57
INTRODUCTION The measurement of drug concentrations in blood samples provides useful clinical information on the relationship between drug administration and drug efficacy (or) toxicity 1 . In order to ensure the efficacy and safety of drug therapy, the outcome of therapy must be monitored. Therapeutic drug monitoring is useful to identify the causes of unwanted (or) unexpected responses, prevent unnecessary diagnostic testing, improve clinical outcomes and even save lives. Information obtained can be used to determine (or) adjust a dosage regimen, evaluate a drug response, aid in the assessment of toxicity, check compliance, minimize the cost of hospitalization and decrease the risk associated with medical- legal problems 2 . THERAPEUTIC DRUG MONITORING (TDM): Definition 3 : It is the process of quantifying drug concentrations in patients and using these measurements to design individualized dosing regimen (dose, formulation, route and frequency of administration. The aim of TDM is to obtain 4 :

INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Embed Size (px)

Citation preview

Page 1: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

INTRODUCTION

The measurement of drug concentrations in blood samples provides useful clinical information

on the relationship between drug administration and drug efficacy (or) toxicity1. In order to

ensure the efficacy and safety of drug therapy, the outcome of therapy must be monitored.

Therapeutic drug monitoring is useful to identify the causes of unwanted (or) unexpected

responses, prevent unnecessary diagnostic testing, improve clinical outcomes and even save

lives. Information obtained can be used to determine (or) adjust a dosage regimen, evaluate a

drug response, aid in the assessment of toxicity, check compliance, minimize the cost of

hospitalization and decrease the risk associated with medical-legal problems2.

THERAPEUTIC DRUG MONITORING (TDM):

Definition3:

It is the process of quantifying drug concentrations in patients and using these

measurements to design individualized dosing regimen (dose, formulation, route and frequency

of administration.

The aim of TDM is to obtain4:

(1) An increased proportion of serum drug concentrations (or assays) with in the therapeutic

range.

(2) Better therapeutic responses and

(3) A reduction in toxic effects.

TDM refers to the individualization of dosage by maintaining plasma (or) blood drug

concentrations with in a target range.

There are two major sources of variability between individual patients in drug response.

These are variation in the relation ship between:

(1) Dose and Plasma concentration (Pharmacokinetic variability)

(2) Drug concentration at the receptor and the response (Pharmacodynamic variability)

Page 2: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

By adjusting doses to maintain plasma drug concentration with in a target range, variability in

the pharmacokinetic phase of drug action is greatly reduced.

A more appropriate description for the optimum use of drug concentration in clinical practice

is Target concentration intervention5.

The rational series of steps involved in achieving the desired outcome in an individual is known

as the target concentration strategy. These steps are

1. Select a target concentration.

2. Predict clearance and volume of distribution values for the patient based on population

pharmacokinetic parameters and observable individual characteristics.

3. Calculate loading dose and maintenance dose to achieve the target concentration.

4. Administer the doses and measure drug concentrations.

5. Use the measured concentrations to predict individualized values of clearance and volume of

distribution for the patient.

6. If appropriate, revise the target concentration for the individual based on the clinical

assessment.

The target concentration should be considered as a surrogate effect, i.e. a convenient substitute

for the desired therapeutic outcome. The target concentration can provide some assurance that

treatment is adequate even if the therapeutic benefit cannot be observed. By intervening with a

dose adjustment to achieve a specific target concentration, a major source of the variability in the

dose response relationship can be reduced, i.e. the variability in concentration when the same

dose is given to different people due to interindividual differences in pharmacokinetics. The

collaborative team approach encourages optimum use of the therapeutic skills of all health

professionals involved.

CONCEPT OF TDM

If there is a concentration-effect relationship, then the measurement of plasma or blood drug

concentration, appropriately sampled, with adequate dosing and clinical history available, allows

rational interpretation of results and subsequent dosage adjustment

Page 3: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

WHEN SHOULD TDM BE USED AND FOR WHICH DRUGS

Therapeutic drug monitoring is indicated in clinical situations in which an expected

therapeutic effect of a drug has not been observed or in cases where drug toxicity related to high

toxic plasma drug concentration is suspected. The characteristics of drugs, which make them

suitable for, or make them require, therapeutic drug monitoring are

Marked pharmacokinetic variability

Compliance

Age- neonates, children, elderly

Physiology-gender, pregnancy

Disease- hepatic, renal, cardiovascular, respiratory

Drug interactions

Environmental influences on drug metabolism

Genetic polymorphism of drug metabolism

Concentration related therapeutic and adverse effects

Narrow therapeutic index

Defined therapeutic (target) concentration range

Desired therapeutic effect difficult to monitor

FACTORS AFFECTING THE THERAPEUTIC RESPONSE OF A PATIENT TO A CERTAIN

DRUG REGIMEN6

A number of factors may affect serum drug concentrations and need to be considered when

interpreting TDM results

Patient demographics

The Patient’s age, sex, body weight and ethnicity should be considered when interpreting TDM

results. Age, sex and lean body weight are particularly important for renally cleared drugs as

knowledge of these allows calculation of creatinine clearance. Ethnicity may be an important

consideration for TDM of some hepatically cleared drugs.

Page 4: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Dosage regimen and duration of therapy

For a drug, which has recently been commenced, sufficient time should elapse to allow steady

state to be achieved before TDM is performed. If loading dose has not been given, this means at

least 5 half - lives of the drug should elapse to allow steady- state to be achieved before TDM is

performed.

Sampling Time

The serum concentration of a drug depends on the time when the blood drawn for a TDM assay

was sampled in relation to the last dose.

The time and date of last dose, and the time and date of blood sampling therefore need to be

known for drugs with a short half - life. Samples should be drawn immediately before the next

dose i.e., a trough level. For drugs with long half-life, samples may be drawn at any time during

the post distribution phase once steady state has been achieved. As the time to reach peak

concentrations shows great variability, peak levels are not performed routinely in clinical

practice.

