and effective in treating specific disease states. These concentrations
are known as the therapeutic range for the drug (see Table 10-1).When a drug is
administered at a fixed dosage to numerous subjects, the blood concentrations
achieved vary greatly due to biological variation. However it is possible to have a
reasomable
Clinically, digoxin concentrations below 0.8 ng ml will elicit a subtherapeutic
effect. Alternatively, when the digoxin concentration exceeds 2.0 ng ml side
effects occur (nausea and vomiting, abdominal pain, visual disturbances). Drugs
like digoxin possess a narrow therapeutic index because the concentrations that

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-2

Dosage Regimen (Healthy, Aged, and Diseased Patients)

may produce toxic effects are close to those required for therapeutic effects. The
importance of considering both pharmacokinetics and pharmacodynamics is clear.
TABLE 10-1. Average

therapeutic drug concentration

DRUG

RANGE

digoxin

0.8-2.0 ng ml

gentamicin

2-10 g ml l

lidocaine

1-4 g ml

lithium

0.4-1.4 mEq L

phenytoin

10-20 g ml

phenobarbitol

10-30 g ml

procainamide

4-8 g ml

quinidine

3-6 g ml

theophylline

10-20 g ml

Note that drug concentrations may be expressed by a variety of units.


Pharmacokinetic factors that cause variability in plasma drug concentration are:

Drug-drug interaction
patient disease state
physiological states such as age, weight, sex
drug absorption variation
differences in the ability of a patient to metabolize and eliminate the drug

If we were to give an identical dose of drug to a large group of patients and then
measure the highest plasma drug concentration we would see that due to individual
variability, the resulting plasma drug concentrations differ. This variability can be
attributed to factors influencing drug absorption, distribution, metabolism, and
excretion. Therefore, drug dosage regimens must take into account any disease
altering state or physiological difference in the individual.
Therapeutic drug monitoring optimizes a patients drug therapy by determining
plasma drug concentrations to ensure the rapid and safe drug level in the therapeutic range.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-3

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Two components make


up the process of
therapeutic drug
monitoring:

Assays for determination of the drug concentration in plasma


Interpretation and application of the resulting concentration data to develop a safe and effective
drug regimen.

The major potential advantages of therapeutic drug monitoring are the maximization of therapeutic drug benefits and the minimization of toxic drug effects. The
formulation of drug therapy regimens by therapeutic drug monitoring involves a
process for reaching dosage decisions.

10.1.2

THERAPEUTIC MONITORING: WHY DO WE CARE?


The usefulness of a drugs concentration vs. time profile i based on the observation
that for many drugs there is a relationship between plasma concentration and therapeutic response. There is a drug concentration below which the drug is ineffective, the Minimum Effective Concentration (MEC), and above which the drug has
untoward effects, the Minimum Toxic Concentration (MTC). That defines the
range in which we must attempt to keep the drug concentration (Therapeutic
Range).
The data in Table 10-1 are population averages. Most people respond to drug concentrations in these ranges. There is always the possibility that the range will be
different in an individual patient.
For every pharmacokinetic parameter that we measure, there is a population average and a range. This is normal and is called biological variation. People are different. In addition to biological variation there is always error in the laboratory assays
that we use to measure the parameters and error in the time we take the sample.
Even with these errors, in many cases, he therapy is better when we attempt to
monitor the patients plasma concentration to optimize therapy than if we dont.
This is called therapeutic monitoring. If done properly, the plasma concentrations
are rapidly attained and maintained within the therapeutic range throughout the
course of therapy. This is not to say all drugs should be monitored. Some drugs
have a such a wide therapeutic range or little to no toxic effects that the concentrations matter very little. Therapeutic monitoring is useful when:

a correlation exists between response and concentration


the drug has a narrow therapeutic range
the pharmacological response is not easily assessed
there is a wide inter-subject range in plasma concentrations for a given dose

In this era of DRGs, where reimbursement is no longer tied to cost, therapeutic


monitoring of key drugs can be economically beneficial to an institution. A recent
study (DeStache 1990) showed a significant difference with regard to days in the

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-4

Dosage Regimen (Healthy, Aged, and Diseased Patients)

hospital between the patients on gentamicin who were monitored (and their dosage
regulated as a consequence) vs. those who were not. With DRGs the hospital was
reimbursed a flat fee irrespective of the number of days the patient stayed in the
hospital. If the number of days cost less than what the DRG paid, the hospital
makes money. If the days cost more than the hospital loses money. This study
showed that if all patients in the hospital who were on gentamicin were monitored,
the hospital would save $4,000,000. Thats right FOUR MILLION per year. I
would say that would pay my salary, with a little left over, and that is only one
drug!
The process of
therapeutic monitoring
takes effort.

First the MD must order the blood assays.


Second, someone (nurse, med tech, you) must take the blood.
Someone (lab tech, you) must assay the drug concentration in the blood.
You must interpret the data
You must communicate your interpretation and your recommendations for dosage regimen
change to the MD. This will allow for informed dosage decisions.

You must follow through to ensure proper changes have been made.
You must continue the process throughout therapy. Therapeutic monitoring, in many cases, will
be part of your practice. It can be very rewarding

Thus, if we have deterimined the therapeutic range, we could use pharmacokinetics to determine the optimum dosage regemin to maintain the patients plasma concentration within that range.

10.1.3

STEADY STATE
It is rare that a drug is given only once. Most therapies consist of multiple doses
of several days duration, if not several years. It is necessary, therefore, to be able
to asess plasma concenterations, both the peak which much be at or below the
MTC and the trough which must be at or above the MEC for the drug to be effective under these conditions. Thus when we dose a patient, the concentration profile must be within the Therapeutic Range during the entire time that the patient is
taking the drug. We can calculate the plasma concentrations in the followin manner. In the simplest model, suppose we give a drug by IV Bolus (because the math
is simpler). The equation which would result be
Cp = Cp 0 e

I.V. Bolus Multiple Dose

( kt )

D ( kt)
= ---- e
V

(EQ 10-1)

The peak would be

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-5

Dosage Regimen (Healthy, Aged, and Diseased Patients)

(EQ 10-2)

1
D
Cp max = ---V

If we allowed the drug to be eliminated for

hours, the trough would be:

1
D ( ( k ) )
Cp min = ---- e
V

(EQ 10-3)

Upon giving a second dose, prior to the complete removal by the body of the first
dose the Cp max 2 would be the second dose plus what was left from the first dose:
2
D
D ( ( k ) )
Cp max = ---- + ---- e

V V

and the

Cp min

would be

Cp max

times

( ( k ) )

(EQ 10-4)

, thus:

2
( ( k ) )
( ( 2k ) )
: Cp min = D
---- e
+ D
---- e

After n doses, the

Cp max

(EQ 10-5)

would be:

n
( ( k ) )
( ( ( n 1 ) k ) )
Cp max = D
---- + D
---- e
+ +D
---- e
V V
V

while the

Cp min

(EQ 10-6)

would be:

n
( ( k ) )
( ( 2k ) )
( ( nk ) )
Cp min = D
---- e
+ D
---- e
+ +D
---- e
V

V
V

(EQ 10-7)

Subtracting equation 10-7 from equation 10-6 to eliminate the series yields:
n
( nk )
n
D
D ( ( nk ) ) D
= ---- ( 1 e
)
Cp max Cp min = ---- ---- e

V
V
V

Cp min

is also

Cp max e
n

( k )

(EQ 10-8)

which means that


n

Cp max Cp min = Cp max Cp max e

( k

= Cp max ( 1 e

Equating equation 10-8 and equation 10-9 and solving for


( nk

( k )

Cp max

(EQ 10-9)

yields:

n
e
Cp max = D
---- 1---------------------------V 1 e( k )

At large n, e ( nk ) 0 and thus the steady state maximum,


Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 10-10)

Cp max

ss

, is:
10-6

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Cp max

and the steady state minimum,

ss

1
= D
---- ------------------------V 1 e( k )

Cp min

Cp min

ss

ss

(EQ 10-11)

, is :
( k

D
e
= ---- ------------------------V 1 e ( k )

(EQ 10-12)

In order to make a general equation set from equation 10-11 and equation 10-12,
let N be the number of half lives in a dosing interval,
k = ( ln ( 2 ) ) t 1 2

. Substituting into the function

( k )

, yields

, and

N = --------t1 2
( k

N
= 1
---
2

and

thus equation 10-11 becomes:


Cp max

ss

1 = D
---- -------------------V
1 N
1 ---
2

(EQ 10-13)

and equation 10-12 becomes :

Cp min

ss

1
---
2

= D
---- --------------------V
1 N
1 ---
2

(EQ 10-14)

The average drug concentration under these conditions would be equivalant to the
steady state concentration attained by an infusion of the same rate, i.e. if we were
to give a multiple dose at 200 mg every four hours (q4h) or 400 mg every eight
hours (q8h), the average that would be attained would be equivelaent to the steady
state plasma concentration attained by giving an infusion at 50 mg/hr, (Q = 50 mg/
hr), and thus, in the infusion, the
k = 0.693 N

ss

Q
= ---------kV

and in multiple dosing

D
Q = ---

, and

so:
Cp

Oral Multiple Dosing


(Approximation)

Cp

ss
avg

D
D
1.443 D
= ------------------- = ------------------------------ = ---------------------V K V 0.693 N
NV

(EQ 10-15)

Similar equations, although more complex, can be derived for multiple dose oral
products. However, if we were agreed to live with some error these equations, with
some modifications could be used to approximate multiple dose oral products.
The error on both calculated Cp maxss and Cp minss would be in the direction of
safety, i.e. the calculated Cp maxss would be higher and the calculated Cp minss would
be lower that their respective multiple dose oral calculations. Thus, if the simpler

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-7

Dosage Regimen (Healthy, Aged, and Diseased Patients)

equations place the Cp max ss and C minss within the Therapeutic Range, the oral multiple dose equations would also. The two modifications would be to the Dose. Bioavailability, f, must be considered, and if the drug given is not the drug measured in
the blood, the salt factor, S, the difference in the molecular weight of the two compounds must be taken into account, (i.e.

MW measured
S = -----------------------------MW given

). Thus, when amino-

phyline, which is a complex consisting of two theophyline molecules and an


ethylinedyamine molecule is given, but theophyline is measured, the salt factor,
MW Theo 2 180.17
- = ------------------------ = 0.857
S = ----------------------MW Amino
420.44

. Thus for oral multiple dose, we can approximate

for
using IV bolus equations as such :
Cp max

Cp

ss
avg

ss

SfD
1
= ------------------ --------------------N
V
1 1---
2

SfD
SfD
1.443 S f D
= ------------------- = ------------------------------ = -----------------------------------V K V 0.693 N
N V

Cp min

ss

1
---
2

(EQ 10-16)

(EQ 10-17)

S f D
= ------------------ --------------------V
1 N
1 ---
2

(EQ 10-18)

because errors involved with this approximation both in maximum and minimum
calculations (peak and trough) place the drug further within the Therapeutic
Range, i.e. the real peaks are lower than the calculated peaks and the real troughs
are higher than the calculated troughs, thus both errors are on the side of safety.
Dosing Interval

The object of pharmacokinetics is to optimize therapy. By definition, that is to


maintain the plasma concentration of the drug within the therapeutic range for the
duration of the therapy presuming that is needed. Thus, the concentrations must
stay within the MTC and MEC or
ss

Cp max
N
MTC-----------= ------------------= 2 max
ss
MEC
Cp min

(EQ 10-19)

by deviding equation 10-18 into equation 10-16 and simplifying. Given these limits, the maximum dosing interval, max , is obtained by solving for Nmax:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-8

Dosage Regimen (Healthy, Aged, and Diseased Patients)

MTC-
Ln -----------MEC
= -------------------------Ln2

N max
and since

max
N max = ---------t1 2

(EQ 10-20)

by definition,
max

= N max t1 2

(EQ 10-21)

is not necessarily the dosing interval of choice, but it is the maximum dosing
interval attainable without sustained or controlled release delivery systems.
max

Accepable dosing intervals are those which result in a dose being given at the same
time of the day, every day. Imagine, if you will, the chaos on the nursing floor if
the dose for a given drug were every 15 hours. Compliance, none too high when
the patient is given a reasonable dosing interval, would go straight to the toilet if
we asked the patient to take a tablet every 5.3 hours. What would be optimal would
be to tie the takeing of the drug with an activity that occurs the same time every
day for once a day therapy, or at least dose the same time every day. Thus, for
multiple daily doses, the only regimens that work are those which when devide
into 24 hours give unit answers: QD = 24/1 = q24h; BID = 24/2 = q12h, TID = 24/
3 = q8h; QID = 24/4 = q6h; q4h; q3h; q2h. These result in decreasing orders of
patient compliance (unless the patient is really motivated to take the drug every 2
hours - forget it.) Thus the maximum acceptable dosing interval would be the largest acceptable dosing interval below the max . So, for example if max is 15.7
hours, the maximum acceptable dosing interval would be 12 hours. we could also
dose every 8, 6 or 4 hours if necessary.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-9

Dosage Regimen (Healthy, Aged, and Diseased Patients)

10.2 Diseases - Dosing the Compromised Patient


As previously discussed in the chapter on clearance, diseases result in changes in
clearance. These are routinely as a consequence of changes in organ function or
blood flow to the organ. Diseases which cause a change in clearance, do so by
changing either the elimination rate constant, K, or the volume of distribution, Vd,
or both. Thus the fractional change in total body clearance :
Cl tot K V
F Cl tot = -------------= -----------------KV
Cl tot
Where X* indicates new or changed variable.

(EQ 10-22)

In general, if

0.80 < F Cl < 1.2 ,


tot

changes in dosage regimen are not necessary. In order to return a previously controlled healthy pateint back to the therapeutic range, a general rule of thumb is suggested as an initial starting point. The desease modifies K (as t1/2) and V. As
pharmacists, we can modify D and . These variables are paired in the above
equations (equation 10-16 and equation 10-18), D with V (in
with

(in

N = --------t1 2

S----------------fD
V

) and

t1 2

). If the physiological change is a change in V, the pharmacist

would recommend a change in D proportionally, and if the half life changes, the
pharmacist would recommend a change in the dosing interval proportionally.
Remenber, the object is to get the plasma concentrations back to where they were
prior to the illness. The only problem is that we are limited to these recommended
changes being incremental and not continuous. That is a change in dose is limited
to the available dosage forms and strengths and a change in dosing interval is
limeted to the accepatable dosing interval.
Protein Binding

If the drug is highly protein bound, the object would be to get the free concentration back to what it was prior to illness. Consequently, equation 10-16, equation
10-17, and equation 10-18 would be rewritten thus:
Cp

Cp

ss
fu S f D
ss
1
= f u Cp
= ------------------------- -------------------N
V
max free
max
1 1---
2

ss
fu S f D fu S f D
ss
= fu C p
= -------------------------= -----------------------------VK
V 0.693 N
avg free
avg

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 10-23)

(EQ 10-24)

10-10

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Cp

1
---
2

ss
fu S f D
ss
= fu Cp
= ------------------------- -------------------N
V
min free
min
1 1---
2

(EQ 10-25)

Some interesting and unexpected things result from these relationships. Since
plasma or blood concentrations are usually measured, if a drug is highly protein
bound and the desease results in upsetting that equilibrium, you might see toxicity
resulltling from normal or even subtherapeutic measured concentrations. More on
that in the chapter on protein binding.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-11

Dosage Regimen (Healthy, Aged, and Diseased Patients)

10.3 Problems

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-12

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Alprazolam
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 1)

AHFS 00:00.00
GPI: 0000000000

Juhl, R. et al., "Alprazolam pharmacokinetics in alcoholic liver disease", Journal of Clinical Pharmacology, Vol.24, (1984), p. 113
- 119.

Alprazolam is an anti-anxiety agent which is metabolized to 4-hydroxy and -hydroxy metabolites. In this
study, patients with cirrhosis of the liver and healthy patients were each given doses of 1.0 mg of Alprazolam. The following data is for healthy patients.
PROBLEM TABLE 10 - 1.

Alprazolam

Dose

1.0 mg BID
1.16 L/kg
1.22
529.3

AUC

Assume that your patient weighs 70 kg when answering the following:


1.

Find k.

2.

Find the MRT.

3.

Find the .

4.

Find the AUMC.

5.

Find .

6.

What is N?

7.

What is the patient's maximum plasma concentration, , under this dosage regimen.

8.

What is the patient's average plasma concentration, , under this dosage regimen.

9.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-13

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Cefixime
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 2)

AHFS 00:00.00
GPI: 0000000000

Faulkner, R. et al., "Pharmacokinetics of cefixime after once-a-day and twice-a-day dosing to steady state", Journal of Clinical
Pharmacology, Vol.27, (1987), p. 807 - 812.

Cefixime is a broad-spectrum cephalosporin which is active against a variety of gram positive and gram negative bacteria. In this study, patients received a 200 mg oral dose of cefixime twice daily.
PROBLEM TABLE 10 - 2.

Cefixime

Dose

200 mg BID
3.3 hours
286
32

AUC

14.12

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-14

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Cefpodoxime
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 3)

AHFS 00:00.00
GPI: 0000000000

Borin, M. et. al., "Pharmacokinetics and tolerance studies of cepodoxime after single-and multiple-dose oral administration of cefpodoxime proxetil", Journal of Clinical Pharmacology, Vol.31, (1991), p. 1137 - 1145.

Cefpodoxime proxetil is a third-generation, broad-spectrum cephalosporin which is given by the oral route. It is a prodrug which is converted in vivo to cefpodoxime which inhibits bacterial cell wall synthesis by binding to penicillinbinding proteins.
PROBLEM TABLE 10 - 3.

Cefpodoxime

Dose

100 mg BID
2.1 hours
271
79.1

AUC

6.9

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-15

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Cefprozil
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 4)

AHFS 00:00.00
GPI: 0000000000

Lode, H. et. al., "Multiple-dose pharmacokinetics of cefprozil and its impact on intestinal flora of volunteers", Antimicrobial
Agents and Chemotherapy, Vol.36, (1992), p. 144 - 149.

Cefprozil is a broad-spectrum cephalosporin which is given by the oral route. In this study subjects received 500 mg
doses of cefprozil every twelve hours for eight days.
PROBLEM TABLE 10 - 4.

Cefprozil

Dose

500 mg q. 12 hours
55.11minutes
310.25
277.50

AUC

27.80

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-16

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Clobazam
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 5)

AHFS 00:00.00
GPI: 0000000000

Greenblatt, D. et. al., "Reduced single-dose clearance of clobazam in elderly men predicts increased multiple-dose accumulation",
Clinical Pharmacokinetics, Vol.8, (1983), p. 83 - 94.

Clobazam is an agent used in the treatment of anxiety. In this study, patients received 10 mg dose of clobazam daily.
PROBLEM TABLE 10 - 5. Clobazam

Dose

10 mg daily
180 hours

AUC

1.

Find k.

2.

Find MRT.

3.

Find the ?

4.

Find the AUMC.

5.

Find .

6.

What is N?

7.

What is the patient's maximum plasma concentration, , under this dosage regimen.

8.

What is the patient's average plasma concentration, , under this dosage regimen.

9.

What is the patient's minimum plasma concentration, , under this dosage regimen.

10.
Your patients renal function drops to 50% of normal. What would be a new dosing regimen under these conditions? (Assume that you want to keep < 110% of the normal and that you want to keep > 90% of the normal .)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-17

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Maloprim

(Problem 10 - 6)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Edstein, M., Rieckmann, K., and Veenendaal, J., "Multiple-dose pharmacokinetics and in vitro antimalarial activity of dapsone
plus pyrimethamine (Maloprim ) in man", British Journal of Clinical Pharmacokinetics, Vol.30, (1990), p.259 - 265.

Maloprim is an agent which contains both dapsone and pyrimethamine. In this study, healthy volunteers
were given 100 mg of dapsone plus 12.5 mg pyrimethamine weekly. The following data is for dapsone:
PROBLEM TABLE 10 - 6. Maloprim

Dose

100 mg weekly
22.6 hours

AUC

35.0

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-18

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Doxycycline

(Problem 10 - 7)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Shmuklarsky, M. et. al., "Failure of doxycycline as a causal prophylactic agent against Plasmodium falciparum malaria in healthy
nonimmune volunteers", Annals of Internal Medicine, Vol.120, (1994), p. 294 - 298.

Doxycycline is an antibiotic which has been recommended for prevention of malaria in people traveling to areas
endemic to chloroquine-resistant P. falciparum malaria who are unable to take mefloquine. This study determined that
doxycycline is not effective for this use. Volunteers were given 100 mg doses of doxycycline daily for 10 days.
PROBLEM TABLE 10 - 7.

Doxycycline

Dose

100 mg daily
21.9 hours

AUC

40.7

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-19

Dosage Regimen (Healthy, Aged, and Diseased Patients)

DQ-2556
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 8)

AHFS 00:00.00
GPI: 0000000000

Nakashima, M. et. al., "Phase I study of DQ-2556, a new parenteral 3-quaternary ammonium cephalosporin antibiotic", Journal of
Clinical Pharmacology, Vol.33, (1993), p. 57 - 62.

DQ-2556 is a new broad-spectrum cephalosporin which is active against many bacteria including Pseudomonas aeruginosa. Subjects in this study were each given a 2000 mg infusion of DQ-2556 over 5 minutes every 12 hours for a total
of 9 doses.
PROBLEM TABLE 10 - 8.

DQ-2556
Dose

2000 mg infusion over 5 minutes


17.6 L
8.5
7.1

AUC

241.0

1.

Find Cl.

2.

Find k.

3.

Find the MRT.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-20

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Erythropoetin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 9)

AHFS 00:00.00
GPI: 0000000000

Gladziwa, U., et al., "Pharmacokinetics of epoetin (recombinant human erythropoietin) after long term therapy in patients undergoing haemodialysis and haemofiltration", Clinical Pharmacokinetics, Vol.8, (1983), p. 83 - 94.

Erythropoetin is a regulatory hormone of red blood cells. In this study patients with end-stage renal disease were given
150 U/kg of epoetin three times a week.
PROBLEM TABLE 10 - 9.

Glipizide

Dose

150 U/kg t.i.w.


7.7hours

Cl

5.4

Assuming that your patient weighs 65 kg, please determine the following:
1.

Find k.

2.

Find MRT.

3.

Find the .

4.

Find the AUC.

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-21

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Flecainide
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 10)

AHFS 00:00.00
GPI: 0000000000

Forland, S. et al., "Flecainide pharmacokinetics after multiple-dosing in patients with impaired renal function", Journal of Clinical
Pharmacology, Vol.28, (1988), p. 727 - 735.

Flecainide acetate is a class 1C anti-arrhythmic agent which is used in the treatment of ventricular and supraventricular
arrhythmias. In this study, subjects were given doses of 100mg of flecainide orally twice daily.
PROBLEM TABLE 10 - 10. Flecainide

Dose

100 mg BID
7.4 L/kg
486
89

AUC

3.429

Assume that your patient weighs 70 kg when calculating the following:


1.

Find Cl.

2.

Find k.

3.

Find the MRT.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-22

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Glipizide
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 11)

AHFS 00:00.00
GPI: 0000000000

Kradjan, W. et al., "Glipizide pharmacokinetics: effects of age, diabetes, and multiple dosing", Journal of Clinical Pharmacology,
Vol.29, (1989), p. 1121 - 1127.

Glipizide is a second-generation oral hypoglycemic agent used in the treatment of non-insulin-dependent (type II) diabetes. In this study, both diabetic and non-diabetic elderly men were each given doses of 2.5 mg of glipizide daily for
five days. The data for the non-diabetic group is given below.
PROBLEM TABLE 10 - 11.

Dose

2.5 mg daily
4.0 hours
0.47

AUC

2325.4

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-23

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Lomefloxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 12)

AHFS 00:00.00
GPI: 0000000000

Hunt, T. and Adams, M., "Pharmacokinetics and safety of lomefloxacin following multiple doses", Diagn Microbiol Infect Dis,
Vol.12, (1989), p. 181 - 187.

Lomefloxacin is a quinolone antibiotic which is useful against both Gram-positive and Gram-negative bacteria. It is
used in the treatment of urinary tract infections and lower respiratory tract infections. A dose of 400 mg of lomefloxacin was given twice daily to healthy patients.
PROBLEM TABLE 10 - 12. Lomefloxacin

Dose

400 mg BID
7.32 hours

AUC

61.67

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-24

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Loratadine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 13)

AHFS 00:00.00
GPI: 0000000000

Radwanski, E. et al., "Loratadine: multiple-dose pharmacokinetics", Journal of Clinical Pharmacology, Vol.27, (1987), p. 530 533.

Loratadine is an antihistamine which is orally active. In this study, healthy, male volunteers were each given a 40-mg
loratadine capsule daily for ten days.
PROBLEM TABLE 10 - 13. Loratadine

Dose

40 mg daily
14.4 hours

AUC

96.0

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-25

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Methamphetamine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 14)

AHFS 00:00.00
GPI: 0000000000

Cook, C., et al., "Pharmacokinetics of oral methamphetamine and effects of repeated daily dosing in humans", Drug Metabolism
and Disposition, Vol.20, (1992), p. 856 - 861.

Methamphetamine is a CNS stimulant which is used in the treatment of attention deficit disorder and obesity. In this
study, subjects were given a 0.125 mg/kg dose of methamphetamine daily.
PROBLEM TABLE 10 - 14. Methamphetamine

Dose

0.125 mg/kg daily


8.46 hours
65.0
212

Assume that your patient weighs 70 kg when calculating the following:


1.

Find k.

2.

Find MRT.

3.

Find the .

4.

Find the AUC.

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-26

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Mexiletine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 15)

AHFS 00:00.00
GPI: 0000000000

Gillis, A. and Kates, R., "Clinical pharmacokinetics of the newer antiarrhythmic agents", Clinical Pharmacokinetics, Vol.9, (1984),
p. 375 - 403.

Mexiletine is a class Ib antiarrhythmic agent. In this study, volunteers each received a 1600 mg dose orally each day.
PROBLEM TABLE 10 - 15. Mexiletine

Dose

1600 mg daily
380 L
681

1.

Find k.

2.

Find the MRT.

3.

Find the .

4.

Find the AUC.

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-27

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Moxisylyte
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 16)

AHFS 00:00.00
GPI: 0000000000

Costa, P. et al., "Multiple-dose pharmacokinetics of moxisylyte after oral administration to healthy volunteers", Journal of Pharmaceutical Sciences, Vol.82, (1993), p. 968 - 971.

Moxisylyte is an -adrenergic blocker which has been used in Europe for some times as a vasodilator in the
treatment of such disease states as age-associated mental impairment, acrocyanosis, Raynaud's syndrome, vascular cochlearvestibular disorders, glaucoma, and benign prostatic hyperplasia.
PROBLEM TABLE 10 - 16. Moxisylyte

Dose

240 mg BID
2.28 hours

AUC

11186

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-28

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Naproxen
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 17)

AHFS 00:00.00
GPI: 0000000000

Ouweland, F. et. al., "Hypoalbuminaemia and naproxen pharmacokinetics in a patient with rheumatoid arthritis", Clinical Pharmacokinetics, Vol.11, (1986), p. 511 - 515.

The pharmacokinetics parameters of naproxen were looked at in patients with rheumatoid arthritis in this study. A
patient received a dose of 500 mg of naproxen orally twice daily.
PROBLEM TABLE 10 - 17. Naproxen

Dose

500 mg BID
9.0 L

AUC

1134

1.

Find Cl.

2.

Find k.

3.

Find the MRT.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-29

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Nisoldipine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 18)

AHFS 00:00.00
GPI: 0000000000

Harten, J. et al., "Influence of renal function on the pharmacokinetics and cardiovascular effects of nisoldipine after single and
multiple dosing", Clinical Pharmacokinetics, Vol.16, (1989), p. 55 - 64.

Nisoldipine is a second-generation calcium-channel blocker which is under investigation for use as an anti-hypertensive agent. Nisoldipine is mainly eliminated through liver metabolism with metabolites being excreted mainly in the
urine but also in the feces. The systemic clearance of nisoldipine depends greatly on liver blood flow.
PROBLEM TABLE 10 - 18. Nisoldipine

Dose

10 mg BID orally
7.9 hours

AUC

5.2

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the ?

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-30

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Pefloxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 19)

AHFS 00:00.00
GPI: 0000000000

Bruno, D. et al., "Bayesian versus NONMEM estimation", , Vol. , (19 ), p. 657 - 668.

Pefloxacin is an antibiotic used to treat patients who are in the intensive care unit. For this study, patients were given a
400 mg dose of pefloxacin twice daily for eight days.
PROBLEM TABLE 10 - 19. Pefloxacin

Dose

400 mg BID
21.3 hours

Cl

3.77

1.

Find k.

2.

Find MRT.

3.

Find the ?

4.

Find the AUC.

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-31

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Phenylpropanolamine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 20)

AHFS 00:00.00
GPI: 0000000000

Scherzinger, S., Rowse, R., and Kanfer, I., "Steady state pharmacokinetics and dose-proportionality of phenylpropanolamine in
healthy subjects", Journal of Clinical Pharmacology, Vol.30, (1990), p. 372 - 377.

Phenylpropanolamine is a sympathomimetic agent which is used both for its action as a nasal decongestant and its
action as an anorexiant. In this study healthy volunteers were given doses of 25 mg every four hours for a total of
seven doses. It was found that phenylpropanolamine is 77% renally excreted.
PROBLEM TABLE 10 - 20. Phenylpropanolamine

Dose

25 mg q. 4 hours
4.71 hours
4.08 L/kg
0.5

1.

Find k.

2.

Find MRT.

3.

Find Cl.

4.

Find the AUC.

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-32

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Promethazine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 21)

AHFS 00:00.00
GPI: 0000000000

Taylor, G. and Houston, J., "Changes in the disposition of promethazine during multiple dosing in rabbits", Journal of Clinical
Pharmacology, Vol.37, (1985), p. 243 - 247.

Promethazine is an agent used as an anti-histamine and a sedative. In this study rabbits weighing 2.7 to 3.3 kilograms
each received a 10 mg/kg dose of promethazine every 24 hours for 14 days.
PROBLEM TABLE 10 - 21. Promethazine

Dose

10 mg/kg
249 minutes
65.0

1.

Find k.

2.

Find MRT.

3.

Find the ?

4.

Find the AUC.

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-33

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Rufloxacin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 22)

AHFS 00:00.00
GPI: 0000000000

Mattina, R. et al., "Pharmacokinetics of rufloxacin in healthy volunteers after repeated oral doses", Chemotherapy, Vol.37, (1991),
p. 389 - 397.

Rufloxacin is a broad-spectrum, fluoroquinolone antibiotic. In this study a patient was given a loading dose of 300 mg
of rufloxacin followed by 150 mg of rufloxacin daily for five days.
PROBLEM TABLE 10 - 22. Rufloxacin

Dose

150 mg daily
104 L
41
10

AUC

121.5

1.

Find k.

2.

Find the MRT.

3.

Find the .

4.

Find the AUMC.

5.

Find .

6.

What is N?

7.

What is the patient's maximum plasma concentration, , under this dosage regimen.

8.

What is the patient's average plasma concentration, , under this dosage regimen.

9.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-34

Dosage Regimen (Healthy, Aged, and Diseased Patients)

Velnacrine (HP 029)


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 10 - 23)

AHFS 00:00.00
GPI: 0000000000

Puri, S. et al., "Multiple dose pharmacokinetics, safety, and tolerance of velnacrine (HP 029) in healthy elderly subjects: a potential therapeutic agent for Alzheimer's disease", Journal of Clinical Pharmacology, Vol.30, (1990), p. 948 - 955.

Velnacrine is an investigative agent which has central cholinergic action and may be beneficial in the treatment of
Alzheimer's disease. Healthy, elderly, men were given doses of 100 mg twice daily in this study. It was found that 30%
of the velnacrine dose was excreted unchanged.
PROBLEM TABLE 10 - 23. Velnacrine

Dose

(HP 029)

100 mg BID
2.4 hours

AUC

809.5

1.

Find k.

2.

Find the MRT.

3.

Find Cl.

4.

Find the .

5.

Find the AUMC.

6.

Find .

7.

What is N?

8.

What is the patient's maximum plasma concentration, , under this dosage regimen.

9.

What is the patient's average plasma concentration, , under this dosage regimen.

10.

What is the patient's minimum plasma concentration, , under this dosage regimen.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-35

Dosage Regimen (Healthy, Aged, and Diseased Patients)

10.4 Answers
Alprazolam
1.

0.0631 h-1

2.

15.85 hours

3.

10.98 hours

4.

8387.81

5.

12

6.

1.093

7.

23.19

8.

16.26

9.

10.876

Cefixime
1.

0.210 h-1

2.

4.76 hours

3.

14.164 L/h

4.

67.435 L

5.

62.224

6.

12

7.

3.64

8.

3.225

9.

1.177

10.

0.259

Cefpodoxime
1.

0.330 h-1

2.

3.03 hours

3.

14.49 L/h

4.

43.91 L

5.

20.905

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-36

Dosage Regimen (Healthy, Aged, and Diseased Patients)

6.

12

7.

5.714

8.

2.322

9.

0.575

10.

0.0442

Cefprozil
1.

0.0126 min-1

2.

76.51 minutes

3.

17.99 L/h

4.

23.83 L

5.

36.84

6.

12

7.

13.065

8.

20.98

9.

2.317

10.

2.45

Maloprim
1.

0.0307 h-1

2.

32.6 hours

3.

2.857 L/h

4.

93.16 L

5.

1141.17

6.

168

7.

7.435

8.

1.08

9.

0.208

10.

6.25

Doxycycline
1.

0.0317 h-1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-37

Dosage Regimen (Healthy, Aged, and Diseased Patients)

2.

31.595 hours

3.

2.457 L/h

4.

77.629 L

5.

1285.92

6.

24

7.

1.096

8.

2.42

9.

1.696

10.

1.133

DQ-2556
1.

8.299 L/h

2.

0.4715 h-1

3.

2.12 hours

4.

1.47 hours

5.

511.11

6.

12

7.

8.163

8.

114

9.

20.083

10.

0.398

Erythropoetin
1.

0.09 h-1

2.

11.11 hours

3.

3599.24 mL

4.

30092.6

5.

334291.14

6.

72

7.

9.35

8.

2713.24

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-38

Dosage Regimen (Healthy, Aged, and Diseased Patients)

9.

417.98

10.

4.156

Glipizide
1.

0.173 h-1

2.

5.77 hours

3.

1.075 L/h

4.

6.204 L

5.

13419.37

6.

24

7.

8.

0.4094

9.

96.893

10.

6.396

Flecainide
1.

29.16 L/h

2.

0.0563 h-1

3.

17.76 hours

4.

12.31 hours

5.

60.91

6.

12

7.

0.975

8.

0.393

9.

0.286

10.

0.200

Lomefloxacin
1.

0.0947 h-1

2.

10.56 hours

3.

6.486 L/h

4.

68.497 L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-39

Dosage Regimen (Healthy, Aged, and Diseased Patients)

5.

651.27

6.

12

7.

1.639

8.

8.6

9.

5.139

10.

2.76

Loratadine
1.

0.0481 h-1

2.

20.77 hours

3.

416.67 L/h

4.

8656.17 L

5.

1994.38

6.

24

7.

1.67

8.

6.746

9.

10.

2.125

Methamphetamine
1.

0.082 h-1

2.

12.21 hours

3.

46.7 L

4.

2.244

5.

27.383

6.

24

7.

2.837

8.

213.74

9.

93.48

10.

29

Moxisylyte

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-40

Dosage Regimen (Healthy, Aged, and Diseased Patients)

1.

0.304 h-1

2.

3.29 hours

3.

0.0215 L/h

4.

70.574 mL

5.

36794.61

6.

12

7.

5.263

8.

3.492

9.

0.9322

10.

0.0909

Naproxen
1.

0.441 L/h

2.

0.049 h-1

3.

20.412 hours

4.

14.149 hours

5.

23147.21

6.

12

7.

0.848

8.

124.98

9.

94.5

10.

69.43

Mexiletine
1.

0.1075 h-1

2.

9.3 hours

3.

6.45 hours

4.

39.16

5.

364.17

6.

24

7.

3.723

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-41

Dosage Regimen (Healthy, Aged, and Diseased Patients)

8.

4.256

9.

1.632

10.

0.345

Nisoldipine
1.

0.0877 h-1

2.

11.397 hours

3.

1923.08 L/h

4.

21.92 mL

5.

59.27

6.

12

7.

1.52

8.

700.7

9.

433.3

10.

244.5

Pefloxacin
1.

0.0325 h-1

2.

30.73 hours

3.

115.85 L

4.

106.1

5.

3260.4

6.

12

7.

0.563

8.

10.68

9.

8.84

10.

7.23

Phenylpropanolamine
1.

0.147 h-1

2.

6.795 hours

3.

36.03 L/h

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-42

Dosage Regimen (Healthy, Aged, and Diseased Patients)

4.

0.694

5.

4.715

6.

7.

0.849

8.

229.5

9.

173.5

10.

127.4

Promethazine
1.

0.167 h-1

2.

5.987 hours

3.

