SlideShare a Scribd company logo
Prepared By: Dolly Sadrani
Department of Pharmaceutics
IInd Sem M.Pharma
1
CONTENTS
 Plasma Drug Concentration Time Profile
 Pharmacokinetic Parameter
 Pharmacodynamic Parameter
 Zero, First Order & Mixed Order Kinetic
 Rates & Order Of Kinetics
 Pharmacokinetic Models
 Application Of Pharmacokinetic
 Reference
 Questions
2
Pharmacokinetics
 Pharmacokinetics is define as the kinetics of
drug absorption, distribution, metabolism and
excretion (KADME) and their relationship with
the pharmacologic, therapeutic or toxicologic
response in human and animals.
 The applications of pharmacokinetic principles
in the safe and effective management of
individual patient is called as clinical
pharmacokinetics.
3
Pharmacodynamics
 The branch of pharmacology concerned with the
effects of drugs and the mechanisms of their
action.
 In particular, pharmacodynamic is the study how
a drug affects an organism, whereas
pharmacokinetics is the study of how the
organism affects the drug.
4
Plasma Drug Concentration-Time
Profile
 A Direct relationship exists between the
Concentration of drug at the site of action
(biophase) and the concentration of drug in
plasma.
 A typical plasma drug concentration- time curve
obtained after a single oral dose of a drug and
showing various pharmacokinetic and
pharmacodynamic parameters.
5
 Such a profile can be obtained by measuring the
concentration of drug in plasma sample taken at
various intervals of time after administration of
a dosage form and plotting the concentration of
drug in plasma (Y-axis) versus the
corresponding time at which the plasma sample
was collect (X-axis).
6
7
Pharmacokinetic Parameter
 Pharmacokinetic Parameter that describe the
plasma level-time curve and useful in
determining the bioavailability of a drug from
formulation are:
1. Peal Plasma Concentration (Cmax): The point of
maximum Concentration of drug in plasma is
called as the peak and the Concentration of
drug at peak is known as peak plasma
Concentration.
o It is also called as peak high Concentration and
maximum drug Concentration.
8
o Cmax is expressed in mcg/ml.
o The peak level depends upon the administered
dose and rate of absorption and elimination.
o The peak represents the point of time when
absorption rate equal to elimination rate of drug.
9
2. Time of peak Concentration (tmax)
o The time for drug to reach peak Concentration in
plasma is called as the time of peak
Concentration.
o It is expressed in hours and is useful in
estimating the rate of absorption.
o Onset time and Onset of action are dependent
upon tmax.
10
3. Area Under the Curve (AUC)
o It represents the total integrated area under the
plasma level-time profile and expresses the total
amount of drug that comes into the systemic
circulation after its administration.
o AUC is expressed in mcg/ml . Hours
o It is the most important parameter in evaluating
the bioavailability of a drug from its dosage
form as it represents the extent of absorption.
11
o AUC is also important for drugs that are
administered repetitively for the treatment of
chronic conditions like asthma and epilepsy.
12
Pharmacodynamic Parameter
1. Minimum Effective Concentration (MEC)
o It is define as the minimum Concentration of
drug in plasma required to produce the
therapeutic effect.
o It reflects the minimum Concentration of drug at
the receptor site to elicit the desired
pharmacologic response.
o The Concentration of drug below the MEC is
said to be in the subtherapeutic level.
13
o In case of antibiotics , the term minimum
inhibitory Concentration (MIC) is used. It
describe the minimum Concentration of
antibiotic in plasma required to kill or inhibit
the growth of microorganisms.
2. Maximum Safe Concentration (MSC)
o Also called as minimum toxic Concentration
(MTC) , it is the Concentration of drug in
plasma above which adverse or unwanted
effects are precipitated.
14
o Concentration of drug above MSC is said to be
in the toxic level.
3. Onset of Action
o The beginning of pharmacologic response is
called as onset of action.
o It occurs when the plasma drug Concentration
just exceed the required MEC.
15
4. Onset Time
o It is the required for the drug to start producing
pharmacologic response.
o It corresponds to the time for the plasma
Concentration to reach MEC after
administration of drug.
5. Duration of Action
o The time period for which the plasma
Concentration of drug remains above the MEC
level is called as duration of drug action.
16
6. Intensity of Action
o It is maximum pharmacologic response produced
by the peak plasma Concentration of drug.
o Also called as peak response.
7. Therapeutic Range
o The drug Concentration between MEC and MSC
represents the therapeutic range.
17
Rate, Rate Constant and Order of
Reaction
 Pharmacokinetic is the mathematical analysis of
processes of ADME.
 The movement of drug molecules from the site
of application to the systemic circulation,
through various barriers, their conversion in to
another chemical form and finally exit out of the
body can be expressed mathematically by the
rate at which they proceed, the order of such
processes and the rate constant.
18
 The velocity with which a reaction or a process
occurs is called as its rate.
 The manner in which the Concentration of drug
influences the rate of reaction or process is
called as the order of reaction or order or
process.
Drug A Drug B
 The rate of forward reaction is expressed as
- dA
dt
 Negative sign indicates that the Concentration of
drug A decreases with time T.
19
 As the reaction proceeds, the Concentration of
drug B increases and the rate of reaction can
also be expressed as:
dB
dt
 Experimentally, the rate of reaction is
determined by measuring the decreases in
Concentration of drug A with time t.
20
 If C is the Concentration of drug A, the rate of
decreases in C of drug A as it is changed to B can
be described by a general expression as a
function of time t.
dc = -K Cn
dt
where, K = rate constant
n = order of reaction
If n = 0, it’s a zero-order process,
If n = 1, it’s a first-order process
21
 The three commonly encountered rate processes
in a physiological system are-
- Zero-order process
- First-order process
- Mixed-order process.
 The pharmacokinetics of most drugs can be
adequately by Zero- and first-order processes.
22
Zero-Order Kinetics (Constant Rate
Processes)
If n= 0 dC = -K0 C0 = -K0 (1.0)
dt
where K0 = Zero-order rate constant (mg/ml)
 The Zero- Order process can be defined as the
one whose rate is independent of the
Concentration of drug undergoing reaction i.e.
the rate of reaction cannot be increased further
by increasing the Concentration of reaction.
23
Rearrangement of equation (1.0) yields:
dC = -K0 dt (1.1)
Integration of equation (1.1) gives:
C – C0 = -K0 t
or
C = C0 - K0 t (1.2)
where, C0 = Concentration of drug at t = 0
C = Concentration of drug yet to
undergo reaction time t.
24
 Equation (1.2) is that of a straight line and states
that the Concentration of reactant decreases
linearly with time.
 A plot of C versus t yields such a straight line
having slope – K0 and y- intercept C0.
25
Zero-Order Half-life
 Half-life (t1/2) or Half-time is defined as the time
period required for the Concentration of drugs to
decrease by one-half.
When t = t1/2
C = C0/2
and the equation (1.2) becomes :
C0 = C0 – k0 t1/2
2 (1.3)
26
Solving (1.3) we get :
t1/2 = C0 = 0.5 C0
2 K0 K0 (1.4)
Equation (1.4) shows that the t1/2 of a Zero-order
process is not constant but proportional to the
initial Concentration of drug C0 and inversely
proportional to the Zero-order rate constant K0.
 Zero-order equation do not require logarithmic
transformation.
27
Examples of Zero-order processes are :
 Metabolism/ protein-drug binding and enzyme or
carrier mediated transport under saturated
conditions. The rate of metabolism, binding or
transport of drug remains constant as long as its
Concentration is in excess of saturating
Concentration.
 Administration of a drug as a constant rate i.v.
infusion
28
 Controlled drug delivery such as that form i.m.
implants or osmotic pump.
First-Order Kinetic (Linear Kinetics)
If n=1 Equation (1. ) becomes
dC = K C
dt (1.5)
Where, K= First order rate constant (in time-1 or
per hour)
29
 From Equation it is clear that a first-order
process is the one whose rate is directly
proportional to the Concentration of drug
undergoing reaction. (greater the Concentration,
faster the reaction.)
 First-order process is said to be follow linear
kinetics.
30
Rearrangement of equation (1.5) yields:
dC =-Kdt
C (1.6)
Integration of equation (1.6)
ln C = ln C0 –Kt (1.7)
Equation (1.7) can also be written in exponential
form as:
C = C0 e-Kt (1.8)
Where e = natural log base.
31
 The First-Order process is also called as
monoexponential rate process. Thus, a first-order
process is characterized by logarithmic or
exponential kinetics i.e. A constant fraction of
drug undergoes reaction per unit time.
since ln = 2.303 log, equation (1.7) can be written
as
log C = log C0 – Kt
2.303 (1.9)
 A semilogarithmic plot of equation (1.9) yields a
straight line with slope = - K/2.303 and y –
intercept = log C0
32
33
First-Order Half-Life
 Substituting the value of C = C0/2 at t1/2 in
equation (1.9) and solving it yields :
t1/2 = 0.693
K (1.10)
Above equation shows that, in contrast to Zero-
Order process, the Half-life of a First-order
process is constant and independent of initial
drug Concentration i.e. Irrespective of what the
initial drug Concentration is, the time required
for the Concentration to decreases by One-half
remains the same.
34
 Most pharmacokinetic processes viz.
Absorption, distribution and elimination follow
first-order kinetics.
Mixed-Order Kinetics (Nonlinear Kinetics)
 A mixture of both first-order and zero-order
kinetics is observed in such cases and therefore
the process is said to be follow mixed-order
kinetics.
35
 Deviation from an originally linear
pharmacokinetic profile are observed , the rate
process of such a drug is called as nonlinear
kinetics.
 Mixed-order kinetics is also termed as dose-
dependent kinetics as it is observed at increased
or multiple doses of some drugs.
 Observed in :
- Drug absorption (Vitamin C)
- Drug distribution ( naproxen)
- Drug elimination (riboflavin)
36
Pharmacokinetic Analysis Of
Mathematical Data:
Pharmacokinetic Models
 Drug movement with in body is a complex
process.
 The two major approaches in the quantitative
study of various kinetic processes of drug
disposition in the body are :
- Model Approach
- Model-independent approach (Non
compartmental analysis)
37
Methods for analysis of Pharmacokinetic Data
Model Approach Model-Independent Approach
Compartment Non Compartmental Analysis
model
- Mammillary
- Catenary
Physiological
model
- Perfusion-limited
- Diffusion-limited
Distributed Parameter
model
38
Pharmacokinetic model Approach
 Model are used to describe change in drug
concentration in the body with time.
 A model is a hypothesis that employs
mathematical terms to concisely describe
quantitative relationship.
 Pharmacokinetic models provide concise means
of expressing mathematically or quantitatively,
the time course of drug throughout the body and
compute meaningful pharmacokinetic
parameter.
39
Application of Pharmacokinetic
Models:
 Characterizing the behaviour of drugs in
patients.
 Predicting the concentration of drug in various
body fluids with any dosage regimen.
 Predicting the multiple-dose concentration
curves from single dose experiments.
 Calculating the optimum dosage regimen for
individual patients.
40
 Evaluating the risk of toxicity with certain
dosage regimens.
 Correlating plasma drug Concentration with
pharmacological response.
 Evaluating the bioequivalence/ bioinequivalence
