Biopharmaceutics & Pharmacokinetics (Thakur)
Biopharmaceutics & Pharmacokinetics (Thakur)
Biopharmaceutics & Pharmacokinetics (Thakur)
BIOPHARMACEUTICS &
PHARMACOKINETICS
B.Pharm, Semester-VI
According to the syllabus based on ‘Pharmacy Council of India’
Dr. N. Deattu
M. Pharm, Ph.D
Assistant Professor
College of Pharmacy, Madras Medical College, Chennai
Published by:
Thakur Publication Pvt. Ltd.
H.O.-645B/187, Abhishekpuram, Jankipuram Extension,
Lucknow-226021
Mob.: 9415584997/98, 9235318591/94/95/96/97/22/17/24.
Website: www.tppl.org.in E-mail: thakurpublication@gmail.com
Preface
It gives us immense pleasure to place before the B.Pharm Sixth Semester
pharmacy students the book on “Biopharmaceutics & Pharmacokinetics”.
This book has been written strictly in accordance with the current syllabus
prescribed by Pharmacy Council of India, for B.Pharm students. Keeping in view
the requirements of students and teachers, this book has been written to cover all
the topics in an easy -to-comprehend manner within desired limits of the
prescribed syllabus, and it provides the students fundamentals of
pharmacokinetics of drugs, different pharmacokinetic parameters to be
determined from compartment models, multi -compartment models, and non -
linear pharmacokinetics which are required by them during their pharmaceutical
career.
All efforts have been made to keep the text error -free and to present the subject
in a student friendly and easy to understand. However, any suggestions and
constructive comments would be highly appreciated and incorporated in the
future edition.
Website, www.tppl.org.in
* *
* *
Acknowledgement
It is with great privilege that I acknowledge my sincere gratitude to Dr. R.
Jayanthi, Dean, Madras Medical College, Chennai and Prof. K. Chinnaswamy
for their constant motivation and support. I take this opportunity to express my
heartfelt thanks to Prof. Dr. M. Nagaraj , Dr. S. Manivannan , Prof. Dr. A.
Jerad Suresh , Prof. Dr. N. Narayanan and Dr. Narmada Sambasivam , for
their suggestions and encouragement. I sincerely thank Mr. J. Jayseelan who
has always been an inspiration for me. I convey my gratitude wholeheartedly to
Thakur Publication Pvt . Ltd. for the efforts taken in the publication of this
book.
-Dr. N. Deattu
I wish to thank Thakur Publication for giving me this opportunity for writing
this book. I thank the Management of Mohamed Sathak A.J.College of
Pharmacy, Sholinganallur, Chennai , for providing me suitable working
environment to carry out my work in a smooth and efficient manner.
I would like to thank a lot to Thakur Publication Pvt. Ltd. and their entire team
for their timely assistance and coordination
* *
-5-
Syllabus
Module 01 10Hours
Introduction to Biopharmaceutics
Absorption
Mechanisms of drug absorption through GIT, factors influencing drug
absorption though GIT, absorption of drug from Non per oral extra -
vascular routes.
Distribution
Tissue permeability of drugs, binding of drugs, apparent, volume of drug
distribution, plasma and tissue protein binding of drugs, factors affecting
protein-drug binding. Kinetics of protein binding, Clinical significance of
protein binding of drugs.
Module 02 10Hours
Elimination
Drug metabolism and basic understanding metabolic pathways renal
excretion of drugs, factors affecting renal excretion of drugs, renal
clearance, Non-renal routes of drug excretion of drugs.
Module 04 08 Hours
Multicompartment Models
Two compartment open model. IV bolus.
Kinetics of multiple dosing, steady state drug levels, calculation of
loading and mainetnance doses and their significance in clinical settins.
Module 05 07 Hours
Nonlinear Pharmacokinetics
Introduction.
Factors causing Non-linearity.
Michaelis-menton method of estimating parameters, Explanation with
example of drugs.
* *
-6-
Contents
Chapter 1: Introduction to Chapter 2: Introduction to
Biopharmaceutics and Absorption Distribution
1.1. Biopharmaceutics 9 2.1. Distribution 45
1.1.1. Introduction 9 2.1.1. Introduction 45
1.1.2. Applications of 9 2.1.2. Tissue Permeability of Drugs 46
Biopharmaceutics 2.1.3. Physiological Barriers 48
1.2. Absorption 10 2.1.4. Factors Affecting Drug 51
1.2.1. Introduction 10 Distribution
1.2.2. Mechanisms of Drug 10 2.1.5. Volume of Distribution 52
Absorption Through GIT 2.1.6. Apparent Volume of 52
1.2.2.1. Passive Diffusion 11 Distribution
1.2.2.2. Carrier-Mediated Transport 12 2.1.7. Binding of Drugs 53
1.2.2.3. Facilitated Diffusion 12 2.1.7.1. Plasma Protein Binding of 55
1.2.2.4. Active Transport 13 Drugs
1.2.2.5. Pore Transport 14 2.1.7.2. Binding of Drugs to Blood 56
1.2.2.6. Ionic or Electrochemical 14 Cells
Diffusion 2.1.7.3. Tissue Binding of Drugs 57
1.2.2.7. Ion-Pair Transport 15 2.1.8. Factors Affecting Protein - 59
1.2.2.8. Endocytosis 15 Drug Binding
1.3. Factors Influencing Drug 16 2.1.8.1. Drug-Related Factors 59
Absorption through GIT 2.1.8.2. Protein/Tissue Binding - 59
1.3.1. Introduction 16 Related Factors
1.3.2. Physicochemical Factors 16 2.1.8.3. Drug Interactions 60
1.3.3. Pharmaceutical Factors 26 2.1.8.4. Patient-Related Factors 62
1.4. Absorption of Drugs from 31 2.1.9. Kinetics of Protein Binding 62
Non per Oral Extra - 2.1.10. Clinical Significance of 64
Vascular Routes Protein Binding of Drugs
1.4.1. Introduction 31 2.2. Summary 65
1.4.2. Sublingual/Buccal Route 31 2.3. Exercise 67
1.4.3. Intravenous (IV) Route 32
1.4.4. Intramuscular (IM) Route 33 Chapter 3: Introduction to
1.4.5. Subcutaneous (SC) Route 34 Elimination
1.4.6. Inhalational Route 34 3.1. Drug Metabolism 67
1.4.7. Transdermal Route 35 3.1.1. Introduction 67
1.4.8. Topical Route 35 3.1.2. Organs Involved in Drug 68
1.4.9. Rectal Route 37 Metabolism
1.4.10. Vaginal Route 37 3.1.3. Enzymes Involved in Drug 68
1.4.11. Intraocular Administration 38 Metabolism
1.4.12. Absorption of Drugs from 39 3.1.4. Factors Affecting Drug 68
Common Non per Oral Metabolism
Routes of Drug 3.2. Basic Understanding of 69
Administration Metabolic Pathways
1.5. Summary 40 3.2.1. Introduction 69
1.6. Exercise 43 3.2.2. Phase I Metabolic Pathways 70
3.2.2.1. Hydrolysis Reaction 70
* *
-7-
* *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 9
1.1. BIOPHARMACEUTICS
1.1.1. Introduction
Drugs are used in the diagnosis, cure, mitigation, treatment, or prevention of
diseases. They are given in various dosage forms, such as solids (tablets,
capsules, etc.), semisolids (ointments, creams, etc.), liquids, suspensions,
emulsions, etc., for systemic or local therapeutic activity. Drug products are drug
delivery systems that deliver and release drug at the action site so that they
produce the desired therapeutic effect. These products are also designed to ful fil
the patient’s requirements including palatability, convenience, and safety.
Release of drug substance from the drug product either for local drug action or
for plasma drug absorption for systemic therapeutic action defines the drug
product performance . Advancements in pharmaceutical technology and
manufacturing have led to the development of safer, more effective, and more
patient convenient quality drug products.
1.2. ABSORPTION
1.2.1. Introduction
The desired therapeutic objective can be achieved if the drug product delivers the
active drug at an optimal rate and extent. If a proper biopharmaceutical design is
maintained, the rate and extent of drug absorption (or bioavailability) or the
systemic delivery of drug into the body can be altered from rapid and complete
absorption to slow and sustained absorption; however, this depends on the
desired therapeutic action. The events that occur after a solid dosage form (a
tablet or a capsule) is administered until its absorption in the systemic circulation
are shown in the figure 1.1.
GI lumen GI barrier Blood
Figure 1.1: Sequence of Events in the Absorption of Drugs after Orally Administered
Solid Dosage Forms
These events comprise of the following four steps:
1) Disintegration of the drug product,
2) De-aggregation and release of the drug,
3) Dissolution of the drug in aqueous fluids at the absorption site, and
4) Movement of the dissolved drug throug h the gastrointestinal membrane into
the systemic circulation and away from the absorption site.
A drug molecule gets absorbed from the GIT and enters the systemic circulation
only if it effectively penetrates all the intestinal regions. Once the drug ent ers the
solution, its absorption is governed by the following three important factors:
1) The physicochemical properties of the drug molecule,
2) The properties and components of the GI fluids, and
3) The nature of the absorbing membrane.
drug absorption), the drug molecule diffuses from the GI fluids to enter the
absorbing membrane surface. A drug molecule gets absorbed from the GIT and
enters the systemic circulation only if it effectively penetrates all the intestinal
regions.
The pathways of drug transport show that drug absorption from the GI lumen
into the systemic circulation involves the passage of drug molecules across
several cellular membranes and fluid regions in the mucosa, i.e., the
gastrointestinal-blood barrier. The GIT epithelium lining is the major cellular
barrier to the a bsorption of drugs from the GIT. The drug molecules in GI fluids
should cross the unstirred aqueous layer, mucus layer, and glycocalyx to reach
the apical cell membrane.
Lipid Bilayer
(Cell Membrane)
Intracellular space
Time
Figure 1.2: Passive Diffusion of a Drug
The kinetic energy of drug molecules is responsible for the movement of drug.
Since no energy source is required, the process is called passive diffusion, during
which the drug in the aqueous solution at the absorption site, partitions and
dissolves in the lipid material of the membrane and leaves it by dissolving in an
aqueous medium at the inside of the membrane.
* *
12 Biopharmaceutics & Pharmacokinetics
Where,
dQ/dt = Rate of drug diffusion (amount/time) or the rate of appearance of
drug in blood.
D = Diffusion coefficient of the drug through the membrane (area/time).
A = Surface area of the absorbing membrane for drug diffusion (area).
Km/w = Partition coefficient of the drug between the lipid membrane and the
aqueous GI fluids (no units).
(CGIT – C) = Concentration gradient (amount/volume), i.e., difference in the
concentration of drug in the GI fluids and plasma.
h = Membrane thickness (length).
Carrie
Carrier
r
Dissociation
of complex
B12-IF-carrier Free
complex vitamin B12
Active transport utilises energy, unlike passive transport that does not use any
type of energy. If it uses chemical energy from ATP, it is termed as primary
active transport. If it uses an electrochemical gradient, it is termed as secondary
active transport.
The significance of active transport pro cess in the absorption of nutrients and
drugs is more than the facilitated diffusion, and both the processes differ in the
following aspects:
1) In active transport, drug is transported from a region of lower concentration
to a region of higher concentration, i.e., against the concentration gradient or
uphill transport.
2) Active transport is an uphill process, thus energy is required in the work done
by the carrier.
3) Active transport process requires energy, thus it can be inhibited by
metabolic poisons (like flu orides, cyanides, di -nitrophenol, lack of oxygen,
etc.) that interfere with energy production.
* *
14 Biopharmaceutics & Pharmacokinetics
Carrier
Drug
ATP
surface carries negative charge. Cationic drugs, due to the repulsion between
similarly charged molecules, show electrostatic repulsion on the outer surface of
the membrane. Thus, the molecules with a high kinetic energy can only cross the
membrane barrier. On t he other hand, cationic drugs inside the membrane
undergo significant interaction with the negatively charged intracellular
membrane, and create the electrical gradient, causing electrical diffusion.
Electrochemical diffusion is the function of electrical field as well as the
concentration difference across the membrane, and this process lasts till
equilibrium is achieved.
+ – – +
Drug Endogenous Ion-pair
Drug
ions
Membrane Blood
Figure 1.6: Ion Pair Transport of Cationic Drug
1.2.2.8. Endocytosis
Endocytosis is a minor transport mechanism in which the extracellular materials
are engulfed within a segment of the cell membrane to form a saccule or a
vesicle, which is then pinched -off intracellularly ( figure 1.7). Thus, this process
is also called as corpuscular or vesicular transport.
Outside Membrane Inside
cell cell
Macromolecule
Process of Vesicle
engulfing
Types of Endocytosis
The process of endocytosis is of the following two types:
1) Phagocytosis (Cell Eating): This process involves adsorptive uptake of
solid particulates.
2) Pinocytosis (Cell Drinking ): This process i nvolves uptake of fluid solute;
for example , orally administered Sabin polio vaccine and large protein
molecules are absorbed by pinocytosis.
At times, transcytosis occurs in which an endocytic vesicle is transferred from
one extracellular compartment to another.
Drug Solubility
Dissolution Rate
Wetting
Chemical Forms
Drug pKa and Lipophilicity and GI pH
Drug pKa and Gastrointestinal pH
Lipophilicity and Drug Absorption
Particle Size and Effective Surface Area
of the Drug
Polymorphism and Amorphism
Hydrates/Solvates
1) Drug Solubility: Almost all the factors that influence dissolution rate, also
affect the drug solubility either directly or indirectly. The dissolution rate is
considered to be directly related to drug solubility. An empirical relation
used for predicting the dissolution rate of a drug from its solubility is:
dc
R= = 2.24 Cs Where R = Dissolution rate of the drug.
dt
Bioavailability problems can be avoided if the dr ug exhibits a minimum
aqueous solubility of 1%.
* *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 17
[C]
x Drug [Cs]
Blood
GI Fluid Membrane
Figure 1.8: Dissolution Process of Drug in GIT
* *
18 Biopharmaceutics & Pharmacokinetics
Dissolution rate is the amount of substance that goes into the solution per
unit time under standard conditions of temperature, pressure, and solvent
composition. Dissolution is a dynamic process and involves mass transfer.
This theory assumes that the surface area of the dissolving solute remains
constant during dissolution; however, this is practically not possible.
Hixson and Crowell modified the Noyes -Whitney equation to explain
the effects of change in the surface area:
W01/3 W1/3 = Kt ….. (4)
Where,
W0 = Initial powder weight.
W = Powder weight at time t.
K = Constant which is function of particle density, viscosity,
diffusion coefficient, Cs and x.
Equation (4) is also termed as Hixson and Crowell’s Cubic Root Law.
ii) Danckwert’s Model: This model of dissolution does not consider the
formation of diffusion layer and the solid surface is constantly exposed
to fresh solvent causing mass transfer. It is assumed that the dissolution
medium and the solid surface are under turbulence. T he agitated
dissolution medium comprises of a mass of eddies or pockets, which are
continuously exposed to the solid surface, absorb the solute by diffusion,
and carry it to the bulk of the solution. The contact of fresh pockets with
the solid surface tran sfers the mass to the bulk, and does not allow the
formation of diffusion layer.
* *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 19
: Fresh pocket
: Drug dissolved in
pocket
Figure 1.9: Surface Renewal Process of Dissolution
iii) Limited So lvation Theory: This theory states that an intermediate
concentration less than saturation exist at the interface due to salvation
mechanism, which is the function of solubility than that of the diffusion.
In the dissolution of crystals, the different faces of a crystal have
different interfacial barrier. Limited solvation theory is demonstrated by
the following equation:
G = Ki (Cs – C) ….. (6)
Were,
G = Dissolution rate per unit area.
Ki = Effective interfacial transport constant.
3) Wetting: Good wettability results in particle size reduction. Aggregation of
powder and air adsorption on powder surface is minimised by adding a
wetting agent. Surfactants and hydrophilic polymers have been added in
dosage forms for enhancing drug dissolution and bioavailability.
2.5
Plasma drug concentration
1.0
0.5
0 2 4 6 8 10 12
Time
Figure 1.10: Effect of PEG on Bioavailability of Phenytoin
* *
20 Biopharmaceutics & Pharmacokinetics
iii) Salt Formation for Increasing Absorption: Drugs are mostly weak
acids or bases. The salts of such drugs have different solubility than their
parent drugs. The solubilit y of such drugs can be easily enhanced by
converting them into their salt forms. Some examples are:
Drugs Salts of the Drug
Chloramphenicol Chloramphenicol succinate
Menadiol Menadiol phosphate
Oxazepam Oxazepam semisuccinate
Testosterone Testosterone phosphate
Tetracycline Tetralysine
Theophylline Diprophylline
a) The aluminium salt of aspirin undergoes very slow dissolution in the
GIT due to the deposition of insoluble aluminium on the surface of
solid particles. Due to this reason, the drug is only partially absorbed.
b) Dissolution rate of phenobarbital tablets is better than that of the
tablets of sodium salt of phenobarbital because the former undergoes
rapid disintegration while the latter swell and dissolve slowly from
the surface.
c) Heptabarbital in tablet form attains the peak plasma level (C max) in
1.5-5 hours (t max). However, its sodium salt attains C max at 0.4 -2
hours (t max). Bioavailability of heptabarbital is up to 17% more than
that of its sodium salt due to the precipitation of sodium salt of the
drug in crystalline form.
5) Drug pKa and Lipophilicity and GI pH (pH Partition Theory): Brodie
gave the pH partition theory to explain the absorption process of drug from
GIT and its distribution across the biological membranes. This theory states
that the absorption of drug compounds with molecular weight more than 100
Daltons involves transportation across the biomembrane by passive diffusion,
and this process depends on:
i) The dissociation constant (pKa) of the drug,
ii) The lipid solubility (Ko/w) of unionised drug, and
iii) The pH at absorption site.
* *
22 Biopharmaceutics & Pharmacokinetics
Drugs are mostly either weak acids or weak bases; thus, their ionisation
degree is influenced by the pH of biological fluid. If the pH on either side of
the membrane is different, the compartment whose pH favour s greater
ionisation of drug will have greater amount of drug. Also, the unionised or
un-dissociated fraction of the drug, if is sufficiently lipid -soluble, will
permeate the membrane by passive diffusion until equilibrium is attained
between the concentra tions of unionised drug on either side of the
membrane.
The pH partition theory lies on the following assumptions:
i) GIT acts as a lipoidal barrier to the transport of drug.
ii) The absorption rate of a drug is directly proportional to the fraction of
unionised drug.
iii) Higher the lipophilicity (K o/w) of the unionised drug, better is its
absorption.
10pH pKa
% of drug unionised = 100 – 100 ….. (9)
110pH pKa
Rb = pKa pHPlasma
….. (14)
C Plasma 1 10
* *
24 Biopharmaceutics & Pharmacokinetics
If the pH ranges from 1 to 8 in the GIT, from 1 to 3 in the stomach, and from
5 to 8 in the intestine (duodenum to colon), certain generalisations about
ionisation and absorption of drugs can be made from the pH -partition
hypothesis.
solvates is more than the non -solvates, for example, n-pentanol solvate of
fluorocortisone and succinyl sulfathiazole and the chloroform solvate of
griseofulvin have greater water solubility than their non -solvated forms like
polymorphs. Solvates differ from each other in terms of their physical
properties.