70.05 L

4.

2.56

5.

15.35

6.

24

7.

5.78

8.

436.19

9.

106.84

10.

7.92

Rufloxacin
0.0237 h-1
42.28 hours
29.30 hours
5136.6
24
0.819
3.33
2.54
1.89
Velnacrine

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-43

Dosage Regimen (Healthy, Aged, and Diseased Patients)

1.

0.289 h-1

2.

3.462 hours

3.

123.53 L/h

4.

427.73 L

5.

2802.87

6.

12

7.

8.

241.33

9.

67.46

10.

7.54

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10-44

CHAPTER 11

Multicompartment Modeling

Author: Michael Makoid


Reviewer: Phillip Vuchetich

OBJECTIVES
1.

This chapter is not completed.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-1

Multicompartment Modeling

11.1 Executive Summary

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-2

Multicompartment Modeling

11.2 Equations

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-3

Multicompartment Modeling

11.3 PHARMCOKINETICS: MAMMILLARY MODELS

For many drugs the equilibrium between drug concentrations in different tissues is
not achieved rapidly. Thus, one of the assumptions of the one-compartment open
model sometimes becomes invalid. A more complex mammillary open model is
often necessary to describe mathematically the plasma concentration data (for
example) seen after the administration of some drugs. The simplest mammillary
open model is a two-compartment open model: for example:

Compartment One (central compartment) can be sampled through the blood (or
plasma, or serum). It may consist of organs or tissues which, being highly perfused
with blood, are in rapid equilibrium distribution with the blood.

Compartnent Two (peripheral compartment) cannot normally be sampled. It may


consist or organs or tissues which, being poorly perfused with blood, are in slow
equilibrium distribution with the blood.

The Body is the sum of both compartments.


Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-4

Multicompartment Modeling

1. Biexponential Properties of Two-Compartment Open Model

Following an intravenous bolus injection, the plasma concentration against time


profile has two phases:

a. Initial phase - ( - phase)

b. Terminal phase - ( - phase)

On semilogarithmic paper the terminal phase is linear, indicating that initial distribution has been completed and that equilibrium has been attained. The terminal
half-life ( t 1 2 ) can be measured from the terminal phase.

2. Intravenous Bolus Administration: Plasma Concentration Data


For a one-compartment open model,

Cp = ( Cp )o e

kt

(EQ 10-26)

i.e., the concentration of drug in the plasma declines exponentially with time

For a two-compartment open model,


Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-5

Multicompartment Modeling

Cp = ( A1 e

+ B1 e

(EQ 10-27)

i.e., the concentration of drug in the plasma declines biexponentially with time

2.1 Symbols

A 1 and B1 are intercept constants ( M L )

and are hybrid rate constants ( T )

V 1 is the apparent volume of unchanged drug distribution in compartment one


3

(L )

k 10, k 12 , and , k 21 are micro rate constants ( T )

2.2 Relationships (for reference, except Eq. 3)

= 0.5 [ ( k 10 + k 12 + k 21 ) + ( k 10 + k 12 + k 21 ) 4k 10k 12 ]

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-6

Multicompartment Modeling

= 0.5 [ ( k 10 + k 12 + k 21 ) ( k 10 + k 12 + k 21 ) 4k 10k 12 ]
D- ( k 21 )
A 1 = ---- ---------------------V1 ( )
( k 21 )
D- --------------------B 1 = ----V1 ( )
A 1 + B1 = ( Cp ) o

(EQ 10-28)

2.3 Obtaining Pharmacokinetic Parameters by Feathering


By convention, >
a. Plot C p against t on semilogarithmic paper

b. Find t 1 2 from the linear terminal phase: see Intravenous Administration,


section A1.4a

c. Calculate the terminal hybrid rate constant ( ) ; in reality it contains both distributive ( k 12 and k 21 ) and elimination ( k 10 ) factors.

0.693
= ------------t1 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 10-29)

11-7

Multicompartment Modeling

d. Draw a straight line through the linear terminal elimination phase and extralpolate this line to t = 0. The intercept is equal to B1 .

e. Read estrapolated plasma concentrations ( C p ) from the plot at times equal to


those given for values of C p which are prior to the terminal phase.

f. At each of these times calculate:

( C p )diff = C p C p

g. Plot ( C p )diff against t (see Eq.8) on semilogarithmic paper. The is a feathered line and should decline linearly.

h. Find the half-life of the plot. It wil refer to the initial phase. Calculate,

0.693 = -----------------------half life

i. Measure the intercept of the feathered line; it will equal to A 1 (Note that usually A1 = B1 , even theoetically).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-8

Multicompartment Modeling

j. Calculate ( C p )o from Eq. 3

k. Calcultate V1 by
Xo
D
= -----------------V1 = ------------( Cp )o A1 + B 1

(EQ 10-30)

.
Theory

When t is large, e

<e

. Hence, Eq. 2 becomes

Cp = B1 e

(EQ 10-31)

i.e., when t is large, the concentration of the drug in the plasma declines exponentilly with time.

The extrapolated plasma concentrations are


Cp = B1 e

(EQ 10-32)

Substituting from Eqs. 2 and 7a into Eq. 5,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-9

Multicompartment Modeling

( C p ) diff = A 1 e

(EQ 10-33)

i.e., the difference between observed and extrapolated drug concentrations in the
plasma declines exponentially with time.
Note (for reference only)

It is usually not informative to determine the microrate constant; but see one use
under the note on dosage regimens.

B 1 + A1
k 21 = ------------------------A 1 + B1

k 10 = k 21

k 12 = + k 10 k 21

2.4 Clearance and Volume

If model-independant equations can be used to define these terms, this is preferred.

a. Systemic Clearance (Cl) may be calculated by,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-10

Multicompartment Modeling

D Cl = ---------------------
( AUC )o

(EQ 10-34)

b. The volume terms are complex than in a one-compartment open model. There
are two terms of interest:

The apparent volume of distribution in compartment one ( V1 )


This is calculated using Eq. 6.

The apparent volume of distribution at pseudo-distribution equilibrium ( V )

This volume may be defined only in relation to the terminal phase ( phase), when
initial distribution has been completed.
D
V = -------------------------
( AUC )o

As V requires calculation of the total area under the plasma concentration against
time curve it is sometimes known as Varea .

c. Comparing Eqs. 9 and 10,

Cl = V

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 10-35)

11-11

Multicompartment Modeling

It may also be shown that,

Cl = k 10V 1

(EQ 10-36)

This follows as systemic clearance is always given by the elimination rate constant
out of the body multiplied by the apparent volume of distribution in the compartment from which drug leaves the body. Comparing Eqs. 11 and 12,

k 10
V = ------V
1

(EQ 10-37)

Note that k 10 (the elimination rate constant) is not the same as (the terminal
hybrid rate constant).

2.5 Bioavailability

Find ( AUC )0 using trapezoidal rule and, if necessary, the calculation for the terminal area.

t C
( AUC )o = ( AUC )o + -----p

(EQ 10-38)

This is a model-independent equation.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-12

Multicompartment Modeling

2.6 Dosage Regimens

The maintenance dose (D) is given by the same model-independent equation as


before,

D = ( C p ) ss Cl

Where ( C p )ss has its same previous definition.

The loading dose ( D L ) achieves a steady-state condition quite rapidly, but only
after initial distribution has been completed. It is given by the previous equation.

D
D L = -------------------------- 0.693N
1e

(EQ 10-39)

As may be expected, equations relating ( C max )ss and ( C min ) ss to ( C p )ss are as
before,

Note (reference only)

All dosage regimen equations strictly apply only when,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-13

Multicompartment Modeling

- 1 + k-----12
------ = 1

k 10
k 21

For digoxin Eq. 17 has a value of 0.947


For warfarin Eq. 17 has a value of 0.990
For cephalexin Eq. 17 has a value of 0.846

This is why, despite the fact that an open two-compartment model is better description of the pharmacokinetic of these drugs, a simple open-compartment model may
often be assumed for dosage regimen purposes.

3. Intravenous Bolus Administration: Compartment Two

It is not normally possible to measure drug concentrations in compartment two.


However, the mass of drug can be predicted based on the ddrug concentrations
observed in compartment one.

2.6 Dosage Regimens

X2 = B2 ( e

(EQ 10-40)

k 12D
Where B2 = ----------------( )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-14

Multicompartment Modeling

Note that the equations forms bears a similarity to that seen for plasma concentrations after oral administration inot a one compartment open model.

When t is large, e

<e

. Hence, Eq. 18 becomes,

X2= B2 e

(EQ 10-41)

This is comparted to the mass modification of Eq. 7,


X1= V1B1 e

(EQ 10-42)

Thus, when t is large the masses of drug in each compartment decline exponentially, and in parallel, with time. This indicates that initial distribution has been
completed and equilibrium attained.
If the value of X 2 reflects drug concentrations at the active site, the time of maximum concentration (and maximum pharmacological effect) is:
ln ( )
t max = --------------------

(EQ 10-43)

4. Others Dosage Forms


The equations become complex and it is therefore difficult to obtain useful parameter values without th eaid of a computer. Fortunately, because the complexity of
the equations is greater than the experimental accuracy of the assays warrants,
drugs that strictly require a mammillary model can be described adequately by an
open one compartment for the purposes of calculating dosage regimens.
4.1 Intravenous Infusion
The plasma concentrations at first rise faster than an open one compartment model
profile would suggest. Later, the rise is slower. The decline, following the cessation of infusion, is biexponential.
4.2 Oral Administration
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-15

Multicompartment Modeling

At a time just after t max the plasma concentration may exhibit a nose, when
compared to the profile of an open one-compartment model.

SELECTED REFERENCES

Riegelman, S., Loo, J.C.K., and Rowland, M., Shortcomings in pharmacokinetic


analysis by conceiving the body to exhibit properties of a single campartment, J.
Pharm . Sci., 57, 117-123 (1968).

Riegelmen, S., Loo, J.C.k., and Rowland, M., Concept of a volume of distribution
and possible errors in evaluation of this parameter, J. Pharm. Sci., 57, 128-133
(l968).

Benet, L.Z. and Ronfeld, R.A., Volume terms in pharnacokinetics, J. Pharm. Sci.,
58, 639-641 (l969).

Gibaldi, M Nagashima, R., ant Levy, G., Relationship between drug concentrations in plasma or serum and amount of drug in the body, J. Pharm. Sci., 58, 193197 (1969).

Metzler, C.M Usefulness of the two-compartent open model in pharmacokinetics,


J. Amer. Stat. Assn., 66, 49-54 (1971).

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-16

Multicompartment Modeling

Gibaldi, M. and Perrier, D., Drug eliminatin and apparent volume of distribution in
multicompartment systems, J. Pharm. Sci., 61, 952-954 (1972).

Gillette, J.R., The importance of tissue distribution in pharmacokinetics, J. Pharmacokinetics. Biopharm., 1, 497-520 (1973).

DRUG DISPOSITION: VOLUME TERMS

As apparent volumes of distribution are proportionality constants, and not physiological volumes, more than one term is of value.

1. Apparent Volume of Sampled Compartment ( V1 )

This relates the concentration of drug in the sampled compartment with the mass
of drug present in that compartment.

It may be measured after an intravenous bolus dose:


D
V 1 = ------------( Cp )o
or

D
V1 = --------------------------
K ( AUC )o

(EQ 10-44)

(EQ 10-45)

It may be measured after an intravenous infusion by:


( X1 ) ss
Q
V 1 = -------------- = ------------------( C p ) ss K ( C p ) ss
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

(EQ 10-46)

11-17

Multicompartment Modeling

2. Apparent Volume at Pseudo-Distribution Equilibrium ( V )

This volume term (sometimes known as the apparent volume of distribution of the
drug in the body) requires the assumption that the drug is evenly distributed
throughout the body. The assumption is not true in practice. Thus V can only be
defined in relation to the terminal phase ( -phase) when equilibrium has been
attained; the equation is analogous to Eq. 2.

D
V = -------------------------
( AUC )o

(EQ 10-47)

3. Relationships Between Apparent Volumes


By secondary alebraic definition, a clearance (Cl) is always given by the first-order
rate constant for removal of drug from the body multiplied by the apparent volume
of distribution on the drug in the compartment from which the drug leaves the
body:
Cl r = k u V 1

(EQ 10-48)

Cl m = k m V1

(EQ 10-49)

Cl s = KV 1

(EQ 10-50)

However, systemic clearance is measured by


D Cl s = ---------------------
( AUC ) o

(EQ 10-51)

Comparing Eqs. 4 and 8, and rearranging,


Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-18

Multicompartment Modeling

Cl s = V

(EQ 10-52)

K
V = ---V
1

(EQ 10-53)

Comparing Eqs. 7 and 9,

Selected References

Riegelman, S., Loo, J.C.K., and Rowland, M., Concept of a volume of distribution
and possible errors in evaluation of this parameter, J. Pharm. Sci., 57, 128-133
(1968).

Benet, L.Z. and Ronfeld, R.A., Volume terms in pharmacokinetics, J. Pharm. Sci.,
58, 639-641 (1969).

Gibaldi, M., Nagashima, R., and Levy, G., Relationship between drug concentrations in plasma or serum and amount of drug in the body, J. Pharm. Sci., 58, 193197 (1969).

Perrier, D. and Gibaldi, M., Relationship between plasma or serum drug concentration and amount of drug in the body at steady state upon multiple dosing, J. Pharmacokin. Biopharm., 1, 17-22 (1973).

Oie, S. and Tozer, T.N., Effect of altered plasma protein binding on apparent volume of distribution, J. Pharm. Sci., 68, 1203-1205 (19793).

Li;vxrX LLrLLxLS

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-19

Multicompartment Modeling

Drug is usuallo sampled from the centr->l compartment, designated compartment


one.

1. Laplace Transforn for Compartment One

As,l

(in)(dS 1)

where As,l is Laplace Transform for mass of drug in comPartment one

s is the Laplace Operator in is the input function

dS,l is ehe disposition function for compartment one

2 InDut Functions
.

N ehat input need not necessarily be to compartment one.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-20

Multicompartment Modeling

2.1 IV Bolus

(in) - D
where D is the dose

2.2 IV Infusion

Q (l-e sb)

where Q is the zero-order infusion rate, b-t when e<T, b-T when eiT, and T is the
eime of cessation of infusion.

2-3 First-order Absorption

(in) . <

(s+ka)

where ka is first-order absorption rate constant, ant F is the fraction of D ultimately


reaching the general circulation.

2-4 Dissolution and Absorpcion (type 1)


(in) - krkaFD

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-21

Multicompartment Modeling

(s+kr) (s+ka)

where kr is tirst-order dissolution rate constant.

2 5 Dissolution and Absorption (tvpe 2)

(la)

ka(l-e sb)

s(s+k>)

where ko is zero-order dissolution rate, ceasing at time T.

2-6 Others
These may be formed by adtition of functions 2-1 through 2-5

e.g., (in) - D + Q(l-e~Sb)

This denotes the simultaneous commencement of an I.V. bolus and infusion.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-22

Multicompartment Modeling

3. Disposition Function for Comcartment One


A driving force compartment has one or more exit rate constanes; for

instance, in compartment i, ehe sum of ehe first-order exit rate constants

is Ei.

As,~

n~

kca

a +

where q is the compartment into which input occurs, n is the number of driving
force compartments,

i,j, and m are counters (maximum value of n),

kql is the first-order rate constant for transfer of drug from input compartment eo
compartment one,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-23

Multicompartment Modeling

kl; and kjl are the first-order rate constants for drug transfer from compartment 1 to
compartment j, ant vice-versa.

3-1 Using the disposition function


(a) If q-l, ehen kqlsl

(b) Tr (Pi) and fT (Pm) are coneinued produces. ~e value of Pi (or Pm) equals one
when thc counter i (or m) takes on a forbidden number. For example, i-l is forbidden in the numerator, ant m-l and m-j are forbidden in the denominaeor. 3-Z
E.camples

(a) one-compartmene open model (n-l,q=l)

ds,l = 1

(Eq.l)

(b) Two-compart:.ent open models (n-2.q=1)


(s+E~)

ds , 1

(s+El) (s+E2) - kl2kx

(c) Three-compartmene open models(n-3,q-2)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-24

Multicompartment Modeling

dS,l

k21 (s+E3)

(Eq.2)

(s+El) (s+E2) (s+E3) - kl2k21 (s+E3) - kl3t31 (5tk2)

(Eq.3)

3-3 Simplifying the Denominator The number of exponeneial terms in ehe final
ineegrated equation will be equal eo the number of driving force compartments
(n). This is also equal to the maximum power to which the Laplace operator (s)
would nppear if

the denominator were multiplied out. Hence, the denominator is simplified n to


become iXl (S+ki), where ki is a composite first-order rate constane.

(a) ds,l

(b) d5 l

(c)

ts.l

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-25

Multicompartment Modeling

(s+kl)

(s+E2)

(s+kl) ts I k2)

k21 (s+E3)

( s+kl ) ( s+k2 ) ( s+k3 )

(Eq.la)

(Eq.2a)

(Eq.3a)

The exact meaning of [i for any model depends on the equalities evident in the
denominaeors. Example for (b):

(s+kl) (s+k2) - (s+El) (s+E2) - kl2k

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-26

Multicompartment Modeling

4. Method of Partial Fractions Ihis method is used to solve (integrate) a Laplace


Iransform providing there are no repeating factors in the denominator. Example:
no 52 or (s+ki)2

l 1 Prepare che Laplace Transform


Example: I.V. bolus into compartment one of two compartment model

( s+kl ) ( S+k2 )

4-2 Obtaining the Roots of Denominator Factors

If the factor is s, the rooc is zero

If ehe faceor is (s+ki), the root is

4-3 Ridden-Hand Method

(a) Deal with each factor of ehe tenominator in turn.

(b) Cover the factor with a finger, and remember its root.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-27

Multicompartment Modeling

(c) Wherever the Laplace operator(s) occurs in the uncovered transform, subseitute
the root for s.

(d) Multiply the resule by eses again substituting ehe rooe for s

(e) After doing (b) through (d) for each factor, simplify.

Example:

X1 D(-kl+E2)e-klt - + D(-k2+E2)e~k2t

(-kl+k2)(-k7+kl )

or C1 - D (kl-Ez)e kl + D (E2-k2)e 2

V1 (kl k2 )

or

C1 - Ale 1 + A2e-k2t

In this example the meaning of A1, A2, kl, k2, and E2 depend on the form

of the two-compartmental model.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-28

Multicompartment Modeling

5. Laplace Transform for Peripheral Compartments

This is obtained by the following procedure, which is analogous eo that

employed when using the Laplace Transform table.

(a) Write the differential rate equation.

(b) Take the Laplace Transform of each side of the differential rate equation, using
the table where necessary.

V1 (kl-k2)

(c) Algebraically manipulate the transformed equation until an equation having


onlv one transformed dependent variable on the left-hand side is obtained.

(t) Substitute for anv known transformed dependent variables on tlle right-lland
sidc of the equation.

(e) Solve (integrate) bv the method of partial fractions (tlle hidden 1land), and
simplifv.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-29

Multicompartment Modeling

6. btethod if Denominator Contains the Factor 52 This may apply to terminal


compartments, such as urine, following an I.V. infusion. The hidden hand
method cannot be used for the factor 52 in the denominator as it has no simple
root.

6-1 Example (n=2, q^l, exit from compartmenr one):


aS,U - kloQ.(l-e~sb)(s+Es)

52(5+kl) (s+k2)

where klo is the first-order excretion rate constant from compartment one.

xu ~ kloQ.E2b + ......
klk2

where Xu is the cumulative mass of drug excreted into the urine.


The other factors can be used as before in the hidden-hand method.
6-2 Example (n-3, q-l, exit from compartment one)
aS u kloQ.(l-e 5b)(S+E2)ts+E3)
s2(s+kl)(s+kv)(s+k3)

Xu t kloQ- E2Elb I

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-30

Multicompartment Modeling

klk2k3

REFERENCES

L.Z. Benet, General treatment of linear mammillary models with elimination from
any compartment as used in pharmacokinetics, J. Pharm. Sci., 61, 536-541 (1972)

D.P. Vaughan, D.J.H. Mallard, A. Trainor, and M. Mitchard, General pharmacokinetic equations for linear mammillary models with trug absorption into peripheral
compartments, Europ. J. Clin. Pharmacol., 8, 141-148 (1975).

D.P. Vaughan and A. Trainor, Derivation of general equations for linear mammillary models when ehe drug is administered by different routes, J. Pharmacokin.
Biopharm., 3, 203-218 (1975).

Two-Compartment Model-l

Prior inputs focuset on one-compartmcnt models, but many drugs arc charactetizet
bettcr by multicompartmcnt motek. In the following three inputs, we shall bricfly
tiscuss multico~_nt motek ant prcstnt a few apB plicadons. Multicompartnent motcis are not uset as fo quentlg u the one-compartment model in therapeutic trug
monitonng, panly because they arc more tifficult to construct ant apply.

Gencially, muldeaw models arc appliet when th,e natural log of plasma drug consentration vcrsus time is not lincar afier an intravenous tose or when thc plasma
concentration versus time psfilc cannot bc chu~ by a single cxpooential function
(i.c., C, - CO e~~). Wben the In of plasma concentration vcrsus timc is not a

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-31

Multicompartment Modeling

stnight line, a multicompartmcnt model must bc constructet to tescribe the change


in concentrations over time.

Of the mul

models, tbe two-compartment motel is tnost fxqucntly uset. lunis model usually

of thc weU-perfia

tissues ant penpbexal compartment of less weU Erfuset dssues (such as muscle
ant fat). hgure 23^ shows a diagram of thc two-compartmcnt model afir an intravenous bolus tose, where:

consists of a central

Xfamount of diug in centnl comva XP s amount of drug in psipheral cowt

K,2rate const nt for transfcr of drug from cd-compartment zo petipheral compartment rne subsaipt 12 irldicses tr nsfcr from thc first (cd) to the second
(peripheral) compattments.

K2, - rate constant for tgansfcr of drugfrov peripheral computment to central comp
rtment lbe subscript 21 indicales tr nsfcr from Ulc second (periphaal) to the fint

Xo~

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-32

Multicompartment Modeling

Kl2

K2l

+ Klo

zgre23~ Gr phic reprtsentation of a zwbcompattment model.

(centri) compartments. (Nott h Kz2 and K2,calkd micxnts.)

fintvder climinX aue consunt (similar to tbe Jr uxd paviously), iting elimiXn of
dmg out of tbe caul ~ into urine, feces,

esc~

A log plasma conscatration versus time curve for a two-compattment model shows
a curvilinear profilea atrved potoon followed by a straight li=. This biexponential curve c n bc described by two expoKntial tcrms (Flgure 23B). lEc phases of
the curve may reprcstnt rapid d1stributioo to organs with high blood flow (central
compuenent) and slower distnbution to organs with Ess blood flow (penphcnl
compartmcnt).

Mer thc intovenous injection of a drug that follows a t_ model, thc drug consentrations in all fluids and dmms associated with tbc central compartmcnt declinc morc
rapidly in tbc distributioo phasc thao during the post-diwibubon phasc. ARcr sornc

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-33

Multicompartment Modeling

tirnc, a pseudocquihEutn is attained betwcen thc ccotral compartmcot and thc


dssucs atid fluids of toc pc ipheral compO thc pl sma coocentration vcrsus timc
ptnfile is thco chaserized by a linear pnmcess.

For many drugs, suco as aminoglycosides, thc distributdoo phast is vcry shott (e.g.,
1920 mtn). If serum consentradons are measured after this phase is compited, toe
ceotral compartmcot can be ignorcd and a one-compartrKnt model adequatcly
repttsents the serum coocentratioos observed. However, for drugs such as vancomycin, thc-distribution phase lasts 1-2 hr after an intravenous dosc. If plasma concentrations of vancomycin are determined within the first hour after a dose is
given, thc nonlincar (multiexponential) decline of vancomycin concentrations
must bc considered.

REVIEW PROBLEMS

23.1. In the twocompartment model. Xt wpresents the

23.2. The log plasms concentration vcrsus time curve for a two-compartment
model is reprcsented by a (bicxponential or monoexponential) cuNe. (Sclect one.)

23.3. The first portion of the log plasma concentration ver. sus timc cune. where
the log concentration r;tpidly declincs. is lomwn as the

phasc.

23.4. The final. Iinear portion of the curve is the phase.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-34

Multicompartment Modeling

11- 1
~1

I>Pur 2t 61

70 [ So

~i

~m

~.

F*wf 238 Four st ges of drug distribution nd eliminatioo following rapid intravenous injectiott. Points I, U, m, nd tv (ript) corrcspond to the points oo the plasmx
concentntion curve (leR). Point 1: The injection has just becn compicted, and drug
density io the cd compartment is hipcst. Drug distribution and elimination hve just
begun. Poin~ 11: At midway through tbe distributioo process, the drug density in
thc central compartment is falling r pidly, dulioly owing to rapid drug distributioo
out of the centd ccrnpartment into the peripheral compartment. The density of drug
io the peripheral compartment has not yet mched tht in the central compartmcnt.
Poixt 111: Distribution equilibrium h s been attained, and drug densitics in the
centd and periphed compartments arc appgoximately equal. Drug distribution in
both directioos contioucs to talze place, but the ratio of drug quantitics in toe centel
and peripheral compartments remains constant. At this point, the major determinant of drug disappearance from thc central compartmcnt becomes the elimination
process; previously, drug disappearance W&S determined mainly by distribution.
Poi/s : During this elimination phase, the drug is being -drained from both com-

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-35

Multicompartment Modeling

partmcnts out of the body (via the central companment) at approximatcly the same
rate. (Reproduced, with permission, from Grecnblatt DJ &nd Shader Rl, Phrmacolsinetics in clinical practice, W.B. Saunders, Philadelphia. PA, 198S.)

TwoW M~

lo this input, wc soall apply mathematicaS principles to toe two-compartmcnt


model to calculatc useful poarmacokinetic parameters.

horo tiscussioo of the ooe-comparaneot model, we koow that the climination zte
coostant (J[) is estsmated fxm the slope of the lo pbsma coocentration vcrsus ame
curve. However, in a two-canzrg tnodel, wose the lo plasma coocentration versus
time curve is curvilinear, the slope varics, tepcndiog on waich porioo of toe curve
is cxamined (Flgurc 24A).

In a two compartment model, the tenninal slope from the pos,t-digributive phase of
the curve may bc backextrapolata~ to axnc zero (T). The oegative slope of this line
is teferret-to as beta (O, aot ,B is the tennioal eliminatton tate coostant of the trug.
The iotesept of this lioe on the In plasn coocentration axis is koown as B and is
uset in vanous two~cotnpeneot equations.

Bc~ is similtr to K in to t it vsents the tsminal elim meion r te constant. From it, a
half-life can be cal~ culS

T%, 0.693

is

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-36

Multicompartment Modeling

which is refenet to as the beta haSf-Ute.

lEroughout the ame th_t trug is present in the boty, tistribuiion takes place between
the central ant peripheral compartmenu. We can calculate a ratc of tistribuaion
using the mct rcsidxals. This methot estim tes the cffect of distribution on the
ovcrall plasma concentration curvc and uses thc diZfcrcncc between thc cffect of
climination and thc actual plasma consentrations to determinc thc distribution rate.
In the In concentration versus vime curvc in Flgure 24A, the slope of the initial
portion is determined prim--arily by the distribution rate while thc tenninal portion
is determined primarily by thc climination rate.

E <~

100

50

o
S~

PodwDiattibutlon
Ph~

Timo Figsot 24A Plasma druy concentralions with a two-compartment model atter
an imravenous bolus do*c.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-37

Multicompartment Modeling

The methot of residuals may be used for calculating phamiacoMinctic parameters


of thc two-compartmcnt model. FuSst, b cl:-extrapolate the terminal straight-line
portion of tbe curve (Flgure 24B). If w, s. y, and 2 are actual, detennined concentration time points, let w, ~, y, and 2 xpresent points on the new (extopolated)
line at the same times that tbe aaual points were obscrved. These newly generated
points xpresent the cffect of clim~ ination alone, as if distribution had been instantaneous. Subtnction of the extrapolated points from the corresponding actual
points (ww, X~, etc.) yields a new set of pbsma concenti-ation points for
each time point. If we plot tbese new points with the appmpriate times, we generate a new line, the residual line (hgure 24C).

The slope of tbe xsidual line isst, and alpha (a) is the distribution rate constant
for the two-compartment system. The intrcept of the residual line is A. Therefore,
witb the coocept of residuals, we attempt to separate the two pwocsscs of diseribution and climin~ jon.

Ist us now pn~cood through an exampic, applying thc metbot of xsidtis. Draw tbc
plot for thc following cxampb on somilog gnph paper. A dosc of dnag is ad

10.0
50

:!

Z c 10
o
fL Q

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-38

Multicompartment Modeling

Tlmo F~241R Mcthod of xsiduals.

100
5e

Bz

\ Sbpo = a

s -R

Timo

F;

24C Dctermination of the rcsidual linc.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-39

Multicompartment Modeling

62

InPtJT 24 63

ministered by rapid intravenous injection, and the tollowing concentrations result:

Tkne afttr
Dose (hr)
0.2S
O.S
1.0
I.S
2.0
4.0
8.0
12.0
16 0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-40

Multicompartment Modeling

Plasms
Concentration
(u~/ml)

43
32
20
14
11
6.S
2.8
1.2
0.S2

A linc is trawn connecting the last four points and intcnecting the y-axis. Then, for
the first five points, cxtrapolated values can be cstimated at cach time (0.2S, O.S,
1.0,l.S, and 2.0 hr). If the extrapolated values from the actual plasma concentrations are subtracted, a new set of points is generated (resitual concentration points)
as fts w~

Tlnse dir
Doz (hr)
0.2S
O.S

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-41

Multicompartment Modeling

1.0
1.5
20

Pbsma Concentratlon (>g/ml)

^>e

Exupol ted Residud

14.S

28.S

13.S

18.S

12.3

7.7

1 1.0

3.0

!O.0

I.D

The zsidual concentrations are then plotted (on semilog paper) versus time, and the
slope of that plot equals 1.8 hr~t. When the negative is dropped, this slope
equals sx; we observe from the plot that the intercept (A) of the line is 4S Fg/ml.
We also can estimate a from the slope of the terminal straight-line portion (equal to
0.21 hr~ ~) and 8 (equal to IS 1lg/ml).

Alpha (ex) must be greater than beta (a), indicating that drug removal from plasma
by distribution into tissues proceeds at a greater rate than does drug removal from

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-42

Multicompartment Modeling

plasma by eliminating organs (e.g., kidncys and liver). rhc initial portion of the
plot is steeper than the terminal portion.

REVIEW PR08LEMS

24.1. Dnw a log pbsma concentntion versus timc profile for a drug Oinimed by the
intravenous bolus nmute and best durizZ by a two-companrnent modeJ (Figure
24D).

242. Tbe slope of the tenninal phase of the above. plot equals

243. Tbe inucept of thc tenniial portion on tbe In pbsma concentstion axis is tenned

>.~ sca(g)~ tbe tenninal const nt of the dmg s it leaves the body.

24.S. One w y to calculatc a distnbution zate is to use tbe metbod of

24.C. Tbe fint step in the metaod of residuals is to.

- the telminal straight-line portioa of the curve. 24.7. The extryolatd points aw
subtmed from tbe actuaJ observed at the correspoading times.

24.8. Tbe slope of the residual line equals

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-43

Multicompartment Modeling

24.9. A is tbeof tbe In


plasnu concenamtion axis by tbe
line.

2410. Tbe coocept of residuals attempts to separate tbe two

processes ofand

100

50

ca e

Z c 10
FQ

Fkwe 24D

Tim
ljg

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-44

Multicompartment Modeling

Two-Compartment Model-3

The estimations of A, 8, ss, ant t perforrned in tbe last input are useful for predicing plasrrs concentotions of dmg ch~nzi by a two-compartrnent model. For

awrst rnodel (Flgureo2SA), we know th t thc plaSsma concentration (C) t any


time (t) can be dessnbet by

Cf ^ Ce e t

where CO is the initial concentration and g is ttne climination rate. Thae two-compartment rnodel (Flgure 2SB) is thc surn of two linear components, reprcsenting
distnbution ant elimination (Flgurc 2SC).

In thc sarnc w y, we can dctrminc dnag consentration (C) at ny tinx (t) by iding thc
two linear components. In cach casc, A or B i-s uset for CO, ant ex or z is used for
XY. Therefe

C, s

+- 8 e~*

ThiSs equation is called a biexponential cquation bccausc two cxponents rc


iwaled. With thc onos

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-45

Multicompartment Modeling

wrunt rnodcl (intravenous), wherc:

C. s Ce e ~

2B

100
501
CO
10
S

Timo Fzwe 25A Pls dmg concentrations with a one com putment model aher an
insvenous bolus dose (first order elimination).

100
50

,o

z -, 10
5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-46

Multicompartment Modeling

~c
o

Timc ft 25B Plasma drug concentrations with 3 two compartment model after an
intnvenous boluXs dose sfirst-order elimination) .

100
50

E;

tO

Sz

Tlmo
Fw 25C lOr azmpo~s of a twozxpo~ (>

t) model.

~nentid becausc thc linc is


t tnodel, diffcrcnt volume of

thc equation is

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-47

Multicompartment Modeling

describet by onc exponents

E or thc twowrat dWibution psramctrs cxist thc centol volurnc {V), thc cxtrapolated volunc (V,~ ,), tbc volunc by arca (V,,, also lcnown as V~|), ant thc stcadystatc vohunc of diwibuX (V,,). Each of thcsc voluncs rclste to diffcrcat undertying
assumptioos.

As in thc onc-cornpenent rnotcl, a volunc can bc calculated by

V dose dox
,~ + B Co

For thc two-compartrnent model, this volurne would bc cquivalent to thc volumc
of the central compartment (V). Thc Ve rclates the amount of drug in thc-central
compartmcnt to the concentration in the central compartment. In thc two-compartrnent model, CO is determined by cxtrapolating back to thc y-axis from the upper
or initial straight-line portioo of the plot.

When we calculate the extrapolated volume of distribution (V,x,,,p), we assume


that instantaneous distribution has occurred. The effect of the iniial distribubon
phase is ignored:

da
B

V_

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-48

Multicompartment Modeling

wherc B is the w-intetcept of thc line extrapolated from the terminal portion of the
curve. This volume of distribution determination may not provide a useful volume
term since it ovcrsimplifics the two-compartmcnt model and disregards thc distribution phasc.

Another volumc (V,,O or V~) is detcrmined from the area under the plasma concentration vcrsus time curvc and thc tcrminal climination rate constant. This volumc is related as follows:

114

lNvts S 65

vffi

dox
=

CL

ffi x AUC3

va

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-49

Multicompartment Modeling

This calculation is not subject to thc ovcrsimplification of V,S,,p, but it is affected


by changes in clearance. The V,,,, relates the amount of drug in the body to the
concentration of drug in plasrna in the post-absorpion and postwdisttibuiion phase.

A ffnal volume tenn is the volume of disttibution at steady state (V,,). Although it
is not affected by changes in drug eliminadoo or ckarance, it is more difficult to
calculate. One way to estimate Vs, is to use the two compartment microconstants:

V,Ve + ~2vf

21

or it may be estimated by more complicated methods using AUC.

Since-different methods can be used to calculate the various volumes of distribution of a two-compartment model, you should always specify the metbod used.
When reading a pharrnacolcinetic study, pay particular attenion to the method for
calculating thc volume of distribution.

REVIEW PROBLEMS

2S.1. The terminal eliniination rate constant tn a twoaconF putment model is

2S.2. For the two-compurtment model. complete the equo tion describing the
relazionship of plasma concentration with time: C, -

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-50

Multicompartment Modeling

25.3. (True or False) The equation describing elimination afer an intravenous bolus
dose of a drug charauerized by a two-compartment model lequires two exponential
terms.

2!;.4. A patient is given a 500-mg dose of drug by intravenous injection and the
following plasma concentrations result:

Plssms
Time snerConcentrstion
Dese (hr)ItsSml)

Oo ss
0.7S
lS

3
,6

72.0
46.0
33.0
26.3
20.0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-51

Multicompartment Modeling

16.6
1r.2
9.0
5.0
_.7
n Rr

Plot the points on semilog paper Ithree cycle) and deterTnine the following: a. ~. b.
B. c. Residual concentrations for the first five points. d. A.

. t.

Prodictod Dlssma concentration at 1.2 hr after the

dose.

S- V,.

h. V,, (if AUC = 131.S mg/L x hr and dose SOO mg).