between different formulation of the same drug.
 Estimating the possibility of drug and/or
metabolites accumulation in the body.
 Determining the influence of altered
physiology/disease state on /drug ADME.
 Explaining drug interaction.
41
Types Of Pharmacokinetic Models:
1. Compartment models: Also called as empirical
models
2. Physiological models: are realistic models
3. Distributed parameter model: are also realistic
models
42
Compartment Models
 These models simply interpolate the
experimental data and allows an empirical
formula to estimate the drug concentration with
time
 Compartments are hypothetical in nature and
based on certain assumptions:
- The body is represented as a series of
compartments arranged either in series or
parallel to each other, which communicate
reversibly with each other.
43
- Each compartment is not a real physiological
and anatomical region but a fictitious or virtual
one and considered as a tissue or group of
tissues that have similar drug distribution
characteristics. (Similar blood flow and affinity).
- Within each compartment, the drug is
considered to be rapidly and uniformly
distributed i.e. The compartment is well-stirred.
- The rate of drug movement between
compartment is described by first-order kinetic.
- Rate constants are used to represent rate of entry
into and exit from the compartment.
44
Compartment models are divided in to:
Mammillary model
Catenary model
Mammillary model:
 It consist of one or more peripheral
compartment connected to the central
compartment.
 The central compartment comprises of plasma
and highly perfused tissues such as lungs, liver,
kidneys which rapidly equilibrate with the drug.
 The drug is directly absorbed into this
compartment. 45
 The peripheral compartments or tissue
compartment are those with low vascularity and
poor perfusion.
 Distribution of drugs to this compartments is
through blood.
 Movement of drug between compartments is
defined by characteristic first-order rate constant
denoted by K.
 The subscript indicated the direction of drug
movement K12 ( K-one-two) refers to drug
movement from compartment 1 to compartment
2 and reverse for K21.
46
 The number of rate constants which will appear in
particular compartment model is given by R.
For intravenous administration R=2n-1 (1.11)
For extravascular administration R = 2n (1.12)
Where n= no. Of compartments.
47
48
Catenary Model:
 In this model, the compartments are joined to
one another in a series like compartments of a
train.
 This is not observable physiologically/
anatomically as the various organ are directly
linked to the blood compartment.
 Hence this model is rarely used.
49
Advantages and Application of
compartment modelling Approach
 It is a simple and flexible approach and thus
widely used.
 It gives a visual representation of various rate
processes involved in drug disposition.
 It shows how many rate constants are necessary
to describe these processes.
 It enables the pharmacokinetic to write
differential equations for each of the rate
processes in order to describe drug
concentration change in each compartment.
50
 It enables monitoring of drug concentration
change with time with a limited amount of data.
 It is useful in predicting drug concentration-time
profile in both normal physiological and in
pathological condition.
 It is important in the development of dosage
regimen.
 It is useful in relating plasma drug levels to
therapeutic and toxic effects in the body.
 It is particularly useful when several therapeutic
agent are compared. Clinically, drug data
comparisons are based on compartment models.
51
 Its simplicity allows for easy tabulation of
parameters such as Vd , t1/2.
Disadvantages:
 Extensive efforts are required in the
development of an exact model that predicts and
describes correctly the ADME of a certain
drugs.
 The model is based on curve fitting of plasma
concentration with complex multiexponential
mathematical equations.
 The model may vary within a study population.
52
 The approach can be applied only to a specific
drug under study.
 The drug behaviour with in the body may fit
different compartmental models depending upon
the route of administration.
 Difficulties generally arise when using models to
interpret the differences between results from
human and animal experiments.
 Owing to their simplicity, compartmental
models are often misunderstood, overstretched or
even abused.
53
Physiological models:
 These models are also known as physiologically
based pharmacokinetic models (PB- PK models).
 The number of compartments to be included in
the model depends upon the disposition
characteristics of the drug.
 organ or tissues such as bones that have no drug
penetration are excluded.
 For example lungs, liver, brain and kidney are
grouped as rapidly equilibrating tissues (RET)
while muscles and adipose as slowly
equilibrating tissues (SET).
54
 The rate of drug carried to a tissue/organ or
tissue drug uptake is dependent upon two major
factors:
- rate of blood flow to the organ
- tissue/blood partition coefficient or diffusion
coefficient of drug that governs its tissue
permeability.
 Physiological models are further categorized
into two types:
- Blood flow rate limited models/perfusion rate
limited model : Based on the assumption that the
drug movement within a body region is much
more rapid than its rate of delivery to that region
by the perfusing blood.
55
56
Example, thiopental, lidocaine
- Membrane permeation rate-limited model/
Diffusion limited model: these models are more
complex and applicable to highly polar, ionized
and charged drug, in which case the cell
membrane acts as barrier for the drug that
gradually permeates by diffusion.
57
Advantages over the conventional
compartment modelling
 Mathematical treatment is straightforward.
 Since it is a realistic approach, the model is
suitable where tissue drug concentration and
binding are known.
 Data fitting is not required since drug
concentration in various body regions can be
predicted on the basis of organ or tissue size,
perfusion rate and experimentally determined
tissue-to-plasma partition coefficient.
58
 The model gives exact description of drug
concentration-time profile in any organ or tissue
and thus better picture of drug distribution
characteristics in the body.
 The influence of altered physiology or
pathology on drug disposition can be easily
predicted from changes in the various
pharmacokinetic parameters since the
parameters corresponds to actual physiological
and anatomical measures.
 The method is frequently used in animal because
invasive methods can be used to collect tissue
sample.
59
 Correlation of data in several animal species is
possible and with some drugs, can be
extrapolated to human since tissue concentration
of drug is known.
 Mechanism of ADME of drug can be easily
explained by this model.
Disadvantages:
 Obtaining the experimental data is a very
exhaustive process.
 Most physiological model assume an average
blood flow for individuals subjects and hence
predictions of individualized dosing is difficult.
60
 The number of data points is less than the
pharmacokinetic parameters to be assessed.
 Monitoring of drug concentration in body is
difficult since exhaustive data is required.
61
 Hypothetical /
empirical approach - no
relation with real
physiology or anatomy.
 Experimentally simple
and flexible approach
as far as data collection
is concerned.
 Owing to its simplicity,
it is widely used and is
often the First-model.
 Realistic approach
since it is based on
physiological and
anatomic information.
 Difficult
experimentally since
exhaustive data
collection is required.
 Less commonly used
owing to complexity.
62
Compartment modelling Physiological modelling
 Complex multiexponential
mathematical treatment is
necessary for curve fitting.
 Data fitting is required for
predicting drug
concentration in particular
compartment.
 Used when there is little
information about tissue
 Easy to monitor time
course of drug in body
with limited data.
 mathematical treatment is
straight forward.
 Data fitting is not required
for since drug
concentration in various
tissue is practically
determined.
 Used where tissue drug
concentration binding is
known.
 Exhaustive data is
required to monitor tome
course of drug in body.
63
 Extrapolation from data to
human and viceversa is
not possible.
 Mechanism of drugs
ADME can not be
explained.
 Effect of pathological
condition on drug ADME
can not be determined.
 Frequently used for data
comparison of various
drug.
 Extrapolation of animal
data to human is easy on
the basis of tissue
concentration of drugs.
 Easy to explained drugs
ADME mechanisms.
 Effect of pathology on
drug ADME can be easily
determined.
 Less commonly used for
data comparisons.
64
Distributed Parameter Model
 This model is analogous to physiological model
but has been designed to take in to account:
- Variation in blood flow to an organ,
- Variation in drug diffusion in an organ.
 Such a model is thus specifically useful for
assessing regional differences in drug
concentration in tumours or necrotic tissue.
65
Noncompartmental Analysis
 Also called as model-independent method dose
not required the assumption of specific
compartment model.
 Based on the assumption that the drugs and
metabolites follow linear kinetics and on the
basis, this technique can be applied to any
compartment model.
 The noncompartmental approach, based on the
statistical moments theory involves collection of
experimental data following a single dose of
drug.
66
 If one considers the time course of drug
concentration in plasma as a statistical
distribution curve then,
MRT = AUMC
AUC (1.13)
where
MRT = mean residence time
AUMC = Area under the First-moment curve
AUC = Area under the Zero-moment curve
67
 AUMC is obtained from a plot of product of
plasma drug concentration and time versus time
t from zero to infinity.
 Mathematically it is expressed by equation:
AUMC = t dt (1.14)
 AUC is obtained from a plot of plasma drug
concentration versus time t from zero to infinity.
 Mathematically it is expressed by equation:
AUC = t dt (1.15)
68
69
 Practically, the AUMC and AUC can be
calculated from the respective graphs by the
trapezoidal rule.
 MRT is defined as the average amount of time
spent by the drug in the body before being
eliminated.
Application:
 Used to estimate the important pharmacokinetic
parameters like bioavailability, clearance and
apparent volume of distribution.
70
 Useful in determining half-life, rate of
absorption and first-order absorption rate
constant of the drug.
Advantages:
 Ease of derivation of pharmacokinetic
parameters by simple algebraic equations.
 The same mathematical treatment can be applied
to almost any drug metabolic provided they
follow first-order kinetics.
71
Disadvantages:
 It provides limited information regarding the
plasma drug concentration-time profile.
 The method does not adequately treat non-linear
cases.
72
Questions:
1. Explain the term ‘compartment’ what are the
assumption made in compartment modelling.
Discuss the advantages and disadvantages of
same.
2. What are pharmacokinetic models. Discuss the
importance and types of such model.
3. Give detailed classification of compartment.
73
Reference:
 Biopharmaceutics and clinical Pharmacokinetics
by Milo Gibaldi and Donald Perrier, fourth
edition philadelphia, lea and febiger,1991
 Biopharmaceutics and Pharmacokinetics by PL
MADAN Second edition, Jaypee Brothers
Medical Publishers (P) LTD.
 Biopharmaceutics and Pharmacokinetics A
TREATISE by D. M. Brahmankar, Sunil B.
Jaiswal, Vallabh Prakashan.
 Applied Biopharmaceutics and
Pharmacokinetics by shargel. Land yu Abc,
Second edition applet on century crofts, 1985.
74
Thank you For
Your
Attention!
75