11) Salt Form of the Drug: Drugs are mostly weak acids or bases. The salts of
such drugs have diffe rent solubility than their parent drugs. The solubility of
such drugs can be easily enhanced by converting them into their salt forms.
With weakly acidic drugs, a strong base salt is prepared ( e.g., sodium and
potassium salts of barbiturates and sulphonamides). With weakly basic drugs,
a strong acid salt is prepared ( e.g., hydrochloride or sulphate salts of several
alkaloidal drugs).
v) Compressed Tablets: These are the most common dosage form, but
simultaneously, the most difficult problem is with respect to availability
of a drug for absorption. The main problems arise during the transfer of a
solid drug from a compressed tablet to a solution in the GIT fluid as the
effective surface area of drug is reduced to a greater extent during the
tablet manufacturing process.
In an intact tablet, the sur face area is very limited, i.e., the dissolution
rate is negligible, except for very water -soluble drugs. The absorption of
drug is affected by primary disintegrat ion that ultimately influences the
dissolution process. Tablet fragmentation increases the su rface area in
contact with fluids at the absorption site, resulting in an increased rate of
dissolution. In granular form , the effective surface area of drug is more
than in the intact tablet, however, it is never less than the effective
surface area of the primary drug particles. Compression force also affects
the bioavailability of drug from compressed tablets.
vi) Coated Tablets: Commonly used coating technique for tablets are sugar
coating, film coating, and press coating. Coating forms a physical barrier
between the GI fluid and the tablet (drug). For proper drug dissolution , it
is important that the film of coating dissolves before tablet disintegration.
The disintegration is rate limiting process of drug absorption in coated
tablets, because in vitro disintegration time affects bioavailability.
Film-coated tablets are compressed tablets coated with water -soluble
material as a thin layer or film providing rapid solubility of the film.
Coatings of the tablet have almost negligible effect on the drug
availability as compared to the availability of drug from uncoated core
tablet. Independent of GIT pH, the film coating should dissolve quickly
in the GI fluids.
vii) Enteric-Coated Tablets: An enteric -coat is a special type of film
coating designed to protect the tablet from gastric fluid and to promote or
allow the dissolution of drug in the intes tine. The most important factor
for enteric-coated tablet is the high liability of the coated tablet passing
through the GIT. This problem mainly arises when enteric -coated
products descend to release the drug at some finite time after
administration. Lack of availability of drug can be decreased by the use
of formulations that are based on pH difference between the stomach and
intestine (a realistic assessment of intestina l pH should be provided).
High intra- and inter-subject variability is observed in the enteric-coated
preparation due to the variability in gastric emptying.
2) Manufacturing Processes: Processes of tablet manufacturing include:
i) Granulation Method: Conventional method for preparing granules is
wet granulation process. Limitations of granulation are:
a) The liquid used may cause chemical reactions like hydrolysis.
b) Formation of crystal bridge due to the presence of liquid.
c) Harmful effects on thermo degradable drugs due to the drying
process.
* *
28 Biopharmaceutics & Pharmacokinetics
A C D
B
Rate
Compression Force
Figure 1.11: Effect of Compression Force on Dissolution Rate
On increasing the compression force, particles bind more tightly to each
other (Curve A). High pressures may also break the particles and form
smaller particles (Curve C). Sometime, a sum of an y of these two may
result. So, it can be concluded that the effect of the compression force on
the dissolution rate of a tablet would appear to be unpredictable.
Therefore, a proper study should be performed on each formulation to
ensure better dissolution as well as bioavailability.
3) Pharmaceutical Ingredients/Excipients (Formulation Factors): Many
excipients (non-drug components) are added in a formulation. Excipients are
used to enhance acceptability and physicochemical stability during shelf-life,
uniform composition and dosage, optimum bioavailability and function -
ability drug product. Important excipients are:
i) Vehicles: These are mainly:
a) Aqueous vehicles (e.g., water and syrup),
b) Non-aqueous water miscible vehicles ( e.g., propylene glycol,
glycerol, and sorbitol), and
c) Non-aqueous water immiscible vehicles (e.g., vegetable oils).
Miscibility of vehicles with biological fluids is an important factor for
bioavailability. Aqueous and water mis cible vehicles are miscible with
the body fluids imparting quick absorption of drug.
ii) Diluents (Fillers): Hydrophilic diluents ( e.g., starch, lactose, and
microcrystalline cellulose) are used to enhance the dissolution of poorly
water-soluble, hydrophobic drugs ( e.g., steroids). Hydrophobic diluents
(e.g., dicalcium phosphate) are used in the formulation of tablets.
iii) Binders and Granulating Agents: Poor wettable drugs ( e.g.,
phenacetin) can improve dissolution by imparting hydrophilic properties
to the granule surface through hydrophilic binders. Proportion of stro ng
*
binders during tablet formulation is very critical. If binders are used in *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 29
primary cause of surface activity is the reflux of intestinal contents into the
stomach. Dissolution of drug is also affected by fillers and diluents used in
formulation.
5) Product Age and Storage Conditions: Any changes in physicochemical
properties of a drug in dosage form affect the drug absorption. Changes in
physicochemical properties of drug can result due to ageing and alterations in
storage conditions that adversely affect the b ioavailability of drug. In
solutions, drug is precipitated due to alteration in solubility, conversion of
metastable into poorly soluble, stable polym orph during the shelf -life of the
product. In suspensions, changes in particle size distribution decrease the rate
of drug dissolution and absorption.
Ageing and storage conditions greatly affect the disintegration and
dissolution rates of solid dosage forms, especially in tablets. It occurs due to
the hardening of excipients ( e.g., polyvinyl pyrrolidone, aca cia etc.) on
storage, whereas decrease in these parameters is due to the softening of tablet
binder (e.g., carboxymethyl cellulose) during storage. Alterations that occur
during the shelf -life of a dosage form are mainly affected by variations in
humidity and temperature.
2) Salivary Secretion: The drug should be highly lipid-soluble, and should also
be soluble in aqueous buccal fluids. Biphasic solubility of drug is required
for absorption, because absorption will be delayed if the mouth is dry.
3) Saliva pH: Buccal pH of 6 assists the absorption of unionised drugs.
4) Binding to Oral Mucosa: Drugs that bind to oral mucosa have a poor
systemic availability.
5) Storage Compartment: A storage compartment exists in the buccal mucosa
for the slow absorption of drugs, like buprenorphine.
6) Thickness of Oral Epithelium : Sublingual absorption is faster than buccal
absorption since the epithelium region of the former is thinner and immersed
in a larger volume of saliva.
Bacterial or other microbial infections may occur due to the use of syringe and
contamination at the site of injection. Injected drugs cannot be removed from
circulation through emesis or using activated charcoal like in oral delivery.
Merits of Drug Administration by Intravenous (IV) Route
1) 100% bioavailability is achieved.
2) Desired blood concentrations are achieved.
3) Large quantities.
4) Helpful in emergency situations.
5) No first-pass metabolism occurs.
6) Prevent gastric manipulation.
* *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 33
The following factors determine the absorption rate of drugs from intramuscular
sites:
1) Vascularity of the Injection Site: The blood flow rate to muscular tissues in
which drugs are injected is arranged in its decreasing order as - Arms
(deltoid) > Thighs (vastus lateralis) > Buttocks (gluteus maximus). Blood
flow rate is the rate-limiting step in drug absorption from intramuscular sites.
The most rapid absorption occurs from deltoid muscles and slowest
absorption occurs from gluteal region.
2) Lipid Solubility and Drug Ionisation: Highly lipophilic drugs undergo
rapid absorption by passive diffusion, while the hydrophilic and ionised
drugs are slowly absorbed through the capillary pores.
3) Drug Molecular Size: Small molecules and ions directly enter the capillaries
through pores, while the macromolecules enter the lympha tic system. Small
peptides and fluids undergo cytopemphis, in which they cross the
endothelial tissue of blood capillaries and lymph vessels by getting
transported in small vesicles that cross the membrane.
4) Injection Volume and Drug Concentration: A drug i n concentrated form
and large volume injection undergoes faster absorption in comparison to the
drug given in dilute form and small volume injection.
5) pH, Composition and Viscosity of Injection Vehicle: When a drug solution
in acidic or alkaline pH ( e.g., phenytoin, pH 12) or in a non -aqueous solvent
such as propylene glycol or alcohol ( e.g., digoxin) is injected
intramuscularly, the drug precipitates at the injection site after a slow and
prolonged absorption. Viscous vehicles such as vegetable oils also sl ow
down the absorption of drug.
Merits of Drug Administration by Intramuscular (IM) Route
1) Uniform absorption occurs.
2) Onset of action is fast.
* *
34 Biopharmaceutics & Pharmacokinetics
There are three pathways via which solutes can diffuse through the skin:
1) Transcellular (passive diffusion),
2) Intercellular (paracellular),
3) Transappendageal in which drug diffusion occurs through:
i) Hair follicles,
ii) Sweat glands, and
iii) Sebaceous glands.
v) Age: With the ageing of skin, gross histological changes occur. Aged
skin has hardened blood vessels and thus is more prone to allergic and
irritant effects of topically contacted chemicals. Infants (just like adults)
efficiently absorb drug through skin. Their ratio of surface area to body
weight is 3 times that of adults , hence, systemic toxicity of topically
applied drugs is of particular concern in infants.
vi) Skin Microflora: Skin surface holds a microbial population that
promote the biotransformation of topically applied therapeutic agents.
vii) Skin pH: The surface pH of nor mal human skin is 4 -6. A pH gradient
exists within the skin. Permeation of drugs prone to ionisation at skin pH
can be influenced.
viii) Skin Surface Lipids: Skin surface exhibits sebaceous glands that secrete
a mixture of lipids, which form an irregular film on the skin surface (0.4-
4.0mm thick) that can solubilise drug in it and influence its permeation.
ix) Anatomical Site: Different skin regions have differences in thickness of
stratum corneum, presence of appendages, blood circulation, and overall
skin thickness. All these factors have a direct influence on drug permeation.
2) Composition of Topical Vehicle
i) Vehicle or Base: The vehicle incorporating the drug influences drug
absorption. The vehicle in which the drug is dissolved promotes
absorption and not the one in which the drug is dispersed.
ii) Permeation Enhancers: Incorporation of certain chemicals such as
DMSO, propylene glycol, azone, etc., in topical formulations aid drug
penetration.
3) Application Conditions
i) Rubbing: On rubbing the area of drug application, blood circulation to
that area and thus drug penetration is influenced.
ii) Occlusion: Topical preparations produce an occlusive effect that prevents
moisture loss to the atmosphere from the skin and aqueous delivery vehicles.
This trapped endogenous or exogenous m oisture hydrates the stratum
corneum, makes it swell, and thus influences drug permeation across it.
iii) Loss of Vehicle: Loss of vehicle from the application site or
translocation of the applied dose from treated to untreated sites also
influences transdermal penetration. Evaporation of solvents decrease or
increase the drug solubility in the residual phase and thus the drug’s
thermodynamic activity and permeation, depending on the polarities of
the volatile solvents(s) and the drug.
4) External Factors
i) Environment Humidity and Temperature: Higher humidity and
temperature increase the hydration rate, local blood flow, and thus drug
absorption.
ii) Grooming: Frequency and vigour with which one bathes and the type of
soap one uses also contribute to variability in drug absorption.
* *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 37
drugs for systemic use. Drug administration via vaginal route avoids the gut and
hepatic first pass metabolism, reduces gastrointestinal and hepatic sideeffects, and
enables local targeting of drugs to the reproductive organs.
Drug transport across vaginal membrane takes place in three major ways:
1) Transcellularly via concentration-dependent diffusion through cells,
2) Paracellularly mediated via tight junctions, and
3) Vesicular- or receptor-mediated transport.
Drug absorption from vaginal delivery system takes place in two main steps:
1) Drug dissolution in vaginal lumen, and
2) Membrane penetration.
Human eye can usually hold around 10 l of fluid; hence, instilling concentrated
form of drug solution in small volume increases its effectiveness than when
administered in dilute form in large volume. Viscosity enhancers added in the
formulation prolong t he drug’s contact time with the eye, thus increase
bioavailability. Due to the same reason, oily solutions and ointments show
sustained drug action. Sometimes systemic absorption of a drug having low
therapeutic index (such as timolol) precipitates undesir able toxic effects.
Systemic entry of drugs occurs by the absorption r oute into lachrymal duct that
drains lachrymal fluid into the nasal cavity and GIT. This can be prevented by
simply closing the eyelid or by nasolacrimal occlusion in which the fingertip is
pressed on the inside corner of the eye after drug instillation.
1.5. SUMMARY
The details given in the chapter can be summarised as follows:
1) Biopharmaceuticsis defined as, ―study of the interrelationship of
physicochemical properties of the drug, dosage form in which the rug d is given
and the administration route on the rate and extent of systemic drug absorption.‖
2) Biopharmaceutics is also defined as ―study of the factors influencing the
rate and amount of drug that reaches the systemic circulation and the use of
this information to optimise the therapeutic efficacy of drug products.‖
3) The GIT epithelium lining is the major cellular barrier to the absorption of
drugs from the GIT.
4) Passive diffusion (or non-ionic diffusion) is defined as the difference in the
drug concentrati on on either side of the membrane. Concentration or
electrochemical gradient is the driving force for this process.
5) The kinetic energy of drug molecules is responsible for the movement of drug.
6) Passive diffusion can be expressed by Fick’s first law of di ffusion,
according to which the drug molecules diffuse from a region of higher
concentration to lower concentration until equilibrium is achieved, and the
rate of diffusion is directly proportional to the concentration gradient across
the membrane.
* *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 41
22) Limited solvation theory states that an intermediate concentration less than
saturation exist at the interface due to s olvation mechanism, which is the
function of solubility than that of the diffusion.
23) Good wettability results in particle size reduction.
24) The solubility of drugs can be easily enhanced by converting them into their
salt forms.
25) Brodie gave the pH partition theory to explain the absorption process of
drug from GIT and its distribution across the biological membranes.
26) The dissociation constant (pKa) of drug and the pH of body fluid at the
absorption site influence the amount of unionised drug.
27) Weakly acidic drugs with pKa > 8 are unionised at all pH values and hence
their absorption rate is rapid and independent of gastrointestinal pH.
28) Weakly acidic drugs with pKa ranging from 2.5 to 7.5 undergo pH -
dependent absorption. At acidic conditions of stomach (pH > pKa), they exist
in unionised form and undergo better absorption.
29) Strongly acidic drugs with pKa < 2.5 are ionised in the entire pH range of
GIT and hence are poorly absorbed.
30) Weakly basic drugs with pKa < 5.0 are ionised at all pH values and hence
undergo rapid and pH-independent absorption.
31) Weakly basic drugs with pKa ranging from 5 to 11 undergo pH-dependent
absorption. At alkaline conditions of the intestine, they exist in unionised
form and undergo better absorption.
32) Strongly basic drugs with pKa > 11 remain ionised in the entire pH range
of GIT and hence are poorly absorbed.
33) If a drug exists in unionised form, it will still undergo poor absorption,
provided its lipid solubility is low, i.e., it has a low value of Ko/w.
34) The lipid solubility of a drug can be determined from its oil/water partition
coefficient value (Ko/w).
35) A solid drug’s particle size and surface area are inversely proportional.
Smaller the drug particle, greater is its surface area.
36) Enantiotropic polymorphs can be reversibly changed into another form by
altering the temperature or pressure.
37) Monotropic polymorphs are unstable at all temperatures and pressures.
38) Stoichiometric type of adducts in which the solvent molecules are
incorporated in the crystal lattice of the solid are termed solvates, and the
trapped solvent is termed solvent of crystallisation.
39) Solvates can exist in different crystalline forms, which are known as
pseudopolymorphs, and the phenomenon is termed pseudopolymorphism.
40) If the solvent in association with the drug is water, the solvates are termed
hydrates, which are the most common solvate forms of drugs.
41) Aqueous solubility of the anhydrous form of a drug, i.e., anhydrates, is more
than that of the hydrates.
42) Non per Os or Oral indicates drug administration routes other than the oral
*
route, which bypasses the GIT and enter the systemic circulation. *
Introduction to Biopharmaceutics and Absorption (Chapter 1) 43
1.6. EXERCISE
17) Aqueous solubility of the _______ is more than that of the _______ .
18) Drug transport across vaginal membrane takes place _______ , _______ , and
vesicular- or receptor-mediated transport.
Answers
1) False 2) True 3) True
4) True 5) False 6) False
7) False 8) True 9) False
10) Electrochemical gradient 11) Concentration gradient 12) Pinocytosis
13) Limited solvation theory 14) > 8
15) Oil/water partition coefficient value 16) Drug particle
17) Anhydrates and hydrates 18) Transcellularly and paracellularly
* *
Introduction to Distribution (Chapter 2) 45
CHAPTER Introduction to
2 Distribution
2.1. DISTRIBUTION
2.1.1. Introduction
The processes which lower the plasma drug concentration are termed
disposition. For the disposition of drugs, mainly two processes are involved:
1) Distribution: This process involves the reversible transfer of a drug between
ompartments. Drug distribution is also defined as the reversible transfer of a
drug between one compartment and another.
2) Elimination: This process involves irreversible loss of drug from the body.
Elimination is further divided into two processes, namely biotransformation
(metabolism) and excretion.
Tissue receptor or
site of action
Excretion
Distribution is a passive transport process and the driving fo rce for this
process is obtained from the difference of concentration gradient between the
blood and extravascular tissues. By the diffusion process, the drug
concentration increases in tissues until it reaches equilibrium where the
amount of drug entering the tissues becomes equal to the amount of drug
draining out from the tissues. At equilibrium state, the drug concentration in
the tissues depends on the rate at which the drug is distributed in the tissues
and on the drug concentration in the plasma.
Plasma concentration Tissue concentration
* *
46 Biopharmaceutics & Pharmacokinetics
Drug distribution does not occur uniformly throughout the body because different
tissues get the drug from the plasma at different rates and in different
concentrations.
Unionised drug
(low Ka and large Ko/w)
Ionised drug + +
(large Ka and low Ko/w) Pore
In case of polar drugs in which permeability is the rate -limiting step in the ir
distribution, effective partition coefficient of drug is the driving force and it
is calculated as follows:
Fraction unionised K o/w of
Effective K o/w …..(1)
at pH 7.4 unioniseddrug
The degree up to which effective partitio n coefficient influences rapid ity of
drug distribution can be explained by the following example (table 2.1):
Table 2.1: Distribution of Acidic Drugs in CSF
Drugs Relative Acidity Effective Ko/w at Relative Rate of
pH 7.4 Distribution
Thiopental Weaker acid 2.0 80
Salicylic acid Stronger acid 0.0005 1
Where,
D = Drug diffusion coefficient in the membrane.
K = Lipid/water partition coefficient.