~ A#=W{~ SGS 2

PRi4CTlCE SET 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-52

Multicompartment Modeling

The following pxbiems are for your xview. Dcfinitions of symbols ant Icey equations = pxvitet hc~:

~w climinton nte cowt

C, s pbSllk tn~ _jUSt Aher a single inuvenous injectioQ

- bese for the nxl log f~|eion - 2@71S

- nte of tose ~ion (msy be cxptesset as


milligsams per bour in the sense of a coQtinuous
infusion or u trug tose divitot by itifusion tune
for intemtittent infusions)

voluxne of tistribution

C_t - pcalc p1ssfu a ocentntis

C_ tnmugn plsCOOCCD

a steaty state

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-53

Multicompartment Modeling

a stcady st te t duFn of intsvenous inAlsion

For muldpk dose, intennittent, intnvenous bolus injeciiOQ g stF st~

k V (I - e~t)
C_Cw e~t

For muletplffbse~ intctmiu, intnvenow itafi~ioo

C - t (I ~ e t~)
~ VK (Ie~tD

C_ - Cp e~tt~4

Fot contitiuous infusion before stee st te is reacbed:


C - V^r (I - C-t)

For continuous infusion t stcaty stac

C.4. 4
Vt Ws

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-54

Multicompartment Modeling

PS2*1. A 60-kg patient is begun on a continuous intravenous infusion of theophylline at 40 mgtbr. a Forty-eight hours after beginning ttte infusion, the plasma concentniion is 12 ~/ml (12 mg/L). If we assume that tbis COncentRtiOD is tbe stcady
state, wb t is the tbeoobylline clearansc?

b. If the volume of distribution is cstimated to be 30 L, what are the X and halflife?

c. Since we kww V and t. what would the concentntion bc 10 hr aftcr beginning the
infusion?

d. ff the idision i continuet for 3 days ant then discondauet, what woult tD plamS
consentm tion be 12 hr ~r stopping the infwiont

e. If tne infusion is continuct for 3 d ys at 40 mg/ hr ant the stcaty-statc pluma concenttation is 12 Fg/tnl, WDat rate of trug infusion would liltely xsult sn a concentradon of 18 ~/ml?

f. ARcr the iocosed infusion nte above is begun, how bog would it tic to tuch a plu
coo~ cenudon of 18 Fgiml?

PS2< A 60-log p ekat is st rted on 80 mS of gentunicin ewety 6 br in I*br infusiagL

L If this pSt is us led to have an avenge V of 15 L ult a normal half-life of 3 hr,


wtuat will be the pealc plm cocenttadon at stcady stuc?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-55

Multicompartment Modeling

b. After thc fifth tese. a peak plasma concentratlon (dsawn at thc ent of thc infusion) is S Ag/ml ant thc ttnugh consentration (drawn right befott thc sixth tosc) is
0.9 Ag/ml. What is thc patients xtual gentamicin half-life? What is thc xtual volume of disvribution?

c. For this patient, what dosc should bc administen:d to reach a new steady-state
peak gentamicin concentration of 8 Fgiml? At this dosc. what will bc thc steadystate trouQh concentration!

II_q

tFoD~ sU

~ Tvo-ConPart-ent Open Hodel

tatlente ufferln~ chronle tenal fellure often require h _ odiolyele. Drug~ cy be


~dxinletered by Injeetlon Into the wenoue lde of the he odtcl~ser ~chine

Such e rituetlon vax ~ecerlbed by L tourneeu-Scheb t 1 (Int. J. Clln. tharv col IS


116-120 {1972)) for lx pctlente vho received n introvenoue done ot gent _ letn
(90 ng). The neen etru- concentrctleno et sent~nicin (C~) hovvd blexponenttel
deellne vith tl t (t).

Ce

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-56

Multicompartment Modeling

(-~lllter)(oln)
o 60

10

5 75

20

5 25

lO

4 80

40

50

50

3 95

90

3 40

150

3 10

180

2 90

240

2 55

28S

o. Caleelete the ter lnal half-llfe (t~) the hybrid rete eonetento

nd O nd the coefflelente (~1 3 Sl) for gent- leln to theeo he-odlulyal~ putlente

b. tor ubJecte vith noreel renal functlon. the yete-le elearence (Cl~) of gentanletn
le pproalaetel1 0.041 llter/ In. Sec uee 98X ef Cl~ le due to excretlon of
unchanged drug. renel tciluro utll ~rkedly effect ~ent~lcin clecr~nce.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-57

Multicompartment Modeling

Show vhether the petlent~ undergoinfS he odlelyele xhlbit o nor-ol gentcnicln


yett-le elexr~nce.

Calculete the two pperent volune ot dletrtbut10n terwe (V nd V )

nd the ell fnetlon rete eonetont. Uhet frectton of tho gt ted eln ln the body fter 3
hr nlght be la the peripherel ee part~ent7 At vh t tbae doee the gent _ Icin in the
peripherel ee pert~ent rench

~t

4. Under the eondittons of h odSxlyxte. coleulete the doxe tD) of

sentcoleln tlSch vould be dolaletered very S hr {1) In order to ~alnteln n


everogew teady-ctete erus concentretlon of 4 g/llter.

. Wlthout the hc odielyrer two ~ale petlente ach xhlbited creetinine eleorence
(Cle ) of 5 llwin; the norx~l velue le 117 t 20 I/nin. A~using that the deereFeed
Clcr {e due to decresce In glo-eruler filtretloo rcte vhst vould h-ve beeD the renel
clecrxnce (Clr) of gent~elcia In there two patlentn hed they rz cined vithout the heodlolysert Uhct done vould then need to be dsinletered every hr to n Intoin (C~ t
4 tert Co pcro your eouer wStb tho thy~telea e Deek Refer neo.

Vancomycin is an antibiotic used in the treatment of endocarditis in patients allergic to penicillin. It is poorly absorbed orally and acute pain is associated with intermuscular injection. I.V. is the route of choice. After a 1 gm I.V. dose the following
data is observed:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-58

Multicompartment Modeling

Time (hours)Concentration (mcg/ml)

.5

51

1.0

36

1.5

28

2.0

23

3.0

18.5

4o

16

6.0

12.5

8.0

99

12.0

6.25

FIND: a) A1 ;

b) alpha
c) Clearance ;

d) * of drug in peripheral compartment at equilibrium .

Clearance:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-59

Multicompartment Modeling

In normals, Erythromycin is cleared 90% by metabolism and 10% by the kidney.


Also, it is 90% protein bound at therapeutic levels. Welling and Creig (JPS 67,
1057-9,1978) reported an increase in the half life from 2.0 to 2.3 hours while also
reporting an increase in clearance from 275 to 485 ml/min and an increase in Vdss
from 57 to 100 litres when comparing normals to uremic patients. (Uremic patients
suffer from inveased concentration of urea as a result of severe renal failure.) A
physician has just called you and asked you to explain how he could have seen an
increase of 15% in the half life and a 75% increase in clearance at the same time
and what is the impact of this on antibiotic therapy for his uremic patient. In clear
concise English (not techno-bable), prepare a short written answer to this request.
Keep in mind that the man who requested the information is a medical professionat
intelligent, and very busy.

The following data was collected from a normal patient in the revious study (Welling, op. cit.) following an IV bolus injection of 500 mg of erythromycin (as lactobionate salt ).

time (hr)concentration (mcg/ml)timeconcentration

12 4

3 2.6

6.4

4 1.9

4.0

6 1.2
8 0.4

~.

Fmd the peak time in the peripheral compartment.the fraction of the drug in the
peripheral compartment at four hours.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-60

Multicompartment Modeling

Two compartment model

Spectinomycin (TROBICIN^TR^T) is an aminocyclitol antibiotic shown to be


active against most strains of NEISSERIA GONORRHOEAE at a minimum
inhibitory concentration of 20 mcg/ml. The usual adult dose is 2 g (4 g in areas of
known resistance) given I.M. through a 20 gauge needle. Initial studies were done
by the company to determine the pharmacokinetic parameters of the drug. The data
from a single IV Bolus dose of 0.5 g is as follows.

Time (minutes)Concentration
10

63

20

51

30

43

45

35

60

30

120

183

240

7.6

360

3.2

FIND:

(5 points) a) A^V1^V =
(5 points) b) B^V1^V =
(5 points) c) Clearance =

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-61

Multicompartment Modeling

(5 points) d) t max in the peripheral compartment =


(10 points) e) % of drug in peripheral compartment at equilibrium =

Spectinomycin (TROBICINR) is an aminocyclitol antibiotic shown to be active


against most strains of NEISSERIA GONORRHOEAE at a minimum inhibitory
concentration of 20 mcg/ml. The usual adult dose is 2 g (4 g in areas of known
resistance) given I.M. through a 20 gauge needle. Initial studies were done by the
company to determine the pharmacokinetic parameters of the drug. The data from
a single IV Bolus

dose of 0.5 g is as follows.


Time (minutes)Concentration
10

63

20

51

30

43

45

35

60

30

120

18.3

240

7.6

360

3.2

b} B c) Ciearance d) t max in the peripheral compartment e) % of drug in peripheral compartment at equilibrium

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-62

Multicompartment Modeling

The following information is offered from a 142 mg IV bolus dose of grisiofulvin


given to a 73 Kg man.

Time mcg/ml
1

1.67

1.22

.97

.83

.66

.56

12

.42

18

.27

24

.17

30

.11

Find A1, B1, alpha, beta, K10, K12, K21, Peak time in the peripheral
compartment, % in the peripheral compartment at equilibrium.
Can you assume that this drug can be estimated by a one compartment
model upon multiple dosing ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-63

Multicompartment Modeling

Quinidine is current-ly used to treat ventricular and supraventricular arrythmias.- It


is available as a sulfate, gluconate, and polygalactouronate which contain 83%,
62* and 60* by weight free base (pRa 8.6). Qunidino sulfate is available in 200,
260, 300 and 325 mg tablets, while quinidine gluconate is available in 325 mg tablets. The following pharnacokinetic parameters are reported by Ueda:

PARAMETER
C1 renal (ml/min/kg)

C1 hepatic (ml/min/kg) V1 (L/kg)

V beta (L/kg) F (oral)

t 1/2 alpha (min) t 1/2 beta (hr)

MTC (mic/ml) NEC (mic/ml)

Population Ave (+ SD)


0.93 (0.52)
3.26 (1.74)
0.66 (0.38)

2.61 (1.10)
0.71 (0.16)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-64

Multicompartment Modeling

6.69 (4.03)
6.44 (1.63)
8.53 (0.81)

1.85

(0.19)

% bound to protein80.00(2.50)

During the last pharmacokinetic exam you noticed ome cardiac problems.
Wh n you ch-cked with your physician, He prescribes quinidine. What
dose should you be on?

Later, he diagnoses mononucliosis from lack of sleep and poor ating h bits s weli
as cardiac arrythmias. Your liver funcion has dropped to 60* of normal. He prescribes quinidine for you. What is the dose that you should be on ?

ie. -

ll-3

Quinidine is currently used to treat ventricular and supraventricular arrythmias. It


is available as a sulfate, gluconate, and polygalactouronate which contain 83%,
62% and 60% by weight free base (pKa 8.6). Qunidine sulfate is available in 200,

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-65

Multicompartment Modeling

260, 300 and 325 mg tablets, while quinidine gluconate is available in 325 mg tablets. The following pharmacokinetic parameters are reported by Ueda:

PARAMETERPopulation Ave (+ SD)


C1 renal (ml/min/kg)0.93 (0.52)
C1 hepatic (ml/min/kg)3.26 (1.74)
V1 (L/kg)0.66 (0.38)
V beta (L/kg)2.61 (1.10)
F (oral)0.71 (0.16)
t 1/2 alpha (min)6.69 (4.03)
t 1/2 beta (hr)6.44 (1.63)
MTC (mic/ml)8.53 (0.81)
MEC (mic/ml)1.85 (0.19)

% bound to protein 80.00 (2.50)

During the last pharmacokinetic exam you noticed some cardiac problems. When
you checked with your physician, He prescribes quinidine. What dose should you
be on?

Later, he diagnoses mononucliosis from lack of sleep and poor eating habits as
well as cardiac arrythmias. Your liver funcion has dropped to 60% of normal. He
prescribes quinidine for you. What is the dose that you should be on ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-66

Multicompartment Modeling

Methotrexate is a cytolytic used for the treatment of acute leukemia and other
forms of cancer. After a a 400 mg/kg dose the following data was recorded for a 12
y/o boy.

Time (hours)
6
12
18
36
48
60
72
90

Concentration (mcg/ml) 360 70 15

2
1.2
0.46
0.36
0.15

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-67

Multicompartment Modeling

bA) B c) Clearance d) % of drug in peripheral compartment at equilibrium

PROBLEM SET

zumper w~wFS
t

L) v

, ~ s D fv

Patients suffering chronic renal failure often require hemodialysis. Drugs may be
administered by injection into the venous side of the hemodialyzer machine.

Such a situation was described by L*tourneau-Saheb et al (Int. J. Clin. PKinmacol., 15, 116-120 (1977)) for six patients who receivet an intravenous dose 7 gentamicin (90 mg). The mean serum concentrations of gentamicin (Cs) showed a
biexponential decline with time (t).

(~9cyml)

(mRn)

6.60

10

5-75

20

5.25

30

4.80

40

4.50

50

3.95

90

3.40

150

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-68

Multicompartment Modeling

3.10

180

2.90

240

2.55

285
.

(a) Calculate the values of t1/29 B, Bj, ~, A~, and V1. LFtourneau
Saheb et al reported,

tl/2 = 5.50 t 0.77 hr (mean i SD)


B s 0.0022 t 0.0004 min 1
B1 s 4.76 f 0.62 vg/ml

v 0.053 t 0.009 min


A1 a 3.47 s 1.01 pg/ml
V1 s 11.3 ffi 2.0 litre

(b) Calculate C1 and VB


LFtourneau-Sahleb et al reported,

C1 s 40.8 t 7.8 ml/min


VB s 19.2 h 2.7 litre

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-69

Multicompartment Modeling

_1

11-33

98X of the systemic clearance of gentamicin is composed of renal clearance-of


unchanged drug, so that renal failure woult severely alter gentamRcin systemic
clearance. In this case, the use of the hemodialyzer gave a systemic clearance close
to the 41.0 ml/min seen in patients with normal renal function.

(c) Under the conditions of hemodialysis, calculate the dose (D) of gentamicin
which would be administered every 8 hr (t) in order to maintain an Waverage
steady-state serum concentration of 4 ug/ml. What would be (Cmax)ss and
(Cmin)ss?

(d) Without the hemodialyzer two male patients each exhibited a creatinine clearance (Clcr) of S ml/min; the noW l value is 117 + 20 ml/min. Assuming that the
decreased ClCr is due to a decrease in glomerular filtration rate, what would have
been the renal clearance (Clr) of gentamicin in these two patients had they
remalned without the hemodialyzer? What dose would then need to be administered every 8 hr to maintajn (~s) at 4 ug/ml? Compare your answer with the Physicians Desk Reference (5677).

Your third patient comes from Edelman et al (Clin Pbarmacol Therap 35:382-6
(1984)). He studied metotrexate is artbritic patients. The pharmacokinetic parameters gleened from the 10 mg IV data arc:

A1 (mg/L) 0.663 Alpha (hr-1) 059 B1 (mg/L) 0.073 Beta (hr-1) 0.097

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-70

Multicompartment Modeling

15) What is the AUC (0 to inf) (mg / L * hr) ?

a) 0.75 b) 1.08 c) 1.12 d) 1.88e) 9.1

16) What is V1 (L) ?


a) 13.6 b) 15.1 c) 253 d) 36.1e)103.1

17) What is the clearance (L/hr) ?


a) 53 b) 8.9 c) 133 d) 15.4e) 17.2
18) What is V(beta) (L) ?
a) 13.6 b) 253 c) 36.1 d) 45e 55

19) What is the t(max) in the perepheral compartment (hr) ? a) 0 b) 1.8 c) 3.7 d) 5.1
e)6.1

20) What percent of the drug is in the perepheral compartment at cquilibrium ? a)


25 b) 50 c) 75 d) 100 e) 125

Two compartment: It has been proposed (Ionescu et al. Clin Pkin 14:178186,1988) that morphine injectcd dirtctly into thc spioal chord would give significant analgesia. The following is a CSF concentration - time profile resulting from
05 mg/lcg IV bolus dose of Morphine:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-71

Multicompartment Modeling

time

conc.

(min) (mg/L)

time

conc.

(min) (mg/L)

251

120

323

181

240

173

10

142.5

360

82

20

1043

480

2.4

40

75.1

720

1.2

80

48.8

1 a) fraction of remaining drug contained in peripheral compartment at equilibrium. Lb) Can this drug be approximated by a one compartment model ? Support
your contention with calculations. Lc) Calculate a rcasonable dosage regimen for
the above patient to maintain the concentration within the therapeuticwindow of
50 to 5 mg/L.

Two compartment:

(1) It has been proposed that diazepam has anticonvulsant properties above 350
nanograms per milliliter. The following is a concentration - time profile resulting
from 10 mg IV bolus dose of diazepam :

time

conc. time

(hrs)

(ng/ml)(hrs)

0.25

480

6.0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-72

Multicompartment Modeling

0-50

400

8.0

l.o

300

10.0

2.0

170

16.0

3.0

120

24.0

on

llo

l.a) fraction of remaining compartment at equilibrium.

~_

conc.
(ng/ml)
100
90
85
70
53

Dlezzewm

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-73

Multicompartment Modeling

UA tD 20A

AD

Time

drug contained in peripheral

k21 = (1.05 * 116.8) + (0.0325 * 487.6) / (487.6 + 116.8)


= 138.5 / 604.4
= 0.239

klO = (1.05 * 0.0325) / 0.239


= n ls

kl2 = 1.05 + 0.0325 - 0.15 - 0.239


=n7

B2 = (0.7 * lOOOOmic) / (1.05 - 0.0325)


= 6880

V1 = lOOOOmic / 604.4 mic/l


= 16.55

X2 / total = 6880 / t6880 + (16.55 * 116.8mic/1)]

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-74

Multicompartment Modeling

= 0.78

l.b) Can this drug be approximated by a one compartment model? Support your
contention with calculations.

AUC = (487.6 / 1.05) + (116.8 / 0.0325)


= 464.4 + 3594.8
= 4058.2

Since the beta phase contributes more than 80% of the total AUC, the model can
be collapsed to a one compartment model.

3594.8 / 4058.2 = 0.886 = 89%

l.c) Calculate a reasonable dosage regimen for the above patient to maintain the
concentration within the therapeutic window of 350 to 1100 nanograms per ml.

K = 1/MRT = 1 / (111007 / 4058.2) = 1/27.35 = 0.0253 per hr


t 1/2 = 0.693 / 0.0253 = 27.39 hr
2^N^ = 1100 / 350 = 3.14
N = ln 3.14 / ln 2 = 1.65

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-75

Multicompartment Modeling

Tau max = 27.39 * 1.65 = 45.19 hr drop the dosing interval to 24 hours now need
to find a new N

N = 24 / 27.39 = 0.876

Vss = lOOOOmic / (4058.2mic*hr/1 * 0.0253/hr) = 97.25L

To find dose:
l.lOOmg/L = (D / 97.25 L) * tl / (1 - 0.5^0.876^)]
D = 49 mg daily (aggressive therapy)

0.350mg/L = (D / 97.25L) * t 1 / (1 - 0.5^0.876^)](0.5^0.876^)


D = 29 mg daily (conseervative therapy)

Therefore, any dose between 30 and 50 mg a day can be given.

Two Compartment:

We have used theophylline as a test drug in many of our calculations in class. We


assumed that it was a one compartment model. Look at the data from Mitenko
(Clin Pharmacol and Ther,14p509 1974) for intravenous theophylline (Dose 5.6
mg/kg):

sos

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-76

Multicompartment Modeling

theophylilne
.

i ,_

1.
OD

2D

4wD 6D8S

tD

Tlm~

Time (hr.)
0.167
0.333
0.500
0.833
1.0
1.5
2.0
3.0
4.0
6.0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-77

Multicompartment Modeling

8.0

Conc. (mg./L.)

24.7 Estimates of Pharmacokinetic parameters

20.3

are as follows:

18.1

A1 (mg/L)16.1

16.1

B1 (mg/L)17.9

15.6

Alpha (hr-l)4.8

14.3

Beta (hr-l)0.15

13.3

AUC (O to inf) 122.7

11.

AUMC (O to inf) 797.6

9.8

7.3
s.4

1) What is the volume of the central compartment (L/Kg) ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-78

Multicompartment Modeling

*A) 0.165 B) 0.30 C) 0.35 D) 2.9 E) 6.1

2) What is the clearance of theophylline (L/Kg/hr) ?

A) 0.007 *B) 0.046C) 2.7 D) 22 E) 142.4

3) What is the Volume of

distribution in the beta phase (Vbeta)

(L/Kg) ?
A) 0.165 *B) 0.30C) 0.35D) 2.9E) 6.1

4) What percent of the equilibrium ?

A) 12 B) 23

dose is in the peripheral compartment at

*C) 46 D) 69 E) 92 treated as a one compartment model for

5) Can theophylline be dosing purposes ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-79

Multicompartment Modeling

A) No, a larger percentage of the drug is in the peripheral compartment.

B) NO, A1 and C) Yes, alpha D) Yes, the approximately

*E) Yes, the approximately

B1 are about the same value. is bigger than beta contribution of the alpha phase
AUC is the total AUC. contribution of the beta the total AUC.

phase AUC is

6) What is the mean residence time (MRT) for the IV dose (hr) ?

A) 4.25 B) 5 C) 5.75*D) 6.5E) 7.25

For the same dose of an oral product information was obtained:

AUC (O to inf)
AUMC (O to inf)

7) What is the MRT for the

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-80

Multicompartment Modeling

A) 8.0 B) 8.75

122
1400

(TheoDur^TQ^T) the following

oral dose (hr) ?

C) 9.5 D) 10.25*E) 11.5

8) What is the mean absorption time (MAT) for the oral dose (hr)?

A) 1

B) 2

C) 3

D) 4

*E) 5

9) What is the absorption rate constant for theophylline in TheoDur

(hr^T-l^T) ?

A) 0.14 B) 0.17 *C) 0.2 D) 0.25 E) 0.3

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-81

Multicompartment Modeling

Two compartment: The following pharmacokinetic information was obtained from


EA, a

45 y/o, 70 kg, healthy male following an800 mg IV dose of


vancomycin:
A1 60 mg/L
alpha 1.33 hr^-l
B1 20 mg/L
beta 0.129 hr^-l

AUC (mg/L * hr)200


AUMC (mg/L)1237

5) What is your patients vancomycin clearance(L/hr)?

a) 0.25 *b) 4 c) 24.74 d) 45.1 e) 155

6) What is your patients V(beta)(L)?

a) 10 b) 24.74 *c) 31 d) 45.1 e) 60

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-82

Multicompartment Modeling

7) What is your patients V1(L)?

*a) 10 b) 24.74 c) 31 d) 45.1 e) 60

8) What is your patients MRT(iv) (hr)?

a) 0.112 b) 0.162 c) 1.39 d) 4.29 *e) 6.19

9) What is your patients effective rate of elimination (hr^-l)? a) 0.112 *b) 0.162 c)
0.5 d) 4.29 e) 6.19

10) What is your patients Vss (L)?

a) 10 *b) 24.74 c) 31 d) 45.1 e) 60

11) Following an IM dose of vancomycin, the MRT(im) was calculated to be 8.185


hr. What was the absorption half life (hr)? a) 0.112 b) 0.162 *c) 1.39 d) 2 e) 6.19

RP s angina was controlled on 40 mg TID of propranolol. You calculated his pharmacokinetic parameters to be: Vd (L) = 125; T1/2 (hr) = 3.1; Qh (L/hr) = 33; Bioavailability (f) = .7; Bound(%) = 95. Propranolol is essen^_ tially 100%
metabolized.

12) What is his Total body clearance (L/hr)? a) 1.4 b) 14 *c) 28 d) 33 e)40.3

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-83

Multicompartment Modeling

13) What is his hepatic extraction ratio?

a) 0.042b) .42 *c) .85 d) 1

e) 1.22

14) Assuming propranolol to be rapidly absorbed, what is his Cpss

max free concentration (ng/mL)?

*a) 13.4 b) 19.2 c) 188 d) 268 e) 355

15) What is his Cpss max total(bound and free) (ng/mL)?

a) 13.4 b) 19.2 c) 188 *d) 268 e) 355

16) What is his Cpss min free (ng/mL)?

*a) 2.2 b) 3.2 c) 31.4 d) 44.8 e) 59.3

He is now suffering from renal failure. His half life went up to 3.8 hr while his
binding went up to 98.3% because of an increase in AAG, a plasma protein to
which propranolol binds.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-84

Multicompartment Modeling

17) What is his new clearance (L/hr)?

a) 14.4 *b) 21.7 c) 24 d) 28 e) 36.2

18) What is his new volume of distribution (L)?

a) 79 *b) 119 c) 132 d) 153 e) 199

19) What will his new Cpss max free be if we keep him on the same

regimen (ng/mL)?

*a) 5.26 b) 7.5 c) 310 d) 442 e) 554

20) What is his new Cpss max total(ng/ml) ? a) 5.26 b) 7.5 *c) 310 d) 442 e) 554

21) What is his new Cpss min free (ng/mL)?

*a) 1.23 b) 1.75 c) 72.3 d) 103 e) 129

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-85

Multicompartment Modeling

22) The physician sees that the clearance has dropped and consequently the total
plasma concentrations have gone up. He wants to decrease to dose to 40 mg / BID.
What would you recommend? a) Sounds good to me. That will get the Cpss max
total concentration back to it was before he was sick. b) His new clearance was
marginally changed because the drug is cleared by the liver. Id leave it alone. c) I
think the change from TID to BID is a bit much. How about lowering it to 30 mg
instead of 40 mg TID. *d) We need to increase the dose, not lower it. Id recommend 40 mg QID. e) We need to increase the dose, not lower it. Id recommend 80
mg QID.

Two compartment

Methotrexate is a cytolytic used for the treatment of acute leukemia and other
forms of cancer. After a a 400 mg/kg dose the following data was recorded for a 12
y/o boy.

Time (hours)Concentration (mcg/ml)


6

360

12

70

18

15

36

48

1.2

60

0.46

72

036

Q(}

0.15

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-86

Multicompartment Modeling

FIND: (5 points) a) A^V1^V =


(5 points) b) B^V1^V =
(5 points) c) Clearance =
(5 points) d) % of drug in peripheral compartment at equilibrium =

Everything.

Primidone (Mysoline^T@^T) is an effective agent in the treatment of generalized


and complex partial seizures. Although primidone has anticonmlsant activity of its
own, much of the activity comes from the conversion to one of its metabolites,
phenobarbital. For the purposes of this exam, an insignificant error will be introduced bytheuse of intermittent IV caSculations instead of oralformultipledose.
Thefolloving information for primidone and phenobarbital is available:

Primiflnne. Phenabarbital

Therapeutic Range (mg/L)5 - 12 10 - 30


Bioavailabiliq (f)1

0.9

Salt factor (S)1

09

Vd (L/Kg)

0.7

T1/2

0.6

7 hr

5 days

% Excreted undhanged2520
% Metabolized75

80

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-87

Multicompartment Modeling

% Metabolized to Phenobarbital25*~

% Metabolized (non Phenobarb-paths)50**

2.(20 pts) Please prepare a short consult for your M.D. in which you caSculate a
dosage regimen for an 80 kg
patient whidh would l~eep the plasma concentration within the therapeutic range.
Prirnidone comes as 250 mg
scored (can be broken in half) tablets. Indude your average, maximum and minimum primidone as well as
phenobarbital steady-state concentrations in the consult.

3. (25 pts) The patient recently contracted mono. His liver function has been
reduced to 50 % of normal.

Would you recommend a change in your therapy ? If you would, prepare a short
consult for his M.D. as in question #2. Dont forget that changes in hepatic clearance result in changes in both phenobarbital clearance 5~nfl fannation

4. (25 pts) Now that his mono has cleared up the doctor noticed that he has stenosis
of the liver as a consequence of all t_e heavy parting that he did after the kinetics
course. His liver plasma flow has been reduced to 50 % of normal. Would you recommend a dhange in therapy ? If you would, prepare a short consult for his M.D.
as in question #2. Dont forget that dhanges in hepatic dearance result in changes
in both phenobarbital dearance and formation.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-88

Multicompartment Modeling

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-89

Multicompartment Modeling

11.4 Begin

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-90

Multicompartment Modeling

11.5 Problems

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-91

Multicompartment Modeling

Aspirin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 1)

AHFS 00:00.00
GPI: 0000000000

Fu, C., Melethil, S., and Mason, W., "The pharmacokinetics of aspirin in rats and the effect of buffer", Journal of Pharmacokinetics
and Biopharmaceutics, Vol. 19, (1991), p. 157 - 173.

Aspirin is an analgesic/ antipyretic commonly used to relieve minor pain and is used in such conditions as rheumatic
fever, rheumatoid arthritis, and osteoarthritis. The major metabolite of aspirin is salicylic acid. The following set of
data was collected using rats which weighed 250 - 300 g.
PROBLEM TABLE 11 - 1.

Aspirin

weight of rat

275 g

Dose

5 mg/kg IV

A1

8.58

1.07

B1

7.24

0.2

AUC

38.8

AUMC

116.0

1.

What is ?

2.

What is ?

3.

What is your patient's clearance?

4.

What is your patients MRT?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is your patient's ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is ?

14.

What is the in the peripheral compartment?

15.

What percent of the dose is in the peripheral compartment at equilibrium?

16.

Can this drug be treated as a one-compartment model for dosing purposes?

17.

If this drug can be treated as a one-compartment model, what is K ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-92

Multicompartment Modeling

Buprenorphine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 2)

AHFS 00:00.00
GPI: 0000000000

Ohtani, M., et al., "Pharmacokinetic analysis of enterohepatic circulation of buprenorphine and its active metabolite, norbuprenorphine, in rats", Drug Metabolism and Disposition, Vol. 22, (1994), p. 2 - 7.

Buprenorphine is a morphine derivative which has twice the duration of action and 30 times the potency of morphine.
Buprenorphine is partially metabolized to norbuprenorphine which is also active in the body. In this study, buprenorphine was given to rats weighing 280 - 300 g.
PROBLEM TABLE 11 - 2.

Buprenorphine

Weight of rat

290 g

Dose

0.06 mg/kg IV

A1

41

3.89

B1

10

0.271

AUC

48.3

AUMC

135.24

1.

What is the ?

2.

What is the ?

3.

What is the AUC?

4.

What is your patient's clearance?

5.

What is your patients MRT?

6.

What is your patient's ?

7.

What is your patient's V1?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is ?

14.

What is the in the peripheral compartment?

15.

What percent of the dose is in the peripheral compartment at equilibrium?

16.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-93

Multicompartment Modeling

17.

If this drug can be treated as a one-compartment model, what is K ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-94

Multicompartment Modeling

Caffeine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 3)

AHFS 00:00.00
GPI: 0000000000

Shi, J., et al., "Pharmacokinetic-pharmacodynamic modeling of caffeine: Tolerance to pressor effects", Clinical Pharmacology
and Therapeutics, Vol. 53, (1993), p. 6 - 14.

This study looks at the cardiovascular effects of caffeine. Caffeine is known to increase blood pressure upon its withdrawl. This study looks at how tolerance to caffeine and its pressor effects develops and disappears with time.
PROBLEM TABLE 11 - 3.

Caffeine

Patient weight

80 kg

Dose

4 mg/kg oral

A1

10.55

4.9

B1

9.1

0.23

98.4 %

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is your patient's ?

13.

What is ?

14.

What is the in the peripheral compartment?

15.

What percent of the dose is in the peripheral compartment at equilibrium?

16.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-95

Multicompartment Modeling

Cefazolin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 4)

AHFS 00:00.00
GPI: 0000000000

Nightingale, C., et al., "Changes in pharmacokinetics of cefazolin due to stress", Journal of Pharmaceutical Sciences, Vol. 64,
(1975), p. 712 - 714.

Cefazolin is a cephalosporin antibiotic used in the treatment of many types of infections. This study looks at the effect
of stress on the pharmacokinetics of cefazolin. The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 4.

Cefazolin

Patient weight

56.3 kg

Dose

1 g IV

A1

206.48

4.832

B1

122.96

0.573

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-96

Multicompartment Modeling

Ceftazidime
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 5)

AHFS 00:00.00
GPI: 0000000000

Ackerman, B., et al., "Effect of decreased renal function on the pharmacokinetics of ceftazidime", Antimicrobial Agents and Chemotherapy, Vol. 25, (1984), p. 785 - 786.

Ceftazidime is a cephalosporin antibiotic. This study explores the effect of compromised renal function on the pharmacokinetics of ceftazidime. The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 5.

Ceftazidime

Dose

1 g IV bolus

A1

188

8.22

B1

58.2

0.49

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-97

Multicompartment Modeling

Clentiazem
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 6)

AHFS 00:00.00
GPI: 0000000000

Shah, A., et al., "Pharmacokinetics of clentiazem after intravenous and oral administration in healthy subjects", Journal of Clinical Pharmacology, Vol. 33, (1993), p. 354 - 359.

Clentiazem is an derivative of diltiazem which is under investigation for its use in the treatment of angina pectoris and
hypertension. Clentiazem blocks calcium channels resulting in a decrease in peripheral vascular resistance which subsequently leads to a decrease in blood pressure.
PROBLEM TABLE 11 - 6.

Clentiazem

Patient weight

77 kg

Dose

20 mg IV bolus

A1

37.52

2.7

B1

16.17

0.078

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-98

Multicompartment Modeling

Cocaine

(Problem 11 - 7)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Levine, B. and Tebbett, I., "Cocaine pharmacokinetics in ethanol-pretreated rats", Drug Metabolism and Disposition, Vol. 22,
(1994), p. 498 - 500.

This study looks into several reports which claim that the euphoric effects of cocaine can be enhanced when taken in
conjunction with alcohol. This effect may be the result of higher cocaine blood levels or a reduced elimination of
cocaine or a combination of both.
PROBLEM TABLE 11 - 7.

Cocaine

Weight of rat

300 g

Dose

2 mg/kg cocaine (also 1 g/ kg ethanol)

A1

1172.6

0.362

B1

462

0.045

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-99

Multicompartment Modeling

1,2-Diethyl-3-Hydroxypyridine-4-One
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 8)

AHFS 00:00.00
GPI: 0000000000

Epemolu, O., et al., "The pharmacokinetics of 1,2-Diethyl-3-Hydroxypyridine-4-One (CP94) in rats, Drug Metabolism and Disposition, Vol. 20, (1992), p. 736 - 741.

1,2-Diethyl-3-Hydroxypyridine-4-One (CP94) is an iron chelator which is orally active. It is being investigated for use
in the treatment of hemoglobinopathic disorders. In this study, rats weighing 250 - 300 g were given doses 50 mg /kg
intravenously and the following data was collected:
PROBLEM TABLE 11 - 8.

1,2-Diethyl-3-Hydroxypyridine-4-One

Weight of rat

275 g

Dose

50 mg/kg IV

A1

30.9

2.03

B1

8.13

0.38

Assume that the rat ( which weighs 275 g) is suffering from thalassemia and his iron levels are very high. The rat is
prescribed CP94 to restore the iron levels to normal.
1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-100

Multicompartment Modeling

2,2-dimethylaziridine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 9)

AHFS 00:00.00
GPI: 0000000000

Lalka, D., Jusko, W., and Bardos, T., "Reactions of 2,2-dimethylaziridine-type alkylating agents in biological systems II: Comparative pharmacokinetics in dogs", Journal of Pharmaceutical Sciences, Vol. 64, (1975), p. 230 - 235.

The 2,2-dimethylaziridine alkylating agents are used for their antitumor capability as antineoplastic agents. In this
study, male mongrel dogs, weighing 20 - 28 kg, were each given a dose of 12 mg/kg of ethyl bis (2,2-dimethylaziridinyl) phosphinate intraveneously.
PROBLEM TABLE 11 - 9.

2,2-dimethylaziridine

Weight of dog

24 kg

Dose

12 mg/kg IV

A1

42

0.409

B1

8.5

0.095

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-101

Multicompartment Modeling

Flurbiprofen
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 10)

AHFS 00:00.00
GPI: 0000000000

Menzel-Soglowek, S., et al., "Variability of inversion of (R)-flurbiprofen in different species", Journal of Pharmaceutical Sciences,
Vol. 81, (1992), p. 888 - 891.

Flurbiprofen is an anti-inflammatory and analgesic agent. This study compares the pharmacokinetics of the (R)-isomer
of flurbiprofen to the those of the (S)-isomer. The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 10. Flurbiprofen

Weight of rat

260 g

Dose

10 mg/kg IV

A1

48.5

2.33

B1

57.68

0.175

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-102

Multicompartment Modeling

Furosemide
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 11)

AHFS 00:00.00
GPI: 0000000000

Tilsone, W., and Fine, A., "Furosemide kinetics in renal failure", Clinical Pharmacology and Therapeutics, Vol. 23, (1978), p. 644
- 650.

Furosemide is an agent which is used for its diuretic action to treat such conditions as renal and cardiac edema. In this
study, normal subjects were given an intravenous bolus dose of 22 mg of furosemide. Blood samples were taken at various intervals and the following data was obtained:
PROBLEM TABLE 11 - 11. Furosemide

Dose

22 mg IV

A1

2.1

6.9

B1

0.77 -

0.96

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-103

Multicompartment Modeling

Glycyrrhizin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 12)

AHFS 00:00.00
GPI: 0000000000

Tsai, T., et al., "Pharmacokinetics of glycyrrhyzin after intravenous administration to rats", Journal of Pharmaceutical Sceinces,
Vol. 81, (1992), p. 961- 963.