More Related Content

What's hot

METHOD OF RESIDUALS
METHOD OF RESIDUALSMETHOD OF RESIDUALS
METHOD OF RESIDUALS
Divya Pushp
 
Drug Absorption from GI Tract
Drug Absorption from GI TractDrug Absorption from GI Tract
Drug Absorption from GI Tract
silambarasan I
 
Physiological pharmacokinetic models
Physiological pharmacokinetic modelsPhysiological pharmacokinetic models
Physiological pharmacokinetic modelsSanjay Yadav
 
Drug Dissolution
Drug DissolutionDrug Dissolution
Drug Dissolution
Sidharth Mehta
 
Multiple Dosage regimen
Multiple Dosage regimenMultiple Dosage regimen
Multiple Dosage regimen
Md. Mohabbot Hossen
 
Methods For Assesment Of Bioavailability
Methods For Assesment Of Bioavailability Methods For Assesment Of Bioavailability
Methods For Assesment Of Bioavailability
Anindya Jana
 
Non linear Pharmacokinetics
Non linear PharmacokineticsNon linear Pharmacokinetics
Non linear Pharmacokinetics
Areej Abu Hanieh
 
Drug absorption from GIT
Drug absorption from GITDrug absorption from GIT
Drug absorption from GIT
Mahewash Sana Pathan
 