Patient compliance

If the concentration of the drug is lower than expected, the possibility

Of non-compliance should be considered before a dose increase is recommended. The simplest

way to check for non- compliance is to ask the Patient in a non-judgmental way about their

compliance.

Individual capacity to distribute/metabolize/excrete the drug

Patients with renal impairment have a reduced ability to excrete renally cleared drugs, and the

interpretation of TDM for renally cleared drugs such as digoxin and aminoglycosides should

always be made in the context of the patient’s renal function.

Page 5: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Altered protein binding

Conditions such as malnutrition or nephropathy may reduce the concentration of plasma

proteins. For drugs, which are strongly bound to plasma proteins, such as phenytoin, a reduced

albumin level may result in higher concentration of unbound (free) drug. The measurement of

both total drug concentration and free drug concentration can be useful in those situations.

Drug interactions

TDM results should be interpreted in the light of the patient’s concomitant drug therapy. For

example, patients on digoxin may have unexpectedly high digoxin serum concentrations and

develop digoxin toxicity if drugs such as amiodarone, Quinidine or verapamil are commenced

with out reduction in the digoxin dose. The serum concentrations of some hepatically cleared

drugs may be affected by the commencement or cessation of drugs, which either induce or

inhibit hepatic cytochrome P450 isoenzymes.

Pathological factors

The patient’s comorbidites should be taken in to consideration when interpreting TDM results.

Conditions such as vomiting, diarrhea or inflammatory bowel disease can alter the absorption of

drugs, which in turn can alter serum drug concentrations. In patients having hepatic cirrhosis and

tuberculosis, administration of normal doses of rifampicin and isoniazid can lead to elevated

concentrations of the drugs along with increased hepatotoxicity. Malabsorption leads to

decreased serum concentrations.

Alcohol and Tobacco use

Chronic use of alcohol has been shown to cause non-specific hepatic microsomal serum

concentration resulting increased clearance and decreased serum concentrations of hepatically

cleared drugs such as phenytoin. Cigarette smoking increases the hepatic clearance of

theophylline and patients who have recently stopped smoking may be unexpectedly high

theophylline concentrations.

Page 6: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Medication or Sampling errors

In cases where the TDM result is incompatible with drug administration records, the possibility

of a medication or sampling error should be considered.

Laboratory errors

If a laboratory error is suspected, the laboratory should be contacted and asked to repeat the

assay.

CONCEPT OF TARGET CONCENTRATION INTERVENTION

Target concentration intervention (TCI) is proposed as an alternative conceptual strategy to

therapeutic drug monitoring (TDM). It is argued that the idea of a therapeutic range has limited

the interpretation of measured drug concentrations and diminished the anticipated clinical benefit

to patients by use of an oversimplified pharmacodynamic model. TCI on the other hand

embraces pharmacokinetic and pharmacodynamic concepts and uses the idea of a target effect

and associated target concentration to make rational individual dose decisions.

Rational therapeutics

Pharmacokinetics Pharmacodynamics

Target concentration intervention

Page 7: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Dose Effect Concentration

The aim of rational therapeutics is to achieve the correct effect with the correct dose. The

foundation of decision-making is based on the pharmacokinetics and pharmacodynamics, which

provide the rational principles to link dose and effect through drug concentration.

Target concentration intervention can now be placed at the Centre of the therapeutic triangle.

This strategy takes information about doses, concentrations and effects in an individual and

integrates them to estimate more precisely the pharmacokinetic and pharmacodynamic

parameters in that individual. These new values can then be used to predict the consequences of

future dosage decisions and lead to the selection of an appropriate dose to achieve the desired

effect, i.e. rational therapeutics.

Pharmacodynamics: The Concentration-Effect Relationship

Pharmacodynamics is the science linking concentration to effect by defining the maximum effect

of the drug (Emax) and the sensitivity of the target organ (EC50) .The selection of a target

concentration for a drug is based upon what is known of the relationship between concentration

and effects, both desired (therapeutic effects) and undesired (adverse effects).

Guidance in establishing the target concentration can be obtained from knowledge of Emax

and EC50 because these define the extent of the therapeutic response that can be expected and the

steepest part of the concentration- response curve where the most gain can be expected for the

smallest increase in concentration. The target concentration can only be defined on the basis of

pharmacodynamics; it is quite independent of the determinants of concentration, i.e. dose and

pharmacokinetics.

Page 8: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Pharmacokinetics: The Dose-Concentration Relationship

Pharmacokinetics is the science that links dose and concentration by defining the process of drug

distribution (volume of distribution) and elimination (clearance). The selection of dose needed to

achieve the target concentration rests upon pharmacokinetics. The elementary principles (loading

dose, maintenance dose rate) of pharmacokinetics relating dose to concentration emphasize that a

major goal of target concentration intervention is to estimate the volume of distribution and

clearance in an individual. Once this has been done, other useful derived parameters such as the

half-life can be obtained to assist in determining appropriate dosage intervals or the time needed

to reach steady state.

Therapeutic Range and Target Concentration7

The concentration of drug required in the plasma to produce adequate therapeutic response has

been established for several drugs and is referred to as therapeutic range of drug. The

concentration below this range is referred to as sub therapeutic range and the concentration

higher are referred to as being in the toxic range.

Sheiner & Tozer proposed the target concentration strategy (TCS) as an algorithm for rational

dose individualization but this idea has rarely been recognized. The essential feature of the

algorithm is to use a target concentration rather than a range. The goal is to achieve this target

using initial doses based on typical pharmacokinetic parameters for the individual predicted from

patient specific factors (covariates) such as body size and renal function. Drug concentration

measurements are used solely to individualize the pharmacokinetic parameters in order to predict

future dosing. TCS does not aim to have the measured concentration equal to the target

concentration. This is a key distinction from TDM, which aims to have the measured

concentration within the therapeutic range. TCS explicitly uses a PK model to understand what

makes the patient an individual while TDM offers no direct guidance and leaves the prescriber to

use any method to get the concentration within range.