Glycyrrhizin is a component of licorice which is proposed to have anti-inflammatory, anti-hepatotoxic, interferoninducing, anti-viral, and anti-ulcer activity. It also causes pseudoaldosteronism. The following data was approximated
from the graph given in this article.
PROBLEM TABLE 11 - 12. Glycyrrhizin

Weight of rat

275 g

Dose

20 mg/kg

A1

91.23

4.16

B1

69.90 -

0.43

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-104

Multicompartment Modeling

Human Deoxyribonuclease
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 13)

AHFS 00:00.00
GPI: 0000000000

Mohler, M., et al., "Altered pharmacokinetics of recominant human deoxyribonuclease in rats due to the presence of a binding protein", Drug Metabolism and Disposition, Vol. 21, (1993), p. 71 - 75.

Deoxyribonucleases are found in human serum, urine, and a variety of tissues. These endonucleases catalyze the
hydrolysis of DNA to oligonucleotides. It has been suggested that increased levels of serum deoxyribonucleases may
predict malignancies.
PROBLEM TABLE 11 - 13. Human

Deoxyribonuclease

Patient weight

260 g

Dose

1 mg/kg IV bolus

A1

19250 -

8.61

B1

4897

0.229

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-105

Multicompartment Modeling

Human Granulocyte Colony-Stimulating Factor


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 14)

AHFS 00:00.00
GPI: 0000000000

Tanaka, H., and Kaneko, T., "Pharmacokinetic and pharmacodynamic comparisons between human granulocyte colony-stimulating factor purified from human bladder carcinoma cell line 5637 culture medium and recombinant human granulocyte colonystimulating factor produced in Escherichia coli", The Journal of Pharmacology and Experimental Therapeutics, Vol. 262, (1992),
p. 439 - 444.

Human Granulocye Colony-Stimulating Factor (hG-CSF) is used to stimulate the proliferation of precursor cells and
their subsequent differentiation in the bone marrow. This article compares the pharmacokinetics of hG-CSF produced
by two different methods. In the first method, the hG-CSF was obtained from human bladder carcinoma cell line 5637
culture medium. In the second method, the hG-CSF was produced by Escherichia coli.
PROBLEM TABLE 11 - 14. Human

Granulocyte Colony-Stimulating Factor

Weight of rat

250 g

Dose

10 g/kg IV

A1

116.21 0.24 hours

B1

99.228
1.27 hours

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is a?

8.

What is b?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-106

Multicompartment Modeling

Hydrocortisone
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 15)

AHFS 00:00.00
GPI: 0000000000

Derendorf, H., et al., "Pharmacokinetics and oral bioavailability of hydrocortisone", Journal of Clinical Pharmacology, Vol. 31,
(1991), p. 473 - 476.

This study looks at both the two-compartment model pharmacokinetics and the oral bioavailability of hydrocortisone.
The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 15. Hydrocortisone

Dose

20 mg IV

A1

430

13.1

B1

439 -

0.445

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-107

Multicompartment Modeling

Levodopa
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 16)

AHFS 00:00.00
GPI: 0000000000

Sasahara, K., et al., "Dosage form design for improvement of bioavailability of levodopa II: Bioavailability of marketed levodopa
preparations in dogs and parkinsonian patients" Journal of Pharmaceutical Sciences, Vol. 69, (1980), p. 261 - 265.

Levodopa is an agent used in the treatment of Parkinson's disease. This study looks at various dosage forms of
levodopa and compares the pharmacokinetic parameters of each. The following data was approximated from the graph
given in this article.
PROBLEM TABLE 11 - 16. Levodopa

Dose

50 mg IV

A1

8.63

13.3

B1

2.97 -

1.14

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-108

Multicompartment Modeling

Meropenem

(Problem 11 - 17)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Chimata, M., et al., "Pharmacokinetics of meropenem in patients with various degrees of renal function, including patients with
end-stage renal disease", Antimicrobial Agents and Chemotherapy, Vol. 37, (1993), p. 229 - 233.

Meropenem is a carapenem antibiotic which has a broad spectrum of activity. It is used in the treatment of infections
caused by both Gram-positive and Gam-negative bacteria and is active against Enterobacteriaceae and Pseudomonas
aeruginosa. Meropenem is 60% renally and 40% hepatically eliminated.
PROBLEM TABLE 11 - 17. Meropenem

Dose

500 mg IV infusion over 40 minutes

A1

21

1.85

B1

20 -

0.503

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-109

Multicompartment Modeling

N-Methylpyridinium-2-Carbaldoxime Chloride
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 18)

AHFS 00:00.00
GPI: 0000000000

Bodor, N., and Brewster, M., "Problems of delivery of drus to the brain", International Encyclopedia of Pharmacology and Therapeutics, Vol. 120, (1975)

N-methylpyridinium-2-cabaldoxime chloride (2-PAM) is the drug of choice for the treatment of orgaonphosphate poisoning. It is mostly renally excreted. This article considers the fact that this agent is highly hydrophilic and thus has
difficulty reaching the brain. The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 18. N-Methylpyridinium-2-Carbaldoxime

Weight of dog

40 kg

Dose

7.0 mg/kg

A1

5.356

0.28796

B1

35.983

11.586

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

Chloride

11-110

Multicompartment Modeling

Pyrazine Diazohydroxide
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 19)

AHFS 00:00.00
GPI: 0000000000

Vogelzang, N., et al., "Phase I and pharmacokinetic study of a new antineoplastic agent: pyrazine diazohydroxide (NSC 361456)",
Journal of Cancer Research , Vol. 54, (1994), p. 114 - 119.

Pyrazine diabhohydroxide is an agent which forms a reactive pyrazine dizonium ion in vivo which acts to destroy
tumor cells. This study looks at the pharmacokinetic parameters of this agent in advanced cancer patients whose cancer
was not curable by any other type of therapy. They were given a dose of 18 mg/m2/day for 5 days every 4 weeks.
Most of the following data was collected for a 66 year old male subject. The remaining data was approximated from
the graph given in this article.
PROBLEM TABLE 11 - 19. Pyrazine

Diazohydroxide

Patient Body Surface Area

1.82 m2

Dose

18 mg/ m2

A1

8063

0.195

B1

1186

0.0257

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-111

Multicompartment Modeling

Terbinafene
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 20)

AHFS 00:00.00
GPI: 0000000000

Kovarik, J., et al., "Dose-proportional pharmacokinetics of terbinafine and its N-demethylated metabolite in healthy volunteers",
British Hournal of Dermatology, Vol. 126, (1992), p. 8 - 13.

Terbinafene is an antifungal agent which acts by interfering with ergosterol biosynthesis. It is active against Trichophyton, Epidermophyton, and Microsporum. Approximately 70% of an oral dose is absorbed. Terbinafene has an Ndemethylated metabolite which is active. The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 20. Terbinafene

Dose

750 mg

A1

2398

0.511 -

B1

102 -

0.0222 -

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-112

Multicompartment Modeling

Verrucarol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 21)

AHFS 00:00.00
GPI: 0000000000

Barel, S., Yagen, B., and Bailer, M., "Pharmacokinetics of the trichothecen mycotoxin verrucarol in dogs", Journal of Pharmacetuical Seciences, Vol. 79, (1990), p. 548 - 550.

Verrucarol is a toxin which is related to toxins which have anti-tumor activity. This study looks at the pharmacokinetics of verrucarol in dogs. The following data was approximated from the graph given in this article.
PROBLEM TABLE 11 - 21. Verrucarol

Weight of Dog

22.5 kg

Dose

0.4 mg/ kg

A1

126.05

0.0415

B1

540.58 -

0.00946-

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-113

Multicompartment Modeling

2-Chloro-2-deoxyadenosine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 22)

AHFS 00:00.00
GPI: 0000000000

Liliemark, J. and Juliusson, G., "On the pharmacokinetics of 2-Chloro-2-deoxy-adenosine in humans", Cancer Research, Vol. 51,
(1991), p. 5570 - 5572.

Two-Chloro-2-deoxyadenosine is an antitumor agent used in the treatment of hairy cell leukemia and other lymphoproliferative diseases. Infusions of 0.14 mg/kg over 12 hours were administered to 12 patients with various lymphoproliferative diseases for 5 consecutive days. The following data was collected:
PROBLEM TABLE 11 - 22. 2-Chloro-2-deoxyadenosine

Patient weight

65 kg

Dose

0.14 mg/kg over 12 hours

A1

177.0 nM

1.04

B1

21.0 nM

0.10

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-114

Multicompartment Modeling

Felodipine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 23)

AHFS 00:00.00
GPI: 0000000000

Regardh, C., et al., "Pharmacokinetics of felodipine in patients with liver disease", Journal of Clinical Pharmacology, Vol. 36,
(1989), p. 473 - 479.

The pharmacokinetic parameters of felodipine in patients with impaired liver function were investigated in this study.
Felodipine blocks calcium channels resulting in a decrease in peripheral vascular resistance which subsequently leads
to a decrease in blood pressure. Felodipine also works as a diuretic. The bioavailability of felodipine is 15%. It is
highly (99.64%) protein bound and is eliminated almost exclusively by liver metabolism. The following data is for a
patient with liver cirrhosis:
Weight of dog

kg

Dose

mg/kg IV

A1
a

B1
b

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-115

Multicompartment Modeling

Lorazepam
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 24)

AHFS 00:00.00
GPI: 0000000000

Segal, J., et al., "Decreased systemic clearance of lorazepam in humans with spinal cord injury", Journal of Clinical Pharmacology, Vol. 31, (1991), p. 651 - 656.

Lorazepam is a benzodiazepine which is used as an anxiolytic, an anti-convulsant, an anti-emetic, and a sedative-hypnotic agent.
PROBLEM TABLE 11 - 24. Lorazepam

Dose

2.0 mg infused intravenously over 1 - 2 minutes

A1
a

B1
b
AUC
AUMC

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-116

Multicompartment Modeling

Metronidazole
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 25)

AHFS 00:00.00
GPI: 0000000000

Uccellini, D., Morgan, D., and Raymond, K., "Relationships among duration of infusion, dose, dosing interval, and stedy-state
plasma concentrations during intermittent intravenous infusions: studies with metronazole", Journal of Pharmacokinetics and
Biopharmaceutics, Vol. 14, (1986), p. 95 - 106

PROBLEM TABLE 11 - 25. Metronidazole

Patient weight

70 kg

Dose

1.5 g IV infusion

A1
a

2.11

B1
b

0.09

AUC
AUMC

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-117

Multicompartment Modeling

Rhizoxin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 26)

AHFS 00:00.00
GPI: 0000000000

Graham, M., et al., "Preclinical and phase I studie with rhizoxin to apply a pharmacokinetically guided dose-escalation scheme",
Journal of the National Cancer Institute, (1991), p. 494 - 499.

Rhizoxin is a lactone which was obtained from the fungus, Rhizopus chinensis. It has anti-tumor activity
against a broad spectrum of tumor types including LOX melanoma, A549 lung tumors, and MX-1 mammary
tumors. This study looks at dosing of rhizoxin. Patients with nontreatable tumors who had a life expectancy
of more than 12 weeks were given doses of 12 mg/ m2. The following data was approximated from the
graph given in this article.
PROBLEM TABLE 11 - 26. Rhizoxin

Patient Body Surface Area

1.82 m2

Dose

12 mg/ m2

A1

1.55

4.00

B1

0.12

0.116 -

1.

What is ?

2.

What is ?

3.

What is ?

4.

What is your patient's clearance?

5.

What is your patient's ?

6.

What is your patient's V1?

7.

What is ?

8.

What is ?

9.

What is ?

10.

What is ?

11.

What is ?

12.

What is ?

13.

What is the in the peripheral compartment?

14.

What percent of the dose is in the peripheral compartment at equilibrium?

15.

Can this drug be treated as a one-compartment model for dosing purposes?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-118

Multicompartment Modeling

11.5.1

TWO-COMPARTMENT MODEL EQUATIONS

The following set of equations were used to solve the two-compartment model problem set. The problem sets for the
first three drugs have been done for you. The others are done the same way. The answers for all of the problems are in
the back of this packet.
Aspirin
1.
2.
3.
4.
5.

The weight of the rat is 275 g or 0.275 kg.

6.
7.
8.
9.
10.
11.
12.
13.
14.
15.

Yes

_____________________________________________________________________
Buprenorphine
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-119

Multicompartment Modeling

12.
13.
14.
15.

Yes

_____________________________________________________________________
Caffeine
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.

11.5.2

Yes

ANSWERS

Aspirin
1.

8.019

2.

36.2

3.

31.1

4.

2.62 minutes

5.

155.48 mL

6.

86.92 mL

7.

81.57 mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-120

Multicompartment Modeling

8.

0.648 min

9.

3.47 min

10.

0.598 min-1

11.

0.358min-1

12.

0.314min-1

13.

15.82

14.

1.928 minutes

15.

44.1%

16.

Yes

17.

0.381 min-1

Buprenorphine
1.

10.54

2.

36.9

3.

47.4

4.

366.8

5.

2.85 hours

6.

1.35 L

7.

0.34 L

8.

0.178 hours

9.

2.58 hours

10.

0.981 h-1

11.

1.075 h-1

12.

2.11 h-1

13.

51

14.

0.736 hours

15.

74.9%

16.

No

17.

Can't be calculated

Caffeine
1.

2.15

2.

39.57

3.

41.72

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-121

Multicompartment Modeling

4.

7.67

5.

33.35 L

6.

16.28 L

7.

0.141 hours

8.

3.014 hours

9.

2.39 h-1

10.

0.471 h-1

11.

2.266 h-1

12.

19.65

13.

0.655 hours

14.

51.2%

15.

Yes

Cefazolin
1.

42.73

2.

214.59

3.

257.32

4.

3.89

5.

6.78 L

6.

3.035 L

7.

0.143 hours

8.

1.21 hours

9.

2.163 h-1

10.

1.28 h-1

11.

1.96 h-1

12.

329.44

13.

0.50 hours

14.

55.2%

15.

Yes

Ceftazidime
1.

22.87

2.

118.78

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-122

Multicompartment Modeling

3.

141.65

4.

7.0598

5.

14.41 L

6.

4.06 L

7.

0.0843 hours

8.

1.415 hours

9.

2.32 h-1

10.

1.738 h-1

11.

4.65 h-1

12.

246.2

13.

0.365 hours

14.

39.2%

15.

Yes

Clentiazem
1.

13.9

2.

207.3

3.

221.2

4.

90.4

5.

1159.2 L

6.

372.5 L

7.

0.257 hours

8.

8.89 hours

9.

0.868 h-1

10.

0.243 h-1

11.

1.67 h-1

12.

53.69

13.

1.35 hours

14.

47.4%

15.

Yes

Cocaine
1.

3239.2

2.

10266.7

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-123

Multicompartment Modeling

3.

13505.89

4.

44.43

5.

987.2 mL

6.

367.1 mL

7.

1.91 minutes

8.

15.4 minutes

9.

0.1346 min-1

10.

0.1210 min-1

11.

0.1514 min-1

12.

1634.6

13.

8.35 minutes

14.

59.2%

15.

No

1,2-Diethyl-3-hydroxpyridine-4-one
1.

15.22

2.

21.39

3.

36.62

4.

375.5

5.

988.2 mL

6.

352.3 mL

7.

0.341 hours

8.

1.824 hours

9.

0.724 h-1

10.

1.066 h-1

11.

0.62 h-1

12.

39.03

13.

1.016 hours

14.

55.4%

15.

No

2,2-dimethylaziridine
1.

102.7

2.

89.47

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-124

Multicompartment Modeling

3.

192.16

4.

1498.7

5.

15.78 L

6.

5.7 L

7.

1.695 minutes

8.

7.296 minutes

9.

0.148 min-1

10.

0.263 min-1

11.

0.093 min-1

12.

50.5

13.

4.65 minutes

14.

56.5%

15.

No

Flurbiprofen
1.

20.82

2.

329.6

3.

350.42

4.

7.42

5.

42.4 mL

6.

24.5 mL

7.

0.297 hours

8.

3.96 hours

9.

1.35 h-1

10.

0.303 h-1

11.

0.856 h-1

12.

106.18

13.

1.2 hours

14.

42.2%

15.

Yes

Furosemide
1.

0.304

2.

0.802

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-125

Multicompartment Modeling

3.

1.106

4.

19.88

5.

20.71 L

6.

7.67 L

7.

0.1005 hours

8.

0.722 hours

9.

2.55 h-1

10.

2.59 h-1

11.

2.71 h-1

12.

2.87

13.

0.322 hours

14.

62.99%

15.

No

Glycyrrhizin
1.

21.93

2.

162.56

3.

184.5

4.

29.8

5.

69.33 mL

6.

34.13 mL

7.

0.167 hours

8.

1.61 hours

9.

2.048 h-1

10.

0.873 h-1

11.

1.67 h-1

12.

161.13

13.

0.61 hours

14.

50.8%

15.

Yes

Human Deoxyribonuclease
1.

2235.78

2.

21384.3

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-126

Multicompartment Modeling

3.

23620.1

4.

11.01

5.

48.07 mL

6.

10.77 mL

7.

0.0805 hours

8.

3.027 hours

9.

1.929 h-1

10.

1.0223 h-1

11.

5.89 h-1

12.

24147

13.

0.433 hours

14.

28.9%

15.

Yes

Human Granulocyte Colony-Stimulating Factor


1.

40.24

2.

181.81

3.

222.05

4.

11.26

5.

20.62 mL

6.

11.6 mL

7.

2.89 h-1

8.

0.546 h-1

9.

1.625 h-1

10.

0.971 h-1

11.

0.839 h-1

12.

215.44

13.

0.711 hours

14.

43.8%

15.

Yes

Hydrocortisone
1.

32.8

2.

986.5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-127

Multicompartment Modeling

3.

1019.3

4.

19.62

5.

44.1 mL

6.

23.01 mL

7.

0.053 hours

8.

1.56 hours

9.

6.838 h-1

10.

0.853 h-1

11.

5.85 h-1

12.

869

13.

0.267 hours

14.

47.8%

15.

Yes

Levodopa
1.

0.649

2.

2.61

3.

3.25

4.

15.37

5.

13.48 L

6.

4.31 L

7.

0.052 hours

8.

0.61 hours

9.

4.25 h-1

10.

3.56 h-1

11.

6.62 h-1

12.

11.6

13.

hours

14.

68.0%

15.

Yes

Meropenem
1.

11.35

2.

39.76

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-128

Multicompartment Modeling

3.

51.11

4.

9.78

5.

19.45 L

6.

12.20 L

7.

0.375 hours

8.

1.378 hours

9.

1.16 h-1

10.

0.802 h-1

11.

0.391 h-1

12.

41

13.

0.967 hours

14.

37.3%

15.

No

N-methylpyridinium-2-carbaldoxime chloride
1.

18.6

2.

3.106

3.

21.71

4.

12.9

5.

1.11 L

6.

6.77 L

7.

2.41 hours

8.

0.0598 hours

9.

1.752 h-1

10.

1.905 h-1

11.

8.218 h-1

12.

41.339

13.

0.327hours

?14.

45.5%

15.

Yes

Pyrazine Diazohydroxide
1.

41348.7

2.

46147.9

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-129

Multicompartment Modeling

3.

87496.6

4.

0.3744

5.

14.57 mL

6.

3.542 mL

7.

3.55 minutes

8.

26.97 minutes

9.

0.0474 min-1

10.

0.106 min-1

11.

0.0676 min-1

12.

9249

13.

11.97 minutes

14.

75.7%

15.

No

Terbinafene
1.

4692.8

2.

4594.6

3.

9287.4

4.

80.75

5.

3637.6 L

6.

300 L

7.

1.36 h

8.

31.2 h

9.

0.0421 h-1

10.

0.2692 h-1

11.

0.222 h-1

12.

2500

13.

6.42 hours

14.

91.8%

15.

No

Verrucarol
1.

3037.35

2.

57143.8

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-130

Multicompartment Modeling

3.

60181.1

4.

149.5

5.

15.81 L

6.

13.5 L

7.

16.7 minutes

8.

73.3 minutes

9.

0.0155 min-1

10.

0.0253 min-1

11.

0.0101 min-1

12.

666.63

13.

46.15 minutes

14.

28.1%

15.

Yes

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

11-131

CHAPTER 12

Protein Binding

Author: Michael Makoid


Reviewer: Phillip Vuchetich

OBJECTIVES
2.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

12-1

CHAPTER 13

Nonlinear (Michaelis-Menton)
Kinetics

Author: Michael Makoid


Reviewer: Phillip Vuchetich

OBJECTIVES
1.

Given population average patient data, the student will devise (V) a dosage regimen which will maintain plasma concentrations of drug within the therapeutic
range.

2.

Given specific patient information, the patient will justify (VI) the optimal dosage
regimen.

3.

Given patient information regarding organ function, the student will devise (V)
and justify (VI) dosage regimens for the compromised patient.

4.

The student will write (V) a professional consult using the above calculations.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-1

Nonlinear (Michaelis-Menton) Kinetics

13.1 Problems

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-2

Nonlinear (Michaelis-Menton) Kinetics

Cefadroxil
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 27)

AHFS 00:00.00
GPI: 0000000000

Sanchez-Pico, A., et al, "Nonlinear intestinal abosrption kinetics of cefadroxil in the rat", Journal of Pharmacy Pharmacology,
Vol.41, (1989), p. 179 - 185.

Cefadroxil is a cephalosporin antibiotic which is commonly used to treat various infections. It is usually given orally.
This study looks at the pharmacokinetics and bioavailability of cefadroxil in the rat.
PROBLEM TABLE 11 - 27. Cefadroxil

Dose
500 mg

7.31

1000 mg

14.67

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of 10 ?
You recommend changing the patient's dosage regimen to 300 mg/day. What would be your patient's steady state
plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-3

Nonlinear (Michaelis-Menton) Kinetics

CD4
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 28)

AHFS 00:00.00
GPI: 0000000000

Qian, M., et al., "Pharmacokinetic evaluation of drug interactions with anti-human immunodeficiency virus drugs: V. effect of soluble CD4 on 2',3'-dideoxycytidine kinetics in monkeys", Drug Metabolism and Disposition, Vol. 20, (1992), p. 396 - 400.

2',3'-dideoxycytidine in combination with recombinant ST4 has been shown to be effective against HIV (human immunodeficiency virus) in vitro. This study examines whether or not the pharmacokinetics of 2',3'-dideoxycytidine are
affected by administration of CD4 (an immunoglobulin). Doses of each drug were given to male adult monkeys
weighing an average of 4.45 kg. The following data is for ST4 (soluble CD4).
PROBLEM TABLE 11 - 28. CD4

Dose
1.1 mg/kg

10.27

2.2 mg/kg

22.23
Weight of Monkey = 4.45 kg

Find .
Find the maximum clearance for this patient.
What would be the dose needed to acheive a steady-state concentration of 15 ?
You recommend changing the patient's dosage regimen to 1.5 mg/ kg. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-4

Nonlinear (Michaelis-Menton) Kinetics

Methylprednisone
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 29)

AHFS 00:00.00
GPI: 0000000000

Haughey, D, and Jusko W.., "Bioavailability and nonlinear dispositionof methylprednisolone and methylprednisone in the rat",
Journal of Pharmaceutical Sceicnes, Vol. 81, (1992), p. 117 - 121.

Methylprednisone is a corticosteroid which is commonly used in the treatment of medical emergencies such as cardiovascular shock, asthma, and cerebral edema. The following data was obtained for two methylprednisolone doses. The
plasma concentration measurement given for each dose below is that for the central compartment.
PROBLEM TABLE 11 - 29. Methylprednisone

Dose
10 mg

6834

50 mg

71519

Find .
Find the maximum clearance for this patient.
What would be the dose needed to acheive a steady-state concentration of 10,000 ?
You recommend changing the patient's dosage regimen to 30 mg. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-5

Nonlinear (Michaelis-Menton) Kinetics

Mezlocillin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 30)

AHFS 00:00.00
GPI: 0000000000

Jungbluth, G. and Jusko, W., "Dose-dependent pharmacokinetics of mezlocillin in rats", Antimicrobial Agents and Chemotherapy,
Vol. 33, (1989), p. 839 - 843.

Mezlocillin is an antibiotic used to treat various types of infection. It is usually given by the intravenous route and
exhibits dose-dependent (nonlinear) pharmacokinetics. This article compares two intravenous bolus doses, one of 20
mg/kg and one of 200 mg/kg in rats. The following data was calculated from the results of this study.
PROBLEM TABLE 11 - 30. Mezlocillin

Dose
20 mg/kg

158.6

200 mg/kg

294.1
Rat weight = 425 g

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of

200 ?

You recommend changing the patient's dosage regimen to 150 mg/ kg. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-6

Nonlinear (Michaelis-Menton) Kinetics

Naphthol
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 31)

AHFS 00:00.00
GPI: 0000000000

Redegeld, A., Hofman, G., and Noordhoek, J., "Conjugative clearance of 1-naphthol and disposition of its glucuronide and sulfate
conjugates in the isolated perfused rat", Journal of Harmacology and Experimental Therapeutics, Vol. 244, (1988), p. 263 - 267.

1-naphthol is a small phenolic compound which is extensively metabolized by conjugation. This study looks at the
pharmacokinetics of naphthol in a rat.
PROBLEM TABLE 11 - 31. Naphthol

Dose
30 mol

6.79 M

40 mol

8.63 M

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of 7.7 M?
You recommend changing the patient's dosage regimen to 35 mol. What would be your patient's plasma
concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-7

Nonlinear (Michaelis-Menton) Kinetics

Paroxetine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 32)

AHFS 00:00.00
GPI: 0000000000

Sindrug, S., Brosen, K, and Gram, L.., "Pharmacokinetics of the selective serotonin reuptake inhibitor paroxetine: nonlinearity
and relation to the sprateine oxidation polymorphism", Clinical Pharmacology and Therapeutics, Vol. 51, (1992), p. 288 - 295.

Paroxetine hydrochloride (Paxil) is a selective serotonin reuptake inhibitor which is used in the treatment of
depression. Paroxetine is metabolized both by oxidation and conjugation with the conjugated metabolites
excreted in the urine. Paroxetine exhibits dose-dependent (nonlinear) pharmacokinetics. The following
data is for a male diabetic patient who was concurrently taking insulin.
PROBLEM TABLE 11 - 32. Paroxetine

Dose
10 mg daily

1.65

20 mg daily

3.30

30 mg daily

8.25

40 mg daily

13.20

50 mg daily

26.40

60 mg daily

39.60

70 mg daily

66.00

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of 50 ?
You recommend changing the patient's dosage regimen to 36 mg/day. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-8

Nonlinear (Michaelis-Menton) Kinetics

Phenytoin
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 33)

AHFS 00:00.00
GPI: 0000000000

Levine, M., et al., "Evaluation of serum phanyton monitoring in an acute care setting", Therapeutic Drug Monitoring, Vol. 10,
(1988)., p. 50 - 57.

Phenytoin is an agent which is commonly used in the treatment of epilepsy. This drug exhibits nonlinear kinetics.
Phenytoin is mainly eliminated from the body by hepatic cytochrome P-450 metabolism. Several doses of phenytoin
were studied in patients and the data is summarized below:
PROBLEM TABLE 11 - 33.

Phenytoin

Dose

Day

300 mg at bedtime

5.0

12

11.4

49

21.5

(started on day 1)
200 mg BID
(started on day 6)
200 mg BID
(started on day 6)

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of

15 ?

You recommend changing the patient's dosage regimen to 300 mg/day. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-9

Nonlinear (Michaelis-Menton) Kinetics

Phenytoin in the Critically Ill


Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 34)

AHFS 00:00.00
GPI: 0000000000

Boucher, B. et al., "Phenytoin pharmacokinetics in critically ill trauma patients", Clinical Pharmacology and Therapeutics, Vol.
44, (1988)., p. 675 - 683.

Phenytoin is an agent which is commonly used in the treatment of epilepsy. This drug exhibits nonlinear kinetics. This
study looks at several doses of phenytoin in severely ill trauma patients. The data given below is that obtained for one
male, 25 year-old, patient who weighed 85 kg.
PROBLEM TABLE 11 - 34. Phenytoin

in the Critically Ill

Dose
615 mg/ day

10

588 mg/day

8.5

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of 12?
You recommend changing the patient's dosage regimen to 450 mg/ day. What would be your patient's steady-state
plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-10

Nonlinear (Michaelis-Menton) Kinetics

Phenytoin in Pediatrics
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 35)

AHFS 00:00.00
GPI: 0000000000

Bauer, L. and Blouin, R., "Phenytoin Michaelis-Menten pharmacokinetics in caucasian paediatric patients", Clinical Pharmacokinetics, Vol. 8, (1989)., p. 545 - 549.

Phenytoin is an agent which is commonly used in the treatment of epilepsy. This drug exhibits nonlinear kinetics. This
study looks at several doses of phenytoin in pediatric patients of several ages. The data for the 4 to 6 year old patients
is given below.

Dose
7.5 mg/ kg/ day

15

6.5 mg/ kg/day

10

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of

12 ?

You recommend changing the patient's dosage regimen to 4.5 mg/ kg/ day. What would be your patient's steady-state
plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-11

Nonlinear (Michaelis-Menton) Kinetics

Quinalapril

(Problem 11 - 36)

Problem Submitted By: Maya Leicht


Problem Reviewed By: Vicki Long

AHFS 00:00.00
GPI: 0000000000

Elliott, H., et al., "Dose responses and pharmaockinetics for the angiotensin converting enzyme inhibitor, quinapril", Clinical
Pharmacology and Therapeutics, Vol. 52, (1992), p. 260 - 265.

Quinalapril is an angiotensin converting enzyme (ACE) inhibitor which is used in the treatment of hypertension and
heart failure. The optimal dosage regimen for the ACE inhibitors is controversial and this study further investigates
quinalapril's pharmacokinetics at various doses ranging from 0.5 to 20 mg. Quinalapril is a prodrug which is metabolized to its active form, quinalaprilat.

Dose
(of quinalapril)

(of quinalaprilat)

2.5 mg daily

47.5

5.0 mg daily

98.1

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of

75 ?

You recommend changing the patient's dosage regimen to 3.0 mg/day. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-12

Nonlinear (Michaelis-Menton) Kinetics

Vanoxerine
Problem Submitted By: Maya Leicht
Problem Reviewed By: Vicki Long

(Problem 11 - 37)

AHFS 00:00.00
GPI: 0000000000

Ingwersen, S., et al., "Nonlinear multiple-dose pharmacokinetis of the dopamine reuptake inhibitor vanoxerine", Journal of Pharmaceutical Sciences, Vol. 82, (1993)., p. 1164 - 1166.

Vanoxerine is a pre-synaptic dopamine reuptake inhibitor which may be useful as an antidepressant. The bioavailability of vanoxerine is changed by food intake. The bioavailability after fasting is increased 76% by a low-fat meal and
255% by a high-fat meal. In this study, the volunteers were given doses of vanoxerine after eating a standard breakfast
of one bowl of cereal with milk, two slices of toast with sunflower margarine and jam, and one cup of tea.

Dose
25 mg

3.4

75 mg

15.1

125 mg

46.5

Find .
Find the maximum clearance,, for this patient.
What would be the dose needed to acheive a steady-state concentration of

30.0 ?

You recommend changing the patient's dosage regimen to 100 mg. What would be your patient's plasma concentration?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-13

Nonlinear (Michaelis-Menton) Kinetics

13.2 Nonlinear Equations


The following equations were used to solve the questions following each "nonlinear" scenario. Three scenerios have
been completed for you. The answers have been provided for the remainder.
1.
2.
3.
4.
Phenytoin in the Critically Ill
CD4
1. The monkeys had an average weight of 4.45 kg.
2.
3.
4.
Cefadroxil
1.
2.
3.
4.

13.2.1

ANSWERS

Cefadroxil
2144.75
147.2 g/day
683.14 mg
4.38
CD4
135.09
69.28 mg/day
6.92 mg/day
14.4
Methylprednisone

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-14

Nonlinear (Michaelis-Menton) Kinetics

52345.27
86.60 mg/day
13.89 mg/day
27747.1
Mezlocillin
324.95
25.92 mg/day
68.23 mg/day
1676.04
Naphthol
46.14 (M
233.86 (mol
25.61 (mol
43.5 (M
Paroxetine
4.125
45 mg/day
41.6 mg/day
16.5
Phenytoin
4.014
540.85 mg/day
426.7 mg/day
5
Phenytoin in the Critically Ill
3.517
831.3 mg/day
642.88 mg/day
4.15
Phenytoin in Pediatrics
6.67
162.5 mg/day

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-15

Nonlinear (Michaelis-Menton) Kinetics

104.46 mg/day
4.74
Quinalapril
1503.15
81.61 mg/day
3.88 mg/day
57.36
Vanoxerine
20.96
179.08 mg/day
105.4 mg/day
26.5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

13-16

CHAPTER 14

Practice Exams: Section 1

Author: Michael Makoid


Reviewer: Vicki Long

ORGANIZATION
Following are several exams which have been used
in previous classes. These exams cover Chapters 1
through 4. Answers (including the graphs) are
included in a format similar to real exam conditions,
and some exams have the problems worked out
completely.
You are encouraged to work out the exams under
conditions similar to how you will take the exam.
You will want to have a calculator, semi-log graph
paper, and scratch paper available. Before you
begin to answer the questions, you will want to: 1)
create the model from the description of the drugs
pharmacokinetics, 2) graph all of the data as discussed in the chapters. From these graphs the
answers will become available. Your graphs should
look like the graphs included in the answers. Applicable equations vary by exam and are included at the
beginning of each chapter covered by the exam.
Remember where you are in the course: Learn the
tools; get the pharmacokinetic parameters from
patient information. So in this section you must be
able to analyze pharmacokinetic data and extract the
pharmacokinetic parameters. That means: Assess
the pharmacokinetic description of the drug and creBasic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-1

Practice Exams: Section 1

ate a model, develop equations for the model, graph


the data according to those equations, and get the
parameters out. A student commented, You mean,
all we have to do is get the slope and intercept from
a couple of graphs and tell you what they mean?
Yes, that right!
Do not just look at the graphs and the answers and
say, Yes, that makes sense. I can do this. You will
be surprised to find out that what looks easy worked
out is not so easy when it isnt.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-2

Practice Exams: Section 1 : Nifedipine - Section 1

14.1 Nifedipine - Section 1


Masotti et al (J Clin Pcol 1985; 25: 27-35) and Traube et al (ibid 125-9) looked at the hemodynamic pharmacological response of
nifedipine.

14.1.1

NIFEDIPINE DATA

Nifedipine (Procardia ) is a calcium channel blocker which specifically inhibits potential-dependent channels not
receptor-operated channels, preventing calcium influx of cardiac and vascular smooth muscle (coronary, cerebral).
Calcium channel blockers reduce myocardial contractility and A-V node conduction by reducing the slow inward calcium current. They are indicated in angina, cardiac dysrhythmias, and hypertension among others. Nifedipine
appears to be metabolized entirely into an inactive metabolite, an acid and subsequently further metabolized to a lactone. Both the acid and the lactone are excreted into the urine and the feces.
PROBLEM TABLE 11 - 1.