A = Membrane surface area.
Cp = Plasma drug concentration.
Ct = Tissue drug concentration.
h = Membrane thickness.
The negative sign indicates drug movement from inside the blood capillary
into the tissues.
2) Simple Capillary Endothelial Barrier : Capillaries supply blood to most of
the tissues. Capillary endothelium allows passage of drugs (ionised or
unionised of molecular size ˂ 600 Daltons) int o the interstitial fluid. Only
drugs bound to the blood components are restricted because of the large
molecular size of the complex.
3) Cell Membrane : Drug present in ECF is transported by passive diffusion
into the cell. Factors influencing the penetration of drugs into cells are same
as those observed in the gastrointestinal absorption of drugs.
For the transport of drugs, cell membrane acts like a lipid barrier.
Permeability of drugs through cell membrane chec ks the entry into the
cell.
The physicochemical properties that influence permeation of drugs across
such a barrier are illustrated in figure 2.3.
* *
Introduction to Distribution (Chapter 2) 49
Brain
ECF of brain
Glial cells
Basement
Tight junctions membrane
Lipid-soluble drug
Blood Capillary
endothelium
Figure 2.4: Transport of Lipid-Soluble Drug across BBB
Normally, only lipid-soluble drugs can penetrate the interstitial fluid of the brain
and spinal cord, while water-soluble compounds need specific carriers to cross
the endothelial lining. In diffusion process , many transport mechanisms are
involved. Degree of ionisation in plasma and drug lipid solubility determines
the penetration rate of a drug into the brain. Highly lipid-soluble drugs (e.g.,
thiopental) cross BBB immediately, and reach the brain from plasma. Polar
drugs ( e.g., barbital) penetrate the CNS slowly. Weak organic acid s (e.g.,
penicillin G having pKa 2.6) are found in completely ionised form in plasma,
but penetrate the brain at a low rate due to poor lipid solubility.
Approaches to Promote Crossing the BBB
i) Permeation enhancers ( e.g., Dimethyl Sulfoxide or DMSO) are used for
*
increasing penetration. *
50 Biopharmaceutics & Pharmacokinetics
ECF
Capillary
endothelium
Open
junctions Lipid-soluble drug
Drugs may give rise to lethal effects on the following two critical stages
during foetal development:
i) First Trimester: It is the duration when the foetal organs are
developing. At this stage , most of the drugs produce teratogenic effects
(congenital defects), e.g., thalidomide, phenytoin, isotretinoin, etc.
ii) Latter Stages: In the l atter stages of pregnancy, drugs may affect the
physiological functions of body, e.g., respiratory depression by
morphine.
Therefore, it is better to avoid using any drug during the pregnancy period
due to the uncertainty of harmful effects.
Foetal vein
Foetal artery
Foetal membrane
Drug movement (trophoblast + endothelium)
Maternal artery
Maternal vein
3) Obesity: High adipose ti ssue content results in low drug distribution and
perfusion. High fatty acid content alters the binding property of acidic drugs.
4) Diet: Fat-rich diet increases free fatty acid concentr ation in the blood that
affects the binding of acidic drugs, e.g., NSAIDs, albumin, etc.
5) Disease States: Drug distribution is severely affected in diseased conditions:
i) Alteration in albumin and other drug-protein concentration,
ii) Reduced or altered perfusion to organs and tissues, and
iii) Alteration in tissue pH.
In case of enceph alitis and meningitis, the BBB becomes more permeable,
therefore, concentration of ionic antibiotics (e.g., penicillin G and ampicillin)
increases in brain tissues.
6) Drug Interaction: Two or more drugs administered together compete for the
binding site, tend to replace each other, and the free drug may produce lethal
effects, e.g., phenylbutazone and warfarin.
concentration ranges between 0.04 -0.1g%. Many basic drugs bind to it, e.g.,
propranolol, quinidine, disopyramide, imipramine, amitriptyline,nortriptyline,
and lidocaine.
3) Binding of Drugs to Lipoproteins: Depending on the chemical
composition, the molecular weight of lipoproteins varies between 0.2 to 3.4
million. On the basis of their density, lipoproteins are classified into:
i) Chylomicrons,
ii) Very Low Density Lipoproteins (VLDL),
iii) Low Density Lipoproteins (LDL, predominant in humans), and
iv) High Density Lipoproteins (HDL, most dense and smallest in size).
A drug binds to lipoproteins by dissolving the lipid core of the lipoproteins. The
binding capacity of drug depends on the lipid content of their lipid core. This
process of binding of drugs to lipoproteins is non-competitive.Many acidic (e.g.,
diclofenac), neutral e( .g., cyclosporine A) and basic drugs e.g.,
( chlorpromazine)
bind to lipoproteins, of which the basic lipophilic drugs have maximum affinity.
4) Binding of Drugs to Globulins: Many plasma globulins are recognised and
are labelled asfollows:
i) α1-Globulin or Transcortin or α1r CBG (Corticosteroid Binding
Globulin): It binds to number of steroidal drugs, e.g., cortisone,
prednisone, thyroxin, and cyanocobalamin.
ii) α2-Globulin or Ceruloplasmin: It binds to vitamins A D, E and K and
cupric ions.
iii) β1-Globulin or Transferrin: It binds to ferrous ions.
iv) β2-Globulin: It binds to carotenoids.
v) γ-Globulin: It binds to antigens.
dissolved in their lipid core. The presence of active anionic and cationic
groups on the albumin molecules to bind a variety of drugs depends on the
physiological pH (pH of blood, plasma, ECF, etc.).
2) Concentration of Protein or Binding Agent: Among the plasma proteins,
binding mainly occurs with albu min because its concentration is high er than
other plasma proteins. During diseased states, the amount of several protei ns
and tissue components available for binding gets changed.
3) Number of Binding Sites on Binding Agent: Albumin has numerous
binding sites as well as high capacity binding component as compared to
other proteins. Many drugs are capable of binding at more th an one site on
albumin. For example, flucloxacillin, flur biprofen, ketoprofen, tamoxifen,
and dicoumarol bind to both primary and secondary sites on albumin.
more available for elimination by the liver and kidneys ( figure 2.9). If drug
gets metabolised or excreted easily, its displacement causes a significant
reduction in elimination half-life.
Enhanced
Site of action
pharmacologic
Receptor response
Blood
Tissue
Increase in free drug
Storage tissue redistribution
concentration
P-D D
Hepatic
(Displaced drug) Liver clearance
D1 (Displacer)
Renal
Kidney clearance
Displacement is almost insignificant if more selec tive, potent, and low dose
drugs are used.
2) Competition between Drugs and Normal Body Constituents: Among the
normal body constituents, the free fatty acids are the major ones that interact
with a number of drugs that bind primarily to HSA. The level of free fatty
acid increases in several physiologic al (fasting), pathologic al (diabetes,
myocardial infarction, and alcohol abstinence) and pharmacologically
induced conditions (after heparin and caffeine administration).
The fatty acids binding to albumin influence the binding of several
benzodiazepines and propranolol (decreased binding) and warfarin (increased
binding). Binding of b ilirubin to HSA can be compromised by some drugs,
and this is important for neonates who lack efficient BBB and bilirubin
metabolising capacity.
Acidic drugs ( e.g., sodium salicylate, sodium benzoate , and sulphonamides)
have the ability to displace bilirubin from its albumin binding site. T he free
bilirubin is not conjugated by the liver of neonates, and thus crosses the BBB
and precipitates the condition of kernicterus (degeneration of brain and
mental retardation).
3) Allosteric Changes in Protein Molecule: This mechanism is also used to
explain that how the drugs affect protein binding interactions. This process
includes alteration of protein structure by the drug or its metabolite to modify
its binding capacity. The agents producing such an effect are known as
allosteric effectors, e.g., aspirin acetylates the lysine fraction of albumin and
changes its capacity to bind NSAIDs , like phenylbutazone (increased
*
affinity) and flufenamic acid (decreased affinity). *
Introduction to Distribution (Chapter 2) 61
1 [D]
r r
1 1
Slope Slope
NKa N
1 1
N NK
a
1 [D
[D] ] Plot
(d) Hitchcock
(c) Klotz Plot
Figure 2.10: Plots Used for the Study of Protein-Drug Binding. (a) Direct
Plot, (b) Scatchard Plot, (c) Klotz Plot, and (d) Hitchcock Plot
The value of association constant (Ka) and the number of binding sites (N) can be
obtained by plotting equation (10) in four different ways (figure 2.10):
1) Direct Plot: It is made by plot ting r against (D) [ figure 2.10 (a)]. When all
the binding sites are occupied by the drug, the protein is saturated and
plateau is reached. At the plateau, r = N. When r = N/2, [D] = 1/Ka.
2) Scatchard Plot: It is made by transforming equation (10) into a linear form.
* *
Introduction to Distribution (Chapter 2) 63
NK a [D]
Thus, r …..(11)
K a [ D] 1
r + rKa [D] = NKa [D]
r = NKa [D] – rKa [D]
Therefore,
r
NK a rKa …..(12)
[D]
A plot of r/[D] versus r yields a straight line [figure 2.10 (b)] with slope as –
Ka, y-intercept as NKa, and x-intercept as N.
3) Klotz Plot/Lineweaver -Burke Plot (Double Reciprocal Plot): The
reciprocal of equation (10) yields:
1 1 1
…..(13)
r NK a [D] N
A plot of 1/r versus 1/[D] yields a straight line with slope as 1/NK a and y -
intercept as 1/N [figure 2.10 (c)].
4) Hitchcock Plot: It is made by rewriting equation (13) as:
NKa [D]
1 K a [D] …..(14)
r
On dividing both sides by NKa:
[D] 1 [D]
…..(15)
r NK a N
Equation (15 ) is Hitchcock equation as per which a plot of [D]/r versus [D]
yields a straight line with slope 1/N and y-intercept 1/NKa [figure 2.10 (d)].
5) Bacterial infections generally affect the organ cells. The efficiency of a drug
against a given pathogen can be evaluated by the drug’s intrinsic antibacterial
activity and its concentration in the extracellular spaces of the tissues, i.e.,
the site of infections. An antibiotic which is found in greater concentration in
the blood show extensive binding in its compartment and is found in low
concentration at the site of infection.
Another antibiotic with same potency, unbound to plasma proteins , and free
to be distributed may be present in high concentration at the site of infection.
This antibiotic is more clinically effective , despite the fact that it would
produce much lower blood levels than the first antibiotic.
6) The protein binding may also stimulate the probability of competitio n for the
binding sites on a protein molecule, and the amount of bound drug might be
decreased by another drug bound to plasma proteins. Protein binding may
influence the activity, distribution, and elimination of a drug. This
competitive phenomenon may have important clinical effects.
When two drugs are simultaneously given, displacement of drugs from
proteins occurs and increases the rate of biotransformation and elimination.
Due to this property, the compounds that are effective displacers may be used
potentially in case of drug intoxication to decrease the body drug content.
7) Another significant source of variation of free drug concentration in plasma
is the competitive binding between drugs and endogenous substances.
8) In diseased states, the electrolytic balance in the blood is changed, which
further changes the binding of drugs since the activity coefficient of drugs
also change. Some alterations are also observed in the 3D structures of
proteins when the electrolytic balance is disturbed. The effect of a ge on the
binding of drugs is also an important factor because the plasma volume and
its composition vary with age.
9) Physicochemical properties of drugs are also an important factor which plays
a significant role in the binding of drugs to blood components.
For example, tetracycline analogues show a correlation between their
physical properties and disposition characteristi cs. When the drug molecules
become more lipid-soluble, their interaction with proteins increases and their
elimination from the body decreases.
2.2. SUMMARY
The details given in the chapter can be summarised as follows:
1) The processes which lower the plasma drug concentration are termed
disposition.
2) Distribution involves the reversible transfer of a drug between compartme nts.
3) Drug distribution is also defined as the reversible transfer of a drug between
one compartment and another.
4) Elimination involves irreversible loss of drug from the body.
5) Distribution is a passive transport process and the driving force for this
process is obtained from the difference of concentration gradient between
the blood and extravascular tissues.
* *
Introduction to Distribution (Chapter 2) 65
36) A decrease in plasma protein -drug binding, i.e., an increase in unbound drug
concentration, favours tissue redistribution and/or clearance of drugs from
the body (enhanced biotransformation and excretion).
37) An increase in concentration of free or unbound drug increases the intensity
of action (therapeutic/toxic).
2.3. EXERCISE
2.3.1. True or False
1) The processes which lower the plasma drug concentration are termed elimination.
2) Disposition involves reversible transfer of a drug between compartments.
3) Drugs that selectively bind to extravascular tissues have a pparent volume of distribution
larger than their true volume of distribution.
4) Site-I binding site of HSA is known as digitoxin binding site.
5) α2-Globulin binds to vitamins A D, E and K and cupric ions.
6) Basic drugs like imipramine, chlorpromazine and antihi stamines accumulate in the hair
shafts.
7) Metallothionein binds to heavy metals and causes their hepatic accumulation and thus
toxicity.
Answers
1) False 2) False 3) True
4) False 5) True 6) False
7) False 8) Degree of ionisation 9) ml/min/ml of the tissue
10) Tamoxifen binding site 11) Carotenoids 12) γ-Globulin
13) Skin 14) Protein binding
CHAPTER Introduction to
3 Elimination
3.1.1. Introduction
Elimination is the major process for the removal of a drug from the body and the
termination of its action. It is defined as the irreversible loss of drug from the
body. Elimination occurs by t wo processes, i.e., metabolism and excretion. The
body metabolises some drugs chemically. Metabolism may either result in the
formation of inactive substances (metabolites) or substances that may resemble to
the original drug in terms of therapeutic activi ty or toxicity or substances that
may differ from the original drug.
Liver is the main site of metabolism for most drugs, and hence, almost every
drug passes through the liver. For the conversion of prodrug to active metabolites
or for the conversion of active drugs to inactive forms, the drugs should enter the
liver to be acted upon by many enzymes. A specific group of cytochrome P-450
enzymes involves in liver’s primary mechanism for metabolising drugs. The
metabolism rate of many drugs is controlled by the level of these cytochrome P -
450 enzymes. Since the enzymes possess a limited capacity of metabolism, they
are burdened, when the levels of drug present in the blood are high.
* *
Introduction to Elimination (Chapter 3) 71
2) Amides
3) Thioesters
5) Acid Anhydride
* *
72 Biopharmaceutics & Pharmacokinetics
9) N-Hydroxylation
* *
Introduction to Elimination (Chapter 3) 73
During azo reduction, N=N undergoes sequential reduction and gets cleaved
into two primary amines. Four reducing equivalents are required for this
reaction; whereas, six reducing equivalents are required for nitro reduction.
2) Carbonyl Reduction: This reaction is catalysed by carbonyl reductases
present in blood and cytosolic fraction of the liver, kidney, brain, and other
tissues. Reduction of aldehydes i nto primary alcohols and reduction of
ketones into secondary alcohols are examples of carbonyl reduction.
* *
74 Biopharmaceutics & Pharmacokinetics
Reductase
[Fe++]
DR—[Fe++] O2 DR—[Fe++]
(reduced material)
Oxidised
DR+H2O
O2
Figure 3.2: Oxidation by MFO System
* *
76 Biopharmaceutics & Pharmacokinetics
Oxidation Reactions
1) Oxidation of Aromatic Carbon Atoms (Aromatic Hyd roxylation): In this
reaction, arene oxide (epoxide) is formed. It is a reactive intermediate that
undergoes rearrangement to yield arenols, and sometimes catechols and
glutathione conjugates.
* *
78 Biopharmaceutics & Pharmacokinetics
* *
80 Biopharmaceutics & Pharmacokinetics
The N-oxide products are highly water -soluble and get excreted in urine.
But, they are prone to get reduced into the corresponding amine.
iv) N-Hydroxylation: Opposing the basic compounds that form N -oxides,
N-hydroxy formation is displayed by non -basic nitrogen atoms such as
amide nitrogen, e.g., lidocaine.
* *
Introduction to Elimination (Chapter 3) 81
Methyl ethers get rapidly dealkylated than the longer chain ethers, such as
the ones containing n -butyl group. The reaction results in active metabolites,
e.g., conversion of phenacetin to paracetamol and codeine to morphine.
3.2.3.1. Glucuronidation
Glucuronidation is quantitatively the most significant phase II reaction.
Glucuronic acid conjugation occurs only when glucuronic acid gets activated.
The cofactor, Uridine Diphosphate -glucuronic acid (UDP -glucuronic acid) is
required during glucuro nidation, which is also catalysed by UDP -Glucuronyl
Transferases (UGTs, located in the endoplasmic reticulum of liver, kidney,
intestine, skin, brain, spleen, and nasal mucosa) (figure 3.3).
* *
Introduction to Elimination (Chapter 3) 83
UDP-glucuronic acid
3) S-Glucuronides
3.2.3.2. Sulphation
Sulphate conjugation forms a highly water -soluble sulphuric acid ester. This
reaction is catalysed by sulphotransferases (sulphokinases), which are cytosolic
enzymes found in liver, kidney, intestinal tract, lung, platelets, and brain. The
cofactor required for sulphation is 3 '-Phosphoadenosine-5'-Phosphosulphate
(PAPS). Many drugs undergoing O -glucuronidation also undergo sulphation. In
sulphation reaction, just as in glucuronidation, the sulphate is activated before the
reaction with substrate.
3.2.3.3. Acetylation
N-acetylation is a major biotransformation route for drugs with an aromatic
amine or a hydrazine group. This reaction requires acetyl coenzyme A (acetyl
CoA) cofactor and is catalysed by N-acetyltransferases.
* *
Introduction to Elimination (Chapter 3) 85
3.2.3.4. Methylation
The metabolites resulting from methylation are not more polar or water-soluble.
They possess equal or higher pharmacological activity than the parent drug. This
reaction is catalysed by methyl transferase.
Displacement Reaction
Addition Reaction
Conjugation of Heteroatom
Electrophiles are very toxic as they have the ability of binding to macromolecules
(proteins and DNA) and causing cellular damage and mutations. Hence,
conjugation of electrophiles with glutathione is an important detoxification
reaction.
Glutathione conjugates are either eliminated in bile or first they are converted to
mercapturic acid in the kidney and then excreted in urine.
* *
Introduction to Elimination (Chapter 3) 87
3.3.1. Introduction
Excretion removes drugs and/or their metabolites from the body. For cessation of
the pharmacological action of a drug, its excretion, in intact or unchanged form is
important. Kid neys are principal organs for excretion; and excretion through
kidneys is termed as renal excretion. Non -renal excretion is the term used to
describe excretion by other organs except the kid neys. The other organs include
lungs, biliary system, intestine, salivary glands, and sweat glands.
All lipid -soluble drugs are converted into water -soluble compounds by the
metabolic processes. This conversion enables their excretion (from the body). In
case, the drug or its metabolites are water -soluble, it is excreted unchanged.
Therefore, it can be said that for a drug to be eliminated from the body, both
metabolism and excretion are important.