Page 9: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Drugs for which TDM is used commonly

Amiodarone Gentamicin

Amikacin Impiramine

Amytryptylline Phenytoin

Carbamazepine Sodium Valproate

Cyclosporine Theophylline

Digoxin Vancomycin

Fluoxetin

ANALYTICAL METHODS USED TO CARRY OUT TDM

The methods used for the estimation of drugs in a TDM can be classified into three groups.

1) CHROMATOGRAPHY METHODS

a) High Performance Liquid Chromatography (HPLC)

b) Gas Chromatography (GC)

Page 10: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

2) IMMUNOLOGICAL METHODS

a) Enzyme Immunoassay (EIA)

b) Cloned Enzyme Donor Immunoassay (CEDIA)

c) Enzyme Linked Immunosorbent Assay (ELISA)

d) Enzyme Multiplied Immunoassay (EMIT)

e) Solid Phase Enzyme Immunoassay (SPEIA)

f) Fluorescence Polarization Immunoassay (FPIA)

g) Substrate Linked Fluorescence Immunoassay (SLFIA)

h) Radial Partition Immunoassay (RPIA)

3) ONE STEP- DRY CHEMISTRY

This uses dry membrane antibody-coated cellular strips.

POPULATION PHARMACOKINETICS8

Population pharmacokinetics is the study of the sources and correlates of variability in plasma

drug concentrations between individuals representative of those in whom drug will be used

clinically when relevant dosage regimens are administered. Certain patient path physiological

features can regularly alter dose concentration relationships. Population pharmacokinetics seeks

to identify the measurable pathophysiological factors that cause changes in the dose-

concentration relationship and to what degree so that the appropriate dosage can be

recommended.

A conceptual framework within which we can provide a more formal definition of population

kinetics is provided by a so-called hierarchical population model (also called a population model,

a mixed effects model, or a random-effects model). At the first level of the hierarchy, such a

model views pharmacokinetic observations in an individual (such as concentrations of drug

species in biological fluids) as arising from an individual probability model, whose mean is

Page 11: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

given by a pharmacokinetic model (e.g., a biexponential) quantified by individual-specific

parameters, which may vary according to the value of individual-specific time-varying

covariates. The variance of the individual PK observations is also modeled, and the parameters

of this model are additional individual-specific PK parameters.

At the second stage of the hierarchy, the individual parameters are regarded as random variables

and the probability distribution of these (often only the mean and variance) is modeled as a

function of individual-specific covariates. These models and their parameter values are what we

mean by a the population kinetics of a given drug, while the use of study designs and data

analysis methods designed to elucidate population PK models and their parameter values is what

is meant by the population pharmacokinetic approach.

The population pharmacokinetic approach offers the possibility of gaining integrated information

on pharmacokinetics not only from relatively sparse data, but also from dense data (or from a

combination of dense and sparse data) obtained from subjects. The approach allows the analysis

of data from a variety of unbalanced designs as well as from studies that are normally excluded

because they do not lend themselves to the usual forms of pharmacokinetic analysis, such as

concentration data obtained from pediatric and elderly patients, or from data obtained during the

evaluation of the relationships between dose or concentration and efficacy or safety.

The subjects of pharmacokinetic studies are usually healthy volunteers or highly selected

patients. Traditionally, the average behavior of a group (i.e., the mean plasma concentration-time

profile) has been the main focus of interest. Interindividual variability in pharmacokinetics is

viewed by many incorrectly as a nuisance factor that has to be overcome, often through complex

study designs and control schemes, and reduced through restrictive inclusion criteria. Study

design and selection of volunteers that are rigidly standardized so that they are as homogeneous

as possible are typical features of pharmacokinetic investigations. These studies, therefore, are

often performed under artificial conditions that do not represent the intended clinical use of the

drug.

The population pharmacokinetic approach encompasses some or all of the following features

Page 12: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

It seeks to obtain relevant pharmacokinetic information in patients who are representative of

the target population to be treated with the drug.

It recognizes variability as an important feature that should be identified and measured

during drug development or evaluation.

It seeks to explain variability by identifying factors of demographic, path physiological,

environmental, or drug-related origin that may influence the pharmacokinetic behavior of a drug.

It seeks to quantitatively estimate the magnitude of the unexplained part of the variability in

the patient population.

The magnitude of the unexplained (random) variability is important because the efficacy and

safety of a drug may decrease as unexplainable variability increases. Drug levels outside the

target range become more likely, the greater the uncompensated variability in the relationship of

dosage to steady state drug concentration. In addition to interindividual variability, the degree to

which steady state drug concentrations in individuals typically vary about their long-term

average is also important. Concentrations appear to vary due to inexplicable day-to-day or week-

to-week kinetic variability and due to errors in concentration measurement. Estimates of this

kind of variability (residual intrasubject, interoccasion variability) are important for therapeutic

drug monitoring using the empiric Bayes approach. The knowledge of the relationship between

concentrations, response, and physiology is essential to design dosing strategies for rational

therapeutics that may not necessarily require therapeutic drug monitoring.

II. POPULATION METHODS

This discussion of population methods focuses on methods that provide estimates of some or all

of the components of variability.

A. The Two-Stage Approach

The usual method of pharmacokinetic data analysis is the two-stage approach. The first stage is

the estimation of pharmacokinetic parameters through nonlinear regression using an individual’s

experimental data (data rich situation). Individual estimates obtained in the first stage serve as

Page 13: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

input data for the second-stage calculation of descriptive summary statistics on the sample,

typically mean vector and variance-covariance matrix. Analysis of dependencies between

parameters and covariates using classical statistical approaches (linear stepwise regression,

covariance analysis, cluster analysis) may be included in the second stage. The standard two-

stage approach, when applicable, can yield adequate estimates of population characteristics.