Application of pharmacokinetics
Application of pharmacokineticsApplication of pharmacokinetics
Application of pharmacokinetics
Christy George
 
In-Vivo In-Vitro Correlation
In-Vivo In-Vitro CorrelationIn-Vivo In-Vitro Correlation
In-Vivo In-Vitro Correlation
dilip kumar tampula
 
Drug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal TractDrug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal Tract
Feba Elsa Mathew
 
P h partition hypothesis
P h partition hypothesisP h partition hypothesis
P h partition hypothesis
Zahid1392
 
study design for bioavailability and bioequivalence
study design for bioavailability and bioequivalencestudy design for bioavailability and bioequivalence
study design for bioavailability and bioequivalence
Priya Rajput
 
In vitro Dissolution Testing Models
In vitro Dissolution Testing ModelsIn vitro Dissolution Testing Models
In vitro Dissolution Testing Models
Ameer Ahmed
 
bioequivalence study design
bioequivalence study designbioequivalence study design
bioequivalence study design
Juhi Priya
 
IVIVC
IVIVCIVIVC
Bioequivalence 112070804009
Bioequivalence  112070804009Bioequivalence  112070804009
Bioequivalence 112070804009Patel Parth
 

What's hot (20)

METHOD OF RESIDUALS
METHOD OF RESIDUALSMETHOD OF RESIDUALS
METHOD OF RESIDUALS
 
Drug Absorption from GI Tract
Drug Absorption from GI TractDrug Absorption from GI Tract
Drug Absorption from GI Tract
 
Physiological pharmacokinetic models
Physiological pharmacokinetic modelsPhysiological pharmacokinetic models
Physiological pharmacokinetic models
 
Pharmacokinetic models
Pharmacokinetic modelsPharmacokinetic models
Pharmacokinetic models
 
Drug Dissolution
Drug DissolutionDrug Dissolution
Drug Dissolution
 
Multiple Dosage regimen
Multiple Dosage regimenMultiple Dosage regimen
Multiple Dosage regimen
 
Non linear pharmacokinetics
Non linear pharmacokineticsNon linear pharmacokinetics
Non linear pharmacokinetics
 
Methods For Assesment Of Bioavailability
Methods For Assesment Of Bioavailability Methods For Assesment Of Bioavailability
Methods For Assesment Of Bioavailability
 
Non linear Pharmacokinetics
Non linear PharmacokineticsNon linear Pharmacokinetics
Non linear Pharmacokinetics
 
Drug absorption from GIT
Drug absorption from GITDrug absorption from GIT
Drug absorption from GIT
 
Application of pharmacokinetics
Application of pharmacokineticsApplication of pharmacokinetics
Application of pharmacokinetics
 
In-Vivo In-Vitro Correlation
In-Vivo In-Vitro CorrelationIn-Vivo In-Vitro Correlation
In-Vivo In-Vitro Correlation
 
Drug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal TractDrug Absorption from the Gastrointestinal Tract
Drug Absorption from the Gastrointestinal Tract
 
P h partition hypothesis
P h partition hypothesisP h partition hypothesis
P h partition hypothesis
 
study design for bioavailability and bioequivalence
study design for bioavailability and bioequivalencestudy design for bioavailability and bioequivalence
study design for bioavailability and bioequivalence
 
In vitro Dissolution Testing Models
In vitro Dissolution Testing ModelsIn vitro Dissolution Testing Models
In vitro Dissolution Testing Models
 
Bioequivalence protocol
Bioequivalence  protocolBioequivalence  protocol
Bioequivalence protocol
 
bioequivalence study design
bioequivalence study designbioequivalence study design
bioequivalence study design
 
IVIVC
IVIVCIVIVC
IVIVC
 
Bioequivalence 112070804009
Bioequivalence  112070804009Bioequivalence  112070804009
Bioequivalence 112070804009
 

Similar to PHARMACOKINETICS: BASIC CONSIDERATION & PHARMACOKINETIC MODELS

Pharmacokinetics basics Introduced and applications
Pharmacokinetics basics Introduced  and applicationsPharmacokinetics basics Introduced  and applications
Pharmacokinetics basics Introduced and applications
Sumant Saini
 
1.pharmacokinetics
1.pharmacokinetics1.pharmacokinetics
1.pharmacokinetics
PNMallikarjun
 
Harvindar singh
Harvindar singhHarvindar singh
Harvindar singh
Harevindarsingh
 
kinetics (Lec 2).pptx
kinetics (Lec 2).pptxkinetics (Lec 2).pptx
kinetics (Lec 2).pptx
HebaYassin10
 
Basic concepts of pharmacokinetics
Basic concepts of pharmacokineticsBasic concepts of pharmacokinetics
Basic concepts of pharmacokinetics
Anand Addagalla
 
Basic concepts of pharmacokinetics
Basic concepts of pharmacokineticsBasic concepts of pharmacokinetics
Basic concepts of pharmacokinetics
Anand Addagalla
 
Pharacokinetics power point for pharmacy
Pharacokinetics power point  for pharmacyPharacokinetics power point  for pharmacy
Pharacokinetics power point for pharmacy
emebetnigatu1
 
Basic Pharmacokinetics & Compartments Modelling.pdf
Basic Pharmacokinetics & Compartments Modelling.pdfBasic Pharmacokinetics & Compartments Modelling.pdf
Basic Pharmacokinetics & Compartments Modelling.pdf
Ashwin Saxena
 
Basic pharmacokinetics and compartment modelling
Basic pharmacokinetics  and compartment modellingBasic pharmacokinetics  and compartment modelling
Basic pharmacokinetics and compartment modelling
PriyankaDabirBharadk
 
basicpharmacokineticsandcompatmentmodelling-171223090651.ppt
basicpharmacokineticsandcompatmentmodelling-171223090651.pptbasicpharmacokineticsandcompatmentmodelling-171223090651.ppt
basicpharmacokineticsandcompatmentmodelling-171223090651.ppt
TohfatulJinan1
 
TOXICOKINETICS
TOXICOKINETICSTOXICOKINETICS
TOXICOKINETICS
Koppala RVS Chaitanya
 
Pharmacokinetic models
Pharmacokinetic modelsPharmacokinetic models
Pharmacokinetic models
SURYAKANTVERMA2
 
Basics of Pharmacokinetics
Basics of PharmacokineticsBasics of Pharmacokinetics
Basics of Pharmacokinetics
Mohammed Asadullah Jahangir
 
Bioavailability studies lecture7
Bioavailability studies lecture7Bioavailability studies lecture7
Bioavailability studies lecture7homebwoi
 
Pocket Guide: Pharmacokinetics Made Easy - sample
Pocket Guide: Pharmacokinetics Made Easy - sample Pocket Guide: Pharmacokinetics Made Easy - sample
Pocket Guide: Pharmacokinetics Made Easy - sample
McGraw-Hill Education ANZ- Medical
 
Relationship between pharmacokinetics and pharmacodynamics.pptx
Relationship between pharmacokinetics and pharmacodynamics.pptxRelationship between pharmacokinetics and pharmacodynamics.pptx
Relationship between pharmacokinetics and pharmacodynamics.pptx
MdHimelAhmedRidoy1
 
Toxicokinetics in preclinical studies by Priyabrata Panda
Toxicokinetics in preclinical studies by Priyabrata PandaToxicokinetics in preclinical studies by Priyabrata Panda
Toxicokinetics in preclinical studies by Priyabrata Panda
Priyabrata Panda
 
2.pharmacokinetics
2.pharmacokinetics2.pharmacokinetics
2.pharmacokinetics
PNMallikarjun
 
Pharmacokinetic Models
Pharmacokinetic ModelsPharmacokinetic Models
Pharmacokinetic Models
IBNSINA National Medical College
 