Given below are some drawbacks and precautions when c alculating Cl cr from
serum creatinine:
1) Liver dysfunction is associated with a significant over -prediction of Cl cr.
These equations should be used with caution in patients with liver disease.
2) Thin individuals have low serum creatinine concentrations secon dary to
decreased muscle mass, resulting in a significant over-prediction of Clcr.
3) Elderly patients have low serum creatinine concentrations secondary to
decreased muscle mass, leading to a possible over-prediction of Clcr.
Collecting Urine
Tubule
The secretion of bile is a capacity -limited process and gets saturated, since it
is an active process. The process of bile secretion is similar to that of active
renal secretion. The secretion of organic anions, cations, and neutral polar
compounds are linked to the existence of varying transpo rt mechanisms for
the same. If a drug concentration in bile pigments is less than drug
concentration in plasma, the drug is said to have a small biliary clearance and
vice versa. The ratio of concentration of bile to the concentration of plasma
in some cases can give a value approximating 1000. The biliary clearance in
such cases can be 500ml/min or more in value.
Based on their bile/plasma concentration ratios, the compounds excreted in
bile can be categorised into the following three groups:
i) Group A: Compounds with bile/plasma concentration ratio of about 1
are categorised in Group A, e.g., sodium, potassium and chloride ions
and glucose.
ii) Group B: Compounds with bile/plasma concentration ratio of more than 1
(generally ranging from 10 -1000) are categorise d in Group B, e.g., bile
salts, bilirubin glucuronide, creatinine, sulfobromophthalein conju
gates, etc.
iii) Group C: Compounds with bile/plasma concentration ratio of less than 1
are categorised in Group C, e.g., sucrose, inulin, phosphates,
phospholipids, and mucoproteins.
2) Pulmonary Excretion: The process of simple diffusion through lungs aids
in the absorption of gaseous and volatile substances (like general
anaesthetics, e.g., halothane). In the same way, diffusion can also help in
their excretion (they can be excreted into the expired air by diffusion).
Pulmonary blood flow, rate of respiration, solubility of the volatile substance,
etc., constitute the factors that influence the pulmonary excretion of a drug.
Those gaseous anaesthetics that are not very soluble in blood, e.g., nitrous
oxide, show rapid excretion. It is usually seen that gaseous drugs which are
intact undergo excretion; however, their metabolites do not undergo
excretion. Lungs are the organ of excretion for those compounds that are
highly soluble in blood and tissues like alcohol.
3) Salivary Excretion: It involves excretion of drugs through saliva by the
process of passive diffusion. The pH-partition hypothesis forms the basis for
the prediction of salivary excretion of drugs. Though salivary pH ranges from
5.8-8.4, its mean value in human is 6.4. Drugs that exist in unionised forms and
that are soluble in lipids, at the mean pH undergo passive salivary excretion.
For weak acids,
S 1 10( pHsalivapKa ) f plasma
Ra ….. (9)
P 1 10( pHplasmapKa ) f saliva
Where,
fplasma and fsaliva = Free drug fractions in plasma and in saliva, respectively.
For weak acids, the S/P ratios are found to be < 1, while for weak bases they
are found to be > 1. This means that basic drugs are excreted i n saliva in
more amounts as compared to drugs that are acidic in nature. Some drugs
show a high (as high as 0.1%) salivary concentration value. Several drugs
show a fairly constant S/P ratio. Therefore, by detecting their quantity in
salivary excretion, th eir concentration in blood can be determined, e.g.,
caffeine, theophylline, phenytoin, carbamazepine, etc. Some drugs ( e.g.,
lithium) are secreted in saliva actively, and sometimes, their concentration in
saliva is found to be 2 -3 times more than that in t he plasma. Saliva also
actively secretes penicillin and phenytoin.
Salivary
Oral glands
administration
Distribution and
GIT elimination
Faecal Blood
excretion
Figure 3.5: Salivary Cycling of Drugs
The unpleasant after taste in the mouth of a patient, who is on drug treatment,
indicates salivary excretion of the drug. A few cases have been reported
where this process resu lts in adverse effects like black hairy tongue in
patients on antibiotic therapy, gingival hyperplasia in patients on phenytoin,
etc. Salivary secretion is inhibited by some basic drugs, and hence results in
dryness of mouth. A cycling process ( figure 3.5), resembling enterohepatic
cycling can be seen in drugs undergoing salivary excretion, e.g.,
sulphonamides, antibiotics, clonidine, etc.
4) Mammary Excretion: A drug that is excreted in milk can enter in infants
feeding on breast, and therefore, it is significant. Milk contains lactic secretions
synthesised in the extracellular fluid. It also contains rich amounts of fats and
proteins. Lactating mothers secrete nearly 0.5-1 litres of milk per day.
Drug excretion in milk is not a passive process. It depends on:
i) pH-partition behaviour,
ii) Molecular weight,
iii) Lipid solubility, and
iv) Degree of ionisation.
The pH of milk ranges from 6.4 -7.6 and possesses a mean pH of 7.0. Free,
unionised, lipid-soluble drugs show passive diffusion in the alveolar cells of
the mammary gland. The ratio of concentration of drug in Milk to that in
Plasma (M/P) determines the amount of drug excreted in milk. Since milk
bears an acidic nature as compared to plasma, drugs that are weakly basic in
* *
96 Biopharmaceutics & Pharmacokinetics
nature show a greater concentration in milk , just like in case of saliva, and
they possess an M/P ratio of more than 1. For drugs weakly acidic in nature,
the reverse is true. Studies have revealed that the excretion of acidic drugs in
milk is inversely correlated with its molecular weight and par tition
coefficient; while in basic drugs, it is inversely correlated to the degree of
ionisation and partition coefficient.
Drugs that show extensive binding with plasma proteins ( e.g., diazepam) are
secreted in milk in lesser amounts. Drugs that undergo excretion via milk can
bind to the proteins that are present in milk. Excretion of drugs in milk is
usually in amounts less than 1% and the portion consumed by infants are in
amounts that are too less to accomplish either beneficial or toxic levels.
However, certain powerful drugs like barbiturates, morphine, and ergotamine
may induce toxic effects in infants.Interaction of bilirubin with sulphonamides
resulting in jaundice and discoloration of teeth with tetracycline are some
instances showing that excretion of a drug in milk may result in adverse effects.
5) Skin Excretion: The pH-partition hypothesis also regulates the excretion of
drugs through skin via sweat. Drugs and their metabolites that are passively
excreted through the skin are accountable to some extent for urticaria and
dermatitis in addition to other hypersensitivity reactions. Sweat shows the
excretion of compounds like benzoic acid, salicylic acid, alcohol and
antipyrine along with heavy metals such as lead, mercury, and arsenic.
6) Gastrointestinal Excretion: Generally, drugs are seen to be excreted in the
GIT when they have been administered through parenteral route and their
concentration gradient favours passive diffusion. The process of GI excretion
of drugs is opposite to that of GI absorpti on. In GIT, stomach is a specific
site that shows the excretion of water-soluble and ionised forms of drugs that
are either weak acids or bases, e.g., nicotine and quinine. GIT also shows the
absorption and excretion of drugs that are administered orally. Excretions of
drugs in GIT are followed by their re -absorption (into the systemic
circulation) and hence are recycled.
7) Genital Excretion: Drugs may also be excreted in the reproductive tract and
genital secretions. Semen has shown the presence of some drugs.
3.4. SUMMARY
The details given in the chapter can be summarised as follows:
1) Elimination is defined as the irreversible loss of drug from the body.
2) Liver is the main site of metabolism for most drugs.
3) A specific group of cytochrome P-450 enzymes involves in liver’s primary
mechanism for metabolising drugs.
4) The biochemical modification of a drug in the body is termed drug
metabolism or biotransformation.
5) Young children show poor drug metabolism as metabolic enzyme systems
are not developed completely.
6) In comparison to males, the females possess lesser ability for drug metabolism.
* *
Introduction to Elimination (Chapter 3) 97
3.5. EXERCISE
3.5.1. True or False
1) Liver is the main site of metabolism for most drugs.
2) Temperature of the body is inversely proportional to drug metabolism.
3) Drugs containing an aldehyde, ketone, disulphide, sulphoxide, quinine, N -oxide,
alkene, azo or nitro group undergo hydrolysis.
4) S-methylation is catalysed by cytochrome P450 and glutathione-S-reductase.
5) Cytochrome oxidase enzyme, commonly known as Cytochrome P-450 (CYP-450) and
is chemically a protein.
6) Desulphuration reaction involves cleavage of carbon -sulphur bond and formation of
*
a product with C=O bond. *
Introduction to Elimination (Chapter 3) 99
Answers
1) True 2) False 3) False
4) False 5) False 6) True
7) False 8) True 9) True
10) False 11) Biotransformation 12) Carbonyl reductases
13) Liver cells 14) Mixed function oxidase 15) UDP-Glucuronyl Transferases
16) 120-130ml/min 17) Glomerular filtration rate 18) Urinary excretion
19) Inversely 20) Excretion
4.1. BIOAVAILABILITY
Where, (AUC T)test and (AUC T)standard = Total areas under the plasma
concentration-time curves after administering a single dose of the test dosage
form and of the standard dosage form, respectively.
If test and standard dosage forms are administe red in different doses, the dose
size can be corrected as follows:
(AUCT ) abs /D test
Relative
Re lative bioavailab ility
bioavailability ….. (6)
AUCT s tan dard /D s tan dard
Where, D test and D standard = Sizes of the single doses of test and standard dosage
forms, respectively.
Relative bioavailability (alike absolute bioa vailability) can be expressed as a
fraction or a percentage, and can also be determined from urinary excretion data
as follows:
(X u ) test
Relative bioavailability
bioavailability
Re lative ….. (7)
X u s tan dard
Where, (X u)test and (X u)standard = Total cumulative amounts of unchanged drug
excreted in the urine after administering single doses of the test dosage form and
standard dosage form, respectively.
If the test and standard dosage forms are administered in different doses at
different times, the total amount of unchanged drug excreted in th e urine per unit
dose of drug should be used in this equation.
Relative bioavailability is measured to determine the effects of dosage form
differences on systemic bioavailability of a drug. Many dosage form factors can
influence the drug bioavailability , like the type of dosage form ( e.g., tablet,
solution, suspension, and hard gelatin capsule), differences in the formulation of
a particular type of dosage form, and manufacturing variables employed in the
production of a particular type of dosage form.
Given below are the three parameters of plasma level -time studies that are
significant in the determination of bioavailability:
1) Cmax: It is the peak that indicates the point at which the drug concentration in
plasma is maximum. Peak plasma concentration (or peak height
concentration or maximum drug concentration ) is the drug concentratio n
at peak. Cmax is usually expressed in mcg/ml.
Peak level depends on the administered dose and the absorption and
elimination rate of drug. Peak is the point of time when the absorption rate and
the elimination rate of drug becomes equal. The portion of ht e curve to the left
of peak indicates the absorption phase where the drug’s absorption rate is
greater than its elimination rate. The portion of the curve to the right of peak
indicates the elimination phase where the drug’s elimination rate is greater anth
its absorption rate. Peak concentration is related to pharmacological response
and should be more than the Minimum Effective Concentration (MEC) and
less than the Maximum Safe Concentration (MSC).
2) tmax: It is the time the drug requires to reach peak con centration in plasma
after extravascular administration. t max is expressed in hours. Time of peak
concentration is used in estimating the absorption rate of drug. It influences
the onset time and onset of action. t max is used for evaluating the efficacy of
drugs employed in treating acute conditions (pain and insomnia) that can be
treated with a single dose of a drug.
3) AUC: It is the area under the plasma level-time curve that gives a measure
of the extent of absorption or the amount of drug that reaches the systemic
circulation. The extent of bioavailability can be determined by following
equations:
F
AUCoral Div ….. (8)
AUCiv Doral
Fr
AUCtest Dstd ….. (9)
AUCstd D test
* *
Bioavailability and Bioequivalence (Chapter 4) 105
CCss,max
ss,max
Plasma Concentration C
Steady-state
level
C
Css,min
ss,min
Dosing interval
Figure 4.2: Determination of AUC and on Multiple Dosing upto Steady-States
study is used for drugs that get excreted in urine in unchanged form ( e.g., certain
thiazide diuretics and sulphonamides) and for drugs that have urine as the site of
action ( e.g., urinary antiseptics such as nitrofurantoin and hexamine). The
concentration of drug metabolites that get excreted in urine is never considered
for calculations, because a drug before getting absorbed may undergo pre -
systemic metabolism at different stages.
The urinary excretion method involves collecting urine samples at regular
intervals for 7 biological half-lives, analysing the unchanged drug in the collected
sample, and determining the amount of drug excreted at each interval and
cumulative amount ex creted. Each time of sampling, the subject should
completely empty the bladder to avoid errors in the next urine sample that may
result from the residual amount. In the beginning of the study, frequent sampling
is essential to correctly calculate the absorption rate.
Given below are the major parameters that are examined in urinary excretion data
obtained with a single dose study:
1) (dXu/dt)max: It is the maximum urinary excretion rate, obtained from the
peak of plot between excretion rate and midpoint time of urine collection
period. It is analogous to the C max obtained from plasma level studies since
the rate of appearance of drug in urine and its concentration in systemic
circulation are proportional. The value of (dX u/dt)max increases with the
increase in rate of and/or extent of absorption.
2) (tu)max: It is the time for maximum excretion rate. It is analogous to t max of
plasma level data. The value of u(t)max decreases with increase in absorption rate.
(dXu/dt)max
Rate of excretion
(tu)max
Midpoint time of urine collection period
Figure 4.3: Plot of Excretion Rate versus Time. The curve is
analogous to a typical plasma level-time profile obtained after
oral administration of a single dose of drug
3) Xu: It is the cumulative amount of dru
g excreted in urine. It is related to the AUC
of plasma level data. The value of Xu increases with the increase in extent of
absorption.The extent of bioavailability is calculated from
the following equations:
F
X D
u oral iv
X D
..… (12)
u iv oral
F
X D
u test std ….. (13)
r
X D
u std testl
* *
Bioavailability and Bioequivalence (Chapter 4) 107
With multiple dose study to steady -state, the equation for computing
bioavailability is:
X u ,ss test Dstd test
Fr
X u,ss std D testl test
….. (14)
4.2.1. Introduction
Various techniques are available for enhancing the solubility of poorly soluble
drugs. Some of these approaches are as follows:
1) Chemical Modifications:
i) Use of salt forms ii) Co-crystallisation
iii) Co-solvency iv) Hydrotrophy
v) Solubilising agents vi) Nanotechnology
2) Physical Modifications:
i) Particle size reduction
a) Micronisation b) Nanosuspension
c) Sonocrystallisation d) Supercritical fluid process
ii) Modification of the crystal habit
a) Polymorphs
b) Pseudopolymorphs
iii) Complexation - Use of complexing agents
iv) Solubilisation by surfactants
a) Microemulsions
b) Self microemulsifying drug delivery system
v) Drug dispersion in carriers
4.2.3. Co-Crystallisation
With the application of co -crystals or molecular complexes , drug solubility can
be improved. If the solvent forms an integral part of the network structure and at
least two component crystal, it is termed as co-crystal. If the solvent do es not
directly involve itself in the network (as in open framework structures), it is
termed as clathrate (or inclusion complex). A co-crystal is a crystalline material
consisting of two or more molecular and electrically neutral species bound by
non-covalent forces.
Co-crystals are more stable as the co -crystallising agents are solids at room
temperature. Only three co -crystallising agents, i.e., saccharin, nicotinamide and
acetic acid, are recognised as safe (GRAS), thus limiting their pharmaceutical
applications. Co -crystallisation may also occur between two active
pharmaceutical ingredients by using sub -therapeutic amounts of drug substances
(such as aspirin or acetaminophen). Co-crystals can be prepared by evaporating a
heteromeric solution or by grin ding the components together. Another technique
of preparing co -crystals includes sublimation, growth from the melt, and slurry
preparation. Formation of molecular complexes and co -crystals and their
importance is increasing day -by-day as an alternative to salt formation, mainly
for neutral compounds or those having weakly ionisable groups.
4.2.4. Cosolvency
Solubilising the drugs in co -solvents is another technique of solubility
enhancement of poorly soluble drugs. Adding an organic cosolvent to water can
change the drug solubility. Water solubility of weak electrolytes and non -polar
molecules is poor and this can be improved by adding another solvent that alters
the solvent polarity. This process is termed cosolvency, and the solvent used for
increasing solubility is termed a cosolvent.
4.2.5. Hydrotrophy
Hydrotrophy is the increase in water solubility due to the presence of large
amounts of additives. Its mechanism of improving solubility is closely rel ated to
complexation that involves a weak interaction between the hydrotrophic agents
(e.g., sodium benzoate, sodium acetate, sodium alginate, and urea) and the solute.
An example of hydrotrophy is solubilisation of theophylline with sodium acetate
and sodium alginate.
Micronisation
Drug solubility is often intrinsically related to drug particle size. By reducing the
particle size of the drug, its surface area is increased and its dissolution properties
are improved. The conventional methods of particle size reduction (such as
comminution and spray drying) rely on mechanical stress to disaggregate the
active compound. M icronisation is used to increase the surface area for
dissolution.
Nanosuspension
Nanosuspensions are sub -micron colloidal dispersion of pure drug particles,
which are stabilised by adding surfactants. They offer the advantages of
increased dissolution ra te due to larger surface area exposed, and absence of
Ostwald ripening due to the uniform and narrow particle size range obtained
that eliminates the concentration gradient factor.
* *
Bioavailability and Bioequivalence (Chapter 4) 111
Sonocrystallisation
Recrystallisation of poorly soluble materials with liquid solvents and anti-solvents
has been successfully employed for particle size reduction. Sonocrystallisation is a
novel approach for particle size reduction that involves inducing crystallisation
using ultrasound waves at a frequency range of 20 -100kHz. T his technique
enhances the nucleation rate, is an effective means of size reduction, and also
controls size distribution of the active pharmaceutical ingredients. In most cases,
ultrasound waves are used in the range of 20 kHz-5 MHz.
Once the drug particles solubilise in SFs, they may be recrystallised at r educed
particle sizes. SCF process allows micronisation of drug particles within a narrow
range of particle size, often to sub-micron levels. The current SCF processes have
the ability to create nanoparticulate suspensions of particles 5 -2,000nm in
diameter. The most widely employed SCF processing methods for micronised
particles are Rapid Expansion of Supercritical Soluti ons (RESS) and gas anti -
solvent recrystallisation (GAS). These methods are used by the pharmaceutical
industries using carbon dioxide (CO2) as the SCF due to its favourable processing
characteristics like low critical temperature (Tc = 31.10°C) and pressure (Pc =
73.8 bar).
and stability. In the same way, amorphous form of drug is more suitable than the
crystalline form as higher energy is associated with the former and increase in
surface area. The dissolution of different solid forms of drug follows the given
order: Amorphous >Metastable polymorph >Stable polymorph
4.2.10. Complexation
Complexation involves association between two or more molecules to form a
non-bonded entity with a well -defined stoichiome try. Complexation relies on
weak forces such as London forces, hydrogen bonding, and hydrophobic
interactions. The complexing agents commonly used in improving bioavailability
of poorly soluble drugs are given in the table below:
Table 4.1: List of Complexing Agents
Types Examples
1) Inorganic lB
2) Coordination Hexamine cobalt(III) chloride
3) Chelates EDTA and EGTA
4) Metal-Olefin Ferrocene
5) Inclusion Cyclodextrins and Cholic acid
6) Molecular Complexes Polymers
With the approach of solid dispersion using suitable hydrophilic carriers, the
solubility of etoposide, glyb uride, itraconazole, ampelopsin, valdecoxib,
celecoxib, and halofantrine can be improved. The eutectic combination of
chloramphenicol/urea and sulphathiazole/urea are the examples for the
preparation of a poorly soluble drug in a highly water soluble carrier.