Mean estimates of parameters are usually unbiased, but the random effects (variance and

covariance) are likely to be overestimated in all realistic situations. Refinements have been

proposed to improve the standard two-stage approach by bias correction for the random effects

covariance and differential "weighting" of individual data according to its quality and quantity.

B. The Population Approach

The population approach in the context of drug evaluation developed from a recognition that, if

pharmacokinetics and pharmacodynamics were to be investigated in patients, pragmatic

considerations dictated that data should be collected under less stringent and restrictive design

conditions. Considering the complete sample, rather than the individual as a unit of analysis, the

population method of analysis (i.e., analysis according to a hierarchical random effect model)

aims to estimate the distribution of the parameters and their relationships with covariates. The

approach uses individual pharmacokinetic data of the observational (experimental) type, which

are unbalanced and fragmentary, in addition to or instead of conventional pharmacokinetic data

from traditional pharmacokinetic studies characterized by rigid and extensive design. Analysis

according to a hierarchical (non-linear) random effects model9 provides estimates of population

characteristics that define the population distribution of the pharmacokinetic (and/or

pharmacodynamics) parameters. In the mixed-effects modeling context, the collection of

population characteristics is composed of population typical values and population variability

values (generally the variance-covariance matrix). In sparse data situations where estimates of

individual parameters are, a priori, out of reach, an original one-stage or population estimation

approach is required. A population analysis of pharmacokinetic data, therefore, consists of

estimating directly the parameters of the population from the full set of individual concentration

values. The individuality of each subject is maintained and accounted for, even when raw data

are sparse.

Page 14: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

III. WHEN TO PERFORM A POPULATION PHARMACOKINETIC STUDY AND

ANALYSIS

The population approach can help increase knowledge of the quantitative relationships between

drug input patterns, patient characteristics, drug disposition, and responses. The population

approach may be used to estimate population parameters of a response surface model in phases 1

and 2B of clinical drug development, where information is gathered on how the drug will be

used in subsequent stages of drug development and after release. The population approach may

increase the efficiency and specificity of drug development by suggesting more informative

designs and analyses of experiments. Application of the population approach to phase 1 and

perhaps much of phase 2B, where patients are sampled extensively, does not necessarily involve

complex methods of data analysis. The two-stage methods can be used to analyze the data, and

standard regression methods can be used to model dependence of parameters on covariates.

Alternatively, the data from individual studies can be pooled and analyzed using the nonlinear

mixed-effects modeling approach.

The population approach can also be applied to phases 2A and 3 of drug development to gain

information on drug safety (efficacy) and to gather additional information on drug

pharmacokinetics (and pharmacodynamics) in special populations, such as the elderly. It is also

useful in post marketing surveillance (phase 4) studies. Studies performed in phase 3 and 4 of

clinical drug development lend them to the use of the full pharmacokinetic screen study design.

V. STUDY DESIGN AND EXECUTION

Certain preliminary pharmacokinetic information should be known before any population

pharmacokinetics study is undertaken. The drugs major elimination pathways in humans should

be known. Preliminary studies should have established the basic model describing the

pharmacokinetics of the drug. The latter is important because the sparse data collected during

population pharmacokinetic studies may not provide adequate information for the deduction of a

pharmacokinetic model. In addition, a sensitive and specific assay (see Assay section) capable of

measuring all species (parent drug and metabolites) of clinical relevance should be available

before a population pharmacokinetic study is undertaken. When properly performed, population

Page 15: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

pharmacokinetic studies in patients combined with suitable mathematical/statistical analysis

(e.g., using nonlinear mixed-effects modeling) is a valid approach and, on some occasions, an

alternative to extensive studies.

A. Study Design

In the population pharmacokinetics context, there are two broad approaches for obtaining

information about pharmacokinetic variability: (a) trough screen (single or multiple) studies and

(b) full pharmacokinetic screen (experimental population pharmacokinetic) studies. They yield

an increasing amount of information.

1. Single-Trough Screen

A single blood sample is obtained from each patient at or close to the trough of drug

concentrations, shortly before the next dose, and a frequency distribution of plasma or serum

levels in the sample of patients are calculated. Provided that the sample size is large, that assay

and sampling errors are small, and that the dosing regimen and sampling times are identical for

all patients, a histogram of such a trough screen gives a fairly accurate picture of the variability

in trough concentrations in a target population. If these conditions are not met, such histograms

do not represent strict pharmacokinetic variability because the data include many other sources

of random fluctuation with significant contribution to the observed spread. When related with

therapeutic outcome and occurrence of side effects, such histograms can be useful to improve the

knowledge of the optimal concentration range of a given drug.

The relationships of patient characteristics to the trough levels can be explored by simple

statistical procedures such as multiple linear regressions. Although simple, the trough

(pharmacokinetic) screen will only yield information about oral clearance and no other

parameters of interest (e.g., apparent volume of distribution, half-life). Only qualitative, not

quantitative, information will be obtained. Components of variability C interindividual and

residual variability C cannot be separated. This method will identify, qualitatively,

pharmacokinetically relevant factors and their differences among subgroups (subpopulations).

When implementing this sampling strategy, the difficulty of getting patients and physicians to

adhere to the sampling strategy should be kept in mind. Compliance with at least the last two

Page 16: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

doses before trough level measurement is adequate for this type of study, but the drug should be

dosed to steady state. Because of uncertainty in doses and samples, the method can only

reasonably be applied to drugs dosed at intervals less than or equal to one elimination half-life

unless timing of dose and of level can be assured, as in inpatient studies. Large numbers of

subjects would be needed for this type of study because the data would be noisy.

With this design, it is not advisable to contemplate measuring peak observations unless the drug

is given intravenously or is a certain type of sustained release formulation. The time for

achieving maximum concentration depends on rates of all processes of drug disposition and may

vary among subjects. Thus, the simple estimation of peak levels is subject to large uncertainty.