Pharmacological Data

Fall in Diastolic
BP

Cp (ng/mL)

Time (hr)

Fall in Diastolic
BP

15

13.0

10

40

12.3

12

100

10.7

13

200

10.0

7.0

Echizen and Eichelbaum (Clin Pkin 1986; 11:425-49) and Kleinbloesem et al (Clin Pcol Therap 1986; 40: 21-8)
reviewed the pharmacokinetics of Nifedipine. While the drug is not routinely given by IV bolus and does not strictly
conform to a one compartment model, let's treat the data as if those problems can be ignored. The following data is
offered for evaluation:
PROBLEM TABLE 11 - 1. Nifedipine

IV Bolus Profile

Cp
(mcg/L)

Xm1f
(mg)

Xm1u
(mg)

Time (hr)

Cm1
(mcg/L)

0.5

24.7

44.4

139

71.8

.14

.59

65.6

96.5

.44

1.83

Xm2f
(mg)

Xm2u
(mg)

.028

.11

31.1

100

.77

3.25

.073

.29

14.6

94.7

1.1

4.65

.135

.54

12

76.5

1.69

7.10

.291

1.15

24

34

2.77

11.63

.75

2.95

3.6

15.1

1.3

5.0

7 days

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-3

Practice Exams: Section 1 : Nifedipine - Section 1

14.1.2

NIFEDIPINE QUESTIONS
1) dR/dT = slope of the Pharmacological
response vs. time profile (mmHg/hr)
2) dR/d(lnC) = slope of the Pharmacological response vs concentration profile
(mmHg)

18) AUMCiv = Area under the first moment


of the plasma concentration of Nifedipine vs
time curve of IV dose (mic/L*hr2 )

19) MRTiv = Mean Residence time of


Nifedipine given as the IV dose (hr)

3) d(lnC)/dt = slope of the concentration


vs time profile (hr-1 )

20) Xm10 = Mass of acidic metabolite in


the body at time = 0 (mg)

4) kr = rate constant of excretion of Nifedipine into the urine (hr-1 )

21) Xm1inf = Mass of the acidic metabolite in the body at infinite time. (mg)

5) kf = rate constant of excretion of Nifedipine into feces (hr-1 )

22) Vm1 = Volume of distribution of the


acidic metabolite in the body (L)

6) km1 = rate of metabolism of Nifedipine in the body (hr-1 )

23) Cm1 = Concentration of acidic metabolite in the body at time = 0 (mic/L)

7) K1 = elimination rate constant of Nifedipine in

Im1 = Intercept of the acidic metabolite concentration vs time profile = 181.2 mic/L

the body; the summation of all of the ways that it is


removed from the body. (hr-1 )

8) T1/2 of Nifedipine in the body (hr)


9) X0 = Mass of Nifedipine in the body
at time 0 (mg)
10) Xinf = Mass of Nifedipine in the body
at time infinite time (mg)
11) Xuinf = Mass of unchanged Nifedipine in the urine at infinite time (mg)
12) Xfinf = Mass of unchanged Nifedipine in the feces at infinite time (mg)
13) V = Volume of distribution of Nifedipine (L)
14) Cp0 = Concentration of Nifedipine in
the body at time 0. (mic/L)
15) AUCiv = Area under the plasma concentration of Nifedipine vs time curve of
the IV dose (mic/L*hr)
16) First trapazoid of the AUCiv (mic/
L*hr)
17) Last trapazoid of the AUCiv (mic/
L*hr)

24) AUCmet = Area Under the Curve of


the metabolite plasma concentration vs
time profile (mic/L*hr)
AUMCmet = Area Under the First Moment
Curve of the metabolite plasma concentration vs
time profile = 35700 mic/L*hr2

25) Xm1u0 = Mass of acidic metabolite


in urine at time = 0 (mg)
26) Xm1uinf = Mass of acidic metabolite
in urine at time = infinity (mg)
27) Xm1f0 = Mass of acidic metabolite in
feces at time = 0 (mg)
28) Xm1finf = Mass of acidic metabolite
in feces at time = infinity (mg)
29) Km1u = Rate constant of excretion of
acidic metabolite into urine (hr-1 )
30) Km1f = Rate constant of excretion of
acidic metabolite into feces (hr-1 )
31) Km2=Rate constant of formation of
lactone metabolite in body =rate of metabolism of
the acidic metabolite in the body (hr-1 )

32) K2 = Km1f + Km1u + Km2 = sumation of


all of the ways that the acidic metabolite is eliminated from the
body (hr-1 )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-4

Practice Exams: Section 1 : Nifedipine - Section 1

33) Xm20 = Mass of lactone metabolite


in body at time = 0 (mg)

38) Xm2finf = Mass of lactone metabolite


in feces at time = infinity (mg)

34) Xm2inf = Mass of lactone metabolite


in body at time = infinity (mg)

K3 = Km2f + Km2u = the sumation of all of


the ways that the lactone can be eliminated =
0.138 hr.-1

35) Cm2inf = Concentration of lactone


metabolite in the body at time = infinity (0)
36) Xm2u0 = Mass of lactone metabolite
in urine at time = 0 (mg)

39) Km2u = Rate constant of excretion of


lactone metabolite into urine (hr -1 )
40) Km2f = Rate constant of excretion of
lactone metabolite into feces (hr -1 ) .

37) Xm2uinf = Mass of lactone metabolite in urine at time = infinity (mg)


PROBLEM TABLE 11 - 1.

Answer Pool Any number from the answer pool may be used

once, more than once, or not at all


Negative Numbers

Small Numbers

Big Numbers

-0.0001

13

-0.0010

0.010

15.1

-0.0100

0.018

17

-0.0180

0.028

20

-0.0280

0.042

25

-0.0375

0.070

38.9

-0.0750

0.085

50

-0.0850

0.110

85

-0.110

0.138

170

-0.138

0.375

181.2

-0.375

0.750

295

-0.750

1.30

415

-1.30

1.85

434

-1.85

2.0

787

-2.0

2.67

2100

-2.67

3.6

3570

-3.6

5.0

8500

-5.0

7.5

21000

-7.5

8.5

35700

-10

9.9

85000

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-5

Practice Exams: Section 1 : Nifedipine - Section 1

14.1.3

NIFEDIPINE SOLUTIONS
X
km1

Xmf1

Xm1

kmf1

kmu1

Xmu1

kmu2

Xmu2

km2
Xmf2

Xm2

kmf2

Graph #2

Graph #1

- 0.75 mmHg/hr

R vs T

Nifedipine

14

14

13
13

RESPONSE (MMHG)

12

Response (mmHg)

12

11
10

11

10

2 mm Hg

9
8
7
6

0
101

102

103

Concentration (ng/mL)

Graph #4

Nifedipine IV Bolus

10

/mL)

L)

Graph #3

Time (hr)

Nifedipine IV bolus - Metabolite

103

103

CONCENTRATION MIC/L

CONCENTRATION MIC/L

181.2 mic/L = Intercept (given)

100
102

1.85 hr

50

101
2

102

10 hrs

101

Time (hours)

12

16

20

24

Time (hours)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-6

Practice Exams: Section 1 : Nifedipine - Section 1

dR
------- = 0.75mmHg hr
dT

1.

dR
------------ = 2.0mmHg
d ln C

2.

Slope from graph # 2

Slope from graph # 1

dR
------0.75 = 0.375hr 1
dT - = ------------d-----------ln C = -----------2
dR
dT
-----------d ln C

3.

Answer
#1------------------------Answer #2

4.

kr = 0, Nifedipine is not excreted into the urine, it is metabolized entirely K1 = k m1 from pkin description

5.

kf = 0, Nifedipine is not found in the feces, only the metabolites are.

6.

km1, Rate of metabolism of Nifedipine in the body = ---------------- = 0.375 hr

7.

K1, Elimination rate constant of Nifedipine in the body = 0.375hr

K1 = k m1 from pkin description

0.693
1.85hr

from graph #3

from graph #3

T 1 2 = 1.85 hr from graph #3

8.

Xo = ( 3.6 + 15.1 + 1.3 + 5.1 ) = 25mg

9.
10.

Xinf = 0 from pkin description

11.

X u ( inf ) = 0 from pkin description

12.

X f ( inf ) = 0 from pkin description

13.
14.
15.

from pkin description = ( X mu1 + X mu2 + X mf 1 + X mf2 )

Dose- = ------------------------25mg - 1000mic


--------------------- = 85L
V = -----------Cp o 295mic L
1mg
295mic L Cp0 extrapolated back to t = 0 obtained from graph #3
Cp
295 = 787mic L hr
AUC iv = ---------o = ------------0.375
k

as an estimate. Using Trapazoidal rule:

Cp 1 + Cp 2
Cp 2 + Cp 3
Cp 3 + Cp last
Cp last
Cp o + Cp 1
------------------------- t 1 + ------------------------- t2 + ------------------------- t 3 + ------------------------------ t last + --------------
2
2
2
2
K1

+ 139- 2 + 139
+ 65.6 2 + 65.6
+ 31.1- 2 + 31.1
+ 14.6- 2 + ------------14.6 mcg
295
----------------------------------------------------------------------------------------------------------hr
2
2
2
2
0.375 L

{ 434 + 204.6 + 96.7 + 45.7 + 38.9 } mcg


----------hr = 821.9mcg
----------hr
L
L

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-7

Practice Exams: Section 1 : Nifedipine - Section 1

16

17.
18

Cp o + Cp 1
-------------------------- t =
2

295 + 139 )- 2 = 434mic L hr


(---------------------------

Cp L 14.6
---------- = ------------- = 38.9mic L hr
k
0.375
AUMC = AUC MRT = 787 2.67 = 2100mic L hr

estimate. Using the trapazoidal rule:

T1 C p1 + T 2 C p 2
T2 C p2 + T3 C p3
T 3 C p 3 + T last C p last
T last C p last Cp last
T 0 C p o + T1 C p1
---------------------------------------------- t 1 + ---------------------------------------------- t 2 + ---------------------------------------------- t 3 + --------------------------------------------------------- t last + ------------------------------ + ---------------
2
2
2
2
2
K1

K1
0 295 + 2 139
2 139 + 4 65.6
4 65.6 + 6 31.1
6 31.1 + 8 14.6
8 14.6
14.6 mcg 2
--------------------------------------- 2 + ---------------------------------------- 2 + ------------------------------------------ 2 + ------------------------------------------ 2 + ------------------ + ---------------- ----------hr
2
2
2
2
2
0.375

0.375 L
mcg 2
{ 278 + 540.4 + 449 + 303.4 + 311.47 + 103.82 } = 1986.1----------hr
L

19.

MRT IV

1
MRT iv = --- = 2.67hr estimate. Using trapazoidal rule and definitions
k
mcg 2
mcg 2
1986.1----------hr
AUMC est 2100 ----------hr
AUMC trap
L
L
= --------------------------- = ------------------------------------ = 2.42 hr ------------------------ = ------------------------------- = 2.67 hr
mcg
mcg
AUC est
AUC trap
787 ----------hr
821.9----------hr
L
L

20.

X m1 ( o ) = 0 from pkin description

21.

X m1 ( inf ) = 0 from pkin description

22.

Vm1 = 170L

From LaPlace transforms, the equation for the metabolite is


k

k m1 X 0
K2t
K1t
Cp m = --------------------------------------- {e
e
}
( K1 K2 ) V dm

m1
0
- .
the intercept, I, given as 181.2 mic/L in the data = ---------------------------------------

( K1 K2 ) V dm

Thus

k m1 X 0
0.375 25mg
V dm = ----------------------------------------------------= ---------------------------------------------------------------- = 169.6 L
mic
mg
( K1 K2 ) 181.2--------( 0.375 0.07 ) 0.1812------L
L

23.

C m1 ( 0 ) = 0 from pkin description

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-8

Practice Exams: Section 1 : Nifedipine - Section 1

AUC met = 2100mic L hr

24.

PROBLEM TABLE 11 - 1. Nifedipine

Data

Cpm ( n ) + Cpm ( n + 1 )
-------------------------------------------------------2

T ( n )

AUC

AUC

t
0

Time (hr)

Cpm (mcg/L)

0.5

24.7

0 + 24.7
------------------2

0.5

6.175

6.175

44.4

24.7 + 44.4
--------------------------2

0.5

17.275

23.45

71.8

44.4 + 71.8
--------------------------2

58.1

81.55

96.5

71.8 + 96.5
--------------------------2

168.3

249.85

100

96.5 + 100
------------------------2

196.5

446.35

94.7

100 + 94.7
------------------------2

194.7

641.05

12

76.5

94.7 + 76.5
--------------------------2

342.4

983.45

24

34

76.5 + 34
---------------------2

12

663

1646.45

Cpm last
+ ------------------K small

485.7

2132.15

25.

X m1u ( 0 ) = 0 from pkin description

26.

X m1u ( inf ) = 15.1mg given in data

27.

X m1f ( 0 ) = 0 from pkin description

28.

X m1f ( inf ) = 3.6mg given in data

29.

k m1u = 0.042hr

30.

k m1f = 0.01hr

31.

k m2 = 0.018hr

k m1u Xm1u
k m1u
----------- = ------------ = -------------------= 15.1mg
-----------------
1
K2
Xo
25mg
0.07hr
k m1f X m1f
k m1f
---------- = ----------- = -------------------= 3.6mg
--------------1
K2
Xo
25mg
0.07hr
0.07 hr

trap

thus k m1u = 0.042hr

thus k m1f = 0.01hr

= K 2= k m1f + k m1u + k m2 = 0.01 hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

+ 0.042 hr

+ k m2
14-9

Practice Exams: Section 1 : Nifedipine - Section 1

32.

K2 = 0.07hr

From your LaPlace Transforms, you know that the equation is bi-exponential, with one of

the slopes being K1 and the other being K2. The terminal slope of graph #4 is 0.07 hr -1, which is not K1 (0.375 hr -1).
So it must be K2.
33.

X m2 ( 0 ) = 0 from pkin description

34.

X m2 ( inf ) = 0 from pkin description

35.

C m2 ( inf ) = 0 from pkin description

36.

X m2u ( 0 ) = 0 from pkin description

37.

X m2u ( inf ) = 5.0mg given in data

38.

X m2f ( inf ) = 1.3mg given in data

39.

k m2u = 0.11hr

40.

k m2f = 0.028hr

k m2u
k m2u
X m2u
5.0mg
----------------------- = ----------------------------= ---------= -----------------------------------1
K3
X m2u + Xm2f
5.0mg + 1.3mg
0.138hr
k m2f
k m2f
Xm2f
1.3mg ----------------------= --------- = ----------------------------= -----------------------------------1
K3
X
+
X
5.0mg
+ 1.3mg
m2u
m2f
0.138hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-10

Practice Exams: Section 1 : Enalapril - Section 1

14.2 Enalapril - Section 1


Citation

14.2.1

ENALAPRIL DATA

Enalapril (Vasotec ) is an angiotensin converting enzyme (ACE) Inhibitor effective in the treatment of hypertension
and chronic heart failure (CHF). The parent compound is weakly active, while its sole metabolite, Enalaprilat, exhibits almost all of the pharmacological activity. Original work on the pharmacokinetics of Enalapril showed the parent
drug, Enalapril and its metabolite, Enalaprilat, show up in both feces and urine.
The following data was offered regarding the Pharmacological response of Enalaprilat as judged by the fall in Seated
Systolic Blood Pressure (SSBP).
PROBLEM TABLE 11 - 1. Enalapril

- Section 1: Pharmacological Profile

Fall in SSBP
(mmHg)

Time (hr) after


Max

Dose (mg)

Fall in SSBP
(mmHg)

15

12

10

10

10

8.6

12

15

10

18

20

16

24

25

20

30

24

PROBLEM TABLE 11 - 1. Enalapril

- Section 1 IV Bolus Profile from a 5 mg dose

Cpm
(ng/ml)

Cumulative
Enalapril in
urine (mg)

Cumulative
Enalapril
in feces
(mg)

Time
(hr)

Cp
(ng/mL)

29

7.5

0.41

0.12

17

11.6

0.65

0.20

10

13.8

0.80

0.24

5.9

14.9

0.88

0.26

15.2

0.96

0.30

1.0

0.3

14.7

10

13.8

20

9.8

30

6.9

40

4.9

50

3.4

Cumulative
Enalaprilat
in urine
(mg)

Cumulative
Enalaprilat
in feces
(mg)

2.25

1.45

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-11

Practice Exams: Section 1 : Enalapril - Section 1

The intercept of Cp m vs t was found to be 19.8 ng mL and a total of 2.25 mg of enalaprilat was found in the
urine while 1.45 mg was found in the feces.
PROBLEM TABLE 11 - 1. Enalapril

Answer Pool

Negative
Numbers

Small
Numbers

Small
Numbers

Medium
Numbers

Large
Numbers

0.000

0.014

1.00

14.5

100

-0.014

0.021

1.45

19.8

174

-0.021

0.030

.1.90

30.3

528

-0.030

0.035

2.25

33.0

395

-0.035

0.039

3.75

37.5

693

-0.110

0.110

5.00

39.5

834

-0.390

0.150

6.93

50.0

1740

-0.530

0.300

8.00

70.0

5280

-0.700

0.390

9.00

94.0

14400

-0.900

0.530

9.76

97.0

16000

Equation Answer Pool:


A

k mu k m Xo 1 e K2t 1 e K1t
------------------------ --------------------- --------------------( K1 K2 )
K2
K2

( k mf k m Xo ) 1 e K2t 1 e K1t
------------------------ --------------------- --------------------( K1 K2 )
K2
K1

km X 0
K2t
K1t
---------------------------------------
) (e )
( K1 K2 ) Vd m ( e

D.

k mu k m Xo
K2t
K1t
------------------------
) (e )
( K1 K2 ) ( e

km X 0
K2t
K1t
------------------------
) (e )
( K1 K2 ) ( e

K1t

kr X 0 e

k
K1t
------u- X0 ( 1 e
)
K1

X 0 K1t
------ e
V
k
K1t
------f- X0 ( 1 e
)
K1
X0e

K1t

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-12

Practice Exams: Section 1 : Enalapril - Section 1

14.2.2

ENALAPRIL QUESTIONS
Find the equation for:
1) X, the mass of parent drug, Enalapril, in
the body.
2) Xu, the mass of parent drug, Enalapril,
in urine.
3) dX u/dt, the rate of excretion of parent
drug, Enalapril, in urine.
4) Xf, the mass of parent drug, Enalapril, in
feces.
5) Xm, the mass of metabolite, Enalaprilat,
in the body.
6) Xmu, the mass of metabolite, Enalaprilat, in urine.
7) dXmu/dt, the rate of excretion of metabolite, Enalaprilat, in urine.
8) Xmf, the mass of metabolite, Enalaprilat, in feces.
9) Cp, the plasma concentration of parent
drug, Enalapril.
10) Cpm, the plasma concentration of
metabolite, Enalaprilat.

18) kmf, the fecal excretion rate constant


for metabolite, Enalaprilat, in plasma.
19) K1, the elimination rate constant of
parent drug in plasma(the summation of all
processes which remove Enalapril)
20) K2, the elimination rate constant of
metabolite in plasma(the summation of all
processes which remove Enalaprilat).

Find the intercept of the graph of:


21) Cp vs T
22) Cpm vs T (terminal extrapolated line)
23) dXu/dt vs T
24) dXf/dt vs T

Find the slope of the terminal portion of


the graph of:
25) Cp vs T
26) Cpm vs T
27) dXu/dt vs T
28) dXf/dt vs T

Find the value of:


11) dR/dT
12) dR/d(ln(C))

Find the area under the curve of the


graph of

13) d(ln(C))/dT

29) Cp vs T(first trapazoid only)

14) kr, the renal excretion rate constant of


parent drug, Enalapril, in plasma.

30) Cpm vs T(first trapazoid only)

15) kf, the fecal excretion rate constant of


parent drug, Enalapril, in plasma.
16) km, the metabolism rate constant
ofparent drug, Enalapril, in plasma.
17) kmu, the renal excretion rate constant
for metabolite, Enalaprilat, in plasma.

31) Cp vs T(from Tlast to T only)


32) Cpm vs T(from Tlast to T only)
33) Cp vs T
34) Cpm vs T
35) dXu/dt vs T

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-13

Practice Exams: Section 1 : Enalapril - Section 1

36) dXmu/dt vs T

49) XMU0

37) T*Cp vs T

50) XMF0

38) T*Cpm vs T

51) X

Find the value of :

52) X u

39) AUC(enalapril)
40) AUMC(enalapril)

53) X f

41) MRT(enalapril)

54) X m

42) AUC(enalaprilat)

55) X mu

43) AUMC(enalaprilat)

56) X mf

44) MRT(enalaprilat)

57) Cp0

45) X0

58) Cpm0

46) XU0

59) Vd

47) XF0

60) Vdm

48) XM0

14.2.3

ENALAPRIL SOLUTIONS

Enalapril Model
kf
Xf

kr

Xu

km
Xmf

kmf

Xm

kmu
Xmu

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-14

Practice Exams: Section 1 : Enalapril - Section 1

sponse

esponse

RESPONSE DATA

R vs C
24
22
20
18
16
14
12
10
10

10

R vs T

15
14
13
12
11
10
9
8
2

Concentration

10

12

Time (hr)
Enalapril

Enalapril IV Bolus - Metabolite


102

101

101

Concentration (ng/mL)

Concentration (ng/mL)

102

100
0

100
0

Time (hrs)

10

20

30

40

50

Time (hours)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-15

Practice Exams: Section 1 : Enalapril - Section 1

k1t

X = Xoe

k
k1t
X u = ----r- X o ( 1 e
)
k1

dx u dt = k r Xo e

10

k1t

k
k1t
)
X f = -----f Xo ( 1 e
k1
km X o
k2t
k1t
Xm = --------------------( e
e
)
( k1 k2 )
k mu k m Xo 1 e k2t 1 e k1t
Xmu = ---------------------------------------- -------------------( k1 k2 )
k2
k1
dX mu k mu k m Xo k2t k1t
------------- = --------------------- ( e
e
)
dt
k1 k2
( k mf k m X ) 1 e K2t 1 e K1t
o
X mf = ------------------------ --------------------- --------------------K1 K2
K2
K1

C p = C po e

k1t

D- e k1t
= -----Vd

km X 0
K2t
K1t
-
Cp m = --------------------------------------) (e
)
( K1 K2 ) Vd m ( e

11

dR dt = 0.7

12

dR d ( ln c ) = 20

13

d ( ln c )

0.7 = 0.035
dt = ---------20

14

Xu
1
k r = ------ k 1 = 1--- 53 = 0.106 = 0.11hr
5
Xo

15

Xf
1
k f = ------ k 1 = 0.3
------- 0.53 = 0.0318 = 0.03 hr
Xo
5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-16

Practice Exams: Section 1 : Enalapril - Section 1

16

Xmu + X mf
1
2.25 + 1.45
k m = ----------------------- k 1 = --------------------------- 0.53 = 0.39 hr
Xo
5

17

Xmu
1
2.25
k 2 = ---------- 0.035 = 0.21hr
k mu = -----------------------

3.7
Xmu + Xmf

Xmf
1
1
1.45
k 2 = ---------- 0.035hr = 0.014 hr
k mf = -----------------------
3.7
Xmu + Xmf

18

19

K1 = 0.53hr

20

K2 = 0.035hr

= slope of Cp vs T = k m + kf

= terminal slope of graph Cp m vs T = k mu + k mf

Cp ( 0 ) = 50ng mL

21
22

I = 19.8 ng mL

23

dX
--------u- vs T
dt

I = 0.53

k f X0 = ( 0.03 5 ) = 0.15ng mL

24

PROBLEM TABLE 11 - 1.

T mid

Time

Urinary Rate Set up

Xu

Xu

Xu

---------T

Xu

---------T

0.5

0.41

0.41

0.41 1

0.41

1.5

0.65

0.24

0.24 1

0.24

2.5

0.80

0.15

0.15 1

0.15

3.5

0.88

0.08

0.08 1

0.08

0.96

0.08

0.08 2

0.04

25

0.53 hr

26

0.035 hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-17

Practice Exams: Section 1 : Enalapril - Section 1

44

16000
--------------- = 30.3hr
528

45

X 0 = 5mg

29 )- 1 = 39.5ng mL hr
---------------------2

46

Xu ( 0) = 0

47

Xf ( 0 ) = 0

(0 +

48

Xm ( 0 ) = 0

49

Xmu ( 0 ) = 0

27

0.53 hr

28

0.53 hr

29
( 50 +

30

7.5 ) 1 = 3.75ng mL hr
--------------------2
31

2
---------- = 3.77ng mL hr
0.53

50

Xmf ( 0 ) = 0

32

3.4 - = 97ng mL hr
-----------0.035

51

X = 0

52

Xu = 1mg

33

94 ng mL hr

53

34

1 19.8 -----------0.035

1 - = 528ng mL hr
--------0.53

35

dX
AUC --------u- vsT = 1mg
dt

36

dX mu
AUC ------------ vsT = 2.25mg
dt

37

174 ng mL hr

38

16000 ng mL hr

39

94 ng mL hr

Xf = 0.3mg = ( 5 1 2.25 1.45 )

54

Xm = 0

55

X mu = 2.25mg

56

X mf = 1.45mg

57

Cp 0 = 50ng mL

58

Cp m ( 0 ) = 19.8ng mL
dose- = 100L
Vd = -----------Cp ( 0 )

59
2

40

174 ng mL hr

41

1.9 hr

42

528 ng mL hr

43

16000 ng mL hr

60

( km X0 )
( 0.39 5
)
Vd m = --------------------------= ----------------------------------------------------( K1 K2 ) I ( K1 K2 ) ( 0.0198
2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-18

Practice Exams: Section 1 : Ciprofloxacin Section 1

14.3 Ciprofloxacin Section 1


14.3.1

CIPROFLOXACIN DATA

Ciprofloxacin (Cipro ) is a fluoroquinolone, synthetic broad spectrum antibacterial agent. It is reasonably well
absorbed orally and is excreted into the urine and is metabolized to four known metabolites which are also excreted
into the urine. Before an accurate analytical assay was worked out for ciprofloxacin, a microbiological assay was used
to determine the pharmacokinetics. This assay utilized the ability of ciprofloxacin to kill microorganisms grown on an
agar plate and create a clear zone of no growth, the zone of inhibition. The following data was collected from that procedure: (Intercept for the extrapolated metabolite graph was 0.372 mg/L)
PROBLEM TABLE 11 - 1.

Data Set 0ne

Data Set Two

Pharmacological Response
Zone -mm

Time (hrs)

Pharmacological Response
zone - mm

Concentration Spiked plasma

27

4.7

0.01

23.2

13.5

0.05

20.4

17.3

0.1

18.5

10

19.5

0.15

5.2

24

21

0.2

23.5

0.3

28

0.7

PROBLEM TABLE 11 - 1. Data

Time
(Hrs)

Cp
(mg/L)

0.5

from 500 mg IV Bolus Cipro.

Cpm
(mg/L)

Interval
(hours)

Cipro (mg)
in urine

0.045

0-4

184

0.077

4-8

92

2.53

0.115

8 - 12

46

2.12

0.129

1.79

0.128

0.119

1.26

0.108

0.893

0.084

10

0.062

12

0.045

16

0.023

18

0.016

20

0.012

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-19

Practice Exams: Section 1 : Ciprofloxacin Section 1

14.3.2

CIPROFLOXACIN QUESTIONS

From the above information find the pharmacokinetic parameters:


1) K (elimination rate constant of Cipro) (hr-1 )
2) kr (urinary excretion rate constant of Cipro)(hr-1 )
3) km (metabolism rate constant of Cipro)(hr-1 )
4) kmu (urinary excretion rate constant of the metabolites)(hr-1 )
5) Cp0 (IV bolus Cipro) (mic/mL)
6) Vd (Cipro)(L.)
7) Vdm (metabolites)(L.)
8) AUC (Cp vs t for Cipro by IV bolus) (mic/mL * hr)
9) AUMC (t*Cp vs t for Cipro by IV bolus (mic/mL * hr2)
10) MRT (Cipro)(hr)
11) AUC (dXu/dt vs t(mid) for Cipro by IV bolus) (mg)
12) Xinf

(Cipro in the body at infinite time)

13) Xuinf

(Cipro in the urine at infinite time)

14) Xminf

(metabolites in the body at infinite time)

15) Xmu inf


16) T1/2

(metabolites in the urine at infinite time)

(Cipro)(hr)

17) Clearance (Cipro)(L/hr)


18) dR/dT (mm/hr)
19) dR/d(lnC) (mm)
20) d(lnC)/dt (Cipro)(hr-1)
21 Slope of Cp vs T ([Cipro] vs t) on semi-log paper
22 Slope of dXu/dt vs T(mid) vs t on semi-Log paper
23 Slope of terminal portion of Cpm vs T on semi-log paper
24) Slope of stripped portion of Cpm vs T on semi-log paper
25) Equation for the rate of formation of the metabolites in the urine (dXmu /dT vs T)
a) kmu * Xo * (e-Kt - e-kmut ) / (kmu - K)
b) kmu * km * Xo * (e-Kt - e-kmut ) / (K - kmu)
c) km * Xo * (e-Kt - e-kmut ) / (kmu - K)
d) kmu * km * Xo * {(1-e-Kt )/K- (1-e-kmut )/kmu} / (kmu - K)
e) kmu * km * Xo * (e-Kt - e-kmut ) / (kmu - K)
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-20

Practice Exams: Section 1 : Ciprofloxacin Section 1

14.3.3

CIPROFLOXACIN SOLUTIONS

Ciprofloxacin Model
X

ku

Xu

kmu

Xmu

km
Xm

Ciprofloxacin

Ciprofloxacin

30

20
15
10
5
0
5

10

15

20

25

Time (hr)

100

100

10-1

10 -1

10-2
4

Time (hours)

10 0

Concentration (mic/mL)

Ciprofloxacin Urine data


102

Concentration (mic.mL)

10 -1

Ciprofloxacin Metabolite data

101

10 -2

10

15

20

dXu/dt (mg)

Ciprofloxacin

Concentration (mic/mL)

Response (zone - mm)

Response (zone - mm)

25

30
25
20
15
10
5
0

101

Time (hours)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10

Tmid (hours)

14-21

Practice Exams: Section 1 : Ciprofloxacin Section 1

K = 0.173hr

1.

2.

R
hr- = 0.13hr 1
k r = -----0- = 65.13mg
-----------------------------X0
500mg
1

k m = 0.043hr

k mu = 0.462hr

Cp 0 = 3.57mic mL

Vd = 140L

Vd m = 200L

9.

AUMC = 119.7mic mL hr

10

MRT = 5.8hr

11

dX u
AUC = 375mg ---------vs
( t mid )
dt
X = 0

13

X u = 375mg

14

Xm = 0

16

T 1 2 = 4.0hr

17

Cl = 24.2L hr

18

dR
------- = 0.95mm hr
dT

19

dR = 5.5mm
----------------d ( ln C )

20

d----------------( ln C ) = 0.173hr 1
dt

21

Slope of Cp vs T = 0.173

22

Slope

of

dX u
--------- vs ( t mid ) = 0.173
dt
23

Slope of terminal portion of

Cp m vsT = 0.173
24

Slope of stripped portion of

Cp m vsT = 0.462
25

k mu k m X 0
Kt
kmut
---------------------------- (e e
)
( k mu K )

12

X mu = 125mg

AUC = 20.7mic mL hr
8
Cp vs T

15

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-22

Practice Exams: Section 1 : Methylphenidate Section 1

14.4 Methylphenidate Section 1


Citation

14.4.1

METHYLPHENIDATE DATA

Methylphenidate (MP) (Ritalin) is an effective stimulant in the treatment of narcolepsy in adults and attention deficit
syndrome in children. It is entirely metabolized to the inactive metabolite, Ritalinic Acid (RA), by the liver which is
subsequently excreted unchanged into the urine. Pharmacological response was measured objectively using sleep
latency as measured by encephalography. Sleep latency is the time required to fall asleep in a darkened quiet room.

PROBLEM TABLE 11 - 1.

Pharmacological Response Data


Time (hr)

Response (min)

Cp (mcg/mL)

Response (min)

0.5

22

22.5

16

18

1.5

10

15

10

PROBLEM TABLE 11 - 1. Plasma

Profiles
(Intercept of extrapolated RA line 0.21
mic/mL)
Plasma Vs. Time Profile
10 mg MP given by IV Bolus

Time (hr)

Cp (MP)
(mcg/mL)

Cpm (RA)
(mcg/mL

0.5

0.091

0.022

0.067

0.029

1.5

0.049

0.030

0.035

0.028

2.5

0.026

0.024

0.019

0.020

0.010

0.012

0.003

0.004

-------

0.0013

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-23

Practice Exams: Section 1 : Methylphenidate Section 1

14.4.2

METHYLPHENIDATE QUESTIONS:
From the above information find the pharmacokinetic parameters:
1) K (elimination rate constant of MP) (hr

-1

2) kr (urinary excretion rate constant of


MP)(hr-1 )
3) km (metabolism rate constant of
MP)(hr-1 )
4) kmu (urinary excretion rate constant of
(RA)(hr-1 )

25) d(lnCp)/dt (MP)(hr-1)


26) The slope of Cp vs T ([MP] vs t) on
semi-log paper
27) The equation for Cp vs T ([MP] vs t)
on semi- log paper
28) The slope of terminal portion of
dXmu/dt vs T(mid) on semi-Log paper
29) The equation for dXmu/dt vs T(mid)
on semi- log paper
30) The slope of terminal portion of Cpm vs

5) Cp0 (IV bolus MP) (mic/mL)

T ([RA] vs t) on semi-log paper

6) Vd (MP)(L.)

31) The equation for Cpm vs T ([RA] vs t)


on semi- log paper

7) Vdm (RA)(L.)
8) AUC (Cp vs t for MP by IV bolus) (mic/
mL * hr)
9) AUMC (t*Cp vs t for MP by IV bolus
(mic/mL * hr2)

32) The slope of stripped portion of Cpm vs


T ([RA] vs t) on semi-log paper

33) The equation for Xm

(bar) (Laplace domain

equation for RA in the body)

34) The equation for Xm (RA in the body)

10) MRT (MP)(hr)


11) AUC (dXu/dt vs t(mid) for MP by IV
bolus) (mg)
12) AUC (dXmu vs t(mid) for RA)(mg)
13) X0 (MP in the body at zero time)
14) Xm0 (RA in the body at zero time)
15) Xmu0 (RA in the urine at zero time)
16) X1hr (MP in the body at 1 hr.)
17) Xm1hr (RA in the body at 1 hr)
18) Xinf

(MP in the body at infinite time)

19) Xuinf

(MP in the urine at infinite time)

20) Xminf

(RA in the body at infinite time)

21) T1/2

(MP)(hr)

22) Clearance (MP)(L/hr)


23) dR/dT (min/hr)
24) dR/d(lnCp) (%)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-24

Practice Exams: Section 1 : Methylphenidate Section 1

14.4.3

METHYLPHENIDATE SOLUTIONS

X
km
Xm

kmu

Xmu

R vs C

Methyl Phenidate

24

Response

25
20

22
20
0

18

15

Response (min)

16

10

14
12

5
10

0.5

1.0

1.5

10

2.0

10

Concentration

Time (hr)

MP IV Bolus - Metab

100

100

10-1

10-1

Concentration (mic/mL)

Concentration (mic/mL)

Methyl Phenidate IV Bolus

10-2
0

Time (hours)

10-2
0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

Time (hours)

14-25

Practice Exams: Section 1 : Methylphenidate Section 1

K = 0.63hr

kr = 0

k m = 0.63hr

k mu = 0.90hr

Cp 0 = 0.125mic mL

6
pool)

Vd = 80L (change in answer

pool)

AUC = 0.20mic mL hr

T 1 2 = 1.1hr

22

Cl = 50.4L hr

23

dR
------- = 11.4min hr
dT

24

dR - = 17.5 min
-------------------d ( ln Cp )

25

d-------------------( ln Cp )- = 0.63hr 1
dT

Vd m = 115L (change in answer

21

26

The slope of Cp vs T 0.63hr

27

The equation for Cp vsT

Cp = Cp 0 e
28

9.

AUMC = 0.315mic mL hr
10

MRT = 1.57hr

The slope of terminal portion of

29

The equation for

dX mu
k mu k m X0 K1t
(e
e
------------- vsT mid = ----------------------------( K2 K1 )
dT
30

Cp m vsT ( RA )vsT = 0.94

X0 = 10mg

14

Xm ( o ) = 0

15

Xmu ( 0 ) = 0

32

16

X1hr = 5.3 (change in answer

0.79

31

The equation for

km X0
K1t
Cp m vsT ( RA ) = ------------------------(e
e
( K2 K1 )

pool)

Xm1hr = 2.8 (change in answer

pool)

33
34

X = 0

19

Xu = 0

20

Xm = 0

The slope of stripped portion

km X0
Xm = --------------------------------------------( s + K1 ) ( s + K2 )
The equation for

km X 0
K1t
K 2t
Xm = ------------------------(e
e
)
( K2 K1 )

18

the slope of the terminal portion of

13

17

k t

dX mu
------------ vsT mid = 0.63
dT

11
AUC = 0 MP is not excreted in
the urine
12
AUC = 10mg All of RA is
excreted in the urine

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-26

Practice Exams: Section 1 : Adinazolam - Section 1

14.5 Adinazolam - Section 1


Fleishaker [Pharm Res 8 (2): 162-7(1991),Pharm Res 6 (5): 379-86(1989),Psychopharm 99:34-9(1989),Psychopharm 105:1815(1991)];Kroboth [J Clin Pharmacol 31:580-6(1991)]; Wagner [Biopharm Drug Dis 8:405-25(1987)].

14.5.1

ADINAZOLAM DATA

Adinazolam (AD) is a triazolobenzodiazepine prodrug which has been shown to have antidepressant activity through
its metabolite, N-desmethyladinazolam (NDMAD). Both AD and NDMAD are excreted into the urine. The following
data regarding AD and NDMAD was collected from the works cited. AD appears to interfere with mental ability. One
of the tests was the ability to Substitute Digits for Symbols (DSST) and the interference of that process was the
response measured below:
PROBLEM TABLE 11 - 1.

Pharmacological Response

% Pharm Response

Time (hrs)

% Pharm Response

NDMADC Conc (ng/,L)

59.7

33

200

45.3

48

300

29.5

59

400

15.5

68

500

12

74

600

From the above information find the pharmacokinetic parameters:


PROBLEM TABLE 11 - 1.