Poorly Water Soluble
Drug
Disintegration Disintegration
Drug molecule
Amorphous Precipitation
When drug precipitates as an amorphous form in the ine rt carrier, amorphous
precipitation occurs. Higher energy state of the drug in this system produces
much greater dissolution rates than the corresponding crystalline forms of the
drug.
* *
Bioavailability and Bioequivalence (Chapter 4) 115
The in vitro studies are performed to analyse the rate or extent of drug dissolution
or release, while the in vivo studies help to study the plasma drug concentration
or amount of drug absorbed. IVIVC is performed to obtain drug dissolution
results from two or more products so that the simila rity or dissimilarity of
expected plasma drug concentration profiles can be determined. It is important to
know how to establish similarity or dissimilarity of in vivo response, i.e., plasma
drug concentration profiles b efore deducing a relationship betwee n the results
obtained from in vitro and in vivo studies. The method used to establish similarity
or dissimilarity of plasma drug concentration profile s is called bioequivalence
testing. There are well -defined guidelines and standards for establishing
bioequivalence between drug profiles and products.
The dissolution apparatus has g radually evolved from a simple beaker type to a
highly versatile and fully automated instrument. The dissolution apparatuses can
be classified into two principal types based on the absence or presence of sink
conditions:
1) Closed-Compartment Apparatus: It i s a limited -volume apparatus that
operate under non -sink conditions. The dissolution fluid is restrained to the
container size. Example, beaker type apparatuses such as the rotating basket
and the rotating paddle apparatus.
2) Open-Compartment (Continuous Flow-Through) Apparatus: In this, the
dosage form is contained in a column which is brought in continuous contact
with fresh, flowing dissolution medium (perfect sink condition).
A third type of apparatus, called dialysis system , is used for very poorly
aqueous soluble drugs that require large volume of dissolution fluid for
maintenance of sink conditions.
3) Rotating Basket Apparatus (Apparatus 1): It was first described by
Pernarowski et al. It is a closed -compartment, beaker type apparatus
containing a cyl indrical glass vessel (one litre capacity) with hemispherical
bottom. This vessel is partially immersed in a water bath to maintain 37 C
temperature. A cylindrical basket of 22 mesh (meant to hold the dosage form)
is centrally placed in the vessel such tha t it is 2cm above the bottom of the
vessel. The basket is rotated by a variable speed motor through a shaft (figure
4.6a). While withdrawing samples, the basket should remain in motion.
4) Rotating Paddle Apparatus (Apparatus 2): It was first described by Levy
and Hayes . Its assembly is same as that for Apparatus 1, with the only
difference that instead of the rotating basket , a paddle is used as a stirrer
(figure 4.6b). The dosage form is sunk to the bottom of the vessel. Sinkers
prevent the floating of ca psules and other floatable forms. Such preparations
are attached with a small, loose, wire helix to prevent them from floating.
5) Reciprocating Cylinder Apparatus (Apparatus 3): It comprises of a set of
cylindrical flat -bottomed glass vessels fitted with rec iprocating cylinders
(figure 4.6c). It is used for dissolution testing of controlled-release bead-type
(pellet) formulations.
6) Flow-Through Cell Apparatus (Apparatus 4): It comprises of a reservoir
for the dissolution medium and a pump that forces the diss olution medium
through the cell holding the test sample (figure 4.6d). It is used in either:
i) Closed-mode where the fluid is re-circulated and is of fixed volume, or
ii) Open-mode where the fluids are continuously replenished.
* *
Bioavailability and Bioequivalence (Chapter 4) 119
specification in very rare cases exceeds 10% of the dissolution of the pivotal
clinical batch, if IVIV correlations are not present. But in the presence of
IVIVC, a range of specifications are applicable based on the prediction of
concentration-time profiles of test batches bioequivalent to the reference batch.
The dissolution specifications can be established in the presence of an IVIVC
that starts after obtaining the reference (pivotal clinical batch) dissolution
profile. Dissolution of batches with different dissolution properties (including
slowest and fastest batches) are used along with the IVIVC model, and the
concentration-time profiles should be deduced using a suitable convolution
method. Specifications should be established in such a way that all batches
with dissolution profiles between the fastest and slowest batches are
bioequivalent and less optimally bioequivalent to the reference batch.
5) Future Biowaivers: During drug development, various changes are made in
the formulations due to various reasons, for example, unexpected problems
in stability, development, availability of better materials, better processing
results, etc. If an established IVIVC is present, bioequivalence studies can be
avoided by using the dissolution profile from the changed formulation, and
predicting the in vivo concentration-time profile. Such a type of established
profile can be used as a surrogate for the in vivo bioequivalence study. This is
a cost-saving approach as it reduces drug development spending and speedy
implementation of post -approval changes. The post -approval changes may
range from minor (e.g., change in non-release-controlling excipient) to major
(e.g., site change, equipment change, change in manufacture method, etc.).
6) IVIVC in Parenteral Drug Delivery: IVIVC is also applicable to parenteral
dosage forms that are either injected or implanted (such as controlled -release
particulate syst ems, depot system, implants, etc.); but, the success rate of
such development of IVIVC for parenteral dosage forms is very low due to
multiple reasons. For establishing a correlation between the in vitro and in
vivo data, sophisticated modelling techniques are needed which are
unpredictable and unavoidable.
7) Biowaivers: A validated IVIVC procedure can be used for justification of a
biowaiver in filings of a Level 3 (or Type II in Europe) variation, either
during scale -up or during post -approval, and also for line extensions ( e.g.,
different dosage strengths). The primary condition before granting a
biowaiver is that the prediction of in vivo performance of the product with
the modified in vitro release rate should remain bioequivalent with the
originally test ed product (i.e., the new dissolution rate remains within the
IVIVC based biorelevant corridor). The FDA guidance puts forward the
following categories of biowaivers:
i) Biowaivers without an IVIVC,
ii) Biowaivers using an IVIVC: Non-narrow therapeutic index drugs,
iii) Biowaivers using an IVIVC: Narrow therapeutic index drugs,
iv) Biowaivers when in vitro dissolution is independent of dissolution test
conditions, and
v) Situations for which an IVIVC is not recommended for biowaivers.
* *
122 Biopharmaceutics & Pharmacokinetics
Given below are some other parameters that are used in the bioequivalence study:
1) Normalised C max: Cmax and t max show significant intra -subject variability,
and hence normalised C max is used in some cases. It is calculated by the
following equation:
C
Normalised Cmax = max …..(15)
AUC
Studies indicate that normalised C max in comparison to C max shows less intra-
subject variability.
2) Mean Residence Time (MRT): It is the time a drug molecule spends in the
body before getting excreted. MRT can be calculated by first determining the
Area Under the Moment Curve (AUMC) by the following equation:
t
t C t C
AUMC0-t i i i-1 i-1 t i t i-1 ..... (16)
i 1 2
Equation (16) indicates that AUMC can be calculated in the same manner as
AUC, with the only difference that in the latter drug plasma concentration is
multiplied by time. AUMC 0-t is calculated from AUMC 0-, and then MRT is
calculated by the following equation:
AUMC0-
MRT ..... (17)
AUC0-
3) Plasma Trough Fluctuation (%): It is used in the bioequivalence study of
sustained release formulations, which are designed such that they maintain
steady-state plasma drug concentration for extended time periods. Hence, in
bioequivalence study of sustained release formulations, the steady -state
plasma drug concentrations obtained from two drug products are compared.
Cmin is the lowest plasma drug concentration just before the next dose, and %
PTF is the % change in plasma drug concentration between two administered
doses. It is calculated by the following equation:
C max C min
%PTF ….. (18)
C average
Where, Caverage = Average plasma drug concentration during the dosingeriod.
p
analyses include calcu lating AUC to the last quantifiable concentration and
to infinity, tmax, Cmax, elimination rate constant ( k), elimination half-life (t1/2),
and other parameters for each subject. For multiple -dose studies,
pharmacokinetic analyses include calculating the s teady-state AUC, t max,
Cmin, Cmax, and % fluctuation for each subject. Proper statistical evaluation
should be performed on the estimated pharmacokinetic parameters.
3) Statistical Evaluation of the Data: Bioequivalence is determined by
comparing population averages of a bioequivalence metric, such as AUC and
Cmax. This approach is termed average bioequivalence , and involves
calculating a 90% confidence interval for the ratio of averages (population
geometric means) of the bioequivalence metrics for the test a nd reference
drug products. Bioequivalence can be established if the calculated confidence
interval falls within a prescribed bioequivalence limit (usually 80 -125% for
the ratio of the product averages).
Another approach is termed individual bioequivalenc e (proposed by the
FDA) that requires a replicate crossover design for estimating within -subject
variability for the test and reference drug products, and subject -by
formulation interaction.
Bioequivalence can be proved only if there is no statistical dif ference
between the bioavailability of the test product and the reference product.
There are many statistical approaches (or parametric tests) that are used for
comparing the bioavailability of drug from the test dosage form and from the
reference dosage f orm. Many of these approaches assume that the data are
distributed according to a normal distribution or “bell-shaped curve”.
The distribution of many biological parameters ( Cmax and AUC) has a longer
right tail than observed in a normal distribution. The true distribution of these
biological parameters may also be difficult to establish because small number
of subjects are used in a bioequivalence study. The distribution of data that
has been transformed to log values resembles a normal distribution compared
to the distribution of non -log-transformed data. Therefore, for determining
bioequivalence, log transformation of the bioavailability data ( e.g., Cmax,
AUC) is performed before statistical data evaluation.
The obtained bioequivalence data is statisti cally evaluated by the following
two methods:
i) Analysis of Variance (ANOVA): This is a statistical procedure in which
the data is tested for differences within and between treatment and control
groups. A bioequivalent product should not produce any signific ant
differences in the tested pharmacokinetic parameters.
AUC(0-t), AUC (0-∞), tmax, and Cmax obtained for each treatment or dosage
form are usually tested. Other metrics of bioavailability have also been
used for comparing the bioequivalence of two or more formulations. The
ANOVA may evaluate variability in subjects, treatmen t groups, study
period, formulation, and other variables, depending on the study design. If
the data shows large variability, the mean difference for each
* *
Bioavailability and Bioequivalence (Chapter 4) 127
4.5. SUMMARY
The details given in the chapter can be summarised as follows:
1) Bioavailability is the rate and extent of an administered dose of drug that
reaches the systemic circulation in unchanged form.
2) The drug dose given to the patient i s the administered dose . The dose
available to the patient is the bioavailable dose.
3) The absolute bioavailability of a given drug from a dosage form is the
fraction (or percentage) of the administered dose absorbed into the systemic
circulation in unchanged form.
4) Relative bioavailability can be determined by comparing the bioavailability
of a drug from a test dosage form with the same drug administered in a
standard dosage form.
5) Cmax is the peak that indicates the point at which the drug concentration in
plasma is maximum. It is usually expressed in mcg/ml.
6) tmax is the time the drug requires to reach peak concentration in plasma after
extravascular administration. It is expressed in hours.
7) AUC is the area under the plasma level -time curve that gives a me asure of
the extent of absorption or the amount of drug that reaches the systemic
circulation.
8) Urinary excretion studies , performed to assess bioavailability, rely on the
principle that the urinary excretion of unchanged drug and the plasma
concentration of drug are directly proportional.
9) (dXu/dt)max is the maximum urinary excretion rate, obtained from the peak of
plot between excretion rate and midpoint time of urine collection period.
10) (tu)max is the time for maximum excretion rate.
11) Xu is the cumulative amount of drug excreted in urine.
12) The method of acute pharmacologic response has a disadvantage that the
pharmacologic response may vary and thus an accurate correlation between
the measured response and drug available from the formulation cannot be
established.
* *
Bioavailability and Bioequivalence (Chapter 4) 129
13) The therapeutic response method involves observing the clinical response
of a drug formulation in patients who are suffering from the disease for
which the drug is intended to be used.
14) If the solvent forms an integral part of the network struct ure and at least two
component crystal, it is termed as co-crystal.
15) If the solvent does not directly involve itself in the network (as in open
framework structures), it is termed as clathrate (or inclusion complex).
16) Water solubility of weak electrolytes a nd non -polar molecules is poor and
this can be improved by adding another solvent that alters the solvent
polarity. This process is termed cosolvency, and the solvent used for
increasing solubility is termed a cosolvent.
17) The system of cosolvent involves solvent blending in which the interfacial
tension between the aqueous solution and hydrophobic solute is reduced.
18) Hydrotrophy is the increase in water solubility due to the presence of large
amounts of additives.
19) Jack H. Shulman first used the term microemulsion in 1959.
20) The term solid dispersion refers to the dispersion of one or more active
ingredients in an inert carrier in a solid state, prepared by the melting (fusion)
method, solvent method, or fusion solvent method.
21) A eutectic mixture of a sparing ly water -soluble drug and a highly water -
soluble carrier is thermodynamically regarded as an intimately blended
physical mixture of two crystalline components.
22) Solid solution is a binary system comprising of a solid solute molecularly
dispersed in a solid solvent. Solid solutions are also termed molecular
dispersions or mixed crystals as the two components crystallise together in
a homogeneous one phase system.
23) In continuous solid solution , the drug and the carrier are miscible in all
proportions.
24) In discontinuous solid solution, solubility of each component in the other is
limited.
25) In substitutional crystalline solid solution, if the drug and carrier molecules
are almost of same size, the drug molecule substitutes for the carrier
molecules in its crystal lattice.
26) In interstitial crystalline solid solution, if the size of drug molecule is 40%
or less than the size of carrier molecules, the drug molecules occupy the
interstitial spaces in the crystal lattice of carrier molecules.
27) A glass solution is a hom ogenous system in which a glassy or a vitreous of
the carrier solubilises drug molecules in its matrix.
28) United State Pharmacopoeia (USP) defined IVIVC as “the establishment of
a rational relationship between a biological property or a parameter derived
from a biological property produced by a dosage form, and a
physicochemical property or characteristic of the same dosage form”.
* *
130 Biopharmaceutics & Pharmacokinetics
4.6. EXERCISE
Answers
1) False 2) True 3) False
4) False 5) True 6) True
7) False 8) True 9) False
10) Administered dose and bioavailable dose 11) mcg/ml
12) Xu 13) Molecular dispersions & mixed crystal
14) D 15) Therapeutic equivalence
16) Mean residence time 17) Individual bioequivalence
18) ANOVA
* *
132 Biopharmaceutics & Pharmacokinetics
* *
Pharmacokinetics (Chapter 5) 133
CHAPTER Pharmacokinetics
5
5.1. PHARMACOKINETICS
5.1.2. Objectives
The objectives of pharmacokinetics are as follows:
1) The main objective of pharmacokinetics is to measure the extent of drug
absorption, distribution, biotransformation, and excretion in intact, living
animals or humans. The information obtained from such studies is used to
determine the effect of alterations in dose, dosage regimen, administration
*
route, and physiologic state on drug accumulation and disposition. *
134 Biopharmaceutics & Pharmacokinetics
5.1.3. Applications
Following are the applications of pharmacokinetics:
1) Equations of pharmacokinetics are used to determine the drug bioavailability.
2) Using the principles of pharmacokinetics the dosing pattern of any d rug can
be fixed.
3) The principles of pharmacokinetics are also used to determine the dosing
pattern of controlled release dosage forms.
4) In case of kidney failure, the drug dose that should be given can be measured
by using pharmacokinetic principles.
5.2.1. Introduction
Many mathematical models are used to describe the rate processes of drug
absorption, distribution, metabolism, and excretion. These models are also used
to develop various equations which analyse drug concentrations in bod y with
respect to time. Since drug concentrations depend on time, the drug concentration
and time variables are termed as dependent and independent variables,
respectively. Direct measurement of pharmacokinetic parameters is not possible ;
thus, can be dete rmined through in vitro experiments using the given set of
dependent and i ndependent variables, which are collectively termed as data.
These data are used to design a pharmacokinetic model and test its validity.
If it is found that the model does not appropriate ly fit all the experimental
observations, a new and a more complex model (hypothesis) is devised and tested
for validity. It should always be known that the pharmacokinetic data should not
replace the clinical observations in patient and sound judg
ement by the clinician.
These compartments are not real physiological or anatomic region , but are
hypothetical or virtual. These compartments represent the bio system and are
assumed to comprise of tissues with similar blood flow and affinity for drug. A
hypothetical model comprising of one, two or maximum three functional
compartments (arranged serially or parallelly to each other) is used to analyse the
kinetics of most of the drugs. It is also considered that the rate of drug movement
between compartments (i.e., entry and exit) follow first-order kinetics.
5.2.2.1. Types
Compartment models are divided into the following two categories based on their
arrangement, i.e., whether parallel to each other or in a series:
1) Mammillary Model: This is the most common compartment modelto be used
in pharmacokinetic studies. In this model , one or more peripheral
compartments are connected to a central compartment , consisting of plasma
and highly perfused tissues in which the drug undergoes rapid distribution. The
administered drug firstly reaches the central compartment and then distributes
to all the other compartments connected to the central compartment. Since the
major organs involved in drug elimination (i.e., kidney and liver) are found in
the central compartment, drug eliminationis assumed to occur from there.
Several types of compartment models have been shown in figure 5.1.
Compartment 1 is the plasma or central compartment, and compartments
2, 3 and 4 are the tissue or peripheral compartments . The letter K
indicates pharmacokinetic rate constants, and the numbers describe the
movement direction of drug between the compartments. For example, K12
indicates the rate constant with respect to drug move ment from compartment
1 to compartment 2.
Parameters of Mammillary Model
Model 1 (figure 5.1) can be described by two parameters, i.e., the volume of
compartment and the elimination rate constant (K). In model 4, the
pharmacokinetic parameters include the volume of compartment 1 and 2 and
*
the rate constants Ka, K13, K12 and K21 for total six parameters. *
136 Biopharmaceutics & Pharmacokinetics
Ka K
1
K12
Ka
1 2
K21
K13
5.2.2.3. Disadvantages
Following are the disadvantages of compartment models:
1) All the physiologic al functions or the anatomic structure of the species
cannot relate to the compartment model. Therefore, many assumptions have
to be made to interpret the data obtained from compartment models.
2) Exhaustive effort is required to find an exact model that can be used to
predict and describe the ADME of a drug under consideration.