Sampling peak levels also yields information on variability of largely irrelevant kinetic processes

for drugs for which effects relate to steady-state mean concentrations, or the area under the

concentration curve.

2. Multiple-Trough Screen

In this design, two or more blood samples are obtained near the trough of steady-state

concentrations from most or all patients. In addition to relating blood concentrations to patient

characteristics, it is possible now to separate interindividual and residual variabilities. Since

patients are studied in greater detail, this design requires fewer subjects, and the relationships to

patient characteristics can be evaluated with higher precision. To estimate interindividual

variability of the oral clearance, nonlinear mixed-effects modeling should be used. When using

pharmacokinetic models for parameter estimation, a sensitivity analysis10 should be required to

fix a parameter such as absorption rate constant to estimate other parameters and to determine

the fixed parameter value that has the least effect on the estimation of the remaining parameters.

The drawbacks of the single-trough screen design apply here. Although the estimates of

intersubject and residual variability may or may not be biased, they may not be precise unless a

large number of patients are studied.

3. Full Pharmacokinetic Screen

With this approach, blood samples are drawn from subjects at various times (typically 1 to 6 time

points) following drug administration .The objective is to obtain, where feasible, multiple drug

Page 17: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

levels per patient at different times. This approach permits an estimation of pharmacokinetic

parameters of the drug in the study population and an explanation of variability using the

nonlinear mixed-effects modeling approach. The full pharmacokinetic screen (experimental

population pharmacokinetic) study should be designed to explore the relationship between the

pharmacokinetics of a drug and demographic/pathophysiological features of the target population

(with its subgroups) for which the drug is being developed. A full pharmacokinetic screen study

requires careful design considerations. Population pharmacokinetic analysis is useful for looking

at influences of pathophysiological conditions on parameters of a model with a well-established

structure. The qualitative aspects of the model should be well known before embarking on a

population study.

The objective for carrying out a population pharmacokinetic study should be clearly defined

since this will determine the study design. When designing a population pharmacokinetic study,

the practical limitations of sampling times, number of samples/subject, and number of subjects

should be considered. Preliminary information on variability from pilot studies make it possible,

through simulation, to anticipate certain fatal study designs as well as informative ones.

Simulation studies can optimize design features for accurate and precise estimation of population

pharmacokinetic parameters. Optimization of sampling design is critical to efficient experimental

design when there are severe limitations on the number of subjects and/or samples/subject, as in

pediatrics and the elderly .The use of informative study designs for population pharmacokinetic

studies that yield informative data is encouraged .The use of Bayesian designs for pediatric

patients where adult data may serve as informative priors may be appropriate. Such a study

should include enough patients in important subgroups to ensure accurate and precise parameter

estimation and the detection of any subgroup differences.

B. Importance of Sampling Individuals on More Than One Occasion

The variance of an individuals PK observations about the individual-specific PK model on a

given occasion (i.e., the intra-individual variability; see Introduction) can conceptually be

factored into two components: variability of PK observations due to variability of the PK model

from occasion to occasion (inter-occasion variability), and variability of PK observations about

the individual PK model appropriate for the particular occasion (noise; PK model

Page 18: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

misspecification). To be sure, some inter-occasion variability may be explained by inter-occasion

variation in individual time-varying covariates, but to the extent that it is not, it represents, along

with the noise, the irreducible uncertainty in predicting, and hence controlling drug

concentrations. Drugs with narrow therapeutic indices and large inter-occasion variability, for

example, will be very difficult to control. If a population PK study consists of PK observations

solely from individuals each studied on only a single occasion, the inter-occasion variability will

appear incorrectly in the inter-individual variability term and not in the intra-individual

variability term. This may lead to inappropriate optimism about the ability to control individual

therapy within the therapeutic range by using feedback (e.g., therapeutic drug monitoring, or

simply adjusting dose according to observed drug effects), and also to a fruitless search for inter-

individual covariates that might explain the spuriously inflated) inter-individual variability. It is

of utmost importance to avoid this artifact by ensuring that at least a moderate subset of subjects

contributing data to a population PK study contributes data from more than one occasion. Indeed

if this is done, one may hope to separately estimate the components of intra-individual

variability.

C. Simulation

Simulation of a planned study offers a powerful tool for evaluating and understanding the

consequences of different study designs. Shortcomings in study design result in the collection of

uninformative data. Simulation can reveal the effect of input variables and assumptions on the

results of a planned population pharmacokinetic study. Simulation allows study designers to

assess the consequences of the design factors chosen and the assumptions made. Thus,

simulation enables the pharmacometrician to better predict the results of a population study and

to choose the study design that will best meet the study objectives. It is important to simulate a

population pharmacokinetic study using alternative study designs to determine the most

informative design, given the study objectives, before initiating such studies. Simulation is a

useful tool to provide convincing objective evidence of the merits of a proposed study design and

analysis.

Page 19: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

D. Study Protocol

Two types of protocol may be considered depending on the setting in which a population

pharmacokinetic study is to be performed. If it is added on to a clinical trial (as can be envisaged

in most situations), the population study should be carefully interwoven with the existing clinical

protocol to ensure that it does not compromise the primary objectives of the study. Every effort

should be made to convince investigators of the relevance of including a population

pharmacokinetic study. Graphical displays of simulation results may help to achieve this

objective. In addition, a population pharmacokinetic study protocol should be written since a

population study can also be defined as evaluating data from existing data and/or data coming

from more than one study. When a population study is a stand-alone study, a comprehensive

protocol should be prepared. The population pharmacokinetic study as part of the clinical

protocol and the population pharmacokinetic study protocol are discussed briefly.

i) Clinical Protocol

The objectives of the population pharmacokinetic study should be clearly defined. These

objectives should be secondary to the primary clinical study objectives or primary when they

would not compromise the study in question. The criteria for sampling subjects and methods for

data analysis (described in the population pharmacokinetic study protocol) should be clearly

stated. The data to be used for population analysis should be defined, including patients and

subgroups to be used and covariates to be measured. The sampling design should be specified

and any subgroup stratification should be defined. In a multicenter trial, it may sometimes be

necessary to obtain extensive data from some centers and sparse data from others. This type of

data collection can be useful for informative data analysis protecting against model

misspecification and it should be specified in the protocol. The data analysis plan should be

described in advance in the protocol as accurately as possible. Statements such as a

pharmacokinetic screen will be performed or a data will be analyzed using NONMEM are

inappropriate because they do not convey information on the study objective or how the analysis

will be carried out.