Kinetics of stability studies
Kinetics of stability studiesKinetics of stability studies
Kinetics of stability studies
ROHIT
 

Similar to PHARMACOKINETICS: BASIC CONSIDERATION & PHARMACOKINETIC MODELS (20)

Pharmacokinetics basics Introduced and applications
Pharmacokinetics basics Introduced  and applicationsPharmacokinetics basics Introduced  and applications
Pharmacokinetics basics Introduced and applications
 
1.pharmacokinetics
1.pharmacokinetics1.pharmacokinetics
1.pharmacokinetics
 
Harvindar singh
Harvindar singhHarvindar singh
Harvindar singh
 
kinetics (Lec 2).pptx
kinetics (Lec 2).pptxkinetics (Lec 2).pptx
kinetics (Lec 2).pptx
 
Basic concepts of pharmacokinetics
Basic concepts of pharmacokineticsBasic concepts of pharmacokinetics
Basic concepts of pharmacokinetics
 
Basic concepts of pharmacokinetics
Basic concepts of pharmacokineticsBasic concepts of pharmacokinetics
Basic concepts of pharmacokinetics
 
Pharacokinetics power point for pharmacy
Pharacokinetics power point  for pharmacyPharacokinetics power point  for pharmacy
Pharacokinetics power point for pharmacy
 
Basic Pharmacokinetics & Compartments Modelling.pdf
Basic Pharmacokinetics & Compartments Modelling.pdfBasic Pharmacokinetics & Compartments Modelling.pdf
Basic Pharmacokinetics & Compartments Modelling.pdf
 
Basic pharmacokinetics and compartment modelling
Basic pharmacokinetics  and compartment modellingBasic pharmacokinetics  and compartment modelling
Basic pharmacokinetics and compartment modelling
 
basicpharmacokineticsandcompatmentmodelling-171223090651.ppt
basicpharmacokineticsandcompatmentmodelling-171223090651.pptbasicpharmacokineticsandcompatmentmodelling-171223090651.ppt
basicpharmacokineticsandcompatmentmodelling-171223090651.ppt
 
TOXICOKINETICS
TOXICOKINETICSTOXICOKINETICS
TOXICOKINETICS
 
Pharmacokinetic models
Pharmacokinetic modelsPharmacokinetic models
Pharmacokinetic models
 
Basics of Pharmacokinetics
Basics of PharmacokineticsBasics of Pharmacokinetics
Basics of Pharmacokinetics
 
Bioavailability studies lecture7
Bioavailability studies lecture7Bioavailability studies lecture7
Bioavailability studies lecture7
 
Pocket Guide: Pharmacokinetics Made Easy - sample
Pocket Guide: Pharmacokinetics Made Easy - sample Pocket Guide: Pharmacokinetics Made Easy - sample
Pocket Guide: Pharmacokinetics Made Easy - sample
 
Relationship between pharmacokinetics and pharmacodynamics.pptx
Relationship between pharmacokinetics and pharmacodynamics.pptxRelationship between pharmacokinetics and pharmacodynamics.pptx
Relationship between pharmacokinetics and pharmacodynamics.pptx
 
Toxicokinetics in preclinical studies by Priyabrata Panda
Toxicokinetics in preclinical studies by Priyabrata PandaToxicokinetics in preclinical studies by Priyabrata Panda
Toxicokinetics in preclinical studies by Priyabrata Panda
 
2.pharmacokinetics
2.pharmacokinetics2.pharmacokinetics
2.pharmacokinetics
 
Pharmacokinetic Models
Pharmacokinetic ModelsPharmacokinetic Models
Pharmacokinetic Models
 
Kinetics of stability studies
Kinetics of stability studiesKinetics of stability studies
Kinetics of stability studies
 

Recently uploaded

Portfolio.pdf
Portfolio.pdfPortfolio.pdf
Portfolio.pdf
garcese
 
White wonder, Work developed by Eva Tschopp
White wonder, Work developed by Eva TschoppWhite wonder, Work developed by Eva Tschopp
White wonder, Work developed by Eva Tschopp
Mansi Shah
 
Exploring the Future of Smart Garages.pdf
Exploring the Future of Smart Garages.pdfExploring the Future of Smart Garages.pdf
Exploring the Future of Smart Garages.pdf
fastfixgaragedoor
 
CA OFFICE office office office _VIEWS.pdf
CA OFFICE office office office _VIEWS.pdfCA OFFICE office office office _VIEWS.pdf
CA OFFICE office office office _VIEWS.pdf
SudhanshuMandlik
 
一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理
一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理
一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理
7sd8fier
 
Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...
Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...
Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...
Mansi Shah
 
Expert Accessory Dwelling Unit (ADU) Drafting Services
Expert Accessory Dwelling Unit (ADU) Drafting ServicesExpert Accessory Dwelling Unit (ADU) Drafting Services
Expert Accessory Dwelling Unit (ADU) Drafting Services
ResDraft
 
Top Israeli Products and Brands - Plan it israel.pdf
Top Israeli Products and Brands - Plan it israel.pdfTop Israeli Products and Brands - Plan it israel.pdf
Top Israeli Products and Brands - Plan it israel.pdf
PlanitIsrael
 
Design Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinkingDesign Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinking
cy0krjxt
 
一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理
一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理
一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理
h7j5io0
 
RTUYUIJKLDSADAGHBDJNKSMAL,D
RTUYUIJKLDSADAGHBDJNKSMAL,DRTUYUIJKLDSADAGHBDJNKSMAL,D
RTUYUIJKLDSADAGHBDJNKSMAL,D
cy0krjxt
 
Common Designing Mistakes and How to avoid them
Common Designing Mistakes and How to avoid themCommon Designing Mistakes and How to avoid them
Common Designing Mistakes and How to avoid them
madhavlakhanpal29
 
Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...
Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...
Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...
ameli25062005
 
一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理
一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理
一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理
h7j5io0
 
一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理
一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理
一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理
n0tivyq
 
一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理
一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理
一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理
smpc3nvg
 
一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理
一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理
一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理
708pb191
 
Design Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinkingDesign Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinking
cy0krjxt
 
一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理
一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理
一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理
smpc3nvg
 
Transforming Brand Perception and Boosting Profitability
Transforming Brand Perception and Boosting ProfitabilityTransforming Brand Perception and Boosting Profitability
Transforming Brand Perception and Boosting Profitability
aaryangarg12
 

Recently uploaded (20)

Portfolio.pdf
Portfolio.pdfPortfolio.pdf
Portfolio.pdf
 
White wonder, Work developed by Eva Tschopp
White wonder, Work developed by Eva TschoppWhite wonder, Work developed by Eva Tschopp
White wonder, Work developed by Eva Tschopp
 
Exploring the Future of Smart Garages.pdf
Exploring the Future of Smart Garages.pdfExploring the Future of Smart Garages.pdf
Exploring the Future of Smart Garages.pdf
 
CA OFFICE office office office _VIEWS.pdf
CA OFFICE office office office _VIEWS.pdfCA OFFICE office office office _VIEWS.pdf
CA OFFICE office office office _VIEWS.pdf
 
一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理
一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理
一比一原版(NCL毕业证书)纽卡斯尔大学毕业证成绩单如何办理
 
Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...
Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...
Between Filth and Fortune- Urban Cattle Foraging Realities by Devi S Nair, An...
 