3) Compartment modelling is based on curve fitting of plasma concentration
with multi-exponential mathematical equations.
4) This approach is not a common procedure and is applicable only for some
specific drugs.
5) The behaviour of drug in body can fit more than one c ompartmental model
depending on the drug administration route.
6) Sometimes, it becomes difficult to explain the differences between results
obtained from human and animal experiments using compartment models.
7) A variation in the study model can be observed.
AUC
t 2 t 1 C1 C2
Concentration × Time
2
AUM
C
Concentration
AUC
t 4 t 3 C3
C1
2
AUC
Trapezoid
C2
C3
Time t1 t2 t3 t4
Figure 5.3: AUC and AUMC Plots
If the time course of drug concentration in plasma is considered as a statistical
distribution curve:
AUMC
MRT ..... (1)
AUC
* *
138 Biopharmaceutics & Pharmacokinetics
After plotting a graph between the product of plasma drug concentration and time
(C.t) versus time (t) from zero to infinity ( figure 5.3), the value of AUMC is
obtained, that can be mathematically represented as:
AUMC C t dt ..... (2)
0
From a plot of plasma drug concentration versus time from zero to infinity, the
value of AUC is obtained, which is mathematically represented as:
AUC C dt ……(3)
0
The values of AUMC and AUC can be calculated from their respective graphs
using the trapezoidal rule in which the curve is divided by a series of vertical
lines int o a number of trapezoids, followed by calculating the area of each
trapezoid separately and then adding them together.
MRT is the average amount of time a drug spends in the body before being
eliminated. Thus, it can be said that MRT is the statistical mo ment analogy of
half-life (t 1/2). Practically, MRT defines the time for the elimination of 63.2% of
the intravenous bolus dose. The values will always be greater when the drug is
administered in any form or through any route other than the intravenous bolus.
The total area under the curve obtained from a plot of drug concentration
versus time is the zero moment of a drug concentration in a plasma -time profile.
The total area under the curve obtained from a plot of product of drug
concentration and time versus time (AUMC0) is the first moment of a plasma
concentration-time profile.
* *
Pharmacokinetics (Chapter 5) 139
Concentration
Time (hrs.)
Figure 5.4: Plots of Drug Concentration (g/ml) ( ) and Drug
Concentration-Time (g-hr/ml) (X) versus Time, during and after an
Hour Constant Rate Intravenous Infusion. The Area under the Drug
Conc. versus Time Plot to Infinity is AUC0, the Area under the Drug
Conc. Time versus Time Plot to Infinity is AUMC0.
During the calculation of various pharmacokinetic parameters, the plasma
concentration versus time data is plotted ( figure 5.4) and the AUC from t = 0 to
the last sampling time (t*) is calculated using the trapezoidal rule. The values of
(C) × (t) (i.e., product of concentration of drug and time) are plotted against time
(figure 5.4). The area under the (C) × (t) versus t plot from t = 0 to the last
sampling time (t*) is called the first moment of drug concentration with
respect to time (AUMC).
The area under the curve from t* to for both the curves can be calculated using
appropriate equations to obtain AUC t* and AUMC t* . By adding these areas to
t*
AUC 0 t* and AUMC0 , the total areas under zero moment and first moment
curves, i.e., AUC
0 and AUMC0 , respectively are obtained. The above obt ained
5.2.3.2. Advantages
Following are the advantages of non-compartmental analysis:
1) The simple algebraic equations are used for easy derivation of the
pharmacokinetic parameters.
2) Similar mathematical treatment can be used for any drug or metabolite,
provided that they follow first-order kinetics.
3) A detailed description of drug disposition is not needed.
5.2.3.3. Disadvantages
The main drawback of non-compartmental analysis is that it offers limited
information about the plasma drug concentration -time profile and most of the
time it deals with averages.
parts. Drug uptake into organs is de termined by binding the drug in these tissues.
In contradiction of the tissue volume of distribution, the actual tissue volume is
used. There are many tissue organs in the body, and each tissue volume and its
drug concentration should be estimated. The phy siological model potentially
predicts the accurate tissue drug concentration, which the compartment model
fails to do.
A physiological pharmacokinetic model, despite the limitation, provides a better
understanding of the changes in drug distribution from one animal species to
another by the physiological factors. Other major differences are:
1) Data fitting is not required in perfusion model. Drug concentrations in
various tissues depend on the organ tissue size, blood flow, and
experimentally determined dru g tissue-blood ratios (i.e., drug partition
between tissue and blood).
2) Due to some pathophysiological conditions, blood flow, tissue size, and the
drug tissue -blood ratio may vary. Effect of these variations on drug
distribution should be considered in physiological pharmacokinetic models.
3) Physiological pharmacokinetic models can be applied to several species,
and human data can be extrapolated with some drugs. In compartment
models, the volume of distribution is a mathematical concept with no
relation to t he blood volume and blood flow, thus extrapolation is not
possible. So far, many drugs (like digoxin, lidocaine, methotrexate, and
thiopental) have been described using perfusion models. It is not possible to
estimate the tissue levels of these drugs using compartment models;
however, they describe blood levels well. A perfusion model is shown in
figure 5.5.
QH
Heart
QM
Muscle
QS
SET
QR
RET
Ke QK
Kidney
Km QL
Liver
5.2.4.1. Types
The physiological models are categorised into the following two types:
1) Blood Flow Rate -Limited Models: These models are more popular and
common in use. They rely on the assumption that drug movement in a body
region is more rapid than its delivery rate to that region by the perfusing
blood. Ho wever, this assumption is applicable to low molecular weighed,
poorly-ionised and highly lipophilic drugs ( e.g., thiopental, lidocaine, etc.)
having high membrane permeability.
2) Membrane Permeation Rate -Limited Models: These models are more
complex. They are used for highly polar, ionised and charged drugs. In these
models, the cell membrane acts as a barrier for the drug that gradually
permeates by diffusion; thus, these models are also called diffusion-limited
models. Equations for these models are very co mplicated due to the time lag
in equilibration between the blood and the tissue.
5.2.4.2. Advantages
The physiological pharmacokinetic models have the following advantages over
the conventional compartment models:
1) Their mathematical treatment is direct.
2) They do not require data fitting; drug concentration in various body regions
can be estimated based on the organ or tissue volume, perfusion rate and
experimentally determined tissue-to-plasma partition coefficient.
3) They provide accurate description of drug concentration -time profile in any
organ or tissue, and thus, provide a better insight of drug distribution
characteristics in the body.
4) They can easily predict the effect of altered physiology or pathology on drug
distribution from the changes in various pharmacokinetic parameters that
correspond to actual physiologic and anatomic measures.
5) They can correlate data in several animal species and with some drugs, and
then can extrapolate to humans.
5.2.4.3. Disadvantages
The physiological pharmacokinetic models have the following disadvantages:
1) Obtaining the experimental data using these models is a very exhaustive
process.
2) Most of them assume an average blood flow for individual subjects, h ence
making the prediction of individualised dosing is difficult.
3) The number of data points in these models is less than the pharmacokinetic
parameters to be assessed.
4) They can monitor drug concentration in body with much difficulty since
*
exhaustive data is required. *
142 Biopharmaceutics & Pharmacokinetics
5.3.1. Introduction
One-compartment open model is the simplest model that describes the body as a
single, kinetically homogeneous unit having no barriers to drug movement. Also
in this model, final distribution equilibrium between the drug in plasma and other
body fluids is rapidly attained and maintained all the times. This model applies
only to those drugs that rapidly distribute throughout the body.
The anatomical reference compartment is the plasma and the drug concentration
in plasma represents the drug concentration in all body tissues, i.e., any change in
plasma drug concentration indicates a proportional change in drug concentration
throughout the body. However, the model does not assume that the plasma drug
concentration is equal to drug concentration in other body tissues. The term open
indicates that the input ( absorption) and output (elimination) are unidirectional
and the drug can be eliminated from the body. Figure 5.6 shows s uch a one -
compartment model.
Ka Blood and KE Metabolism
Drug Other Body
Input Tissues Output Excretion
(Absorption) (Elimination)
Figure 5.6 Representation of One-Compartment Open Model showing
Input and Output Processes
The following assumptions help in deriving mathematical equations for one -
compartment open model:
1) Drug absorption from the absorption sit e may be explained by the first -order
kinetics.
Explanation: Most drugs are absorbe d by passive diffusion governed by the
first-order process, i.e., the drug absorption rate and the drug concentration at
absorption site are proportional. Drugs which are absorbed by active
transport, facilitated diffusion, etc., do not follow this assumption.
2) A drug on entering the systemic circulation rapidly distributes to other body
fluids and tissue, thus a dynamic equilibrium is rapidly achieved between the
drug in the blood and the drug in other tissues.
Explanation: The drug distributing to highly p erfused tissues, like heart,
lungs, liver, kidney, etc., can distribute instantaneously and attain
equilibrium between the drug levels in plasma and other body fluids and
tissues. If the drug is distributed to poorly perfused tissues, the time required
for its distribution and equilibrium is considered. Hence, this simple model is
not used for pharmacokinetic analysis of the data obtained with such drugs.
3) Any changes that occur in the drug plasma levels indicate proportional
changes in the tissue drug levels.
Explanation: A dynamic equilibrium exists between the drug concentration
in plasma and drug concentration in tissues. Thus, a change in plasma drug
level reflects a proportional change in the drug levels in tissues.
* *
Pharmacokinetics (Chapter 5) 143
4) Drug elimination from the body follo ws apparent first -order kinetics and its
rate constant (K) is known as an apparent first-order rate constant.
Explanation: Drug elimination from the body occurs by different processes,
like renal, biliary, biotransformation, excretion in the expired air, etc.
The general expression for rate of drug presentation to the body is as follows:
dX
Rate in (absorption) Rate out (elimination) ..... (4)
dt
Since the rate in or absorption is absent, equation (4) becomes:
dX
Rate out ..... (5)
dt
If the rate out or elimination follows first-order kinetics:
dX
KE X ..... (6)
dt
Where, KE = First-order elimination rate constant.
X = Amount of drug in the body remaining to be eliminated at time (t).
The negative sign indicates that the drug is lost from the body. Now, the various
related pharmacokinetic parameters can be estimated.
log C2
K log C log C
1 2
Concentration
Slope E
2.303 2.303( t t )
log C1
1 2
t1/2 t1/2
t1 t2
Time
Figure 5.7 (a) Cartesian Plot of a Drug that follows One-Compartment Kinetics and
given by Rapid I.V. Injection, and (b) Semi-logarithmic Plot for the Rate of
Elimination in a One-Compartment Model
* *
Pharmacokinetics (Chapter 5) 145
Thus, C o, K E, and t 1/2 can be readily obtained from log C versus t graph.
Elimination of drug from the body is the sum of urinary excretion, metabolism,
biliary excretion, pu lmonary excretion, and other involved mechanisms. Thus,
KE is an additive property of rate constants for each of these processes and is
called the overall elimination rate constant.
KE = Ke + Km + Kb + Kl + …… ..... (12)
If the number of rate constants involved and their values are known, evaluating
the fraction of drug eliminated by a particular route becomes easier. For
example, if a drug is eliminated by urinary excretion and metabolism, the
fraction of drug excreted unchanged in urine (F e) and the fraction of drug
metabolised (Fm) is given as:
Ke
Fe ..... (13)
KE
K
Fm m ..... (14)
KE
Time
Figure 5.8: Graphical Representation for
Linear Trapezoidal Method to Estimate
Let us assume that f(t) is a function thatAUC
describes plasm a
drug concentration -
time curve, and (t) is a function that is linear between two su ccessive plasma
drug level-time points.
The AUC expressed by (t) ( t ) dt will be approximately t ( t ) dt
tn
tn
0
t 0
tn
The integral t (t ) is expressed as the sum of total number of trapezoids into
0
Based on equation s (28) and (29), equation (27) can be expressed as:
tn t t t t t t
t 0 (t ) dt 1 2 0 (C0 C1 ) 2 2 1 (C1 C2 ) ... n 2 n 1 (Cn 1 Cn ) …..(30)
n 1
t11 t i
( t ) dt
tn
Thus, t 0
i 0 2
(Ci Ci 1 ) …..(31)
The rate of change in the amount of drug in the body (dX/dt) at any time during
infusion, is the difference between the zero -order rate of drug infusion (R 0) and
the first-order rate of elimination (–KEX):
dX
R o KEX ..... (32)
dt
Since X = V dC, equation (3 3) can be trans formed into concen tration terms as
follows:
Ro
C (1 e K E t ) ..... (34)
K E Vd
C R o (1 e K E t ) ..... (35)
Cl T
* *
Pharmacokinetics (Chapter 5) 149
Since the drug amount in body is zero at the beginning of constant rate infusion,
no elimination occurs. With time, the amount of drug in body gradually increases
(elimination rate < infusion rate) up to a point after which the plasma drug
concentration reaches a constant value, i.e., steady-state, plateau or infusion
equilibrium (elimination rate = infusion rate) (figure 5.9).
Plasma Drug Concentration
Infusion rate
(2RO)
CSS Steady-state
Infusion stopped
Infusion rate
(RO) When plotted on a semi-log
graph yields a straight line with
Infusion time (T) slope = –KE/2.303
Time
Figure 5.9: Plasma Concentration -Time Profile for a Drug given by Constant Rate I.V.
Infusion (the two curves indicate different infusion rates Ro and 2Ro for the same drug)
At steady -state, the rate of change of amount of drug in the body is zero, so
equation (38) becomes:
Zero = Ro – KE Xss
Or KE Xss = Ro ..... (36)
On transforming and rearranging equation (36) to concentration terms:
Ro R Infusion rate
Css = o , i.e., ..... (37)
K E Vd ClT Clearance
Where, XSS and CSS = Amount of drug in the body and plasma drug concentration
at steady-state, respectively.
The value of K E (and t1/2) can be obtained from the slope of straight line obtained
from a semi-log plot (log C versus t) of plasma concentration-time data collected
at the time when the drug is no more infused. Alternatively, K E can be calculated
from the data collected during infusion to steady-state.
On substituting R0/ClT = CSS from equation (37) in equation (35):
C = CSS (1 – e–KEt) ..... (38)
On rearranging equation (38):
CSS C K E t
e ..... (39)
CSS
On transforming equation (39) into log form:
C C KEt
Log SS ..... (40)
CSS 2.303
A semi -log plot of (C SS – C)/CSS versus t yields a straight line with slope of
–KE/2.303 (figure 5.10).
* *
150 Biopharmaceutics & Pharmacokinetics
C C
log ss
Css K E
Slope
2.303
t
Figure 5.10: Semi-log Plot to Compute KE from
Infusion Data Up to Steady-State
The time to reach steady-state concentration depends on the elimination half -life.
An increase in infusion rate will increase the plasma concentration attained at
steady-state. If since the beginning of infusion (t 1/2), n is the number of half -lives
passed, equation (38) can be written as:
C = CSS [1 − (t1/2)n] ..... (41)
The percent of C SS achieved at the end of each t 1/2 is the sum of C SS at previous
t1/2 and the concentration of drug remaining after a given t1/2.
These two parameters can also be computed from the total area under the curve
till the end of infusion:
R oT R T
AUC o CssT ….. (42)
K E Vd ClT
Zero-order absorption process has a constant absorp tion rate, and is independent
of Amount Remaining to be Absorbed (ARA). Its regular ARA versus t plot is
linear with slope equal to absorp tion rate while its semi -log plot shows an ever -
increasing gradient with time. On the other hand, first -order absorption process
shows a decline in the rate with ARA, i.e., absorption rate depend on ARA; its
regular plot is curviline ar and semi -log plot is a straight line with slope equal to
absorption rate constant.
* *
Pharmacokinetics (Chapter 5) 151
Log ARA
Zero-order
ARA
Zero-order
First-
order
First-order
Time Time
a) Cartesian Plot b) Semi-log Plot
Figure 5.11: Distinction between Zero-Order and First-Order Absorption
Processes. (a) Regular Plot and (b) Semi-log Plot of Amount of Drug
Remaining to be Absorbed (ARA) versus Time (t)
After extravascular administration, the rate of change in the drug amount in body
(dX/dt) is the difference between the rate of input (absorption, dX ev/dt) and rate
of output (elimination, dXE/dt).
dX/dt = Rate of absorption – Rate of elimination
dX dX ev dX E
..... (43)
dt dt dt
The plasma concentration -time profile of a drug following one -compartment
kinetics shows the absorption phase, post -absorption phase, and elimination
phase (figure 5.12).
Cmax
Plasma drug concentration
Post-absorption phase
Absorption
phase
Elimination phase
Time
Figure 5.12: Absorption and Elimination Phases of Plasma
Concentration-Time Profile Obtained After Extravascular
Administration of a Single Dose of Drug
During the absorption phase, the absorption rate > the elimination rate.
dX ev dX E
..... (44)
dt dt
At peak plasma concentration, the absorption rate = the elimination r ate, and
the change in amount of drug in the body is zero.
dX ev dX E
..... (45)
dt dt
During the post-absorption phase, some drug still remains to be absorbed at the
extravascular site, and the elimination rate > the absorption rate.
dX ev dX E
..... (46)
dt dt
After the completion of drug absorption , the absorption rate be comes zero and
the plasma level-time curve shows only the elimination phase.
* *
152 Biopharmaceutics & Pharmacokinetics
As in the case of several controlled drug delivery systems, the drug absorption
rate is constant and continues till the amount of drug at the absorption site ( e.g.,
GIT) gets depleted. The equations explaining the plasma concen tration-time
profile for constant rate intravenous infusion are also applicable to this model.
Equation (53) shows that as K a becomes greater than K E, t max becomes smaller
since (Ka – KE) increases faster than log Ka/KE.
By substituting equation ( 53) in equation (4 9), the value of Cmax can be
obtained. However, this can be expressed in a simpler way as follows:
F X o K E t max
C max e ..... (54)
Vd
It is shown that at Cmax, when Ka = KE, tmax = 1/KE. Hence, equation (54) reduces to:
F X o 1 0.37 F X o
C max e ..... (55)
Vd Vd
Since, FXo/Vd represents Co after I.V. bolus, the maximum plasma concentration
that can be attained after extravascular administration is 37% of the maximum
level that can be attained w ith I.V. bolus in the same dose. If bioavailability is
<100%, lower concentration will be attained.
A plot of log C versus t yields a straight line with slope –KE/2.303 (half-life can
be computed from KE). Elimination rate constant can also be deduced from
urinary excretion data.