Page 20: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

If possible, special case report forms that can be easily understood by investigators should be

designed to meet the needs of the pharmacokinetic evaluation.

ii) Population Pharmacokinetic Study Protocol

The practical details of the pharmacokinetic evaluation should be described in a population

pharmacokinetic study protocol, although the principles may be specified in the clinical study

protocol in a general way. The primary (same as that in the clinical protocol) and secondary

objectives should be clearly stated. The secondary objectives should be those that enable the data

analyst to search for the unexpected, after the primary objectives have been addressed. The

sampling design, data assembly, data checking procedure, and procedures for handling missing

data and data anomalies should be clearly spelled out in the protocol. The data to be used for

population analysis should be defined, including patients and subgroups to be used and

covariates to be measured. The sampling design should be specified and any subgroup

stratification should be defined11. Real-time data assembly would permit population

pharmacokinetic data analysis to be performed before the end of a clinical trial and would make

it possible to include the results in the filing of the new drug application (NDA). If drug-drug

interactions are to be characterized, the protocol should prespecify whether to determine the

effect of the presence or absence of a specific concomitant medication or the total daily dose of

the concomitant medication or the plasma concentration of the potentially interacting medication.

If food effect is to be evaluated, the time of sampling in relation to food intake, and the

composition of food, should be specified in the protocol. Also, the procedure for analyzing the

data (and validation when appropriate) should be specified.

Distinguishing between clinically relevant and statistically significant covariates is important.

Time variant covariates represent particular problems. In this case, several measurements should

be made during the course of the study and, if this information is found to be incomplete, model-

based techniques may be used for imputation between available data (See Handling of Missing

Data). This also applies to time invariant covariates. Sensible methods of dealing with missing

data should be predefined in the data analysis plan of the protocol. The issue of interoccasion

variability should be recognized and addressed in long-term studies. It is understandable that

population pharmacokinetic data analysis, as a modeling exercise, cannot be planned to the

Page 21: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

fullest detail. However, the protocol should include study objectives; patient inclusion/exclusion

criteria and pharmacokinetic evaluability criteria; sampling design; data handling and checking

procedures; initial assumptions for modeling; a list of possible covariates to be investigated and

the rationale for choosing them; and whether a sensitivity analysis and a validation procedure is

envisaged.

E. Study Execution

A population pharmacokinetic study should be conducted according to current good clinical

practice and good laboratory practice standards. The sampling strategy and the recording of

samples should be part of good clinical practice and the handling of samples part of good

laboratory practice. Error in recording sampling times relative to dosing history could result in

biased and imprecise parameter estimates, depending on the nature and degree of the error. Every

effort should be made to ensure that study subjects and clinical investigators comply with study

protocol. To improve compliance, the protocol should not be overly complicated and blood-

sampling times should be convenient to both clinical staff and patients. The necessity of blood

sampling should be carefully explained to patients and investigators. Instructions provided to the

investigators should be clear and concise. Adequate monitoring by the sponsor while the study is

ongoing should back up these measures. Adequate resources should be available for optimal

sample preparation, storage at the investigator site, and transportation and storage of biological

samples prior to analysis.

Noncompliance with drug intake can be a source of confounding and lead to inappropriate

interpretation of study results. Special care should be taken to use methodologies that are as

objective as possible to reconstruct dosage history. Communication between all parties involved

is essential for the successful conduct of a population study, especially if the study is part of a

large-scale clinical trial.

VI. ASSAY

Page 22: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Correct evaluation of pharmacokinetic data depends on the accuracy of the analytic data

obtained. Clinical investigators and their staff should be educated on the importance of sample

timing, recording, proper labeling, and handling of samples. The accuracy of analytical data

depends on the criteria used to validate the method. Consequently, drug and/or metabolite(s)

stability, assay sensitivity, selectivity, recovery, linearity, precision, and accuracy should be

carefully scrutinized before samples are analyzed. The importance of using validated assay

methods for analyzing pharmacokinetic data cannot be over emphasized.

VII. DATA HANDLING

A. Data Assembly and Editing

Real-time data assembly prevents the problems that generally arise when population

pharmacokinetic data are stored until the end of a clinical trial. Real-time data assembly permits

an ongoing evaluation of site compliance with the study protocol and provides the opportunity to

correct violations of study procedures and policy. Evaluation of pharmacokinetic data can

provide the data safety monitoring board with insights into drug exposure safety evaluations and

drug-drug interactions. Adequate policies and procedures should be in place for studying blind

maintenance. Real-time data assembly creates the opportunity for editing the concentration-time

data, drug dosing history, and covariate data necessary to meet the pharmacokinetic objectives of

a clinical trial Data assembly to create a population pharmacokinetic database after study

completion may result in delays that are incompatible with the time course of the development

program. It is important, therefore, to specify in the study protocol the use of real-time data

analysis.

Data editing means using a set of procedures for detecting and correcting errors in the data. The

procedures should be planned before study initiation and predefined in the study protocol.

Criteria for declaring data usable or unusable (e.g., time of blood sampling missing, dosing

information with no associated concentrations, concentrations with missing dosing information)

should be spelled out in the study protocol.