Five mg IV bolus dose of AD yieled: (intercept of extrapolated metabolite line is 150 ng/mL)

Plasma Data

Urine Data

Time
(hr)

Cp (AD)
(ng/mL)

0.5

47

40

15.4

28

3
4

14.7

5
6

Cpm(NDMAD)
(ng/mL)

Time (mid)
t

AD Ecretion Rate
(mg/hr)

0.5

0.56

0.48

23.5

0.34

27.3

0.18

28

0.09

27
7.5

24.9

20

10

15

12

11

15

7.5

18

4.2

20

2.8

22

1.9

24

1.2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-27

Practice Exams: Section 1 : Adinazolam - Section 1

14.5.2

ADINAZOLAM QUESTIONS

1) K (elimination rate constant of AD) (hr-1 )


2) kr (urinary excretion rate constant of AD)(hr-1 )
3) km (metabolism rate constant of AD)(hr-1 )
4) kmu (urinary excretion rate constant of NDMAD)(hr -1 )
5) Cp0 (IV bolus AD) (ng/mL)
6) Vd (AD)(L.)
7) Vdm (NDMAD)(L.)
8) AUC (Cp vs t for AD by IV bolus) (ng/mL * hr)
9) AUMC (t*Cp vs t for AD by IV bolus (ng/mL * hr2)
10) MRT (AD)(hr)
11) AUC (dXu/dt vs t(mid) for AD by IV bolus) (mg)
12) Xinf

(AD in the body at infinite time)

13) Xuinf

(AD in the urine at infinite time)

14) Xminf

(NDMAD in the body at infinite time)

15) Xmu inf


16) T1/2

(NDMAD in the urine at infinite time)

(AD)(hr)

17) Clearance (AD)(L/hr)


18) dR/dT (%/hr)
19) dR/d(lnC) (%)
20) d(lnC)/dt (NDMAD)(hr-1)
21 Slope of Cp vs T ([AD] vs t) on semi-log paper
22 Slope of dXu/dt vs T(mid) vs t on semi-Log paper
23 Slope of terminal portion of Cpm vs T ([NDMAD] vs t) on semi-log paper
24) Slope of stripped portion of Cpm vs T ([NDMAD] vs t) on semi-log paper
25) Xm

(bar) (Laplace domain equation for NDMAD in the body)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-28

Practice Exams: Section 1 : Adinazolam - Section 1

PROBLEM TABLE 11 - 1. Answer

Pool

Numbers

LaPlace Forms

-1.33

-0.1

10

100

Xo/(s+K)

-1.55

-0.133

0.1

1.33

12.5

120

Xo/(s+km)

-2

-0.15

0.133

1.5

20

167

Xo/(s+kmu)

-3.33

-0.2

0.15

30

200

km * Xo / ((s+km) * (s+kmu))

-4

-0.333

0.2

37.5

375

kmu * Xo / ((s+km) * (s+kmu))

-5

-0.5

0.333

40

425

km * Xo / ((s+km) * (s+K ))

-6.93

-0.693

0.5

50

500

kmu * Xo / ((s+K ) * (s+kmu))

-7.5

-0.73

0.693

55.6

630

km * Xo / ((s+K ) * (s+kmu))

-8.25

-0.85

0.75

6.67

66.7

775

kmu*km*Xo / ((s+K ) * (s+kmu))

-9

-0.95

0.84

7.5

75

930

kr*Xo / ((s+K ) * (s+kmu))

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-29

Practice Exams: Section 1 : Adinazolam - Section 1

14.5.3

ADINAZOLAM SOLUTIONS

ku

Xu

kmu

Xmu

km
Xm

onse %

R vs T
60

Adinazolam metabolite
80

50

70
40

Response (%P-col)

60

30

20

10
2

Time (hr)

50
40
30
10 2

10 3

Concentration (ng.mL)

AD IV Bolus - Metab

Adinazolam IV Bolus

102

10 2

101

Concentration (ng/mL)

Concentration (ng/mL)

10 1

10 0
0

Time (hours)

100
0

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10

15

20

25

Time (hours)

14-30

Practice Exams: Section 1 : Adinazolam - Section 1

K = 0.333hr

k r = 0.133hr

k m = 0.2hr

k mu = 0.2hr

Cp 0 = 55.6ng mL

23

Slope of the terminal portion of

Cp m vsT = 0.2
1

24
0.333
25

6.

Vd = 90L

Vd m = 50L

AUC = 167ng mL hr
AUMC = 500ng mL hr

10

MRT = 3.0hr

11

AUC m = 2.0mg

12

X = 0

13

X u = 2.0mg

14

Xm = 0

15

Xmu = 3.0mg

16

T1 2 = 2hr

17

Cl = 30L hr

18

dR
------- = 7.42 %hr
dT

19

dR = 37.5
----------------%
d ( ln C )

20

d----------------( ln C ) = 0.2hr 1
dt

21
0.333

Slope of

dX u
---------vsT( mid ) = 0.333
dt

22

Slope of the stripped portion = -

km X0
X m = --------------------------------------( s + K ) ( s + k mu )

Slope of Cp vs T([AD] vs t) = -

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-31

Practice Exams: Section 1 : Labetalol - Section 1

14.6 Labetalol - Section 1


Saotome, et. al (J. Clin. Pcol 1993,33:979- 988)

14.6.1

LABETALOL DATA

Labetalol is a selective - blocking and nonselective -blocking agent used in the treatment of hypertension. It is
excreted unchanged into the urine and extensively metabolized. The metabolites are excreted into the urine and bile.
Labetalol reduces diastolic blood presure.
PROBLEM TABLE 11 - 1. P-col

Resp vs Time (hrs) P-col Resp v Labetalol Conc

Pharmacological Response Profiles


Decrease in BP
(mm Hg)

Time
(hr)

Decrease in BP
(mm Hg)

Cp
(ng/mL)

22.2

14.5

100

20.3

16.5

160

18.5

19.5

275

16.6

22.5

525

PROBLEM TABLE 11 - 1.

Data from a 200 mg IV Bolus dose (Intercept of extrapolated Metabolite data = 22.7 mg/hr)

Plasma Data

Urine Data
Time
(mid)

Labetalol
Excretion rate

Metabolite
Excretion Rate

Time
(hrs)

Cp

(mg/hr)

(mg/hr)

0.5

364

0.5

1.82

5.1

331

1.66

8.2

274

1.5

187

128

2.5

88

9.8
1.37

10.6
10.7
10.5

0.94

9.5

0.64

7.0

4.9

12

2.3

18

0.74

24

0.24

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-32

Practice Exams: Section 1 : Labetalol - Section 1

14.6.2

LABETALOL QUESTIONS
From the above information find the pharmacokinetic parameters:
1) K (elimination rate constant of labetalol) (hr-1 )
2) K2 (elimination rate constant of labetalol metabolites (hr-1 )
3) kr (urinary excretion rate constant of
labetalol)(hr-1 )
4) km (metabolism rate constant of labetalol)(hr-1 )
5) kmu (urinary excretion rate constant of
Metabolites of labetalol)(hr-1 )
6) kmb (biliary excretion rate constant of
Metabolites of labetalol)(hr-1 )
7) Cp0 (IV bolus labetalol) (ng/mL)
8) Vd (labetalol)(L.)

21) Clearance (labetalol)(L/hr)


22) dR/dT (%/hr)
23) dR/d(lnC) (%)
24) d(lnC)/dt (labetalol)(hr-1)
25 Slope of Cp vs T ([labetalol] vs t) on
semi-log paper
26 Slope of dXu/dt vs T(mid) vs t on
semi-Log paper
27 Slope of terminal portion of Cpm vs T
([Metabolites of labetalol] vs t) on
semi-log paper
28) Slope of stripped portion of Cpm vs T
([Metabolites of labetalol] vs t) on
semi-log paper
29) Xm

(bar) (Laplace domain equation for Metabolites


of labetalol in the body)

9) AUC (Cp vs t for labetalol by IV bolus)


(ng/mL * hr)

30) dXmu / dT (equation for the rate of


excretion of metabolites into the urine)

10) AUMC (t*Cp vs t for labetalol by IV


bolus (ng/mL * hr2)

31) Fraction
unchanged.

11) MRT (labetalol)(hr)

32) Fraction of labetalol eliminated as


metabolites in the urine.

12) AUC (dXu/dt vs t(mid) for labetalol by


IV bolus) (mg)
13) AUC (dXmu/dt vs t(mid) for Metabolites (mg))

of

labetalol

excreted

33) Fraction of labetalol eliminated as


metabolites in the bile.

14) AUC (dXmb/dt vs t(mid) for Metabolites (mg))


15) Xinf

(labetalol in the body at infinite time (mg))

16) Xuinf

(labetalol in the urine at infinite time (mg))

17) Xminf

(Metabolites of labetalol in the body at infinite time (mg))

18) X muinf (Metabolites of labetalol in the urine at infinite time (mg))


19) X mbinf

(Metabolites of labetalol in the bile at infi-

nite time (mg))

20) T1/2

(labetalol)(hr)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-33

Practice Exams: Section 1 : Labetalol - Section 1

14.6.3

LABETALOL SOLUTIONS

ku

Xu

kmu

Xmu

km
Xmf

kmf

Xm

Labatalol
24
22
20

Response (mmHg)

Response (mmHg)

Labatalol
23
22
21
20
19
18
17
16
8

Time (hr)

18
16
14
10 2

103

Concentration (ng.mL)

Labatalol Metabolite

Labatalol

Labatalol

102

103

10 1

101

10 0

102

Time (hours)

10-1
0

10

15

20

Time (hours)

25

Excretion rate (mg/hr)

101

Excretion Rate (mg/hr)

Concentration (ng/mL)

100

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10 -1
0

Tmid (hours)

14-34

Practice Exams: Section 1 : Labetalol - Section 1

K1 = 0.190hr

2.

K2 = 0.792hr
k r = 0.01hr

k mu = 0.38hr

k mb = 0.41hr

Cp 0 = 400ng mL

24

1
d ( ln C )
----------------- = 0.190hr
dt

25

Slope of Cp vs T = 0.190hr

26

Slope of

dX u
--------- vsT( mid ) vs ( t ) = 0.190
dt

27
Slope of terminal portion
of Cp m vsT = 0.190
28

Vd = 500L
AUC = 2105ng mL hr

dR
----------------- = 4.90 %
d ( ln C )

23

k m = 0.18hr

Slope of stripped portion of

Cp m vsT = 0.79
km X 0
X m = ----------------------------------------( s + K1 ) ( s + K2 )

29

10

AUMC = 11072ng mL hr
11

MRT = 5.26hr

12

AUC = 10.5mg

13

AUC = 91.2mg

14

AUC = 98.3mg

15

X = 0

16

X u = 10.5mg

17

Xm = 0

18

Xmu = 91.2mg

19

Xmb = 98.3mg

20

T1 2 = 3.64hr

21

Cl = 95L

22

dR
------- = 0.93 %hr
dT

30

X mu ( k mu k m X 0 ) K2t K1T
= ---------------------------------- ( e
)
--------e
dt
( K1 K2 )
31

Fraction of labetalol excreted

10.5
200

unchanged: ---------- 100 = 5.25 %


32
Fraction of labetalol excreted as
metabolites in urine:

91.2
---------- 100 = 45.6 %
200

33
Fraction of labetalol eliminated as
metabolites in bile:

98.3
---------- 100 = 49.15 %
200

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-35

Practice Exams: Section 1 : Zidovudine Section 1

14.7 Zidovudine Section 1


14.7.1

ZIDOVUDINE DATA

Zidovudine (ZDV - formerly azidithymidine (AZT)) is used to treat HIV positive patients with AIDS and AIDS related
complex (ARC). The in vitro virustatic concentration is 250 mic/mL. While the pharmacokinetics of ZDV is complex,
we can approximate it with the following model. ZDV is excreted unchanged into the urine to UZDV. It is metabolized by hepatic glucuronidation to an inactive metabolite, GZDV, and also metabolized by intracellular phosphorylation to the active metabolite, ZDV-TP. Both metabolites are excreted into the urine (UGZDV and UZDV-TP). The
anti-retroviral activity was shown in the following data:
PROBLEM TABLE 11 - 1. ZDV-TP

Response vs time and response vs concentration data

Pharmacological Response Data


Concentration
ZDV-TP (ng/mL)

Activity
%

Time
(min)

Activity
%

100

80

100

90

76.8

30

100

75

68.5

60

100

50

52.3

120

92

25

24.5

240

82

360

72

480

62

PROBLEM TABLE 11 - 1. 100

mg dose ZDV given by IV Bolus to 70 KG patient

Plasma Data
Time
(hours)

Cp ZDV
(ng/mL)

Urine Data

Cpm GZDV
(ng/mL)

Interval
(hrs)

UZDV Collected
(mg)

0.25

40

0-1

9.2

0.5

75

1-2

4.8

100

2-3

2.5

125

4-6

0.75
1

315

1.5

Rate of
excretion data

AUC
UGZDV (mg)
60

150

170

160

91

160

48

145

13

100

65

10

40

12

25

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-36

Practice Exams: Section 1 : Zidovudine Section 1

Please use the following nomenclature:


D

= Dose of ZDV

= elimination rate constant of ZDV

kr = excretion rate constant of ZDV


kmu1 = excretion rate constant of ZDV-TP
kmu2 = excretion rate constant of GZDV
km1 = metabolism rate constant of ZDV to ZDV-TP
km2 = metabolism rate constant of ZDV to GZDV

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-37

Practice Exams: Section 1 : Zidovudine Section 1

14.7.2

ZIDOVUDINE QUESTIONS
1) Using Laplace transforms, find the
equation for the rate of excretion of GZDV
into the urine. (For this question use the
following answer set:
a=

I only

b=

III only

15) What is the terminal slope (*-1) of


ln(dUZDV/dt) vs t (hr^-1)?
16) What is the intercept of ZDV vs t (ng/
mL)?

c = I + II only

17) What is the intercept of dUZDV/dt vs t


(mg/hr)?

d = II + III only

18) What is the intercept of the extrapolated line from plasma GZDV vs time data
(ng/mL)?

e = All three

14) What is the terminal slope (*-1) of


ln(GZDV) vs t (hr^-1)?

(kmu2*km2*D/(K-kmu2))*(exp(-K*t))

II (kmu2*km2*D/(K-kmu2))*(exp(kmu2*t))

19) What is the intercept of the extrapolated line from dUGZDV/dt vs time data
(mg/hr)?

III -(kmu2*km2*D/(K-kmu2))*(exp(K*t))

20) What is the Cp0 of ZDV (ng/mL)?

2) Using linear regression on the pharmacological response / concentration profile,


what was the Summation of XY?

22) What is the volume of distribution of


GZDV (L)?

3) What is d%R/d(lnC)(%)?

24) What is the half life of ZDV (hr)?

4) What is d%R/dt (%/hr)?

25) What is the MRT of ZDV (hr^-1)?

5) What is K (hr^-1)?

26) What is the AUC of the first trapazoid


of ZDV vs time (ng/mL*hr)?

6) What is kr (hr^-1)?

21) What is the volume of distribution of


ZDV (L)?

23) What is the clearance of ZDV (L/hr)?

27) What is the AUC(from 0 to infinity) of


the IV bolus ZDV plasma data (ng/
mL*hr)?

7) What is km1 (hr^-1)?


8) What is km2 (hr^-1)?
9) What is kmu1 (hr^-1)?
10) What is kmu2 (hr^-1)?

28) How much ZDV is in the body at infinite time (mg)?

11) What is the terminal slope (*-1) of


ln(ZDV) vs t (hr^-1)?

29) How much GZDV is in the body at


infinite time (mg)?

12) What is the terminal slope (*-1) of


ln(ZDV-TP) vs t (hr^-1)?

30) How much ZDV-TP is in the body at


infinite time (mg)?

13) What is the terminal slope (*-1) of


ln(ZDV-TP) vs t (hr^-1)?

31) How much ZDV is in the urine


(UZDV) at infinite time (mg)?
32) How much GZDV is in the urine
(UGZDV) at infinite time (mg)?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-38

Practice Exams: Section 1 : Zidovudine Section 1

33) How much ZDV-TP is in the urine


(UZDV-TP) at infinite time (mg)?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-39

Practice Exams: Section 1 : Zidovudine Section 1

14.7.3

ZIDOVULDINE SOLUTIONS

Xm1

km1

ku

Xu

kmu2

Xmu2

km2

kmu1
Xmu1

Xm2

Zidovudine

Zidovudine

110
100
90

Response (% Activity)

Response (% Activity)

80
70
60
0

100

200

300

400

500

Time (min)

Zidovudine

10 1

102

Concentration (ng/mL)

Zidovudine Urine data

Zidovudine Metobolite
103

102

102

102

101

101

101

100
0

10 12

Time (hours)

UZVD (mg)

Time (hours)

Concentration (ng/mL)

103

0 1 2 3 4 5 6

Concentration (ng/mL)

90
80
70
60
50
40
30
20

10-1
0

Tmid (hours)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-40

Practice Exams: Section 1 : Zidovudine Section 1

1.

Answer is D - II and III

1289.2

40%

-0.08333%hr
1

K = 0.630hr

k r = 0.125hr

k m1 = 0.25hr

457.5ng mL hr

27

952ng mL hr

28

29

30

31

20mg

32

60mg

33

20mg

k m2 = 0.25hr

k mu1 = 0.125hr

10

k mu2 = 0.378hr

11

0.630hr

12

0.250hr

13

Skip - same question as #12

14

0.25hr

15

0.630hr

1
1

1
1

16

600ng mL

17

12.6mg hr

18

500ng mL

19

12.6mg hr

20

600ng mL

21

166.7L

22

200L

23

26

105L hr

24

1.1hr

25

1.6hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-41

Practice Exams: Section 1 : Fosinopril Section 1

14.8 Fosinopril Section 1


Kostis et al Fosinopril: Pharmacokinetics and pharmacodynamics in congestive heart failure Clin Pcol Ther 58(6) 660-5
(1995); Hui et al Pharmacokinetics of fosinopril in patients with various degrees of renal function Clin Pcol Ther 49(4) 457 -66
(1991)

14.8.1

FOSINOPRIL DATA

Fosinopril Sodium is a phosphinic prodrug of the angiotensin converting enzyme (ACE) inhibitor fosinoprilat effective
in the treatment of hypertension and chronic heart failure (CHF). The parent compound is weakly active, if at all. Its
sole metabolite, fosinoprilat, exhibits almost all of the pharmacological activity. After administration, fosinopril is
entirely converted to the active fosinoprolat by esterases in the liver. Unlike other ACE inhibitors, elimination of fosinoprilat is divided equally between renal and hepatic pathways (liver metabolism). The following information was
obtained from a 70 Kg male.
Wherever necessary, please use the following symbols:
Cp = plasma concentration of parent drug, fosinopril
Cpm1 = plasma concentration of metabolite, fosinoprilat.
Cpm2= plasma concentration of metabolite of fosinoplilat.
X = amount of parent compound, fosinopril, in the body
Xm1 = amount of metabolite, fosinoprilat, in the body
Xm2 = amount of metabolite of fosinoprilat in the body
Xu = cumulative amount of parent drug in urine.
Xmu1 = cumulative amount of metabolite, fosinoprilat, in urine.
Xmu2 = cumulative amount of metabolite of fosinoprilat in urine.
ku = renal excretion rate constant of parent drug in plasma.
kmu1 = renal excretion rate constant for metabolite, fosinopril, in plasma.
kmu2 = renal excretion rate constant for metabolite of fosinopril in plasma.
kf = fecal excretion rate constant of parent drug in plasma.
kmf1 = fecal excretion rate constant for metabolite, fosinopril, in plasma.
kmf2 = fecal excretion rate constant for metabolite of fosinopril in plasma.
km1 = metabolism rate constant of fosinopril in plasma.
km2 = metabolism rate constant of fosinoprilat in plasma.
K1 = elimination rate constant of parent drug in plasma = summation of all processes which remove fosinopril.
K2 = elimination rate constant of metabolite1 in plasma = summation of all processes which remove fosinoprilat.
K3 = elimination rate constant of metabolite2 in plasma = summation of all processes which remove the metabolite of fosinoprilat.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-42

Practice Exams: Section 1 : Fosinopril Section 1

PROBLEM TABLE 11 - 1. Pharmacological

Response data (Six hours after dosing with fosinopril)

Time
(hr)

Fall in SSBP
(mm Hg)

Fall in SSBP
(mg Hg)

Dose
(mg)

22

10

21

11

20

19

16

40

11

17

22

80

13

16

PROBLEM TABLE 11 - 1.

Plasma Data
(from7.5 mg Fosinoprilat IV Bolus)

Urine Data
(from 20 mg Fosinopril IV bolus)
(Intercept 0.855 mg/hr)

Tme
(hours)

Cp
(Fosinoprilat)
(ng/mL)

Interval
(hr)

Amount (Fosinoprilat)
(mg)

387

0-1

0.158

360

1-2

0.362

335

2-3

0.467

311

3-4

0.513

269

4-6

1.05

6 - 10

1.84

10 - 14

1.42

14 - 22

1.84

22 - 26

0.592

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-43

Practice Exams: Section 1 : Fosinopril Section 1

14.8.2

FOSINOPRIL QUESTIONS
From the parent compound, Fosinopril,
given by IV bolus, find the equation for:
1) X, the mass of parent drug, Fosinopril,
in the body
) Xu, the mass of parent drug, Fosinopril,
in urine.

18) K1, the elimination rate constant of parent drug in


plasma , the summation of all processes which remove
Fosinopril.

19) K2, the

elimination rate constant of metabolite in


plasma , the summation of all processes which remove
Fosinoprilat.

3) dXu/dt, the rate of excretion of parent


drug, Fosinopril, in urine.

For fosinoprilat IV, find the value of :

4) Xf, the mass of parent drug, Fosinopril, in feces.

21) AUMC(fosinoprilat)

20) AUC(fosinoprilat)

5) Xm1, the mass of metabolite, Fosinoprilat, in the body

22) MRT(fosinoprilat)

6) Xm1u, the mass of metabolite, Fosinoprilat, in urine.

24) T 1 2

7) dXm1u/dt, the rate of excretion of


metabolite, Fosinoprilat, in urine.

25) Cp0(fosinoprilat) for iv dose (ng/mL)

8) Xm1f, the mass of metabolite, Fosinoprilat, in feces.

27) Cp of Fosinoprilat at eight hours


after the IV dose

9) Cp, the plasma concentration of parent


drug, Fosinopril
10) Cpm1, the plasma concentration of
metabolite, Fosinoprilat.

23) Ke for fosinoprilat (hr-1)


for Fosinoprilat (hr)

26) Vd for Fosinoprilat (L)

For fosinoprilat given as IV fosinopril,


dXm1/dt, find the value of :
28) AUC(fosinoprilat)(mg)
29) AUMC(fosinoprilat)

Find the value of:

30) MRT(fosinoprilat)

11) dR/dT

31) K2 for fosinoprilat (hr-1)

12) dR/d(ln(Cpm1))

32) T ?

13) d(ln(Cpm1))/dT
14) ku, the renal excretion rate constant of parent drug,

for Fosinoprilat (hr)

33) K1 for fosinopril (hr-1)

Fosinopril, in plasma.

15) km1, the metabolism rate constant of parent drug,


Fosinopril, in plasma.

16) kmu1, the renal excretion rate constant for metabolite, Fosinoprilat, in plasma.

17) km2, the metabolism rate constant for the metabolite of Fosinoprilat in plasma.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-44

Practice Exams: Section 1 : Fosinopril Section 1

PROBLEM TABLE 11 - 1. Answer

Pool

Minus

Small

Med.

Large

Equations

More equations

-0.037

1.35

155

None

((km1 X0/Vdm )/(K1-K2))(e-K2t-e-K1t )

-0.050

0.037

5.0

232

X0e-K1t

((km1 X0/Vdm )/(K1-K2))(e-K1t-e-K2t )

-0.074

0.06

7.4

420

(X0/Vd)e-K1t

((kmu1 X0/Vdm )/(K1-K2))(e-K2t-e-K1t)

-0.082

0.074

8.2

840

ku X0e -K1t

((kmu1km1 X0)/(K1-K2))(e-K2t-e-K1t )

-0.10

0.135

9.4

940

kf X0e-K1t

((kmu1km1 X0)/(K1-K2))(e-K1t-e-K2t )

-0.135

0.5

10

5676

(k u/K1) X0(1-e-K1t)

((kmu1 X0)/(K1-K2))(e-K2t-e-K1t )

-0.37

0.6

12

13500

(k f/K1) X0(1-e-K1t)

((kmf1km1 X0)/(K1-K2))((1-e-K2t)/K2-(1-e-K1t )/K1)

-0.50

0.74

13.5

15700

((km1 X0)/(K1-K2))(e-K2t-e-K1t)

((kmf1km1 X0)/(K1-K2))((1-e-K1t)/K1-(1-e-K2t )/K2)

-0.6

0.82

15.5

18000

((km1 X0)/(K1-K2))(e-K1t-e-K2t)

((kmu1km1 X0)/(K1-K2))((1-e-K2t )/K2-(1-e-K1t)/K1)

-0.74

0.94

18

76700

((kmu1 X0)/(K1-K2))(e-K2t-e-K1t)

((kmf2km1 X0)/(K1-K2))((1-e-K2t)/K2-(1-e-K1t )/K1)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-45

Practice Exams: Section 1 : Fosinopril Section 1

14.8.3

FOSINOPRIL SOLUTIONS
ku=0

K1= km1
K2= kmu1+km2

km1

K3= kmu2
kmu1
Xm1

Xmu1

km2
kmu2
Xm2

Xmu2

Response

R vs Ln(c)
25

20

Response

15

10

R vs T
23

22

21

20

19

18

17

16

0
10

10

15
0

Concentration

10

12

14

Time (hr)

Fosinoprilat in Urine

Excretion Rate (mg/hr)

Fosinoprilat

Concentration ng/mL

10

10

-1

10

102
0

Time (hr)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

10 15
Time (hr)

20

25

14-46

Practice Exams: Section 1

1) X, the mass of parent drug, Fosinopril, in the body

B - X0e-K1t

) Xu, the mass of parent drug, Fosinopril, in urine.

A - none(no drug goes there)

3) dXu/dt, the rate of excretion of parent drug, Fosinopril, in urine

A = none(no drug goes there)

4) Xf, the mass of parent drug, Fosinopril, in feces.

A = none(no drug goes there)

5) Xm1, the mass of metabolite, Fosinoprilat, in the body

H = (km1X0)/(K1-K2)(e-K2t-e-K1t)

6) Xm1u, the mass of metabolite, Fosinoprilat, in urine.

( k mu1 k m1 X 0 ) 1 e K2t 1 e K1t


-------------------------------- --------------------- ---------------------
K1
( K1 K2 ) K2
(k

X )

7) dXm1u/dt, the rate of excretion of metabolite, Fosinoprilat, in urine.

K2t
K1t
mu1 m1 0
-{e
e
}
D = -------------------------------

8) Xm1f, the mass of metabolite, Fosinoprilat, in feces.

A = none(no drug goes there)

9) Cp, the plasma concentration of parent drug, Fosinopril

X
C = -----0- e K1t
Vd

10) Cpm1, the plasma concentration of metabolite, Fosinoprilat.

K2t
K1t
m1 0
-{e
e
}
A = -------------------------------

( K1 K2 )

( K1 K2 )V d

Find the value of:


11) dR/dT

I = -0.6 mm Hg/hr

12) dR/d(ln(Cpm1))

D = 8.2 mm Hg

13) d(ln(Cpm1))/dT

C = -0.074 hr-1

14) ku, the renal excretion rate constant of parent drug, Fosinopril, in plasma.

A = none(no drug goes there)

15) km1, the metabolism rate constant of parent drug, Fosinopril, in plasma.

F = 0.5 hr -1

16) kmu1, the renal excretion rate constant for metabolite, Fosinoprilat, in plasma.

B = 0.037 hr -1

17) km2, the metabolism rate constant for the metabolite of

B = 0.037 hr -1

Fosinoprilat in plasma.

18) K1, the elimination rate constant of parent drug in plasma , the summation of all processes
19) K2, the

which remove Fosinopril.

F = 0.5 hr -1

elimination rate constant of metabolite in plasma , the summation of all processes which remove Fosinoprilat. D

= 0.074 hr -1

For fosinoprilat IV, find the value of :


20) AUC(fosinoprilat)

F = 5676 ng/mL

21) AUMC(fosinoprilat)

J = 76700 mg/mL*hr2

22) MRT(fosinoprilat)

H = 13.5 hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-47

Practice Exams: Section 1

23) Ke for fosinoprilat (hr-1)

D = 0.074 hr -1

24) T 1 2

E = 9.4 hr

for Fosinoprilat (hr)

25) Cp0(fosinoprilat) for iv dose (ng/mL)

C = 420 ng/mL

26) Vd for Fosinoprilat (L)

J = 18 L

27) Cp of Fosinoprilat at eight hours after the IV dose

B = 232 ng/mL

For fosinoprilat given as IV fosinopril, dXm1/dt, find the value of :


28) AUC(fosinoprilat)(mg)

F = 10 mg

29) AUMC(fosinoprilat)

A = 155 mg*hr

30) MRT(fosinoprilat)

I = 15.5 hr

31) K2 for fosinoprilat (hr-1)

D = 0.074 hr -1

32) T ?

E = 9.4 hr

for Fosinoprilat (hr)

33) K1 for fosinopril (hr-1)

F = 0.5 hr -1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-48

Practice Exams: Section 1

14.9 Omeprazole
14.9.1

OMEPRAZOLE DATA

Omeprazole (mw: 345.42) is a gastric proton-pump inhibitor which decreases gastric acid secretion. It is effective in
the treatment of ulcers and esophageal reflux. In normal patients, 20% of the omeprazole is excreted into the feces and
80% of the omeprazole dose is excreted as an inactive metabolite into the urine. In the study by Anderson, et. al., eight
patients were given 20 mg, IV bolus doses of omeprazole. The patients had a mean body weight of 70 kg. Blood and
urine were collected at various intervals throughout the study and the following data was obtained:
TABLE 14-2 Pharmacological

Response

% Acid Secretion
Inhibition

Time (min)

% Acid Secretion
Inhibition

Concentration
(ng/mL)

100

120

74

100

240

62

.6

75

330

56

.5

63

360

47

.35

56

375

27

.15

50

390

25

450

10

570

600

720

TABLE 14-3 Plasma

profile of parent and urine profile of metabolite


Intercept = 4 mg/hr
AUMCmet = 96 mg*hr

Parent Compound

Time (hr)

Cp (ng/mL)

Urine Collection
Interval (hr)

Metabolite
collected (mg)

0.5

336

0-1

1.19

205

1-2

2.07

75

2-3

2.10

28

3-5

3.44

10

5-7

2.38

7-9

1.62

9 - 11

1.08

11 - 13

0.72

13 - 15

0.48

16 - 20

0.44

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-49

Practice Exams: Section 1

14.9.2

OMEPRAZOLE QUESTIONS
Using LaPlace transforms, from Omeprazole, given by IV bolus, find the equation
for:
1.

1) X, the mass of Omeprazole in the body

2.

) Xu, the mass of Omeprazole in urine.

3.

) dXu/dt, the rate of excretion of Omeprazole in urine.

4.

) Xf, the mass of Omeprazole in feces.

5.

) Xm, the mass of metabolite of Omeprazole in the body

6.

) Xmu, the mass of metabolite of Omeprazole in urine.

7.

) dXmu/dt, the rate of excretion of metabolite of Omeprazole in urine.

8.

) Xmf, the mass of metabolite of Omeprazole in feces.

9.

) Cp, the plasma concentration of parent drug, Omeprazole

10.

) Cpm, the plasma concentration of metabolite of Omeprazole.

Find the value of:


) dR/dT = slope of the Pharmacological response vs. time profile (% / hr)
12. ) dR/d(lnC) = slope of the Pharmacological response vs concentration profile (%)
11.

13.

) d(lnC)/dt = slope of the concentration vs time profile (hr-1 )

14.

) kr = urinary excretion rate constant Omeprazole (hr-1 )

15.

) kf = fecal excretion rate constant Omeprazole (hr-1 )

16.

) km = rate of metabolism of Omeprazole in the body (hr-1 )

17.

) K1 = elimination rate constant of Omeprazole in the body; the summation of all of the ways that it
is removed from the body. (hr-1 )

18.

) T1/2 of Omeprazole in the body (hr)

19.

) X0 = Mass of Omeprazole in the body at time 0 (mg)

20.

) Xinf = Mass of Omeprazole in the body at time infinite time (mg)

21.

) Xuinf = Mass of unchanged Omeprazole in the urine at infinite time (mg)

22.

) Xfinf = Mass of unchanged Omeprazole in the feces at infinite time (mg)

) V = Volume of distribution of Omeprazole (L)


24. ) Cp0 = Concentration of Omeprazole in the body at time 0. (mic/L)
23.

25.

) AUCiv = Area under the plasma concentration of Omeprazole vs time curve of the IV
dose (mic/L*hr)

) First trapazoid of the AUCiv (mic/L*hr)


27. ) Last trapazoid of the AUCiv (mic/L*hr)
26.

28.

) AUMCiv = Area under the first moment of the plasma concentration of Omeprazole vs
time curve of IV dose (mic/L*hr2 )

29.

) MRTiv = Mean Residence time of Omeprazole given as the IV dose (hr)

30.

) Xm = Mass of metabolite in the body at time = 0 (mg)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-50

Practice Exams: Section 1

31.

) Xminf = Mass of the metabolite in the body at infinite time. (mg)

32.

) Vm = Volume of distribution of the metabolite in the body (L)

33.

) Cpm = Concentration of metabolite in the body at time = 0 (mic/L)

34.

) AUCmet = Area Under the Curve of the metabolite excretion rate vs time profile (mg)

35.

) Xmu0 = Mass of metabolite in urine at time = 0 (mg)

36.

) Xmuinf = Mass of metabolite in urine at time = infinity (mg)

37.

) Xmf0 = Mass of metabolite in feces at time = 0 (mg)

38.

) Xmfinf = Mass of metabolite in feces at time = infinity (mg)

39.

) Kmu = Rate constant of excretion of metabolite into urine (hr-1 )

40.

) Kmf = Rate constant of excretion of metabolite into feces (hr-1 )


TABLE 14-4 Omeprazole

Minus

Small Med

Large

-0.8

100

-1

0.1

147

-2

0.2

210

-4

0.3

223

-8

0.4

10

372

-10

0.5

16

444

Answer Pool

Equations
None

X0 e

km X0
K 2 t
K1 t
--------------------------------- (e
e
)
( K 1 K 2 )V dm

K1 t

km X0
K 1 t
K2 t
--------------------------------- (e
e
)
( K 1 K 2 )V dm
k mu X 0
K 2 t
K1 t
--------------------------------- (e
e
)
( K 1 K 2 )V dm

X 0 K 1 t
------ e
Vd
K 1 t

k mu k m X 0 K 2 t
K1 t
----------------------(e
e
)
( K1 K 2 )

K1 t

k mu k m X 0 K 1 t
K2 t
----------------------(e
e
)
( K1 K 2 )

ku X0 e

kf X0 e

ku
K1 t
-----X (1 e
)
K 0
1

-16

0.6

20

556

More Equations

k mu X 0
K 2 t
K1 t
----------------------(e
e
)
( K1 K 2 )

kf
K1 t
-----)
K X0 ( 1 e

k mf k m X 0 1 e K2 t 1 e K 1 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 2 K 1

-20

0.7

25

643

k m X0
K 2 t
K1 t
----------------------(e
e
)
( K1 K 2 )

k mf k m X 0 1 e K1 t 1 e K 2 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 1 K 2

-25

0.8

36

756

k m X0
K 1 t
K2 t
----------------------(e
e
)
( K1 K 2 )

k mu k m X 0 1 e K2 t 1 e K 1 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 2 K 1

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-51

Practice Exams: Section 1

Minus

Small Med

Large

-36

0.9

893

Missing Information. Can not answer question with information available

44

Equations
k mu X 0
K2 t
K1 t
----------------------(e
e
)
( K1 K 2 )

More Equations
k mu k m X 0 1 e K1 t 1 e K 2 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 1 K 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-52

Practice Exams: Section 1

14.9.3

OMEPRAZOLE SOLUTIONS

kf
Xf

X
km
Xm

kmu

80

80

70

70

60

-1

60

Response

Response

Xmu

50

40

50

40

30

30

20

20
320

340

360

380

400

420

440

460

Concentration

10

Time (min)

10

10

Concentration

Concentration

10

10

10

10
0

10

10

12

Time (hr)

10

15

20

Time (hr)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-53

Practice Exams: Section 1

K1 t

1. (B) X = X0 e
2. (A) X u = 0 See model.
3. (A)

dX u
--------- = 0
dt

4. (G)

k
K 1 t
Xf = ------f X 0 ( 1 e
)
K
1

5. (H)

km X0
K 2 t
K1 t
X m = ----------------------(e
e
)
( K1 K 2 )

See model.

k mu k m X 0 1 e K2 t 1 e K1 t
X mu = ---------------------- --------------------- ---------------------
( K 1 K2 ) K2 K1

6. (I)

7. (D)

k mu k m X 0 K2 t K 1 t
dX mu
------------- = ----------------------(e
e
)
dt
( K 1 K2 )

8. (A)

X mf = 0

9. (C)

X
K 1 t
C p = ------0- e
Vd

10.(A)

km X 0
K2 t
K 1 t
C pm = ---------------------------------(e
e
)
( K 1 K 2 )V dm

11.(I)

See model.

dR 0.4% 25%
------- ------------------ -------------dt minute
hr

Estimate from graph. Between 20-80% of maximal

response ONLY!
12.(H)

dR
74%-57%
17%
--------------- = ---------------------------------------------- = ------------- 25%
d ln C p
ng
ng
0.693
ln 1.0 ------ ln 0.5 -----ml
ml

Points taken from sample graph.