During the elimination phase, when absorption is almost over, K a >> K E and the
value of second exponential e – K a t , approaches zero; whereas the first exponential
e – Ka t , approaches a finite value. Hence, equation (59) reduces to:
C A e–KEt …..(60)
t
Figure 5.13: Plasma Concentration-time Profile after Oral Administration of a
Single Dose of a Drug. The Biexponential Curve has been Resolved into its Two
Components, i.e., Absorption and Elimination
By deducting true plasma concentration values, i.e., equation ( 59) from the
extrapolated plasma concentration values, i.e., equation (60), a series of residual
concentration values (C) is obtained:
(C – C) Cτ A e – Kat …..(62)
On taking logarithm of equation (62):
Ka t
log C log A – …..(63)
*
2.303 *
Pharmacokinetics (Chapter 5) 155
A plot of log C versus t yields a straight line with slope –Ka/2.303 and y -
intercept log A (figure 5.13). Absorption half-life can be obtained from Ka using
the relation 0.693/K a. Thus, the method of residuals allows resolution of the bi -
exponential plasma level -time curve into its two exponential components. This
method gives best results if the difference between K a and K E is large (K a/KE
3). In some cases, K E obtained after I.V. bolus of the same drug is much larger
than the K a obtained by the method of residuals ( e.g., isoprenaline). If KE/Ka 3,
the terminal slope gives the value of K a (and not K E), while the slope of residual
line gives the v alue of K E (and not K a). This is called flip-flop phenomenon,
since the slopes of the two lines exchange their meanings.
Ideally, the extrapolated and the residual lines intersect each other on y -axis, i.e.,
at time t = zero, and there is no lag in absorp tion. However, if such an
intersection occurs at time > zero, it indicates time lag , which is the time
difference between drug administration and start of absorption. It is denoted by t 0
and represents the beginning of absorption.
The above method for Ka estimation is a curve-fitting method that is suitable for
drugs that get rapidly and completely absorbed and follow one -compartment
kinetics even when given intravenously. However, if the drug absorption is
affected by gastrointestinal motility or enzymati c degradation, and if the drug
shows multi-compartment characteristics after intravenous administration (which
is true for virtually all drugs), the value of K a computed by curve-fitting method
is invalid even if the drug has undergone absorption by first -order kinetics. The
so obtained K a is the best estimate of first -order disappearance of drug from the
GIT instead of the first-order appearance in the systemic circulation.
Assumptions
The Wagner-Nelson method of calculation does not require a model assump tion
concerning the absorption process. It does require the assumption that:
1) The body behaves as a single homogeneous compartment, and
2) The elimination process of drug follows first-order kinetics.
The amount of drug i n the body is X = V dC. The amount of drug eliminated at
any time (t) can be calculated as:
XE = KE Vd [AUC]ot ..... (65)
On substituting the values of X and XE in equation (64):
XA = Vd C + KE Vd [AUC]ot ..... (66)
The total amount of drug absorbed into the systemic circulation from time zero to
infinity (XA) can be given as:
XA = Vd C + KE Vd [AUC]o ..... (67)
Since at t = , C = 0, equation (67) reduces to:
XA = KE Vd [AUC]o ..... (68)
Log % ARA
t
Figure 5.14: Semi-log Plot of %ARA versus t
according to Wagner-Nelson Method
The fraction of drug absorbed at time t is given as:
X A VdC K E Vd [A C]o C K E [A C]o
t t
..... (69)
XA K E Vd [A C]o K E [A C]o
The Wagner-Nelson method involves collecting blood samples after a single oral
dose at regular intervals of time till the entire amount of drug is elimi nated from
the body. The value of K E is obtained from a plot of log C versus t and [AUC] ot
and [AUC] o are obtained from a plot of C versus t. A semi -log plot of %
unabsorbed (i.e., %ARA) versus t yields a straight line with slope Ka/2.303
(figure 5.14). If a regular plot of the same is a straight line, the absorption is
assumed to be following zero-order kinetics.
Ab C X t /Vc K13AUC 0
t
K13AUC 0
Or, ..... (80)
Abα α
Where,
Ct = Apparent tissue concentration
tn = Time of sampling for sample n
tn-1 = Time of sampling for the sampling point proceeding sample n
(C)tn–1 = Concentration of drug at central compartment for sample n –1
C = Concentration difference at central compartment between two sampling
times
t = Time difference between two sampling times
Fraction of drug unabsorbed
[1 − (Ab/Abα)]
Slope = –Ka/2.303
Time (hours)
Figure 5.15: Plot of the Fraction of Drug Unabsorbed [1 – (Ab/Ab)]
versus Time used to determine the Absorption Rate Constant by Loo-
Riegelman Method for a Drug that Follows a Two-Compartment Model
For example, plasma is sampled at times t = 0 and 0.5 hours, and corresponding
concentrations of the drug in the central compartment are 0 and 2g/ml.
Then, (Ct)tn = 2g/ml and (C)tn –1 = 0, C = 2g/ml, t = 0.5 hours
If the drug is administered via intravenous route for estimating the distribution
and elimination rate constants, only then the absorption rate can be estimated by
Loo-Riegelman method. For drugs that cannot be given intravenously, K a cannot
be calculated by the Loo-Riegelman method.
Xu
From ur inary excretion data, the first -order elimination (and excretion) rate
constants can be determined by using the following two methods:
Rate of Excretion Method
In the rate of excretion metho d, the rate of urinary drug excretion (dX u/dt) is
directly proportional to the amount of drug in the body (X):
dX u
X
dt
dX u
Or, Ke X ...... (82)
dt
Where, K e = first -order urinary excretion rate constant (a proportionality
constant). According to the first-order disposition kinetics:
X = XO e-KEt ..... (83)
On substituting equation (83) in equation (82):
dX u
K e X Oe K E t ..... (84)
dt
Where, XO = Dose administered (intravenous bolus).
On taking logarithm of equation (84):
dX u
log log K e X O K E t log e
dt
1
But, log e = log 2.718 = 0.4343 =
2.303
dX u K t
log log K e XO E ..... (85)
*
dt 2.303 *
Pharmacokinetics (Chapter 5) 161
Equation (85) is used to draw a semi -log plot of rate of excretion (log dX u/dt)
versus time (t) that gives a straight line with slope –KE/2.303 (figure 5.16). The
slope of such an excretion rate versus time plot is re lated to elimination rate
constant (KE), and not to excretion rate constant (Ke). However, the excretion rate
constant can be obtained from the intercept on y -axis (log K e Xo). From K E and
Ke, the elimination half -life and non -renal elimination rate consta nts can be
computed.
log Ke Xo
dX u
log slope = –KE/2.303
dt
Sigma-Minus Method
In sigma-minus method, from equation (84):
dX u KEt
K e XO e
dt ……(86)
dX u KEt
Or, K e XO e dt
dt
On integrating equation (86):
dX u K e XO e
KEt
dt
KEt
e
X u K e XO I ..... (87)
KE
Where, I = Integration constant whose value is obtained from initial conditions ,
i.e., t = 0, Xu = 0.
* *
162 Biopharmaceutics & Pharmacokinetics
Or, I K e X O e0 1
KE
...... (88)
If a drug is infused for a long period to attain steady -state in the plasma, the term
e K E t approaches zero and equation (96) becomes:
K R t K R
Xu e 0 e 2 0 ..... (97)
KE KE
KeR 0
Slope
KE
1/KE
t
Figure 5.17: Regular Plot of Xu versus t during Constant
Rate I.V. Infusion
dt
( Ka KE )
e
dX u K e K a FX O K E t
e K a t …..(100)
* *
164 Biopharmaceutics & Pharmacokinetics
A semi-log graph of Xu XO versus time (t) yields a bi -exponential curve. If K a >
KE, the slope of the terminal linear portion of the curve gives the valu e of K E for
the drug. Equation ( 104) is also used to estimate the absorption rate constant
(Ka) by the method of residuals.
The urinary excretion data obtained after administering a drug orally can be
analysed by the Wagner-Nelson method to evaluate K a by drawing %ARA plots.
In this method, urine samples are collected for sufficient time intervals to ensure
accurate estimation of K E (but should not be collected to time infinity). The
equation obtained relating % ARA with urine excretion rate is as follows:
X dX u / dt K E X u
% ARA 1 A 100 1 100 ..... (104)
Xu K E X u
A semi-log plot of %ARA versus t yields a straight line with slope –Ka/2.303.
Accurate values of K a are obtained from urine excretion data of drugs having
slow rate of absorption. Collecti on of urine samples at very short intervals of
time is difficult for drugs having rapid absorption rate.
5.4. SUMMARY
The details given in the chapter can be summarised as follows:
1) Pharmacokinetics is the study of rate processes invol ved in absorption,
distribution, metabolism, and excretion of a drug.
2) The word pharmacokinetic has been originated from the Greek word pharmakon
which means drug and kinesis which means motion or change of rate.
3) Absorption is the process of movement of u nchanged drug from the site of
administration to systemic circulation or to the site of measurement (i.e.,
plasma).
4) Distribution is the reversible transfer of a drug between the blood and the
extravascular fluids and tissues.
5) Elimination is the major proce ss of drug removal from the body and
termination of its action. It is defined as the irreversible loss of drug from the
body.
* *
Pharmacokinetics (Chapter 5) 165
23) Vd is the measure of the extent of drug distribution and is expressed in litres.
24) Clearance is the theoretical volume of body fluid contain ing the drug (i.e.,
that fraction of appare nt volume of distribution) from which the drug has
been completely removed in a given period of time.
25) Clearance at an individual organ level is termed organ clearance.
26) Planimeter is an instrument used for mechanically measuring the area of
plane figures drawn on rectilinear graph paper.
27) Absorption rate constant can be computed by the method of residuals (or
feathering, peeling, and stripping).
28) Wagner-Nelson method involves determination of K a from % unabsorbed -
time plots and does not require the assumpti on of zero - or first -order
absorption kinetics.
29) Loo-Reigelman method is used for determining the absorption rate constant
(Ka) from plasma concentration -time profile of a drug that obeys two -
compartment model.
30) In the rate of excretion method , the rate of urinary drug excretion (dX u/dt)
is directly proportional to the amount of drug in the body (X).
5.5. EXERCISE
5.5.1. True or False
1) The most traditional and common approach used for pharmacokinetic
characterisation of a drug is non-compartment analysis.
2) In mammillary model, the compartments are joined to each other in a chain or series
3) The total area under the curve from time zero to infinity is the zero moment of a drug
concentration in plasma versus time curve.
4) The total area under the curve obtained from a plot of product of drug concentration
and time versus time is the first moment of a plasma concentration-time profile.
5) Membrane permeation models rely on known anatomic and physiological data.
6) Vd is the measure of the extent of drug distribution and is expressed in hours.
* *
168 Biopharmaceutics & Pharmacokinetics
CHAPTER Multicompartment
6 Models
6.1.1. Introduction
One-compartment model describes the pharmacokinetic profile of many drugs. In
such case, instantaneous dist ribution equilibrium is assumed, and decline in the
amount of drug in the body with time is expressed as elimination by an equation
with a mono -exponential term. Instantaneous distribution, however, is not
possible for larger number of drugs and drug disposition is not mono -exponential
but bi- or multi-exponential. The reason for this is that the body is composed of a
heterogeneous group of tissues, each with different degree of blood flow, drug
affinity, and therefore different equilibration rates.
An ideal pharmacokinetic model should have a rate constant for each tissue
undergoing equilibrium (which is difficult mathematically). The best approach is
to combine the tissues together based on the similarity in their distribution
characteristics. Just like one -compartment models, drug disposition in multi -
compartment models is also assumed to follow first-order kinetics.
Metabolism Elimination
Compartment in Parallel
D1
VP C1 Metabolism
Ka Kel
D Output
Input
Vc C2 Elimination
D2
Figure 6.1: Open Two-Compartment Pharmacokinetic Model. (C1 -Concentration
in Compartment 1; D1 - Amount of Drug in Compartment 1; C2 -Concentration in
Compartment 2; D2 - Amount of Drug in Compartment 2).
Classification of a particular tissue (like brain) into central or peripheral
compartment depends on the drug ’s physicochemical properties. A drug that is
highly lipophilic can cross the BBB, and in this case brain will be included in the
central compartment. On the other hand, a polar drug ca nnot cross the BBB and
in this case brain in spite of being a highly perfused organ will be included in the
peripheral compartment.
Brain
Carcass
Liver
Bile
Gut Tissue
Gut
Lumen
Faecal Matter
Kidney
Skin
Hair
Urine
Figure 6.2: Nine-Compartment Model of Mercury
In figure 6.2, a nine-compartment research model for mercury is shown. Separate
compartments are included for major organ systems. Input to the model is oral
(into the gut) and excretion occurs through hair, urine, and faeces. The model
studied had 18 compartments, nine for each of two chemical forms of mercury.
With the help of computers, the mathematical equations for 9 and 18
compartment models can be easily so lved. However, these large models behave
in a more complex manner than the one- or two-compartment models.
KE
Central compartment
Peripheral compartment
Time
Figure 6.3: Changes in Drug Concentration in the Central (Plasma)
and the Peripheral Compartment after I.V. Bolus of a Drug that Fits
Two-Compartment Model
Drug concentration in the central compartment initially undergoes a rapid decline
due to drug distribution from the central to the peripheral compartment. This
phase is therefore termed the distributive phase . After a while, pseudo -
distribution equilibrium is attained between the two compartments, and then the
subsequent loss of drug from the central compartment becomes slow due to
elimination. This phase of slower rate process is termed the post-distributive or
elimination phase . Contrary to the central compartment, drug concentration in
the peripheral compartment f irst increases and reaches maximum in the
distribution phase. Thereafter, the drug concentration declines in the post -
distributive phase (figure 6.3).
Let K 12 and K 21 be the first -order distribution rate constants representing
reversible drug transfer bet ween the central and peripheral compartments; they
are termed microconstants or transfer constants. Let subscript c and p define
central and peripheral compartments, respectively. The rate of change in drug
concentration in the central compartment is expressed as:
dC c
K 21Cp K12Cc K ECc …..(1)
dt
On extending the relationship X = VdC to equation (1):
dC c K 21X p K12Xc K E Xc
….. (2)
dt Vp Vc Vc
Where,
Xc and Xp = Amountsof drug in the central and peripheral
compartments, respectively.
Vc and Vp = Apparent volumes of the central and peripheral compartments, respectively.
The rate of change in drug concentration in the peripheral compartment is
expressed as:
dC p
K12Cc K 21Cp …..(3)
dt
K X K 21X p
12 c …..(4)
Vc Vp
* *
172 Biopharmaceutics & Pharmacokinetics
Area under the plasma concentration-time curve (AUC) can be obtained as:
A B
AUC …..(16)
The apparent volume of central compartment (Vc) is given as:
X X0
Vd 0 …..(17)
C 0 K E AUC
The apparent volume of peripheral compartment (Vp) is given as:
VK
Vp c 12 …..(18)
K 21
The apparent volume of distribution at steady
-state or equilibrium(Vd,ss) is given as:
Vd,ss = Vc + Vp …..(19)
It is also given as:
X0
Vd ,area …..(20)
AUC
Total systemic clearance is given as:
ClT = Vd …..(21)
The pharmacokinetic parameters can be calculated from the urinary excretion
data as follows:
dX u
K e Vc …..(22)
dt
An equation identical to equation ( 6) can be derived for rate of excretion of
unchanged drug in urine:
dX u
K e Aet K e Be t …..(23)
dt
Equation ( 23) can be resolved into individual exponents by the method of
residuals as described for plasma concentration-time data.
Renal clearance is given as:
ClR = KeVc …..(24)
Methods of Residuals
This method aids in resolving the bi-exponential disposition curve, obtained after
intravenous bolus of a drug that follows two -compartment model, into its
individual exponents. On rewriting equation (9):
Cc Ae – t Be –t …..(25)
The bi -exponential curve in figure 6.4 shows that the initial decline due to
distribution is more rapid than the terminal decline due to elimination, i.e., the
rate constant >> , and hence the term e –t approaches zero more rapidly than
the does e–t. Thus, equation (25) reduces to:
C – Be –βt …..(26)
* *
174 Biopharmaceutics & Pharmacokinetics
Log B
Back extrapolated terminal portion of elimination
phase (log C values)
True plasma concentration curve (log C values)
Residual line (log Cr values)
Slope = – /2.303
Slope = – /2.303
Time
Figure 6.4: Resolution of Bi-exponential Plasma Concentration-Time Curve
by the Method of Residuals for a Drug that Follows Two-Compartment
Kinetics on I.V. Bolus Administration
A semi-log plot of C versus t yields the terminal linear phase of the curve with
slope –/2.303. On back extrapolating to time zero, y -intercept log B is yielded
(figure 6.4). The t ½ for the elimination phase can be obtained from t ½ = 0.693/.
On subtracting extrapolated plasma concentration values of the elimination phase
[equation ( 26)] from the corresponding true plasma concentration values
[equation (25)], a series of residual concentration values (Cr) are obtained.
Cr C – C Ae– t …..(28)
1 K1 2
RO
Central 2 Peripheral
Compartment K2 Compartment
1
KE
The second and the third term in the bracket becomes zero at steady -state (i.e., at
time infinity), and equation (30) reduces to:
R0
Css ..... (31)
Vc K E
The loading dose (X0,L) to obtain Css immediately at the start of infusion can be
calculated from equation (33):
R
X0, L CssVc 0 ..... (33)
KE
Ka 1 K12 2
Central Peripheral
Compartment K21 Compartment
KE
The rate of change in concentration of a drug that enters the body by first -order
absorption process and distributes according to the two-compartment model in
the central compartment is described by an absorption exponent, and the two
usual exponents describing drug disposition. The plasma concentration at any
time (t) is given by equation (34):
C = Ne–Kat + Le–t + Me–t ..... (34)
C = Absorption exponent + Distribution exponent + Elimination exponent
Where, Ka, and have usual meanings; while L, M and N are coefficients.
C oP
A
B
Plasma drug concentration
Slope ( 0.434 )
2.303
Slope ( 0.434 )
2.303
Ka
Slope
2.303
Time
Figure 6.5: Semi-log Plot of Plasma Drug Concentration versus Time for
Two Compartment Model after Extravascular Administration
* *
176 Biopharmaceutics & Pharmacokinetics
When the first dose is administered in an oral multiple dosing regimen, the
plasma drug concentration increases and reaches the peak and then declines.
When the second d ose is administered, plasma drug concentration again
increases to reach a level higher than the first dose, and continues to increase till
a steady state plasma drug concentration is achieved (figure 6.6).
Plasma drug concentration
C
Time
Figure 6.6: Plasma Drug Concentration-Time Profile after Multiple
Dosing Regimen (C – Average Steady State Plasma Drug Concentration)
At steady state, the drug input and o utput will be equal. The amount of drug that
accumulates in the body relative to the first dose is calculated as follows:
1
R ( 0.693 ) / t 1
…..(35)
1 e /2
From first-order kinetics, approximately 90% of steady state will be reached within
four half-lives. However, the time required to reach the steady state depends on the
) depends on:
drug’s half-life. Average steady state plasma drug concentration (C
1) Maintenance dos (X0),
2) Fraction of the dose absorbed (F),
3) Dosing interval (), and
4) Clearance (Cl).
F.X 0
C' …..(36)
Cl
1.44 F X 0 t1/2
C'
Vd τ
AUC
…..(37)
Where, Vd = Volume of distribution.
ACU = Area under the plasma drug concentration-time curve after single
maintenance dose.
From equations (36) and (37) maintenance dose can be calculated as:
C' Cl
X0
F
C' V
…..(38)
1.44 F t1 / 2
Where, the coefficient 1.44 is the reciprocal of 0.693, while AUC is the are a
under the curve after a single maintenance dose. Equation ( 44) is used for
determining the maintenance dose of a drug to achieve a desired concentration.