Expert Accessory Dwelling Unit (ADU) Drafting Services
Expert Accessory Dwelling Unit (ADU) Drafting ServicesExpert Accessory Dwelling Unit (ADU) Drafting Services
Expert Accessory Dwelling Unit (ADU) Drafting Services
 
Top Israeli Products and Brands - Plan it israel.pdf
Top Israeli Products and Brands - Plan it israel.pdfTop Israeli Products and Brands - Plan it israel.pdf
Top Israeli Products and Brands - Plan it israel.pdf
 
Design Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinkingDesign Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinking
 
一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理
一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理
一比一原版(UCB毕业证书)伯明翰大学学院毕业证成绩单如何办理
 
RTUYUIJKLDSADAGHBDJNKSMAL,D
RTUYUIJKLDSADAGHBDJNKSMAL,DRTUYUIJKLDSADAGHBDJNKSMAL,D
RTUYUIJKLDSADAGHBDJNKSMAL,D
 
Common Designing Mistakes and How to avoid them
Common Designing Mistakes and How to avoid themCommon Designing Mistakes and How to avoid them
Common Designing Mistakes and How to avoid them
 
Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...
Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...
Коричневый и Кремовый Деликатный Органический Копирайтер Фрилансер Марке...
 
一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理
一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理
一比一原版(Bolton毕业证书)博尔顿大学毕业证成绩单如何办理
 
一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理
一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理
一比一原版(Glasgow毕业证书)格拉斯哥大学毕业证成绩单如何办理
 
一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理
一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理
一比一原版(Brunel毕业证书)布鲁内尔大学毕业证成绩单如何办理
 
一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理
一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理
一比一原版(UAL毕业证书)伦敦艺术大学毕业证成绩单如何办理
 
Design Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinkingDesign Thinking Design thinking Design thinking
Design Thinking Design thinking Design thinking
 
一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理
一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理
一比一原版(Bristol毕业证书)布里斯托大学毕业证成绩单如何办理
 
Transforming Brand Perception and Boosting Profitability
Transforming Brand Perception and Boosting ProfitabilityTransforming Brand Perception and Boosting Profitability
Transforming Brand Perception and Boosting Profitability
 