13.(B)

d ln C p
1
dR d ln C
25%
1
--------------- = ------- ---------------p = -------------- ----------- = 1 hr
dt
dt
dR
hr
25%

14.(A)

kr = 0

15.(C)

1
Xf K 1
1
4mg 1hr
- = ----------------------------- = 0.2hr
k f = ------------X0
20mg

See model.

0.2hr-1
16.(I) k m =

K 1 k f = 1hr

0.2hr

Also,

= 0.8hr

kf

is 20% of

if you have

K1 ,

or 20% of 1hr-1 =

k f .or

1
X mu K 1
1hr = 0.8hr 1
k m = ---------------= 16mg
-------------------------------X0
20mg

. Also,

km

is 80% of

K1 ,

or 80% of 1hr-1 =

0.8hr-1
17.(A)

d ln C
1
K 1 = ------------------p = 1hr
dt

18.(H)

t 1 2 = 0.693
------------- = 0.693 0.7hr
K1

19.(G) X 0 = 20mg Read introduction.


20.(A) X inf = 0 See model.
21.(A) Xu ( inf ) = 0
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-54

Practice Exams: Section 1

20
X f ( inf ) = --------- 20mg = 4mg
100

22.(C)
23.(I)

20% of parent excreted in feces (read introdction).

Dose- ----------------20mg 20
, 000g- 36L
V d = ----------------------------------C p0
ng
g
556 -------556 -----mL
L

24.(G)

ng- 556 g
-----C p0 556 ------mL
L

25.(G)

AUC IV =

From graph.

C p ( last )
p ( n ) + C p ( n + 1 )
C
--------------------------------------t + -----------------

K1
2

or

C p0
AUC IV = -------K1

556 + 336
336 + 205
205 + 75
75 + 28
AUC IV = ------------------------ 0.5 + ------------------------ 0.5 + --------------------- 1 + ------------------ 1 +
2
2
2
2

or

10
g hr
+ 10
28
------------------ 1 + ------ = 578.75 556 ---------------- 2
1
L
ng
556 ------- hr
mL = 556
AUC IV = ----------------------------L
1hr

26.(D)

+ 336- 0.5 = 223 g


hr 556
-------------------------------------

2
L

27.(E)

10 g hr
------ ----------------1
L

28.(G)

AUMC IV =

tn Cp (n ) + tn + 1 Cp (n + 1 )
C p ( last ) tlast C p ( last )
------------------------------------------------------- t + -----------------+ ---------------------------

2
2
K1
( K1 )

( 556 ) + 0.5 ( 336 -) 0.5 + 0.5


( 336 ) + 1 ( 205 ) 0.5 + 1------------------------------------( 205 ) + 2 ( 75 ) 1 +
AUMC IV = 0---------------------------------------------------------------------------------------

2
2
2
2

g hr
( 75 ) + 3 ( 28 )
3 ( 28 ) + 4 ( 10 )
10 4 ( 10 )
2--------------------------------- 1 + ---------------------------------- 1 + ------ + -------------- = 541.75 556 ------------------

2
1
L
2
2
1
2

hr
556 g
------------------AUMC
L
= ------------------ ----------------------------- 1hr
hrAUC
556 g
---------------L

29.(A)

MRT IV

30.(A)
31.(A)
32.(A)
33.(A)

Xm 0 = 0

34.(F)

AUC met =

or

1 = ------------1 - = 1hr
MRT IV = -----1
K1
1hr

See model.
X m ( inf ) = 0 See model.
V dm =
Can not determine from information given. Need
C pm0 = 0 See model.

Xmu

80
= --------- 20mg = 16mg .
100

C pm

information.

AUC is equal to all drug which goes

through compartment, which is 80% of the 20 mg dose, or 16 mg (read introduction).


35.(A) X mu0 = 0 See model.
36.(F)

80- 20mg = 16mg


X mu ( inf ) = -------100

80% of the 20 mg dose is found as metabolite in

urine (Read introduction).


37.(A) X mf0 = 0 See model.
38.(A) X mf ( inf) = 0 See model.
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-55

Practice Exams: Section 1

39.(C) k mu = K 2 = 0.2hr 1 = is
which is K1 .
40.(A) K m f = 0 See model.

k small

from urine data graph because

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

k l arg e 1hr

14-56

Practice Exams: Section 1

14.10 EXP3312, an Experimental Drug


Wong N, Wong P. (J Pharm Pharmacol 1996; 48: 492-497) investigated the pharmacological and pharmacokinetic response of EXP3312

14.10.1 EXP3312 DATA


EXP3312 is a non-peptide angiotension II AT1-receptor antagonist which has
potential as an oral hypertensive drug. EXP3312 a pro-drug is metabolized to an
active metabolite M1. Experiments with rats show M1 is the active drug. As with
all angiotension II antagonists EXP3312 blocks the renin-angiotension system.
EXP3312 is a synthesized analog of Losartin, but EXP3312 has greater oral
hypotensive potency and has promise in future treatment of human renal hypertension. EXP3312 is still an experimental drug without all pharmacological responses
having been explored. However, we may assume the drug is completely metabolized to the active metabolite (M1). Thereafter, the active metabolite (M1) is
excreted unchanged in the urine.
The following pharmalogical data was obtained using pithed (spinal tap) rats with
an average weight of 350 g. The concentration of drug in each rat is based on average weights of the rats. The measured responses of decrease in mean arterial pressure (MAP decrease) are shown below.The researchers supplied graphical data of
mean arterial pressure in conscious renal hypertensive rats. M1 metabolite was
measured and the results are also shown in the table below.
TABLE 14-5

Response (mm Hg)

M1 administration (mg/kg)

Response (mm Hg)

(MAP) decrease

I.V. bolus

(MAP) decrease

25

0.01

60

36.5

0.03

57.4

48.5

0.1

54.7

59

0.3

49.3

44.5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

Time (hr)

14-57

Practice Exams: Section 1

The parent compound EXP3312 and metabolite M1 levels were measured in the
plasma over time. The data obtained from the active M1 metabolite and the inactive partent drug EXP3312 are shown below. The standard dose for each rat was
0.3mg/kg.
TABLE 14-6

time (hr)

EXP3312

M1

Cp (mic/mL)

Cp (mic/mL)

0.15

0.32

0.3

0.59

0.5

0.4

0.26

1.34

0.11

1.61

0.045

1.26

0.81

10

0.3

12

0.18
I = 3.81 mic/mL

Use the following data was obtained when an intravenous infusion of a different
dose was given to an average rat over 45 minutes.

TABLE 14-7

time (hr)

Plasma concentration
(mic/mL)

0.25

0.139

0.5

0.26

0.75

0.35

0.29

0.12

0.05

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-58

Practice Exams: Section 1

14.10.2 EXP3312 & M1 QUESTIONS:


Using the information given find the pharmacokinetic parameters (CHECK
YOUR UNITS!!!!!!):
1.

dR/dt of M1 active metabolite (mmHg/hr)

2.

dR/d(lnCp) active metabolite (mmHg)

3.

d(lnCp)/dt of M1active metabolite (hr-1)

4.

The slope of Cp vs: t of EXP3312 on semi-log paper (hr-1)

5.

The slope of terminal portion of dXmu/dt vs: t (mid) on semi-log paper (hr-1)

6.

The slope of stripped portion of Cpm vs: t or (Concentration of M1 vs: t) on semi-log paper
(hr-1)

7.

The equation for X in the body.

8.

The equation for Xm or M1 in the body.

9.

K1 (elimination rate constant of EXP3312) (hr-1)

10.

ku (urinary excretion rate constant of EXP3312) (hr-1)

11.

km (metabolism rate constant of M1 (hr-1)

12.

kmu (urinary excretion rate constant of M1 (hr-1)

13.

Cp0 (Initial IV bolus concentration of EXP3312) (mic/ml)

14.

Vd of EXP3312 (L)

15.

Vdm of M1 (L)

16.

AUC (Cp vs: t for EXP3312 by IV bolus)

17.

AUMC (t*Cp vs: t for EXP3312 by IV bolus) (mic/mL* hr2))

18.

MRT for EXP3312 (hr)

19.

AUC (dXu/dt vs: t (mid) for EXP3312 by IV bolus) (mg)

20.

AUC (dXmu vs: t (mid) for M1) (mg)

21.

X0 (EXP3312 in the body at time zero) (mg)

22.

Xmu 0 (M1in the urine at time zero) (mg)

23.

Xm0 (M1 in the body at time zero) (mg)

24.

X at 2hr (EXP3312 in the body at 2 hr.) (mg)

25.

Xm at 2hr (M1 in the body at 2 hr.) (mg)

26.

Xinf (EXP3312 in the body at infinite time) (mg)

27.

Xuinf (EXP3312 in the urine at infinite time) (mg)

28.

Xminf (M1in the body at infinite time) (mg)

29.

T1/2 (EXP3312) (hr)

30.

Clearance of EXP3312 (L/hr)

(mic/mL*hr))

EXP3312 Infusion Questions:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-59

Practice Exams: Section 1

31.

K (elimination rate constant of EXP3312) (hr-1)

32.

T1/2 (EXP3312) (hr)

33.

Volume of distribution (L/kg)

34.

Clearance (L/hr/kg)

35.

How long will it take to reach 95% steady state? (hr)

36.

Q (infusion rate) (mg/hr)

37.

Find the plasma concentration if the infusion in Table 14-7 on page 58 was discontinued at 5
hours instead of 45 minutes. (mic/mL)

38.

Find the time need for the concentration to drop to 0.23 mic/mL after the infusion in question 37
is discontinued (hours).

39.

Determine the new infusion rate necessary to maintain a plasma concentration of Cpss 1.0 mic/
mL of the average rat. (mg/hr)

40.

Find a loading dose for the average rat which would give the Cpss of 1.0 mic/mL immediately
(mg)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-60

Practice Exams: Section 1

TABLE 14-8 Answer

Pool

Minus
ones

Small
ones

Big
ones

Bigger
ones

-0.121

0.55

1.05

-0.26

0.019

0.62

1.15

-0.391

0.039

0.72

1.32

-0.567

0.063

0.80

1.69

-0.621

0.105

0.83

2.63

-0.866

0.146

0.866

3.5

Equations
None
X0 e

K 1 t

X0 K1 t
------ e
Vd

-1.112

0.169

0.94

4.17

km X0
K1 t
K 2 t
---------------------------------(e
e
)
( K 1 K 2 )Vd m
k mu X 0
K2 t
K 1 t
--------------------------------- (e
e
)
( K 1 K 2 )V dm
k mu k m X 0 K 2 t K1 t
----------------------(e
e
)
( K 1 K2 )

K1 t

k mu k m X 0 K 1 t K2 t
----------------------(e
e
)
( K 1 K2 )

ku
K1 t
-----X (1 e
)
K 0
1

km X0
K2 t
K 1 t
---------------------------------(e
e
)
( K 1 K 2 )Vd m

K 1 t

kuX 0 e

kf X0 e

Equations

k mu X 0
K2 t
K1 t
----------------------(e
e
)
( K 1 K2 )

kf
K1 t
-----X (1 e
)
K 0

k mf k m X 0 1 e K 2 t 1 e K 1 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 2 K1

-1.981

0.26

0.95

4.83

km X0
K2 t
K 1 t
----------------------(e
e
)
( K1 K 2 )

k mf k m X 0 1 e K 1 t 1 e K 2 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 1 K2

-2.63

0.42

0.96

7.2

km X0
K1 t
K 2 t
----------------------(e
e
)
( K1 K 2 )

k mu k m X 0 1 e K 2 t 1 e K 1 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 2 K1

-3.71

0.48

0.99

10.1

k mu X 0
K2 t
K 1 t
----------------------(e
e
)
( K1 K2 )

k mu k m X 0 1 e K 1 t 1 e K 2 t
---------------------- --------------------- ---------------------
( K1 K 2 ) K 1 K2

This exam is given early at the request of those students who had conflict with the time agreed upon. They signed a
document stating that they would not share their experience with anyone under pain of prosecution for adademic misconduct which will result minimally in an F for the course. You also must sign this document.
I ___________________________________________ did not comunicate in anyway anything at all regarding this
exam nor give or receive any assistance during the exam upon my honor as a health professional and under pain of
prosecution for adademic misconduct.
Signed__________________________________________________________Date__________________

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-61

Practice Exams: Section 1

14.11 Graph Paper

0
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

14-62

CHAPTER 15

Practice Exams: Exam 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-1

Practice Exams: Exam 2

15.1 Nifedipine: Exam 2


Nifedipine (Procardia @) is a calcium channel blocker which specifically inhibits potential-dependent channels not
receptor-operated channels, preventing calcium influx of cardiac and vascular smooth muscle (coronary, cerebral).
Calcium channel blockers reduce myocardial contractility and A-V node conduction by reducing the slow inward calcium current. They are indicated in angina, cardiac dysrhythmias, and hypertension among others. Nifedipine
appears to be metabolized entirely into an inactive metabolite, an acid and subsequently further metabolized to a lactone. Both the acid and the lactone are excreted into the urine and the feces.

Hepatic blood flow in normals is 1.6 L/min


Renal blood flow in normals is 1.2 L/min

Echizen and Eichelbaum (Clin Pkin 1986; 11:425-49) and Kleinbloesem et al (Clin Pcol Therap 1986; 40: 21-8)
reviewed the pharmacokinetics of Nifedipine. While the drug is not routinely given by IV bolus and does not strictly
conform to a one compartment model, let's treat the data as if those problems can be ignored. The following data is
offered for evaluation:
TABLE 1-1. Nifedipine

Time (hr)

Cp

(mcg/L)

Cm1 (mcg/L)

0.5

24.7

44.4
71.8

IV Bolus Profile

Xm1f (mg)

Xm1u (mg)

.14

.59

Xm2f (mg)

Xm2u (mg)

139

65.6

96.5

.44

1.83

.028

.11

31.1

100

.77

3.25

.073

.29

14.6

94.7

1.1

4.65

.135

.54

12

76.5

1.69

7.10

.291

1.15

24

34

2.77

11.63

.75

2.95

3.6

15.1

1.3

5.0

7 days

TABLE 1-2 Nifedipine

Oral information

Brand

Generic

Route

IV

Oral Capsule

Oral Capsule

Dose (mg)

25

10

10

AUC (ug/L*hr)

785

236

204

AUMC (ug/L*hr2 )

2093

866

816

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-2

Practice Exams: Exam 2 : Nifedipine: Exam 2

15.1.1

NIFEDIPINE QUESTIONS:
Find:

21) MRT (oral generic capsule) (hr)

1) MRT iv (hr)

22) MAT (oral generic capsule) (hr)

2) Ke (elimination rate constant) for Nifedipine (hr-1 )

23) Ka, the apparent absorbtion rate constant, for the generic capsule (hr-1 )

3) T 1/2 for Nifedipine

24) Peak time for the generic capsule (hr)

4) Cp0 for iv dose (ug/L)


5) Vd for Nifedipine (L)

25) Cpmax , the maximum concentration


of the generic oral capsule give as a single
dose (ug/L)

7) Cp of Nifedipine at one hour after the


IV dose

26) Comparative bioavailability of the oral


capsules

8) AUC from 0 to one hour for the IV dose


9) Total Body Clearance of Nifedipine (L/
hr)

Your patient is controlled by 20 mg TID


of the brand name oral capsule when he
is healthy.

10) Renal Clearance of Nifedipine (L/hr)

27) What is his N for that dosing regimen?

11) Hepatic Clearance of Nifedipine (L/hr)

28) Cpssmax for this patient at this dosing


regimen (ug/L)

12) Renal Extraction Ratio


13) Hepatic Extraction Ratio
14) Absolute bioavailability for the brand
name capsule
15) MRT (oral brand name capsule) (hr)
16) MAT (oral brand name capsule) (hr)
17) Ka, the apparent absorbtion rate constant, for the brand name capsule (hr-1 )
18) Peak time for the brand name capsule
(hr)
19) Cpmax , the maximum concentration
of the brand name oral capsule give as a
single dose (ug/L)

29) Cpssavg for this patient at this dosing


regimen (ug/L)
30) Cpssmin for this patient at this dosing
regimen (ug/L)
You want to maintain his plasma concentrations between 110% of Cpssmax
and 90% of Cpssmin . How would you
change the dosage regimen to if your
patient suffered from:
31) stenosis of the kidney (Fr = 0.67).
32) renal failure (Fi = 0.67).
33) stenosis of the liver (Fr = 0.67)
34) cirrhosis of the liver (Fi = 0.67)

20) Absolute bioavailability for the generic


capsule

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-3

Practice Exams: Exam 2 : Nifedipine: Exam 2

TABLE 1-3 Nifedipine

Small

Answer Pool

Medium

Large

Dosing changes:

a) 0.00

a) 1.85

a) 59

a) 10 mg once daily

b) 0.05

b) 2.67

b) 85

b) 10 mg BID

c) 0.33

c) 3.67

c) 92.5

c) 10 mg TID

d) 0.375

d) 4.0

d) 101

d) 10 mg QID

e) 0.65

e) 4.32

e) 124

e) 20 mg once daily

f) 0.75

f) 9.3

f) 147

f) 20 mg BID

g) 0.85

g) 18.5

g) 185

g) 20 mg TID (no change necessary)

h) 1.0

h) 32

h) 202

h) 20 mg QID

i) 1.33

i) 38

i) 248

i) 30 mg once daily

j) 1.57

j) 49

j) 294

j) 30 mg BID

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-4

Practice Exams: Exam 2 : Nifedipine: Exam 2

15.1.2

NIFEDIPINE SOLUTIONS
1.

21.

MRT iv = 2.67hr
k e = 0.375hr

3.

T1 2 = 1.85hr

4.

Cp 0 = 294ug L

5.

Vd = 85L

generic

capsule

generic

capsule

generic

capsule

MRT = 4hr

2.

Oral

22.

Oral

MAT = 1.33hr
23.

Oral

Ka = 0.75hr
24.

Oral generic capsule peak time

6.

skip (error in numbering)

1.85hr

7.

Cp 1 = 202ug L

25.

8.

AUC 0 1 = 248ug L hr

9.

Cl s = 32L hr

10.

Cl r = 0

Peak time = 1.18 (ok)

11.

Cl h = 32L hr

Cpmax = 0.78 (not ok!)

12.

Er = 0

27.

N = 4.32

13.

Eh = 0.33

28.

Cp

14.
Absolute bioavailability for brand
name capsule = 0.75

29.

Cp

15.

30.

Cp

31.

No change

32.

No change

33.

No change

34.

No change

Oral

generic

capsule

Cp max = 38ug L
26.
Comparative bioavailability of the
oral capsules B/A:
AUC = 0.86 (ok)

Oral

brand

name

capsule

MRT = 3.67hr
16.

Oral

brand

name

capsule

brand

name

capsule

MAT = 1.0hr
17.

Oral

Ka = 1.0hr
18.

Oral

ss
ss
ss

max =

185ug L

ave =

59ug L

min =

9.3ug L

brand

name

peak

time

1.57hr
19.

Oral

brand

name

Cp max = 49ug L
20.
Absolute
generic = 0.65

bioavailability

for

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-5

Practice Exams: Exam 2 : Valproate: Exam 2

15.2 Valproate: Exam 2


All questions are internally consistent. Information gained in any one can be used in all others. Note, however, there
is a change in patient status midway through.
Valproate is a carboxylic acid anticonvulsant. Its activity may be related, at least in part, to increase concentrations of the neurotransmitter inhibitor gamma aminobutyric acid in the brain. It is used alone or in combination
with other anticonvulsants. in the prophylactic management of petit mal. It appears to be almost entirely cleared
by liver function with negligible amounts excreted into the urine unchanged. It comes as soft gelatin capsules of 250
mg and enteric coated tablets 250 and 500 mg. The therapeutic range for valproate appears to be between 20 and 100
mic/ml. The volume of distribution is 0.19 L/Kg and your patient is 70 kg.

TABLE 1-4

IV bolus
Dose (mg)
AUC (mg/L*hr)
2

AUMC(mg/L*hr )

Generic

250

250

594

273

253

9428.6

4605.5

4184.6

2.95

Tpeak (hr)
Cpmax
Bioavailability (f)

Brand

500

14.35

1.0

.92

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-6

Practice Exams: Exam 2 : Valproate: Exam 2

15.2.1

VALPROATE QUESTIONS

1) Calculate the MRTiv


a) 11

of Valproate (hr).

b) 15.87 c) 16.54

d) 16.87 e) 17.08

2) Calculate the rate constant of elimination (hr-1 ) in normals ?


a) 0.091

b) 0.063

c) 0.060

d) 0.0593

e) 0.0585

3) Calculate the half life of Valproate (hr)?


a) 7.6

b) 11 c) 11.5

d) 11.7 e) 11.8

4) Calculate the hepatic clearance of valproate (L/hr).


a) 0.0084

b) 0.063

c) 0.84

d) 1.25

d) 2.5

5) Calculate the hepatic extraction ratio of valproate.


a) 0.0063

b) 0.0084

c) 0.063

d) 0.084

e) 0.84

6) What is the maximum N for multiple dosing of Valproate?


a) 0.78

b) 1.65

c) 2.12

d) 2.32

e) 2.5

7) What is the maximum acceptable dosing interval for normal patients (hr)?
a) 8

b) 12

c) 18

d) 24

e) 25.5

8) What is the N if we are going to dose TID?


a) 0.63

b) 0.727

c) 0.842

d) 1.25 e) 1.42

9) If you dosed this patient 500 mg BID with the brand name product, what would be your maximum concentration at
steady state (mg/L)?
a) 87.3

b) 70.9

c) 65.2

d) 52.8

e) 45.8

10) What would be your minimum concentration at steady state (mg/L)?


a) 52.8

b) 32.7

c) 30.1 d) 27.1

e) 22.9

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-7

Practice Exams: Exam 2 : Valproate: Exam 2

11) What would be your average concentration at steady state (mg/L)?


a) 63.8

b) 49.8

c) 47.7

d) 45.8

e) 34.4

12) What loading dose would you give to get to Cpss right away (mg)?
a) 250

b) 500

c) 750

d) 1000

e) 1500

13) If you changed the dosage regimen to 250 mg QID, what would happen to the Cpssmax, Cpssave, Cpssmin?
a) Cpssmax - up, Cpssave - up, Cpssmin - up
b) Cpssmax - down, Cpssave - down, Cpssmin - down
c) Cpssmax - same, Cpssave - same, Cpssmin - same
d) Cpssmax - up, Cpssave - same, Cpssmin - down
e) Cpssmax - down, Cpssave - same, Cpssmin - up

14) Calculate f, the absolute bioavailability of the generic product.


a) 0.67

b) 0.75

c) 0.85

d) 0.93e) 1.0

15) Calculate the comparative bioavailability of the generic product.


a) 0.67

b) 0.75

c) 0.85

16) Calculate the MRToral


a) 11

b) 15.87 c) 16.54

d) 0.93e) 1.0

of the generic product.

d) 16.87 e) 17.08

17) Calculate the MAT of the generic product.


a) 0.67

b) 0.75

c) 0.85

d) 0.93e) 1.0

18) Calculate the Ka of the generic product.


a) 1.5

b) 1.33

c) 1.18

d) 1.08

e) 1.0

19) Calculate the peak time of the generic product.


a) 2.21

b) 2.45

c) 2.59

20) Calculate the Cpmax

d) 2.76

e) 2.95

of the generic product.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-8

Practice Exams: Exam 2 : Valproate: Exam 2

a) 8.1

b) 9.3

c) 10.7

d) 12.6

e) 14.4

21) Is the generic product bioequivalent?


a) Yes
b) No, because the comparative bioavailability is outside the federal guidelines.
c) No, because the ratio of the peak times is outside the federal guidlines.
d) No, because the ratio of the Cpmax s is outside the federal guidlines.
e) No, because the generic fails more than one of the required comparisons.

In patients who are also currently on phenobarbital their intrinsic clearance of valproate increases by 50% as the phenobarbital induces the enzymes which metabolize valproate. Further questions refer to this condition.

22) Calculate his new clearance.


a) 0.0084

b) 0.063

c) 0.84

d) 1.25

d) 2.5

d) 0.059

e) 0.042

23) Calculate his new K.


a) 0.095

b) 0.084

c) 0.063

24) Calculate his new maximum acceptable dosing interval (hr).


a) 8

b) 12

c) 17

d) 18

e) 24

25) What dosage regimen would you recommend to try to maintain his plasma concentrations within 110% of the maximum and 90% of the minimum concentrations attained when he was normal?
a) 750 mg BID

b) 500 mg TID

c) 750 mg TID

d) 250 mg QID

e) 500 mg QID

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-9

Practice Exams: Exam 2 : Valproate: Exam 2

15.2.2

VALPROATE SOLUTIONS

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-10

Practice Exams: Exam 2 : Methyl phenidate

15.3 Methyl phenidate


Methyl phenidate (MP) (Ritalin@) is an effective stimulant in the treatment of narcolepsy in adults and attention deficit
syndrome in children. It is entirely metabolized to the inactive metabolite, Ritalinic Acid (RA), by the liver which is
subsequently excreted unchanged into the urine. The following information was obtained from a 70 Kg male.
QH = 24 mL/min/Kg
QR = 19 mL/min/Kg
TABLE 1-5 Ritalin Data

IV

Brand
Name

Generic

Dose (mg)

10

20

20

AUC (ug/ml*hr)

0.20

0.04

0.035

AUMC (ug/ml*hr2

0.32

0.14

0.1225

TABLE 1-6 Ritalin

Answer Pool

Itty- Bitty

Tiny

Puny

Small

Medium

Large

0.016

0.1

1.1

10

100

yes

0.00016

0.0251

0.125

1.6

20

125

no, Ratio of Tps

0.0005

0.035

0.25

1.75

30

225

no, Ratio of Cmaxs

0.00074

0.042

0.35

1.9

40

375

no, Ratio of AUCs

0.00084

0.05

0.42

2.6

50

435

no, Ratio of AUMCs

0.0016

0.067

0.53

3.5

60

550

no, more than one criterion

0.005

0.075

0.625

4.2

70

675

20 mg TID

0.0074

0.0875

0.727

5.0

80

750

20 mg BID

0.0084

0.091

0.875

7.27

90

875

20 mg QD

0.0090

0.096

0.91

8.75

96

995

20 mg QID

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

Words

15-11

Practice Exams: Exam 2 : Methyl phenidate

15.3.1

METHYL PHENIDATE QUESTIONS:


1) MRT iv (hr)
2) Ke (elimination rate constant) for
Ritalin (hr-1 )

25) Cpmax , the maximum concentration of


the generic oral tablet give as a single dose
(ug/mL)
26) Comparative bioavailability of the oral
tablets

3) T 1/2 for Ritalin (hr)


4) Cp0 for iv dose (ug/mL)
5) Vd for Ritalin (L)
6) Cp of Ritalin at one hour after the IV
dose
7) AUC from 0 to one hour for the IV dose
8) Total Body Clearance of Ritalin (L/hr)

27) Are the tablets bioequivalent?


Your patient is controlled by 20 mg TID of
the brand name oral tablet when he is
healthy. For This patient and this dosage
regimen, what is his:
28) N ?

9) Renal Clearance of Ritalin (L/hr)

29) Cpssmax (ug/mL)

10) Hepatic Clearance of Ritalin (L/hr)

30) Cpssavg (ug/mL)

11) Intrinsic Hepatic Clearance of Ritalin


(L/hr)

31) Cpssmin (ug/mL)

12) Renal Extraction Ratio


13) Hepatic Extraction Ratio
14) Absolute bioavailability for the brand
name tablet
15) MRT (oral brand name tablet) (hr)
16) MAT (oral brand name tablet) (hr)
17) Ka, the apparent absorbtion rate constant, for the brand name tablet (hr-1 )
18) Peak time for the brand name tablet
(hr)

You want to maintain his plasma concentrations between 110% of Cpssmax and 90% of
Cpssmin . How would you change the dosage
regimen to if your patient suffered from:
(in no case was there a change in Vd)
32) stenosis of the kidney (Fr = 0.67)?
33) renal failure (Fi = 0.34)?
34) stenosis of the liver (Fr = 0.67)?
35) cirrhosis of the liver. (Fi = 0.67)?
36) treatment with phenobarbital (Fi =
1.33)?

19) Cpmax , the maximum concentration of


the brand name oral tablet give as a single
dose (ug/mL)
20) Absolute bioavailability for the generic
tablet
21) MRT (oral generic tablet) (hr)
22) MAT (oral generic tablet) (hr)
23) Ka, the apparent absorbtion rate constant, for the generic tablet (hr-1 )
24) Peak time for the generic tablet (hr)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-12

Practice Exams: Exam 2 : Methyl phenidate

15.3.2

METHYL PHENIDATE SOLUTIONS:


1.

19.

MRT iv = 1.6hr

2.

k = 0.625hr

3.

Brand

name

Cp max = 0.0084ug mL

20.

Generic - f = 0.0875

T1 2 = 1.1hr

21.

Generic - MRT = 3.5hr

4.

Cp 0 = 0.125ug mL

22.

Generic - MAT = 1.9hr

5.

Vd = 80L

23.

Generic - Ka = 0.53hr

6.

Cp 1 = 0.067ug mL

24.

Generic - T peak = 1.75hr

25.

Generic

7.

AUC 0 1 = 0.096ug mL hr
Cl s = 50L hr

9.

Cl r = 0

10.

Cl h = 50L hr

11.

Cl int = 100L hr

28.

N = 7.27

12.

Er = 0

29.

Cp

13.

Eh = 0.53

30.

Cp

31.

Cp

32.

20 mg TID

33.

20 mg TID

34.

20 mg BID

35.

20 mg TID

36.

20 mg QID

15.

Cp max = 0.0074ug mL

8.

14.

26.
0.875

Comparative

bioavailability

27.
Yes, the tablets are bioequivalent all parameters are within federal guidelines.

Brand name - f = 0.1


Brand name - MRT = 3.5hr

16.

Brand name - MAT = 1.9hr

17.

Brand name - Ka = 0.526hr

18.

Brand name - Tpeak = 1.74hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

ss
ss
ss

max =

0.0251ug mL

avg =

0.005ug mL

min =

0.00016ug mL

15-13

Practice Exams: Exam 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-14

Practice Exams: Exam 2 : Verapamil

15.4 Verapamil
Verapamil is a calcium channel blocker with vasodilatory and antiarrhythmic effects. It is about 95% metabolized by
the liver with the metabolites showing up in the urine and feces.
Hepatic blood flow in normals is 1.6 L/min
Renal blood flow in normals is 1.2 L/min
TABLE 1-7 Verapamil

Route

Data

IV

Brand

Generic

Oral Tablet

Oral Tablet

Dose (mg)

15

80

80

AUC (ng/mL*hr)

300

480

400

AUMC (ng/mL*hr2 )

1600

2690

2280

TABLE 1-8

Verapamil Answer Pool

Tiny

Small

Medium

Large

Dosing regimens

Bioavailability answers

1.06

26

116

40 mg qd

Yes

0.063

2.15

46

267

40 mf bid

No, tp ratio is not within limits

0.188

2.50

47.5

369

40 mg tid

No, Cpmax ratio is not within limits

0.250

2.70

50

533

40 mg qid

No, AUC ratio is not within limits

0.270

3.69

56

637

80 mg qd

No, f ratio is not within limits

0.300

5.33

61.5

830

80 mg bid

No, ka ratio is not within limits

0.370

5.60

76.5

905

80 mg tid

No, ke ratio is not within limits

0.693

5.70

83

970

80 mg qid

No, MRT ratio is not within limits

0.85

6.37

90.5

1160

160 mg qd

No, Cl ratio is not within limits

0.905

8.30

97

2670

160 mg bid

No, more than one of the required ratios are no within limits

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-15

Practice Exams: Exam 2 : Verapamil

15.4.1

VERAPAMIL QUESTIONS
1) MRT iv (hr)
2) Ke (elimination rate constant) for Verapamil (hr-1 )
3) T 1/2 for Verapamil
4) Cp0 for iv dose (ug/L)
5) Vd for Verapamil (L)
7) Cp of Verapamil at one hour after the IV
dose
8) AUC from 0 to one hour for the IV dose

23) Ka, the apparent absorbtion rate constant, for the generic tablet (hr-1 )
24) Peak time for the generic tablet (hr)
25) Cpmax , the maximum concentration
of the generic oral tablet give as a single
dose (ug/L)
26) Comparative bioavailability of the oral
tablets
27) Are the tablets bioequivalent?

9) Total Body Clearance of Verapamil (L/


hr)

Your patient is controlled by 80 mg TID


of the brand name oral tablet when he is
healthy.

10) Renal Clearance of Verapamil (L/hr)

28) What is his N for that dosing regimen?

11) Hepatic Clearance of Verapamil (L/hr)

29) Cpssmax for this patient at this dosing


regimen (ug/L)

12) Renal Extraction Ratio


13) Hepatic Extraction Ratio
14) Absolute bioavailability for the brand
name tablet
15) MRT (oral brand name tablet) (hr)
16) MAT (oral brand name tablet) (hr)
17) Ka, the apparent absorbtion rate constant, for the brand name tablet (hr-1 )
18) Peak time for the brand name tablet
(hr)
19) Cpmax , the maximum concentration
of the brand name oral tablet give as a single dose (ug/L)
20) Absolute bioavailability for the generic
tablet
21) MRT (oral generic tablet) (hr)
22) MAT (oral generic tablet) (hr)

30) Cpssavg for this patient at this dosing


regimen (ug/L)
31) Cpssmin for this patient at this dosing
regimen (ug/L)
You want to maintain his plasma concentrations between 110% of Cpssmax
and 90% of Cpssmin . How would you
change the dosage regimen to if your
patient suffered from:
32) stenosis of the kidney (Fr = 0.67). (No
change in volume of distribution.)
33) renal failure (Fi = 0.34).(Volume of
distribution is reduced in this disease to
50% of normal)
34) stenosis of the liver (Fr = 0.67) (No
change in volume of distribution)
35) cirrhosis of the liver (Volume of Distribution and the bioavailability are both
doubled while the half life is quadrupled )

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-16

Practice Exams: Exam 2 : Verapamil

15.4.2

VERAPAMIL SOLUTIONS
1.

17.

MRT iv = 5.33hr

Brand

Ka = 3.69hr

2.

k e = 0.188hr

3.

T1 2 = 3.69hr

0.85hr

4.

Cp 0 = 56ug L

19.

5.

Vd = 267L

18.

name

tablet

Brand name tablet - peak time Brand

name

tablet

Cp max = 76.5ug L

6.

skip - error in numbering

20.
Generic - Absolute bioavailability
- f = 0.25

7.

Cp 1 = 47ug L

21.

Generic - MRT = 5.7hr

22.

Generic - MAT = 0.37hr

8.

AUC 0 1 = 50ug L hr

9.

Cl = 50L hr

23.

Generic - Ka = 2.70hr

10.

Cl r = 2.50L hr

24.

Generic - T peak = 1.06hr

11.

Cl h = 47.5L hr

25.

Generic - Cp max = 61.5ug L

12.

Er = 0.063

26.

Comparative bioavailability - 0.85

13.

Eh = 0.49

Correction - should

be added to answer pool


14.
Brand name tablet - absolute bioavailability = 0.30
15.

Brand

name

tablet

name

tablet

MRT = 5.60hr
16.

Brand

MAT = 0.270hr

27.
The tablets are not bioequivalent peak time ratio is not within limits.
28.

N = 2.15

29.

Cp

ss

max =

116ug L

ss

Cp avg = 60ug L (change in


30.
answer pool)
31.

Cp

ss

min =

26ug L

32.
No change in dosing regimen - 80
mg TID
Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-17

Practice Exams: Exam 2 : Verapamil

33.

40 mg TID

35.

40 mg BID

34.
No change in dosing regimen - 80
mg TID

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-18

Practice Exams: Exam 2 : Hydromorphone hydrochloride

15.5 Hydromorphone hydrochloride


Hydromorphone hydrochloride is an analog of morphine which has about seven times the analgesic effect of morphine
when given by IV. It is about 90% metabolized. The following data was obtained by Valner et al J.Clin Pcol 21(1981)
152-6:
TABLE 1-9 Hydromorphone

hydrochloride Data
IV

Oral tablet
Brand

Oral tablet
Generic

Dose (mg)

AUC (ng/mL*hr)

83

87

65

AUMC (ng/mL*hr2 )

291

348

292

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-19

Practice Exams: Exam 2 : Hydromorphone hydrochloride

15.5.1

HYDROMORPHONE HYDROCHLORIDE QUESTIONS

For the IV product, find:


1.

MRTiv.

(291/83=3.5)

2.