Since X = VdC, the body content at steady state is given as:
1.44FX 0 t1 / 2
Xss, av ….(45)
These average values are not arithmetic mean of C ss,max and C ss,min as the plasma
drug concentration undergoes exponential decline.
Equation (47) is true when Ka >> K E and the drug is distributed rapidly. If the
drug is given in travenously or if drug is absorbed rapidly, the absorption phase
can be neglected and equation (47) becomes:
X 0, L 1
R ac
X0
1 e K E
….. (48)
Loading Dose
The ratio of loading dose to maintenance dose (X 0,L/X0) is termed dose ratio. As
a rule, when
1) = t1/2, dose ratio = 2.0.
2) > t1/2, dose ratio < 2.0.
3) < t1/2, dose ratio > 2.0.
Figure 6.7 shows that if loading dose is not optimum, and is either too low or too
high, the steady state is attained in five half -lives just as when no loading dose is
given. Loading dose X0,L Maintenance doses X0
X0 X0 X0 X0
Plasma drug concentration
MSC
Dose ratio = 2
Dose ratio < 2
MEC
In drugs with short half -life (< 2.5 hours) and narrow therapeutic i ndex, e.g.,
heparin, it is not easy to maintain such a level as the dosing frequency must
be less than the half -life. However, some drugs like penicillin with half -life =
0.9 hours may be given less frequently (in every 4 -6 hours) but the
maintenance dose should be larger so that the plasma concentration is
maintained above the minimum inhibitory level.
* *
180 Biopharmaceutics & Pharmacokinetics
A drug with intermediate half-life (3-8 hours) but low therapeutic index should
be given at intervals less than or equal to half -life. A drug having high
therapeutic index should be given at intervals between 1 to 3 half -lives. A drug
with long half -life (> 8 hours) should be giv en once every half -life. Steady state
in such a case can be rapidly attained by administering a loading dose.
A drug with very lon g half-life (> 24 hours), e.g., amlodipine, should be given
once in 24 hours.
6.2. SUMMARY
The details given in the chapter can be summarised as follows:
1) In multi-compartment models, blood/plasma and the highly perfused tissues
(like brain, heart, lung, liver, and kidneys) form the central compartment.
2) In multi -compartment models, drugs administered via intravenous route are
directly introduced into the central compartment.
3) In multi -compartment models, irreversible drug elimination by hepatic
biotransformation or renal excretion occurs from the central compartment.
4) In multi -compartment models, after drug equilibration between central and
peripheral compartments, drug elimination follows first-order kinetics.
5) In multi -compartment models, peripheral compartment c annot be accessed
for direct measurement, and it is not a site of drug elimination or clearance.
6) Central compartment or compartment 1 comprises of blood and highly
perfused tissues (like liver, lungs, kidneys, etc.) that rapidly attain
equilibrium with the drug. Elimination occurs from this compartment.
7) Peripheral or tissue compartment or compartment 2 comprises of poorly
perfused and slow equilibrating tissues (like muscles, skin, adipose, etc.), and
is a hybrid of several functional physiologic units.
8) Methods of residuals aids in resolving the bi -exponential disposition curve,
obtained after intravenous bolus of a drug that follows two -compartment
model, into its individual exponents.
9) The time required to reach steady state depends on the drug’s half-life.
10) If K a >> K E, the drug reaches the plateau in approximately five half -lives;
this is called the plateau principle, which also means that K E determines the
rate at which the multiple dose steady state is reached.
11) Fluctuation is the ratio of C max to C min. Greater the ratio, greater is the
fluctuation.
12) The ratio of loading dose to maintenance dose (X0,L/X0) is termed dose ratio.
13) A drug with intermediate half-life (3-8 hours) but low therapeutic index
should be given at intervals less than or equal to half-life.
14) A drug having high therapeutic index should be given at intervals between 1
to 3 half-lives.
15) A drug with long half-life (> 8 hours) should be given once every half-life.
16) A drug with very long half -life (> 24 hours), e.g., amlodipine, should be
given once in 24 hours.
* *
Multicompartment Models (Chapter 6) 181
6.3. EXERCISE
Answers
1) False 2) False 3) True
4) False 5) True 6) Central compartment
7) Peripheral compartment 8) Half-life 9) Dose ratio
10) > 8 hours
* *
182 Biopharmaceutics & Pharmacokinetics
CHAPTER Non-Linear
7 Pharmacokinetics
In some cases, the rate process of a drug’s ADME depends on carrier or enzymes
that are substrate -specific, have definite capacities, and are susceptible to
saturation at high drug concentration. In these cases, first -order kinetics
transform into a mixture of first -order and zero -order rate processes and the
pharmacokinetic parameters alter with the administered dose. Pharmacokinetics
of such drugs is dose -dependent. Other synonymous terms are mixed -order, non-
linear, and capacity -limited kinetics. Drugs wit h such a pharmacokinetic profile
give rise to variability in pharmacological response.
Drug Absorption
Non-linearity in drug absorption occurs due to the following reasons:
1) When Absorption is Solubility or Dissolution Rate -Limited: When a
drug, e.g., griseofulvin, is administered at higher doses, a saturated solution
of the drug is formed in the GIT or at any other extravascular site and the
absorption rate attains a constant value.
2) When Absorption Involves Carrier -Mediated Transport Systems:
Saturation of the transport system at higher doses of vitamins, e.g.,
riboflavin, ascorbic acid, cyanocobalamin, etc., causes non-linearity.
3) When Pre -Systemic Gut Wall or Hepatic Metabolism Attains
Saturation: Saturation of pre -systemic metabolism of drugs, e.g.,
propranolol, hydralazine and verapamil, at high doses increases
bioavailability.
* *
Non-Linear Pharmacokinetics (Chapter 7) 183
The parameters, F, K a, C max and AUC, undergo a decrease in the first two cases
and undergo an increase in the last case. Changes in gastric emptying and
gastrointestinal blood flow and other physiological factors are some other causes
of non -linearity in drug absorption. Non -linearity in drug absorption is of little
consequence unless availability is significantly affected.
Drug Distribution
Non-linearity in distribution of drugs administered at high doses occurs due to
the following reasons:
1) Saturation of Binding Sites on Plasma Proteins: A fixed number of
binding sites are present on plasma proteins for a particular drug, e.g.,
phenylbutazone and naproxen; therefore, with increase in drug concentration,
the fraction of unbound drug also increases.
2) Saturation of Tissue Binding Sites: With large single bolus doses or
multiple dosing of drugs, e.g., thiopental and fentanyl, the tissue storage sites
undergo saturation.
In both the cases, the free plasma drug concentration increases; however, Vd
increases in the first case and decreases in the last one. Clearance also alters
depending on the extraction ratio of the drug. Clearance of a drug with high
Extraction Ratio ( ER) is increased due to saturation of binding sites. Unbound
clearance of dru gs with low ER is unaffected and the pharmacological response
increases.
Drug Metabolism
Non-linear kinetics of clinical importance is capacity -limited metabolism as
small changes in dose administered produce large variations in plasma drug
concentration at steady state. It is a major source of large inter -subject variability
in pharmacological response.
Saturation of enzyme decreases Cl H and increases C ss; while, the reverse is true
for enzyme induction. Other causes of non -linearity in metabolism are saturation
of binding sites, inhibitory e ffect of the metabolite on enzyme, and pathological
situations such as hepatotoxicity and changes in hepatic blood flow.
Drug Excretion
The two active processes in renal excretion of a saturable drug are:
1) Active Tubular Secretion: After the saturation of carrier system, a decrease
*
in renal clearance of drug, e.g., penicillin G, occurs. *
184 Biopharmaceutics & Pharmacokinetics
Km
C
Figure 7.1: Plot of Michaelis-Menten Equation [Elimination Rate (dC/dt) versus
Concentration (C)]. Initially, the rate increases linearly (first-order) with con-
centration, becomes mixed-order at higher concentration and then reaches
maximum (Vmax) beyond which it proceeds at a constant rate (zero-order)
2) When Km >> C: Under this condition,Km + C = Km and equation (1) reduce to:
dC Vmax C
….. (3)
dt Km
Equation (3) is similar to the one describing first -order elimination of a
drug, where V max/Km = KE. This indicates that the drug concentration in body
resulting from usual dosage regimens of most drugs is below the K m value of
elimination process (however, phenytoin and alcohol are certain exceptions).
3) When Km << C: Under this condition, Km + C = C andequation (1) reduces to:
dC
Vmax ….. (4)
dt
* *
Non-Linear Pharmacokinetics (Chapter 7) 185
Equation (4) is similar to the one describing a zero -order process, i.e., the
rate-process occurs at a constant rate (V max) and is independent of drug
concentration, e.g., metabolism of ethanol.
Log Co
t
Figure 7.2: Semi-log Plot of a Drug given as I.V. Bolus with Non-
Linear Elimination and that Fits One-Compartment Kinetics
A semi-log plot of C versus t yields a curve with a terminal linear portion having
slope –Vmax/2.303Km. Back extrapolation to time zero gives y -intercept of log
C 0 (figure 7.2). This line can be described by the following equation:
Vmax
log C log C 0 ….. (7)
2.303 K m
Equations (6) and (7) are similar at low plasma concentrations. On equating and
simplifying both the equations:
(C 0 C) C0
log ….. (8)
2.303K m C0
The value of Km can be obtained from equation (8). V max can be calculated by
substituting the value of Km in the slope value.
Vmax and Km can also be obtained by determining the rate of change of plasma drug
concentration at different times and by using the reciprocal equation
of (1). Thus:
1 Km 1
….. (9)
dC dt Vmax C m Vmax
* *
186 Biopharmaceutics & Pharmacokinetics
Equations (10) and (11) are rearrangements of equation (9) . Equation (10) is
used to pl ot C m/(dC/dt) versus Cm. Equation (11) is used to plot dC/dt versus
(dC/dt)/Cm. The K m and V max parameters can be determined from the slopes and
y-intercepts of the two plots.
Css
Km
Vmax
2 Vmax
DR
Figure 7.3: Curve for a Drug with Non-Linear Kinetics Obtained by
Plotting the Steady State Concentration versus Dosing Rates
Several measurements should be made at s teady state during dosage with
different doses in order to accurately define the characteristics of a curve.
* *
Non-Linear Pharmacokinetics (Chapter 7) 187
The parameters Km and Vmax can be graphically determined in the following ways:
1) Lineweaver-Burke Plot/Klotz Plot: On taking reciprocal of equation (13):
1 Km 1
….. (14)
DR Vmax C ss Vmax
Equation (14) is similar to equation (9). A plot of 1/DR versus 1/Css yields a
straight line with slope Km/Vmax and y-intercept 1/Vmax (figure 7.4):
1
DR Km
slope
Vmax
1
Vmax
1
C ss
Figure 7.4: Lineweaver-Burke/Klotz Plot for Estimation of Km
and Vmax at Steady State Concentration of Drug
2) Direct Linear Plot : A pair of C ss, viz., Css,1 and C ss,2 obtained with two
different dosing rates (DR1 and DR 2) is plotted (figure 7.5). The points C ss,1
and DR 1 are joined to form a line, and the points C ss,2 and DR 2 are joined to
form a second line. The intersection point of these two lines is extrapolated
on DR axis to obtain Vmax and on x-axis to obtain Km.
DR
Vmax
DR1
DR2
Css,1 Css,2 Km
Css 0 Km
Figure 7.5: Direct Linear Plot for Estimation of Km and Vmax at
Steady-State Concentrations of a Drug Given at Different Dosing Rates
3) Graphical Method : This method of estimating K m and V max involves re -
arranging equation (13) to yield:
K DR
DR Vmax m ….. (15)
Css
A plot of DR versus DR/Css yields a straight line with slope –Km and y -
intercept V max. The pa rameters K m and V max can also be determined
numerically by setting-up simultaneous equations:
Vmax C ss,1
DR 1 ….. (16)
K m C ss , 1
* *
188 Biopharmaceutics & Pharmacokinetics
Vmax C ss, 2
DR 2 ….. (17)
K m C ss , 2
The Time Required to Attain 90% of the True Steady State Plasma
Concentration for Phenytoin
The time required to attain 90% of the true steady state plasma concentration for
phenytoin administered at different rates, where V = 50 L, V max = 500mg/day,
and K m = 4μg/ml ( = 4mg/l) is to be determined. From equation ( 19), the time
required to attain 90% of the steady state concentration can be determined for
various daily doses.
* *
Non-Linear Pharmacokinetics (Chapter 7) 189
K mV
(2.303Vmax – 0.9R ) t 0.9. …..(19)
Vmax – R 2
For 100mg dose:
(4mg L–1 )(50L)
–1 2
(2.303[500mg day –1 ] – 0.9[100mg day–1]) = 1.33 days.
(400mg day )
If the value of K m was 5.7mg/l (and not 4mg/l), it can be said that since K m is the
numerator of equation ( 19), K m and the time to reach 90% of the steady state
phenytoin concentration are directly proportional. Similarly, 0.9(C p)ss is also
directly proportional to K m. Therefore, each value can be multiplied by the factor
(5.7/4.0) to obtain the figures given in table 7.2.
Table 7.1: Time (t0.9) to 90% of Steady State Plasma Concentration (C p)ss Level
as a Function of Daily Dose (R); K m = 4.0mg L–1
–1
R (mg day ) t0.9 (days) 0.9(Cp)ss (mg L–1)
100 1.33 0.90
200 2.16 2.40
300 4.41 5.10
400 15.8 14.4
Table 7.2: Time (t0.9) to 90% of Steady State Plasma Concentration (C p)ss Level
as a Function of Daily Dose (R); K m = 5.7 mg L–1
–1
R (mg day ) t0.9 (days) 0.9(Cp)ss (mg L–1)
100 1.90 1.28
200 3.08 3.42
300 6.28 7.27
400 22.5 20.5
* *
190 Biopharmaceutics & Pharmacokinetics
7.2. SUMMARY
The details given in the chapter can be summarised as follows:
1) After the saturation of carrier system, a decrease in renal clearance of drug,
e.g., penicillin G, occurs.
2) After the saturation of carrier system, an increase in renal clearance of drug,
e.g., water-soluble vitamins and glucose, occurs.
3) Michaelis-Menten equation describes the kinetics of capacity -limited or
saturable processes.
4) A double reciprocal plot or the Lineweaver-Burke plot of l/(dC/dt) versus
1/Cm yields a straight line with slope Km/Vmax and y-intercept 1/Vmax.
5) Lineweaver-Burke plot has a disadvantage that its points are clustered.
Thus, Hanes-Woolf plot and Woolf-Augustinsson-Hofstee plot are used
that give more reliable plots and uniformly scattered points.
7.3. EXERCISE
7.3.1. True or False
1) After the saturation of carrier system, a decrease in renal clearance of drug occurs.
2) After the saturation of carrier system, a decrease in renal clearance of drug occurs.
3) Michaelis-Menten equation describes the kinetics of capacity -limited or saturable
processes.
Answers
1) True 2) False 3) True 4) Km/Vmax and 1/Vmax
5) Lineweaver-Burke plot 6) Hanes-Woolf plot and Woolf-Augustinsson-Hofstee plot
Index
A L
Absorption of Drugs from Non per Oral Extra- Levels of IVIVC, 116
Vascular Routes, 31 Level A Correlation, 116
Apparent Volume of Distribution, 52, 145 Level B Correlation, 116
Area Under Curve, 147 Level C Correlation, 116
Level D Correlation, 117
B Loading and Maintenance Doses, 178
Binding of Drugs, 53 Loo –Riegelman Method, 157
Binding of Drugs to Blood Cells, 56
Bioavailability, 100, 128 M
Bioequivalence, 123, 130 Measurement of Bioavailability, 103
Bioequivalence Study Parameters, 124 Mechanisms of Drug Absorption, 10, 11, 12
Biopharmaceutics, 9, 40 Methods of Residuals, 173, 180
Applications, 9 Methods to Enhance the Dissolution Rates and
Bioavailability of Poorly Soluble Drugs,
C 108
Cell Drinking, 16, 41 Michaelis Menten Equation, 184
Clearance, 146 Multi-compartment models, 168
Clearance and Renal Clearance, 87
Clinical Significance of Protein Binding of N
Drugs, 64 Non-Compartmental Models, 137
Compartment Models, 135 Non-Linear Pharmacokinetics, 182
Catenary Model, 136, 165, 166 Non-Renal Routes of Drug Excretion, 93
D O
Distribution, 45, 65 One Compartment Model after IV Drug
Factors Affecting Drug Distribution, 51 Administration, 190
Drug Metabolism, 67 One-compartment open model, 142, 165
E P
Elimination Half-Life, 145 Pharmacokinetics, 133, 164
Elimination Rate Constant, 143 Phase I Metabolic Pathways, 70
Evaluation of Bioequivalence Data, 125 Phase II Metabolic Pathways, 82
Physiological Barriers, 48
F Placental Barrier, 50
Factors Affecting Protein-Drug Binding, 59 Physiological Models, 139
Factors Affecting Renal Excretion of Drugs, Advantages, 141
91 Disadvantages, 141
Factors Influencing Drug Absorption, 16 Types, 141
Pharmaceutical Factors, 26
Physicochemical Factors, 16 R
Fick’s first law of diffusion, 12, 40, 43 Rate of Excretion Method, 160
Regulatory Requirements for Bioequivalence
I Studies
In vitro Drug Dissolution Models, 117 Review, 132, 167, 181
In Vitro-In Vivo Correlation, 115 Renal Excretion of Drugs, 89
K S
Kinetics of Multiple Dosing, 176 Sigma-Minus Method, 161
Kinetics of Protein Binding, 62 Statistical Moment Theory, 138
Steady State Drug Levels, 177
* *
192 Biopharmaceutics & Pharmacokinetics
Bibliography
Chatwal G.R., Biopharmaceutics and Pharmacokinetics, Himalaya Pub.
Brahmankar D.M., Jaiswal Sunil B. , Biopharmaceutics and
Pharmacokinetics A Treatise, Vallabh Prakashan.
Venkateswarlu V., Khar Roop K., Biopharmaceutics and Pharmacokinetics,
PharmaMed Press.
Kulkarni J.S., Pawar A.P., Shedbalkar V.P., Biopharmaceutics and
Pharmacokinetics, CBS Publishers.
Madan P.L., Biopharmaceutics and Pharmacokinetics, Jaypee Brothers.
Gibaldi Milo, Biopharmaceutics and Clinical Pharmacokinetics , 4 th Edition,
PharmaMed Press.
Notari Robert E., Biopharmaceutics and Clinical Pharmacokinetics: An
Introduction, 4th Edition, Marcel Dekker Inc.
Kar Ashutosh, Essentials of Biopharmaceutics And Pharmacokinetics ,
Elseveir.
Shargel Leon, Yu Andrew, Wu-Pong Susanna, Applied Biopharmaceutics &
Pharmacokinetics, 6th Edition, McGraw Hill Professional.
* *