PHARMACOKINETICS: BASIC CONSIDERATION & PHARMACOKINETIC MODELS

  • 1. Prepared By: Dolly Sadrani Department of Pharmaceutics IInd Sem M.Pharma 1
  • 2. CONTENTS  Plasma Drug Concentration Time Profile  Pharmacokinetic Parameter  Pharmacodynamic Parameter  Zero, First Order & Mixed Order Kinetic  Rates & Order Of Kinetics  Pharmacokinetic Models  Application Of Pharmacokinetic  Reference  Questions 2
  • 3. Pharmacokinetics  Pharmacokinetics is define as the kinetics of drug absorption, distribution, metabolism and excretion (KADME) and their relationship with the pharmacologic, therapeutic or toxicologic response in human and animals.  The applications of pharmacokinetic principles in the safe and effective management of individual patient is called as clinical pharmacokinetics. 3
  • 4. Pharmacodynamics  The branch of pharmacology concerned with the effects of drugs and the mechanisms of their action.  In particular, pharmacodynamic is the study how a drug affects an organism, whereas pharmacokinetics is the study of how the organism affects the drug. 4
  • 5. Plasma Drug Concentration-Time Profile  A Direct relationship exists between the Concentration of drug at the site of action (biophase) and the concentration of drug in plasma.  A typical plasma drug concentration- time curve obtained after a single oral dose of a drug and showing various pharmacokinetic and pharmacodynamic parameters. 5
  • 6.  Such a profile can be obtained by measuring the concentration of drug in plasma sample taken at various intervals of time after administration of a dosage form and plotting the concentration of drug in plasma (Y-axis) versus the corresponding time at which the plasma sample was collect (X-axis). 6
  • 7. 7
  • 8. Pharmacokinetic Parameter  Pharmacokinetic Parameter that describe the plasma level-time curve and useful in determining the bioavailability of a drug from formulation are: 1. Peal Plasma Concentration (Cmax): The point of maximum Concentration of drug in plasma is called as the peak and the Concentration of drug at peak is known as peak plasma Concentration. o It is also called as peak high Concentration and maximum drug Concentration. 8
  • 9. o Cmax is expressed in mcg/ml. o The peak level depends upon the administered dose and rate of absorption and elimination. o The peak represents the point of time when absorption rate equal to elimination rate of drug. 9
  • 10. 2. Time of peak Concentration (tmax) o The time for drug to reach peak Concentration in plasma is called as the time of peak Concentration. o It is expressed in hours and is useful in estimating the rate of absorption. o Onset time and Onset of action are dependent upon tmax. 10
  • 11. 3. Area Under the Curve (AUC) o It represents the total integrated area under the plasma level-time profile and expresses the total amount of drug that comes into the systemic circulation after its administration. o AUC is expressed in mcg/ml . Hours o It is the most important parameter in evaluating the bioavailability of a drug from its dosage form as it represents the extent of absorption. 11
  • 12. o AUC is also important for drugs that are administered repetitively for the treatment of chronic conditions like asthma and epilepsy. 12
  • 13. Pharmacodynamic Parameter 1. Minimum Effective Concentration (MEC) o It is define as the minimum Concentration of drug in plasma required to produce the therapeutic effect. o It reflects the minimum Concentration of drug at the receptor site to elicit the desired pharmacologic response. o The Concentration of drug below the MEC is said to be in the subtherapeutic level. 13
  • 14. o In case of antibiotics , the term minimum inhibitory Concentration (MIC) is used. It describe the minimum Concentration of antibiotic in plasma required to kill or inhibit the growth of microorganisms. 2. Maximum Safe Concentration (MSC) o Also called as minimum toxic Concentration (MTC) , it is the Concentration of drug in plasma above which adverse or unwanted effects are precipitated. 14
  • 15. o Concentration of drug above MSC is said to be in the toxic level. 3. Onset of Action o The beginning of pharmacologic response is called as onset of action. o It occurs when the plasma drug Concentration just exceed the required MEC. 15
  • 16. 4. Onset Time o It is the required for the drug to start producing pharmacologic response. o It corresponds to the time for the plasma Concentration to reach MEC after administration of drug. 5. Duration of Action o The time period for which the plasma Concentration of drug remains above the MEC level is called as duration of drug action. 16
  • 17. 6. Intensity of Action o It is maximum pharmacologic response produced by the peak plasma Concentration of drug. o Also called as peak response. 7. Therapeutic Range o The drug Concentration between MEC and MSC represents the therapeutic range. 17
  • 18. Rate, Rate Constant and Order of Reaction  Pharmacokinetic is the mathematical analysis of processes of ADME.  The movement of drug molecules from the site of application to the systemic circulation, through various barriers, their conversion in to another chemical form and finally exit out of the body can be expressed mathematically by the rate at which they proceed, the order of such processes and the rate constant. 18
  • 19.  The velocity with which a reaction or a process occurs is called as its rate.  The manner in which the Concentration of drug influences the rate of reaction or process is called as the order of reaction or order or process. Drug A Drug B  The rate of forward reaction is expressed as - dA dt  Negative sign indicates that the Concentration of drug A decreases with time T. 19
  • 20.  As the reaction proceeds, the Concentration of drug B increases and the rate of reaction can also be expressed as: dB dt  Experimentally, the rate of reaction is determined by measuring the decreases in Concentration of drug A with time t. 20
  • 21.  If C is the Concentration of drug A, the rate of decreases in C of drug A as it is changed to B can be described by a general expression as a function of time t. dc = -K Cn dt where, K = rate constant n = order of reaction If n = 0, it’s a zero-order process, If n = 1, it’s a first-order process 21
  • 22.  The three commonly encountered rate processes in a physiological system are- - Zero-order process - First-order process - Mixed-order process.  The pharmacokinetics of most drugs can be adequately by Zero- and first-order processes. 22
  • 23. Zero-Order Kinetics (Constant Rate Processes) If n= 0 dC = -K0 C0 = -K0 (1.0) dt where K0 = Zero-order rate constant (mg/ml)  The Zero- Order process can be defined as the one whose rate is independent of the Concentration of drug undergoing reaction i.e. the rate of reaction cannot be increased further by increasing the Concentration of reaction. 23
  • 24. Rearrangement of equation (1.0) yields: dC = -K0 dt (1.1) Integration of equation (1.1) gives: C – C0 = -K0 t or C = C0 - K0 t (1.2) where, C0 = Concentration of drug at t = 0 C = Concentration of drug yet to undergo reaction time t. 24
  • 25.  Equation (1.2) is that of a straight line and states that the Concentration of reactant decreases linearly with time.  A plot of C versus t yields such a straight line having slope – K0 and y- intercept C0. 25
  • 26. Zero-Order Half-life  Half-life (t1/2) or Half-time is defined as the time period required for the Concentration of drugs to decrease by one-half. When t = t1/2 C = C0/2 and the equation (1.2) becomes : C0 = C0 – k0 t1/2 2 (1.3) 26
  • 27. Solving (1.3) we get : t1/2 = C0 = 0.5 C0 2 K0 K0 (1.4) Equation (1.4) shows that the t1/2 of a Zero-order process is not constant but proportional to the initial Concentration of drug C0 and inversely proportional to the Zero-order rate constant K0.  Zero-order equation do not require logarithmic transformation. 27
  • 28. Examples of Zero-order processes are :  Metabolism/ protein-drug binding and enzyme or carrier mediated transport under saturated conditions. The rate of metabolism, binding or transport of drug remains constant as long as its Concentration is in excess of saturating Concentration.  Administration of a drug as a constant rate i.v. infusion 28
  • 29.  Controlled drug delivery such as that form i.m. implants or osmotic pump. First-Order Kinetic (Linear Kinetics) If n=1 Equation (1. ) becomes dC = K C dt (1.5) Where, K= First order rate constant (in time-1 or per hour) 29
  • 30.  From Equation it is clear that a first-order process is the one whose rate is directly proportional to the Concentration of drug undergoing reaction. (greater the Concentration, faster the reaction.)  First-order process is said to be follow linear kinetics. 30
  • 31. Rearrangement of equation (1.5) yields: dC =-Kdt C (1.6) Integration of equation (1.6) ln C = ln C0 –Kt (1.7) Equation (1.7) can also be written in exponential form as: C = C0 e-Kt (1.8) Where e = natural log base. 31
  • 32.  The First-Order process is also called as monoexponential rate process. Thus, a first-order process is characterized by logarithmic or exponential kinetics i.e. A constant fraction of drug undergoes reaction per unit time. since ln = 2.303 log, equation (1.7) can be written as log C = log C0 – Kt 2.303 (1.9)  A semilogarithmic plot of equation (1.9) yields a straight line with slope = - K/2.303 and y – intercept = log C0 32
  • 33. 33
  • 34. First-Order Half-Life  Substituting the value of C = C0/2 at t1/2 in equation (1.9) and solving it yields : t1/2 = 0.693 K (1.10) Above equation shows that, in contrast to Zero- Order process, the Half-life of a First-order process is constant and independent of initial drug Concentration i.e. Irrespective of what the initial drug Concentration is, the time required for the Concentration to decreases by One-half remains the same. 34
  • 35.  Most pharmacokinetic processes viz. Absorption, distribution and elimination follow first-order kinetics. Mixed-Order Kinetics (Nonlinear Kinetics)  A mixture of both first-order and zero-order kinetics is observed in such cases and therefore the process is said to be follow mixed-order kinetics. 35
  • 36.  Deviation from an originally linear pharmacokinetic profile are observed , the rate process of such a drug is called as nonlinear kinetics.  Mixed-order kinetics is also termed as dose- dependent kinetics as it is observed at increased or multiple doses of some drugs.  Observed in : - Drug absorption (Vitamin C) - Drug distribution ( naproxen) - Drug elimination (riboflavin) 36
  • 37. Pharmacokinetic Analysis Of Mathematical Data: Pharmacokinetic Models  Drug movement with in body is a complex process.  The two major approaches in the quantitative study of various kinetic processes of drug disposition in the body are : - Model Approach - Model-independent approach (Non compartmental analysis) 37
  • 38. Methods for analysis of Pharmacokinetic Data Model Approach Model-Independent Approach Compartment Non Compartmental Analysis model - Mammillary - Catenary Physiological model - Perfusion-limited - Diffusion-limited Distributed Parameter model 38
  • 39. Pharmacokinetic model Approach  Model are used to describe change in drug concentration in the body with time.  A model is a hypothesis that employs mathematical terms to concisely describe quantitative relationship.  Pharmacokinetic models provide concise means of expressing mathematically or quantitatively, the time course of drug throughout the body and compute meaningful pharmacokinetic parameter. 39
  • 40. Application of Pharmacokinetic Models:  Characterizing the behaviour of drugs in patients.  Predicting the concentration of drug in various body fluids with any dosage regimen.  Predicting the multiple-dose concentration curves from single dose experiments.  Calculating the optimum dosage regimen for individual patients. 40