K. (1/3.5=0.285

3.

T1/2 (0.693/.285= 2.4 hr)

4.

Cp0 (AUC*K = 83*0.285= 23.7ng/mL

5.

Ku (0.1*0.285=0.029)

6.

Km (0.9*0.285=0.256)

7.

Cls (f*D/AUC=1*2000/83=24L/hr)

8.

Vd (Cls/K=Vd= 24/0.285=84L

9.

Clh (0.9*Cls = 21.6 L/hr

10.

Clr (0.1*Cls = 2.4 L/hr

11.

Eh (21.6 / (24 mL/min * 60 min * 70 Kg /


1000mL/L) = 0.2

12.

Er ( 2.4 / ( 10 ml/min * 60 min * 70 Kg /


1000mL/L) = 0.06

27.

R(Bio)= 0.75 fail

You would like to dose your healthy


patient using the brand name tablet so
that his plasma concentration is in the
therapeutic range of 35 to 5 ng/mL.
Find:
28.

Nmax (ln(35/5)/ln(2) = 2.8

29.

Taumax (2.8*2.4=6.74 hr

30.

Maximum acceptable tau = 6

31.

N (6/2.4 =2.5)

32.

What dosage regimen would you recommend? (4mg q6h)

33.

Cpssmax? (30 ng/mL)

34.

Cpssavg? (14 ng/mL)

35.

Cpssmin? ( 5 ng/mL)

For the Brand name oral tablet, find:


He gets renal stenosis (Fr = 0.5) with no
change in Vd. Find:

13.

MRT (348/87=4.0)

14.

MAT(4 - 3.5 = 0.5)

15.

Ka (1/0.5 = 2.0)

16.

Tp (ln(ka/K)/(ka-K)=1.95/1.715=1.14hr

17.
18.

Absolute bioavailability, f ((87/4)/(83/


2)=0.52
Cpmax (18 ng/mL)

For the Generic oral tablet, find:


19.

MRT ( 292/65=4.5 hr)

20.

MAT (4.5 - 3.5 = 1 hr)

21.

ka (1/MAT = 1/1=1 hr-1)

22.

Tp (ln(1/0.285)/(1-0.285)=1.26/.715=1.76
hr

23.

Absolute bioavailability, f ((65/4)/(83/


2)=0.39

24.

Cpmax (11 ng/mL)

Would you consider the oral tablets to


be bioequivalent? Why or why not?
25.

No R(Tp) = 1.54 fail

26.

R(Cp) = 0.61 fail

36.

FClr0.94

37.

Clr*2.3

38.

FClh1

39.

Clh*21.6

40.

FCls.99

41.

Cls*23.9

42.

K*0.284

43.

t1/2*2.44

44.

Nmax

45.

Taumax6.83

46.

Maximum acceptable tau

47.

N 2.46

48.

What dosage regimen would you recommend? 4mg q6h

49.

Cpssmax? 30

50.

Cpssavg? 15

51.

Cpssmin? 5.5

2.8
6

His stenosis of the kidney clears and he


now goes into renal dysfunction
(Fi=0.5), find:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-20

Practice Exams: Exam 2 : Hydromorphone hydrochloride

52.

FClr.52

77.

Taumax8

53.

Clr*1.23

78.

Maximum acceptable tau 8

54.

FClh2

79.

N6

55.

Clh*21.6

80.

56.

FCls.95

What dosage regimen would you recommend? 4 mg q6h

57.

Cls*22.8

81.

Cpssmax? 32

58.

K*0.271

82.

Cpssavg? 17

59.

t1/2*2.55

83.

Cpssmin? 7.5

60.

Nmax

61.

Taumax

62.

Maximum acceptable tau6

63.

N 2.35

64.

What dosage regimen would you recommend? 4mg q6h

84.

FClr1

85.

Clr*2.4

65.

Cpssmax? 31

86.

FClh.56

66.

Cpssavg? 15

87.

Clh*12

67.

Cpssmin? 6.1

88.

FCls.6

89.

Cls*14.4

90.

K*.171

91.

t1/2*4.04

92.

Nmax 2.8

2.8
7.13

His renal dysfunction clears and now he


suffers from stenosis of the liver
(Fr=0.5), Find:

His stenosis of the liver clears and he


now suffers from liver dysfunction
(Fi=0.05), Find:

68.

FClr1

93.

Taumax11.3

69.

Clr*2.4

94.

Maximum acceptable tau8

70.

FClh.833

95.

N 1.48

71.

Clh*18

96.

72.

FCls.85

What dosage regimen would you recommend?4 mg q8h

73.

Cls*20.4

97.

Cpssmax? 33

74.

K*.243

98.

Cpssavg? 18

75.

t1/2*2.85

99.

Cpssmin?8.4

76.

Nmax 2.8

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-21

Practice Exams: Exam 2 : Hydromorphone hydrochloride

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-22

Practice Exams: Exam 2

15.6 Fosinopril Sodium


Fosinopril Sodium (MW 585) is a phosphinic prodrug of the angiotensin converting enzyme (ACE) inhibitor fosinoprilat (MW 563). After oral administration, fosinopril is slowly and incompletely absorbed, and is converted to the active
fosinoprolat by esterases in the gastrointestinal mucosa and the liver. Unlike other ACE inhibitors, elimination of fosinoprilat is divided equally between renal and hepatic pathways. With the IV dose fosinoprilat was given and measured. With the oral dose fosinopril was given but fosinoprilat was measured. The following information was obtained
from a 70 Kg male. This information was constructed from Kostis et al Fosinopril: Pharmacokinetics and pharmacodynamics in congestive heart failure Clin Pcol Ther 58(6) 660-5 (1995); Hui et al Pharmacokinetics of fosinopril in
patients with various degrees of renal function Clin Pcol Ther 49(4) 457 -66 (1991)
QH =

24 mL/min/Kg Blood

QR = 19 mL/min/Kg Blood
TABLE 1-10 Fosinopril

Data
IV

Brand Name
Tablet

Generic
Tablet

Dose (mg)

7.5

10

10

AUC (ng/ml*hr)

5700

1500

1400

AUMC (ng/ml*hr2)

78000

25000

23100

TABLE 1-11 Fosinopril

Answer Pool:

0.015

0.19

1.3

10

100

yes

0.00016

0.0251

0.0005

0.035

0.21

1.6

13.7

142

no, Ratio of Tps

0.25

1.75

16.6

192

no, Ratio of Cmaxs

0.00074

0.042

0.36

1.9

18

253

no, Ratio of AUCs

0.00084

0.05

0.42

2.8

50

387

no, Ratio of AUMCs

0.0016

0.067

0.53

3.5

67

402

no, more than one criterion

0.005

0.073

0.65

4.0

73

416

10 mg qid

0.0064

0.0875

0.72

5.6

84

750

10 mg tid

0.0084

0.091

0.84

7.0

93

875

10 mg bid

0.0090

0.096

0.93

9.5

96

995

10 mg qd

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-23

Practice Exams: Exam 2

15.6.1

FOSINOPRIL QUESTIONS
1) MRT for fosinoprilat (hr)
2) Ke for fosinoprilat (hr-1)
3) T ?
4) Cp0

for Fosinoprilat (hr)


for iv dose (ng/mL)

5) Vd for Fosinoprilat (L)


6) Cp of Fosinoprilat at one hour after the IV dose
7) AUC from 0 to one hour for the IV dose
8) Total Body Clearance of Fosinoprilat (L/hr)
9) Renal Clearance of Fosinoprilat (L/hr)
10) Hepatic Clearance of Fosinoprilat (L/hr)
11) Intrinsic Hepatic Clearance of Fosinoprilat (L/hr)
12) Renal Extraction Ratio
13) Hepatic Extraction Ratio
14) Absolute bioavailability for the brand name tablet
15) MRT (oral brand name tablet) (hr)
16) MAT (oral brand name tablet) (hr)
17) Ka, the apparent absorption rate constant, for the brand name tablet (hr-1

18) Peak time for the brand name tablet (hr)


19) Cpmax , the maximum concentration of the brand name oral tablet give as a single dose (ng/mL)
20) Absolute bioavailability for the generic tablet
21) MRT (oral generic tablet) (hr)
22) MAT (oral generic tablet) (hr)
23) Ka, the apparent absorption rate constant, for the generic tablet (hr-1

24) Peak time for the generic tablet (hr)


25) Cpmax , the maximum concentration of the generic oral tablet give as a single dose (ng/mL)
26) Comparative bioavailability of the oral tablets
27) Are the tablets bioequivalent?

Your patient is controlled by 10 mg TID of the brand name oral tablet when he is
healthy. For This patient and this dosage regimen, what is his:
28) N ?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-24

Practice Exams: Exam 2

29) Cpssmax (ng/mL)


30) Cpssavg (ng/mL)
31) Cpssmin (ng/mL)

You want to maintain his plasma concentrations between 120% of Cpssmax


Cpssmin

and 80% of

. How would you change the dosage regimen to if your patient suffered from:

(in no case was there a change in Vd)


32) stenosis of the kidney (Fr = 0.67)?
33) renal failure (Fi = 0.34)?
34) stenosis of the liver (Fr = 0.67)?
35) cirrhosis of the liver. (Fi = 0.67)?
36) treatment with phenobarbital (Fi = 1.5)?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-25

Practice Exams: Exam 2 : Fosinopril Sodium

15.6.2

FOSINOPRIL SODIUM SOLUTIONS


19.

Oral

brand

name

tablet

1.

MRT = 13.7hr

2.

k = 0.073hr

3.

T1 2 = 9.5hr

4.

Cp 0 = 416ng mL

21.

5.

Vd = 18L

22.

6.

Cp 1 = 387ng mL

7.

AUC 0 1 = 402ng mL

23.

Generic tablet Ka = 0.36hr

8.

Cl s = 1.3L hr

24.

Generic tablet peak time 5.6hr

Cl r = 0.65L hr

25.

Generic

9.
10.

Cl h = 0.65L hr

11.

Intrinsic Hepatic Clearance

Cp max = 0.073ng mL

20.
Oral generic tablet absolute bioavailability is 0.19

Er = 0.015

13.

Eh = 0.0064

Oral

is

15.

30.

Cp

31.

Cp

32.

10 mg TID

33.

10 mg BID

34.

10 mg TID

35.

10 mg TID

36.

10 mg QID

name

tablet

MRT = 16.6hr
name

tablet

MAT = 2.8hr
17.

Oral

Ka = 0.36hr
18.

brand

tablet

all within 20% range.

Cp

brand

27.
Yes! the tablets are bioequivalent
The AUCs, peak times, and Cp max s are

29.

Oral

tablet

26.
Comparative bioavailability of the
oral tablets = 0.93

14.
Oral brand name tablet absolute
bioavailability is 0.21

16.

generic

Cp max = 0.067ng mL

N = 0.84

brand

tablet

MAT = 2.8hr

28.

Oral

generic

MRT = 16.6hr

0.65L hr
12.

Oral

name

tablet

Oral brand name tablet peak time

5.6hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

ss
ss
ss

max =

253ng mL

avg =

192ng mL

min =

142ng mL

15-26

Practice Exams: Exam 2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-27

Practice Exams: Exam 2

15.7 Remoxipride
Remoxipride (MW 296 - Unionized Base pKa 9.4) is a new antipsychotic of the benzamide type (See figure 1). The
pharmacokinetics were studied by Movin-Osswald and Hammarlind-Udenaes (Brit. J Clin Pcol 1991 32(3) 355ff).
Their results are summarized in table 1. The HCl salt of the drug was given (MW 332.5) to these patients in this study
but the drug concentration was reported as the free base in the plasma. In this study, 25% of the Remoxipride was
excreted unchanged, 75% was metabolized. The hepatic and renal blood flow in these patients was 1.5 and 1.2 L/min
respectively. REMEMBER TO PAY ATTENTION TO UNITS.
FIGURE 4-5.

Remoxipride

TABLE 1-12 Remoxipride

Data
IV bolus

Oral Solution

Tablet A

Tablet B

100

100

Dose (mg)

50

AUMC (umole/
L*hr2 )

145

158.7

319.6

282.6

AUC (umole/L*hr)

20.9

19.8

37.6

31.4

Tp (hr)

3.6

Cpmax (mg/L)

.8

.75

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-28

Practice Exams: Exam 2

15.7.1

REMOXIPRIDE QUESTIONS
1) Find the MRT of the IV product (hr).
2) Find the elimination rate constant of remoxipride (hr-1 ).
3) Find the Cp0 of the IV product (mg/L)
4) Find the volume of distribution of the IV product. (L)
5) Find the half life of remoxipride (hr).
6) Find the clearance of remoxipride (L/hr).
7) Find the renal clearance (L/hr).
8) Find the hepatic clearance
9) Find the renal extraction ratio.
10) Find the hepatic extraction ratio.
11) Find the MRT of remoxipride given as the oral solution (hr).
12) Find the MAT of remoxipride given as oral solution (hr).
13) Find the absorption rate constant of remoxipride given as an oral solution (hr-1 ).
14) Find the bioavailability of the oral solution (f).
15) Find the peak time of the oral solution (hr).
16) Find the MRT of remoxipride given as Tablet A, the brand name product.
17) Find the MAT of remoxipride given as Tablet A.
18) Find the apparent absorption rate constant (ka) of remoxipride given as Tablet A.
19) Find the peak time of Tablet A.
20) Find the single dose Cpmax for tablet A (mg/L)
21) Find the mean dissolution time (MDT) of Tablet A, the brand name product (hr).
22) Is Tablet B, the generic product is bioequivalent to Tablet A. Why or why not?
23) Find N for BID dosing.
24) Find Cpss max for two caps bid for the brand name product.
25) Find Cpss avg for two caps bid for the brand name product.
26) Find Cpss min for two caps bid for the brand name product.
27) Which change in physiological status would result in the most significant change in
the TBC of remoxipride?
a) Changes which effect the flow of blood to the liver.
b) Changes which effect the flow of blood to the kidney.
c) Changes which effect the function of the liver.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-29

Practice Exams: Exam 2

d) Changes which effect the function of the kidney.


Your patient is experiencing that change. That physiological function was up 75% above
normal (F = 1.75).
28) Find his new renal clearance
29) Find his new hepatic clearance.
30) Find his new total body clearance.
31) Assuming now change in volume of distribution, find his new half life.
32 Assuming no change in dosage regimen, find his new N.
33 What dosing regimen would you recommend to return his plasma concentrations back
to normal (within 110% of max and 90% of min)?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-30

Practice Exams: Exam 2

15.7.2

REMOXIPRIDE SOLUTIONS
1.

MRT = 6.93hr

2.

k e = 0.144hr

3.

Cp 0 = 0.88mg L

4.

Vd = 50L

5.

T1 2 = 4.8hr

6.

Cl = 7.28L hr

7.

Cl r = 1.8L hr

8.

Cl h = 5.46L hr

9.

Er = 0.025

10.

Eh = 0.06

11.

Oral solution MRT = 8hr

12.

Oral solution MAT = 1.07hr

13.

Oral solution Ka = 0.935hr

14.

Oral solution bioavailability f = 0.95

15.

Oral solution peak time 2.4hr

16.

Tablet A MRT = 8.5hr

17.

Tablet A MAT = 1.57hr

18.

Tablet A Ka = 0.637hr

19.

Tablet A peak time 3.0hr

20.

Tablet A single dose Cp max = 1.03mg L

21.

Tablet A MDT = 0.5hr

Tablet B is not bioequivalent to Tablet A because peak time and Cp max are out of
federal guidelines.
22.

23.

For dosing BID, N = 2.5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-31

Practice Exams: Exam 2

24.

Cp

25.

Cp

26.

Cp

ss
ss
ss

max =

3.88mg L

avg =

1.85mg L

min =

0.685mg L

27.
The change in physiological status that would result in the most significant change
in TBC of remoxipride
is (C) - changes
which effect the function of the liver.
28.

New renal clearance = no change

29.

New Cl h = 9.0L hr

30.

New Cl s = 10.8L hr

31.

New T1 2 = 3.2hr

32.

New N = 3.75

33.

The dosing regimen to recommend = 200 mg TID

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-32

Practice Exams: Exam 2

15.8 Naproxen
Naprosyn@ (naproxen) is a nonsteroidal anti-inflammatory drug (NSAID) with
analgesic properties. It is well absorbed (f = 0.95) and highly protein bound (98%)
with a volume of distribution of about 10 L and a half-life of 13 hours in normal
adults. It is almost entirely cleared by hepatic function (CLr = 1%) with about onethird being metabolized to the 6-o-desmethylnaproxen (which is further metabolized by conjugation) and two-thirds being conjugated directly. Both the 6-o-desmethyl metabolites as well as the conjugates are inactive.
Normal dosing is 500 mg Naproxen BID. You stock 200 and 500 mg tablets in
your HMO.
For the following conditions, new parameters are given in parentheses.
Concomitant treatment with Probenecid, a uricosuric which increases the urinary
excretion of uric acid, while not interfering with the protein binding, effectively
blocks the hepatic conjugation process reducing the hepatic function (Clinth ) to
one-third of normal.
In chronic renal failure (Fir = 0.1) the protein binding is reduced (94%) because of
uremia. This results in a marginal increase in half-life (14 hr.)
In rheumatoid arthritis, hypoalbuminaemia results in a reduction in protein binding
(97%) and increase in the volume of distribution (13 L).
Elderly patients exhibit a decrease in binding (96%), but no change in half-life or
volume of distribution.
For each of the conditions, (with Probenecid, chronic renal failure, arthritic, and
elderly), please recommend a dosage regimen which would give approximately the
same plasma concentrations of free naproxen obtained in the normal case (+ 10%.)
Constants:
L - renal blood perfusion 70kg 60min
L
Q r = 0.0191 --------------------------- 80 -----blood
min kg
hr
hr
L
min
L
Q H = 0.0238 -------------------- hepatic blood perfusion 70kg 60--------- 100 -----blood
min kg
hr
hr

Extraction ratios are calculated for normals and considered to be constant throughout.

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-33

Practice Exams: Exam 2

15.8.1

NAPROXEN QUESTIONS
TABLE 1-1. Question

Patient Condition

Normal

Numbers For Exam

Probenecid

Chronic

Rheumatoid

treatment

Renal Failure

arthritis

Elderly

Dose(mg)

27

51

75

99

28

52

76

100

fu

29

53

77

101

Vd (L)

30

54

78

102

k (hr-1)

31

55

79

103

T 1/2 (hr)

32

56

80

104

AUC (mg/L*hr)

33

57

81

105

% Cl h

34

58

82

106

% Cl r

35

59

83

107

Cl tot (L/hr)

10

36

60

84

108

Cl h (L/hr)

11

37

61

85

109

Cl r (L/hr)

12

38

62

86

110

Eh

13

Er

14

Cl h
Cl r

Cp

(L/hr)

15

39

63

87

111

(L/hr)

16

40

64

88

112

FR h

17

41

65

89

113

FI h

18

42

66

90

114

FR r

19

43

67

91

115

FI r

20

44

68

92

116

FCL

21

45

69

93

117

(hr)

22

46

70

94

118

23

47

71

95

119

24

48

72

96

120

int

int

ss
max free

g
------ mL

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-34

Practice Exams: Exam 2

Cp

Cp

ss g
-------avg free mL

25

49

73

97

121

g
------ mL

26

50

74

98

122

ss
min free

Answer Pool
S

XL

XXL

XXXL

0.000067

0.0182

0.100

1.0

10.0

100

0.0005

0.0200

0.133

1.1

11.3

200

0.0050

0.0300

0.177

1.4

12.0

303

0.0080

0.0400

0.182

1.5

13.0

356

0.0089

0.0495

0.267

1.6

13.3

500

0.0092

0.0533

0.315

1.7

14.0

595

0.0095

0.0600

0.333

1.8

26.5

891

0.0097

0.0610

0.341

1.9

31.7

1044

0.0098

0.0675

0.528

2.0

38.1

2000

0.0099

0.0700

0.533

2.1

45.0

3000

0.0790

0.577

2.2

50.0

0.0798

0.615

2.9

53.0

0.0857

0.675

3.0

62.0

0.0923

0.700

5.3

80.0

0.0950

0.790

6.2

86.0

0.0970

0.798

8.0

92.0

0.0990

0.857

8.5

97.0

0.0999

0.923

9.0

99.0

0.09997

0.95

9.9

99.97

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-35

Practice Exams: Exam 2

15.8.2

NAPROXEN SOLUTIONS
TABLE 1-1. Question

Patient Condition

Normal

Numbers For Exam

Probenecid
treatment

Chronic

Rheumatoid

Renal Failure

Elderly

arthritis

1. 500 mg

27. 200

51. 500

75. 500

99. 200

2. 0.95

28. 0.95

52. 0.95

76. 0.95

100. 0.95

fu

3. 0.02

29. 0.02

53. 0.06

77. 0.03

101. 0.04

Vd (L)

4. 10

30. 10

54. 31.67

78. 13

102. 10

k (hr-1)

5. 0.0533

31. 0.0182

55. 0.0495

79. 0.061

103. 0.533

T 1/2 (hr)

6. 13

32. 38.1

56. 14

80. 11.3

104. 13

AUC (mg/L*hr)

7. 891

33. 1044

57. 303

81. 595

105. 356

% Cl h

8. 99

34. 97

58. 99.97

82. 99

106. 99

% Cl r

9. 1

35. 3

59. 0.03

83. 1

107. 1

Cl tot (L/hr)

10. 0.533

36. 0.182

60. 1.57

84. 0.798

108. 0.533

Cl h (L/hr)

11. 0.528

37. 0.177

61. 1.57

85. 0.790

109. 0.528

Cl r (L/hr)

12. 0.005

38. 0.005

62. 0.005

86. 0.008

110. 0.005

Eh

13. 0.005

Er

14. 0.000067

Dose(mg)

Cl h
Cl r

(L/hr)

15. 26.5

39. 8.84

63. 26.5

87. 26.5

111. 13.26

(L/hr)

16. 0.267

40. 0.267

64. 0.00889

88. 0.267

112. 0.133

FR h

17. 1

41. 1

65. 1

89. 1

113. 1

FI h

18. 1

42. 0.333

66. 3

90. 1.5

114. 1

FR r

19. 1

43. 1

67. 1

91. 1

115. 1

FI r

20. 1

44. 1

68. 0.1

92. 1.5

116. 1

FCL

21. 1

45. 0.341

69. 2.94

93. 1.5

117. 1

(hr)

22. 12

46. 12

70. 12

94. 12

118. 8

int

int

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-36

Practice Exams: Exam 2

23. 0.923

47. 0.315

71. 0.857

95. 1.06

119. 0.615

g-
------ mL

24. 2.01

48. 1.94

72. 2.01

96. 2.1

120. 2.2

g-
------ mL

25. 1.5

49. 1.74

73. 1.5

97. 1.5

121. 1.78

g
------ mL

26. 1.06

50. 1.55

74. 1.1

98. 1.01

122. 1.42

N
Cp

ss
max free

Cp

Cp

ss
avg free
ss
min free

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-37

Practice Exams: Exam 2

15.8.3
1. Dose = 500 mg (given)
2. Bioavailability (f) = 0.95 (given)
3. Unbound Fraction = 0.02 (given)
4. V d = 10L (given)

0.693
t1 2

5. k = ------------- = 0.0533hr

6. t 1 2 = 13hr (given)

f Dose
k Vd

7. AUC = ------------------- = 891L


8. %Cl h = 100 %Cl r = 99
9. %Cl r = 1 (given)
10. Cl tot = k V d = 0.533L hr
11. Cl h = Cl tot %Cl h = 0.528L hr
12. Cl r = Cl tot %Cl r = 0.005L hr

Cl

13. E h = -------h- = 0.00528


Qh

Cl

14. E r = --------r = 0.000067


Qr

15. Cl h
int

16. Cl r

int

Q Cl h
-----------------Q Cl
= ------------------h- = 26.5L hr
fu
Q Cl
-----------------rQ Cl
= ------------------r = 0.267L hr
fu

17. FR h = 1 (given)
18. FI h = 1 (given)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-38

Practice Exams: Exam 2

19. FR r = 1 (given)
20. FI r = 1 (given)
21. F Cl = 1 (given)
22. = 12hr (given)

t1 2

23. N = --------- = 0.923

24.

Cp

25.

Cp

ss
fu S f D
g1
= ------------------------- -------------------= 2.0 ------N
mL
V
max free
1
1 ---
2
ss
f u S f D fu S f D
g= -------------------------= ------------------------------ = 1.5 ------VK
V 0.693 N
mL
avg free
N

26.

Cp

ss
min free

1---
2
fu S f D
g= -------------------------- -------------------- = 1.1 ------N
mL
V
1 1---
2

27. Dose = mg
28. Bioavailability (f) = 0.95 (no change)
29. Unbound Fraction = 0.02 (no change - given)
30. V d = 10L (given)

0.693
t1 2

31. k = ------------- = 0.0533hr

32. t 1 2 = 13hr (given)

f Dose
k Vd

33. AUC = ------------------- = 891L


34. %Cl h = 100 %Cl r = 99
35. %Cl r = 1 (given)
36. Cl tot = k V d = 0.533L hr
37. Cl h = Cl tot %Cl h = 0.528L hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-39

Practice Exams: Exam 2

38. Cl r = Cl tot %Cl r = 0.005L hr

39. Cl h

int

40. Cl r
int

Q Cl h
-----------------Q Cl
= ------------------h- = 26.5L hr
fu
Q Cl
-----------------rQ Cl
= ------------------r = 0.267L hr
fu

41. FR h = 1 (given)
42. FI h = 1 (given)
43. FR r = 1 (given)
44. FI r = 1 (given)
45. F Cl = 1 (given)
46. = 12hr (given)

t1 2

47. N = --------- = 0.923

48.

Cp

49.

Cp

ss
fu S f D
1
g= ------------------------- -------------------= 2.0 ------N
V
mL
max free
1 1---
2
ss
f u S f D fu S f D
g= -------------------------= ------------------------------ = 1.5 ------VK
V 0.693 N
mL
avg free
N

50.

Cp

ss
min free

1---
2
fu S f D
g= -------------------------- -------------------- = 1.1 ------N
V
mL
1 1---
2

51. Dose = mg
52. Bioavailability (f) = 0.95 (no change)
53. Unbound Fraction = 0.02 (no change - given)
54. V d = 10L (given)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-40

Practice Exams: Exam 2

0.693
t1 2

55. k = ------------- = 0.0533hr

56. t 1 2 = 13hr (given)

f Dose
k Vd

57. AUC = ------------------- = 891L


58. %Cl h = 100 %Cl r = 99
59. %Cl r = 1 (given)
60. Cl tot = k V d = 0.533L hr
61. Cl h = Cl tot %Cl h = 0.528L hr
62. Cl r = Cl tot %Cl r = 0.005L hr

63. Cl h
int

64. Cl r
int

Q Cl h
-----------------Q Cl
= ------------------h- = 26.5L hr
fu
Q Cl
-----------------rQ Cl
= ------------------r = 0.267L hr
fu

65. FR h = 1 (given)
66. FI h = 1 (given)
67. FR r = 1 (given)
68. FI r = 1 (given)
69. F Cl = 1 (given)
70. = 12hr (given)

71. N = --------- = 0.923

t1 2

72.

Cp

ss
fu S f D
1
g= ------------------------- -------------------= 2.0 ------N
V
mL
max free
1 1---
2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-41

Practice Exams: Exam 2

73.

74.

Cp

Cp

ss
f u S f D fu S f D
g= -------------------------= ------------------------------ = 1.5 ------VK
V 0.693 N
mL
avg free
ss
min free

1--- N
2
fu S f D
g= ------------------------- -------------------- = 1.1 ------N
V
mL
1 1---
2

75. Dose = mg
76. Bioavailability (f) = 0.95 (no change)
77. Unbound Fraction = 0.02 (no change - given)
78. V d = 10L (given)

0.693
t1 2

79. k = ------------- = 0.0533hr

80. t 1 2 = 13hr (given)

f Dose
k Vd

81. AUC = ------------------- = 891L


82. %Cl h = 100 %Cl r = 99
83. %Cl r = 1 (given)
84. Cl tot = k V d = 0.533L hr
85. Cl h = Cl tot %Cl h = 0.528L hr
86. Cl r = Cl tot %Cl r = 0.005L hr

87. Cl h
int

88. Cl r
int

Q Cl h
-----------------Q Cl
= ------------------h- = 26.5L hr
fu
Q Cl
-----------------rQ Cl
= ------------------r = 0.267L hr
fu

89. FR h = 1 (given)
90. FI h = 1 (given)

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-42

Practice Exams: Exam 2

91. FR r = 1 (given)
92. FI r = 1 (given)
93. F Cl = 1 (given)
94. = 12hr (given)

t1 2

95. N = --------- = 0.923

96.

Cp

97.

Cp

ss
fu S f D
g1
= ------------------------- -------------------= 2.0 ------N
mL
V
max free
1
1 ---
2
ss
f u S f D fu S f D
g= -------------------------= ------------------------------ = 1.5 ------VK
V 0.693 N
mL
avg free
N

98. Cp

ss
min free

1---
2
fu S f D
g= -------------------------- -------------------- = 1.1 ------N
mL
V
1 1---
2

99. Dose = mg
100. Bioavailability (f) = 0.95 (no change)
101. Unbound Fraction = 0.02 (no change - given)
102. V d = 10L (given)

0.693
t1 2

103. k = ------------- = 0.0533hr

104. t1 2 = 13hr (given)

f Dose
k Vd

105. AUC = ------------------- = 891L


106. %Cl h = 100 %Cl r = 99
107. %Cl r = 1 (given)
108. Cl tot = k V d = 0.533L hr
109. Cl h = Cl tot %Cl h = 0.528L hr

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-43

Practice Exams: Exam 2

110. Cl r = Cl tot %Cl r = 0.005L hr

111. Cl h

int

112. Cl r
int

Q Cl h
-----------------Q Cl
= ------------------h- = 26.5L hr
fu
Q Cl
-----------------rQ Cl
= ------------------r = 0.267L hr
fu

113. FR h = 1 (given)
114. FI h = 1 (given)
115. FR r = 1 (given)
116. FI r = 1 (given)
117. F Cl = 1 (given)
118. = 12hr (given)

t1 2

119. N = --------- = 0.923

120.

Cp

121.

Cp

ss
fu S f D
1
g= ------------------------- -------------------= 2.0 ------N
V
mL
max free
1 1---
2
ss
fu S f D f u S f D
g= -------------------------= ------------------------------ = 1.5 ------VK
V 0.693 N
mL
avg free
N

122. Cp

ss
min free

1---
2
fu S f D
g= -------------------------- -------------------- = 1.1 ------N
V
mL
1 1---
2

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

15-44

CHAPTER 16

Exam 3

Author:
Reviewer:

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

16-1

Exam 3

16.1 Pharmacokinetics Final Exam


Summer 1996

Fluvoxamine is a selective serotonin reuptake inhibitor (SSRI) with antidepressant properties. After oral
administration, the drug is almost completely absorbed from the gastrointestinal tract, however despite complete
absorption, oral bioavailibiliity in man is approximately 50% on account of first-pass hepatic metabolism. Steady-state
plasma concentrations are achieved within 5 to 10 days after initiation of therapy and are 30 to 50 % higher than those
predicted from single dose data. Fluvoxamine displays non-linear steady state pharmacokinetics over the therapeutic
dose range, with disproportionally higher plasma concentrations with higher dosages. Plasma protein binding of fluvoxamine (77%) is low compared with that of other SSRIs.
V.L. is a 39 year old, 110# female suffering from severe depression. She was admitted to the hospital and prescribed 100mg BID of Fluvoxamine but still her depression was uncontrolled at this dose. Her plasma concentration
on this regimen was 20ug/L. After her physician increased her dose to 300 mg BID her plasma concentration was 500
ug/L. V.L.s depression was controlled at this dose, however she was complaining of adverse effects. The therapeutic
range (total drug ug/L) of fluvoxamine is 20-500. Fluvoxamine is metabolized extensively (93%) by the liver to an
inactive metabolite.
1.

What was her clearance on the 100 mg BID regimen (L/day)?

2.

What was her clearance on the 300 mg BID regimen (L/day)?

3.

What was her V max (mg/day)?

4.

What was her K m (mg/L)?

5.

The doctor would like to change her therapy in order to minimize side effects. What dose would you
recommend tolower her plasma concentration to 300 ug/L?

V.L. has major complications from a combined hepatitis B infection and cirrhosis of the liver. As a result her
protein binding is reduced to 66%, her K m changes to 0.03 mg/L and her V max changes to 100mg/day.

6.

What would be her plasma concentration of total fluvoxamine if she maintained the regimen from
question 5 (mg/L)?

7.

What is her free plasma concentation (mg/L)?

8.

What total fluvoxamine plasma concentration would you recommend achieving to get her free fluvoxamine plasma concentration back to that of the regimen in question 5 (mg/L)?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

16-2

Exam 3

9.

What daily dose of fluvoxamine (by I.V. bolus) would you recommend to get her average free fluvoxamine
plasma concentration approximately back to what it was when she was healthy (mg/day)?

Flurbiprofen is a nonsteroidal anti-inflammatory drug (NSAID) which is a potent inhibitor of prostaglandin


synthesis. It was introduced in the U.S. in 1986 for the treatment of osteoarthritis, rheumatoid arthritis, acute gouty
arthritis, and ankylosing spondylitis. Flurbiprofen is stereoselectively and extensively bound to plasma albumin.
Approximately 99% of the drug is metabolized by the liver, with trace amounts excreted in the urine as unchanged
drug. Flurbiprofen is 80% bioavailable. The recommended dosages for flurbiprofen are 50 mg q 4-6 hr as needed for
analgesia and 100-300 mg/day for the treatment of inflammatory conditions.
The following healthy and sick parameters are given for the patient V.L., 110# suffering from severe arthritis.
Her effective hepatic blood flow is 24 mL/min/kg and effective renal blood flow is 15.0 mL/min/kg. Her healthy halflife is 6 hours. Dr. M. recommends 100 mg flurbiprofen BID.

Vd
% Bound Drug

Healthy

Sick

0.15 L/kg

0.19L/kg

99.0%

97.0%

10.

What is her total body clearance of flurbiprofen (L/hr)?

11.

What is the intrinsic hepatic clearance (L/hr)?

12.

What is the hepatic extraction ratio?

13.

What is the Cp max of total flurbiprofen in mg/L?

14.

What is the Cp min of total flurbiprofen in mg/L?

15.

What is the Cp max of free flurbiprofen in mic/L?

16.

What is the Cp min of free flurbiprofen in mic/L?

17.

V.L. is now suffering from chronic renal insufficiency. Bound flurbiprofen has now decreased to 97%

ss
ss
ss
ss

due to significant uremia. What is her new hepatic clearance (L/hr)?


18.

What is her new k?

19.

What is the new N?

20.

What would her dose be now, if you wanted to maintain approximately the same free plasma concentrations as the previous therapy with the largest tau?

21.

The renal insufficiency clears up and she comes down with mono. Her plasma albumin drops to 50% of
normal thus reducing the bound fraction to 96%. What is her new hepatic clearance using the healthy V d ?

22.

What would her dose be now, if you wanted to maintain approximately the same plasma concentrations as
the previous therapy with the largest tau?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

16-3

Exam 3

23.

The mono clears up, but now there seems to be hepatic stenosis. Her plasma flow is reduced to 50% of
normal. What would her dose be now, if you wanted to maintain approximately the same concentrations
as the previous therapy with the largest tau?

Clentiazem is a chlorinated analog of diltiazem. It is currently undergoing clinical evaluation for the treatment
of angina pectoris and hypertension. The primary mechanism responsible for the antihypertensive effect of CLZ is a
decrease in peripheral vascular resistance due to the blockade of calcium channels. The following information was
obtained from a dose of 20 mg Clentiazem given I.V.

Table 2:
37.52ng mL

A1

2.70hr

16.17ng mL

B1

0.078hr

k 10

0.243hr

k 12

1.67hr

k 21

0.868hr

AUMC

2729.6ng mL hr

1
1

1
1
2

24.

Can this data be adequately evaluated by a one compartment model?

25.

What is the volume of the central compartment (L)?

26.

What is the clearance of clentiazem (L/hr)?

27.

What is the MRT of clentiazem (hr)?

28.

What is the Vd eff of clentiazem (L)?

29.

What is the V for clentiazem (L)?

30.

What percent of the clentiazem dose is in the central compartment at equilibrium?

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

16-4

Exam 3

ss

31.

What is the Cp max , and for a 20 mg IV daily dose clentiazem (ng/mL)?

32.

What is the Cp min for the above dosing regimen (ng/mL)?

33.

What is the Cp avg for the above dosing regimen (ng/mL)?

ss
ss

Any number from the answer pool may be used once, more than once, or not at all.
TABLE 2-1 Answer

Pool

A
B
C
D
E
F
G
H
I
J
A
B
C
D
E
F
G
H
I
J

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

16-5

Basic Pharmacokinetics

REV. 99.4.25

Copyright 1996-1999 Michael C. Makoid All Rights Reserved

http://kiwi.creighton.edu/pkinbook/

-1

You might also like