Page 23: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

B. Handling of Missing Data

After assembling data for population analysis, the issue of any missing covariate data should be

addressed. Missing data represent a potential source of bias. Thus, every effort should be made to

fulfill the protocol requirements concerning the collection and management of data. Deletion of

subjects with missing covariates can yield biased and imprecise parameter estimates. Although

caution should be taken when imputing missing values, it is usually better to estimate and impute

a subject missing covariate data values than to delete that subject from the data set. Simple

methods of imputation include the use of median, mean, or mode for missing values. These

methods are biased and inefficient when predictors are correlated. Using maximum likelihood

procedures (i.e., deriving regression models) for predicting each predictor from all other

predictors is a better method. Alternatively, where imputation across a time series is possible,

such a method should be used12. Imputation procedures should be described, including a detailed

explanation of how such imputations were done and of the underlying assumptions made. The

sensitivity of the results of the analysis to the method of imputation of missing data should be

tested, especially if the number of missing values is substantial. So-called multiple imputation, in

which several imputed data sets are analyzed, can be used, where conclusions are of borderline

significance, yet of clinical importance, to remove the optimistic bias from estimates of precision

caused by imputing data and treating it as though it were actually observed.

C. Outliers

The statistical definition of an outlier is, to some extent, arbitrary. The reasons for declaring a

data point to be an outlier should be statistically convincing and prespecified in the protocol. Any

physiological or study-related event that renders the data unusable should be explained in the

study report. Because of the exploratory nature of population analysis, it may be that the study

protocol did not specify a procedure for dealing with outliers. In such a situation, model building

should be performed on the reduced data set (i.e., data set without the outliers) as long as the

conclusions are appropriately restricted to the limited population defined by the outlier-removal

procedure. Including extreme outliers is not a good practice when using least squares or normal-

Page 24: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

theory type estimation methods, as these inevitably have a disproportionate effect on estimates.

Also, it is well known that for most biological phenomena, outlying observations are far more

frequent than suggested by the normal distribution (i.e., biological distributions are heavy-tailed).

Some robust methods of population analysis have recently been suggested, and these may allow

outliers to be retained, without giving them undue weight.

D. Data Type

Two types of data can be used in population analysis: experimental data and observational (or

population) pharmacokinetic data. Experimental data arise from traditional pharmacokinetic

studies characterized by controlled conditions of drug dosing and extensive blood sampling.

Population pharmacokinetic data are collected, most often, as a supplement in a study designed

and carried out for another purpose. The data are characterized by minimal control and few

design restrictions: the dosing history is subject specific, the amount of pharmacokinetic data

collected from each subject varies, the timing of blood sampling in relation to drug

administration differs, and the number of samples per patient, typically 1 to 6, is small.

E. Data Integrity and Computer Software

Data management activities should be based on established standard operating procedures. The

validity of the data analysis results depends on the quality and validity of methods and software

used for data management (data entry, storage, verification, correction, and retrieval), and

pharmacostatistical processing. Documentation of testing procedures for the computer software

used for data management should be available. It is crucial that the software used for population

analysis be adequately supported and maintained.

VIII. DATA ANALYSIS

Population modeling can potentially be used in several phases of new drug development,

including the planning, design, and analysis of studies in the exploratory and confirmatory stages

of new drug development. Thus, the protocol should describe the pharmacokinetic models to be

tested. Careful documentation of modeling efforts, data visualization, model validation (when

appropriate), and data listing should be provided.

Page 25: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Population pharmacokinetic data analysis can be carried out in three interwoven steps: (a)

exploratory data analysis, (b) population pharmacokinetic model development, and (c) model

validation. The data analysis plan should be clearly defined in the study protocol.

A. Exploratory Data Analysis

Exploratory data analysis isolates and reveals patterns and features in the population data set

using graphical and statistical techniques. It also serves to uncover unexpected departures from

familiar models. An important element of the exploratory approach is its flexibility, both in

tailoring the analysis to the structure of the data and in responding to patterns that successive

analysis steps uncover.

Most population analysis procedures are based on explicit assumptions about the data, and the

validity of the analyses depends upon the validity of assumptions. Exploratory data analysis

techniques provide powerful diagnostic tools for confirming assumptions or, when the

assumptions are not met, for suggesting corrective actions. Without such tools, confirmation of

assumptions can be replaced only by hope. Exploratory data analysis should be coupled with

more sophisticated population modeling techniques in the analysis of population

pharmacokinetic data13. Exploratory data analysis performed should be well described in the

population report.

B. Population Pharmacokinetic Model Development

1. Objectives, Hypotheses, and Assumptions

The objectives of the analyses should be clearly stated. The hypotheses being investigated should

be clearly articulated. It is recommended that all known assumptions inherent in the population

analysis be explicitly expressed.

2. Population Model Development

The steps taken (i.e., sequence of models tested) to develop a population model should be clearly

outlined to permit the reproducibility of the analysis. The criteria and rationale for model

building procedures dealing with confounding, covariate, and parameter redundancy should be

Page 26: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

clearly stated. The criteria and rationale for model reduction to arrive at the final population

model should also be clearly explained.

3. Reliability of Results

The reliability of the analysis results should be checked using diagnostic plots; confidence

intervals (standard errors) for key parameters should be checked using nonparametric techniques

(such as the jackknife and the profile likelihood plot (mapping the objective function. This is

appropriate because the possibility of biased results is increased by the complexity of the

population models and by the sparse individual data available. The nonlinearity of the statistical

model and ill conditioning of a given problem can produce numerical difficulties and force the

estimation algorithm into a false minimum. Because the maximum likelihood procedure is

sensitive to bizarre observations, it may be necessary to check the stability of the model .It is

important to evaluate the quality of the results of a population study/analysis for robustness.

Evaluation for robustness may be approached by sensitivity analysis. The use of case deletion

diagnostics is also encouraged. Evidence of robustness renders the results reasonable and

independent of the analyst.

C. Model Validation

Validation can be defined as the evaluation of the predictability of the model developed (i.e., the

model form together with the model parameter estimates) with a learning or index data set on a

validation data set not used for model building and estimation. A model may be valid for one

purpose and not valid for another. The objective of validation is to examine whether the model is

a good description of the validation data set in terms of its behavior and of the application

proposed.