  • 41.  Evaluating the risk of toxicity with certain dosage regimens.  Correlating plasma drug Concentration with pharmacological response.  Evaluating the bioequivalence/ bioinequivalence between different formulation of the same drug.  Estimating the possibility of drug and/or metabolites accumulation in the body.  Determining the influence of altered physiology/disease state on /drug ADME.  Explaining drug interaction. 41
  • 42. Types Of Pharmacokinetic Models: 1. Compartment models: Also called as empirical models 2. Physiological models: are realistic models 3. Distributed parameter model: are also realistic models 42
  • 43. Compartment Models  These models simply interpolate the experimental data and allows an empirical formula to estimate the drug concentration with time  Compartments are hypothetical in nature and based on certain assumptions: - The body is represented as a series of compartments arranged either in series or parallel to each other, which communicate reversibly with each other. 43
  • 44. - Each compartment is not a real physiological and anatomical region but a fictitious or virtual one and considered as a tissue or group of tissues that have similar drug distribution characteristics. (Similar blood flow and affinity). - Within each compartment, the drug is considered to be rapidly and uniformly distributed i.e. The compartment is well-stirred. - The rate of drug movement between compartment is described by first-order kinetic. - Rate constants are used to represent rate of entry into and exit from the compartment. 44
  • 45. Compartment models are divided in to: Mammillary model Catenary model Mammillary model:  It consist of one or more peripheral compartment connected to the central compartment.  The central compartment comprises of plasma and highly perfused tissues such as lungs, liver, kidneys which rapidly equilibrate with the drug.  The drug is directly absorbed into this compartment. 45
  • 46.  The peripheral compartments or tissue compartment are those with low vascularity and poor perfusion.  Distribution of drugs to this compartments is through blood.  Movement of drug between compartments is defined by characteristic first-order rate constant denoted by K.  The subscript indicated the direction of drug movement K12 ( K-one-two) refers to drug movement from compartment 1 to compartment 2 and reverse for K21. 46
  • 47.  The number of rate constants which will appear in particular compartment model is given by R. For intravenous administration R=2n-1 (1.11) For extravascular administration R = 2n (1.12) Where n= no. Of compartments. 47
  • 48. 48
  • 49. Catenary Model:  In this model, the compartments are joined to one another in a series like compartments of a train.  This is not observable physiologically/ anatomically as the various organ are directly linked to the blood compartment.  Hence this model is rarely used. 49
  • 50. Advantages and Application of compartment modelling Approach  It is a simple and flexible approach and thus widely used.  It gives a visual representation of various rate processes involved in drug disposition.  It shows how many rate constants are necessary to describe these processes.  It enables the pharmacokinetic to write differential equations for each of the rate processes in order to describe drug concentration change in each compartment. 50
  • 51.  It enables monitoring of drug concentration change with time with a limited amount of data.  It is useful in predicting drug concentration-time profile in both normal physiological and in pathological condition.  It is important in the development of dosage regimen.  It is useful in relating plasma drug levels to therapeutic and toxic effects in the body.  It is particularly useful when several therapeutic agent are compared. Clinically, drug data comparisons are based on compartment models. 51
  • 52.  Its simplicity allows for easy tabulation of parameters such as Vd , t1/2. Disadvantages:  Extensive efforts are required in the development of an exact model that predicts and describes correctly the ADME of a certain drugs.  The model is based on curve fitting of plasma concentration with complex multiexponential mathematical equations.  The model may vary within a study population. 52
  • 53.  The approach can be applied only to a specific drug under study.  The drug behaviour with in the body may fit different compartmental models depending upon the route of administration.  Difficulties generally arise when using models to interpret the differences between results from human and animal experiments.  Owing to their simplicity, compartmental models are often misunderstood, overstretched or even abused. 53
  • 54. Physiological models:  These models are also known as physiologically based pharmacokinetic models (PB- PK models).  The number of compartments to be included in the model depends upon the disposition characteristics of the drug.  organ or tissues such as bones that have no drug penetration are excluded.  For example lungs, liver, brain and kidney are grouped as rapidly equilibrating tissues (RET) while muscles and adipose as slowly equilibrating tissues (SET). 54
  • 55.  The rate of drug carried to a tissue/organ or tissue drug uptake is dependent upon two major factors: - rate of blood flow to the organ - tissue/blood partition coefficient or diffusion coefficient of drug that governs its tissue permeability.  Physiological models are further categorized into two types: - Blood flow rate limited models/perfusion rate limited model : Based on the assumption that the drug movement within a body region is much more rapid than its rate of delivery to that region by the perfusing blood. 55
  • 56. 56
  • 57. Example, thiopental, lidocaine - Membrane permeation rate-limited model/ Diffusion limited model: these models are more complex and applicable to highly polar, ionized and charged drug, in which case the cell membrane acts as barrier for the drug that gradually permeates by diffusion. 57
  • 58. Advantages over the conventional compartment modelling  Mathematical treatment is straightforward.  Since it is a realistic approach, the model is suitable where tissue drug concentration and binding are known.  Data fitting is not required since drug concentration in various body regions can be predicted on the basis of organ or tissue size, perfusion rate and experimentally determined tissue-to-plasma partition coefficient. 58
  • 59.  The model gives exact description of drug concentration-time profile in any organ or tissue and thus better picture of drug distribution characteristics in the body.  The influence of altered physiology or pathology on drug disposition can be easily predicted from changes in the various pharmacokinetic parameters since the parameters corresponds to actual physiological and anatomical measures.  The method is frequently used in animal because invasive methods can be used to collect tissue sample. 59
  • 60.  Correlation of data in several animal species is possible and with some drugs, can be extrapolated to human since tissue concentration of drug is known.  Mechanism of ADME of drug can be easily explained by this model. Disadvantages:  Obtaining the experimental data is a very exhaustive process.  Most physiological model assume an average blood flow for individuals subjects and hence predictions of individualized dosing is difficult. 60
  • 61.  The number of data points is less than the pharmacokinetic parameters to be assessed.  Monitoring of drug concentration in body is difficult since exhaustive data is required. 61
  • 62.  Hypothetical / empirical approach - no relation with real physiology or anatomy.  Experimentally simple and flexible approach as far as data collection is concerned.  Owing to its simplicity, it is widely used and is often the First-model.  Realistic approach since it is based on physiological and anatomic information.  Difficult experimentally since exhaustive data collection is required.  Less commonly used owing to complexity. 62 Compartment modelling Physiological modelling
  • 63.  Complex multiexponential mathematical treatment is necessary for curve fitting.  Data fitting is required for predicting drug concentration in particular compartment.  Used when there is little information about tissue  Easy to monitor time course of drug in body with limited data.  mathematical treatment is straight forward.  Data fitting is not required for since drug concentration in various tissue is practically determined.  Used where tissue drug concentration binding is known.  Exhaustive data is required to monitor tome course of drug in body. 63
  • 64.  Extrapolation from data to human and viceversa is not possible.  Mechanism of drugs ADME can not be explained.  Effect of pathological condition on drug ADME can not be determined.  Frequently used for data comparison of various drug.  Extrapolation of animal data to human is easy on the basis of tissue concentration of drugs.  Easy to explained drugs ADME mechanisms.  Effect of pathology on drug ADME can be easily determined.  Less commonly used for data comparisons. 64
  • 65. Distributed Parameter Model  This model is analogous to physiological model but has been designed to take in to account: - Variation in blood flow to an organ, - Variation in drug diffusion in an organ.  Such a model is thus specifically useful for assessing regional differences in drug concentration in tumours or necrotic tissue. 65
  • 66. Noncompartmental Analysis  Also called as model-independent method dose not required the assumption of specific compartment model.  Based on the assumption that the drugs and metabolites follow linear kinetics and on the basis, this technique can be applied to any compartment model.  The noncompartmental approach, based on the statistical moments theory involves collection of experimental data following a single dose of drug. 66
  • 67.  If one considers the time course of drug concentration in plasma as a statistical distribution curve then, MRT = AUMC AUC (1.13) where MRT = mean residence time AUMC = Area under the First-moment curve AUC = Area under the Zero-moment curve 67
  • 68.  AUMC is obtained from a plot of product of plasma drug concentration and time versus time t from zero to infinity.  Mathematically it is expressed by equation: AUMC = t dt (1.14)  AUC is obtained from a plot of plasma drug concentration versus time t from zero to infinity.  Mathematically it is expressed by equation: AUC = t dt (1.15) 68
  • 69. 69
  • 70.  Practically, the AUMC and AUC can be calculated from the respective graphs by the trapezoidal rule.  MRT is defined as the average amount of time spent by the drug in the body before being eliminated. Application:  Used to estimate the important pharmacokinetic parameters like bioavailability, clearance and apparent volume of distribution. 70
  • 71.  Useful in determining half-life, rate of absorption and first-order absorption rate constant of the drug. Advantages:  Ease of derivation of pharmacokinetic parameters by simple algebraic equations.  The same mathematical treatment can be applied to almost any drug metabolic provided they follow first-order kinetics. 71
  • 72. Disadvantages:  It provides limited information regarding the plasma drug concentration-time profile.  The method does not adequately treat non-linear cases. 72
  • 73. Questions: 1. Explain the term ‘compartment’ what are the assumption made in compartment modelling. Discuss the advantages and disadvantages of same. 2. What are pharmacokinetic models. Discuss the importance and types of such model. 3. Give detailed classification of compartment. 73
  • 74. Reference:  Biopharmaceutics and clinical Pharmacokinetics by Milo Gibaldi and Donald Perrier, fourth edition philadelphia, lea and febiger,1991  Biopharmaceutics and Pharmacokinetics by PL MADAN Second edition, Jaypee Brothers Medical Publishers (P) LTD.  Biopharmaceutics and Pharmacokinetics A TREATISE by D. M. Brahmankar, Sunil B. Jaiswal, Vallabh Prakashan.  Applied Biopharmaceutics and Pharmacokinetics by shargel. Land yu Abc, Second edition applet on century crofts, 1985. 74