Validation depends on the objective of the analysis. Not all population models may need to be

validated. Population models developed for explaining variability (which does not require dosage

adjustment recommendation) and for providing descriptive information for labeling may be

tested for stability only14. Also, population pharmacokinetic models developed as part of a

population pharmacokinetic /pharmacodynamics model may not need to be validated. However,

the predictive performance of a population pharmacokinetic model developed for dosage

Page 27: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

recommendation as part of labeling should be tested. In such a situation, model validation

procedure should be an integral part of the protocol.

1. Types of Validation

There are two types of validation: external and internal. External validation is the application of

the developed model to a new data set (validation data set) from another study. Internal

validation refers to the use of data-splitting and resampling techniques (cross-validation and

bootstrapping) for validation purposes. External validation provides the most stringent method

for testing a developed model.

Data-splitting

For testing predictive performance, data splitting is an effective method of model validation

when it is not practical to collect a new set of data to test the model. The disadvantage of this

method is that, in the area of linear regression, the predictive accuracy of the model is a function

of the sample size resulting from the data-splitting .To maximize the predictive accuracy, it is

recommended that the entire sample be used for model development and assessment. Data

splitting may not validate the final model if one desires to recombine the index and validation

data sets to derive a refined model for predictive purposes. However, if data splitting is to be

used, a random subset of the data (two-thirds, i.e., the index data set) should be used for model

building and the remaining data should be used for model validation. At the end of the exercise,

the index and validation data sets should be pooled and the final population model fitted to the

data to determine the appropriateness of each covariate retained in the final model.

Cross-validation

Cross-validation is repeated data-splitting. The benefits of cross-validation over data-splitting are

that (1) the size of the model development database can be much larger so that less data are

discarded from the estimation process and (2) not relying on a single sample split reduces

variability. Due to high variation of accuracy estimates, cross-validation is inefficient when the

entire validation process is repeated.

Page 28: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

Bootstrapping

Bootstrapping, an alternative method of internal validation, has the advantage of using the entire

data set for model development. Sample size is critical in pediatric settings where ethical and

medical concerns limit recruitment into studies. The bootstrap resampling procedure can be

useful for evaluating the performance of a population model when there is no test data set.

2. Validation Methods

The issue of validation of population models remains unresolved. The advantages and

disadvantages of methods addressed in the literature and of methods used in applications have

been discussed above. The data analyst should justify the method he/she chooses. Although the

science of validation of population models is still evolving, consideration will be given to well-

described innovative methods of model validation.

Prediction Errors on Concentrations

This is calculated as the difference between observed and model-predicted concentrations. The

mean prediction error is calculated and used as a measure of accuracy and the mean absolute

error (or root mean square error) is used as a measure of precision.

This method can be used when only one sample per subject is obtained. When more than one

observation is obtained per subject, the method is inadequate because prediction errors are not

independent if several concentrations per subject are available .The method does not take into

account the correlation of observations within subjects.

Standardized Prediction Errors

This method15 takes into account variability and correlation of observations within an individual.

The mean standardized prediction error and the variance are calculated, and a t-test

(appropriately a z-test) performed to determine whether the mean is significantly different from

zero and the standard deviation approximates 1. Confidence intervals about the standard

deviation of the standardized prediction errors can be constructed. This test performed on the

Page 29: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

mean of the standardized prediction errors incorrectly assumes that the estimates for the

population parameter values are given without error. The use of the approach is discouraged.

Validation through Parameters

This method avoids the problems encountered in prediction error of concentrations by

performing validation with model parameters. Model parameters are predicted from the

validation data set with or without covariates and bias and precision are calculated for the

predictions.

Plot of Residuals against Covariates

A simple plot of residuals obtained by freezing the final model and predicting into a validation

data set against covariates can yield information on the clinical significance of the model in

terms of a covariate or subpopulation.

A Plot of Residuals against Covariates and Validation through Parameters.

These methods are useful approaches for examining the predictive performance of population

models. When there is no test data set, the bootstrap approach should be used: the mean

parameter values obtained by repeatedly fitting the final population model to at least 200

bootstrap replicate data sets are compared to the final population model parameter estimates

obtained without bootstrap replication.

Posterior Predictive Check

A new technique, the so-called Posterior Predictive Check, may prove useful in determining

whether important clinical features of present and future data sets are faithfully reproduced by

the model.

Page 30: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

IX. POPULATION ANALYSIS REPORT

The report should contain the following: (a) introduction, (b) objectives, hypotheses, and

assumptions (c) assay, (d) data, (e) data analysis methods, (f) results, (g) discussion,

(h) application, (I) appendix, and (j) electronic format files.

A. Introduction

The introduction should briefly state the general intent of the analysis. It should include enough

background information to place the analysis in its proper context within the drug=s clinical

development and to indicate any special features of a population pharmacokinetic study.

B. Objectives, Hypotheses, and Assumptions

The objectives of the analysis, and study where applicable, should be stated. In addition to the

primary objective, any secondary objectives should be explicitly stated. If the analysis was

performed as a result of the implementation of a study protocol, the report should note whether

the objectives were preplanned or were formulated during or after study completion. This is not

necessary for the analysis of pooled data. The assumptions made and the hypotheses tested

should be clearly stated in the report.

C. Assay

This section should contain a description of the assay method(s) used in quantitating drug

concentrations. Assay performance (quality control samples) should also be included. The

validity of the method(s) should also be described.

D. Data

Where data are pooled for analysis, the report should state the studies from which the data were

pooled. The data set should be part of the appendix to the report. The report should contain the

response variable and all covariate information and explain how they were obtained. The report

Page 31: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

should include a description of the sampling design used to collect the plasma samples and a

description of the covariate, including their distributions and the accuracy and precision with

which they were measured. An electronic copy of the data set should be submitted. Data quality

control and editing procedures should be described in this section.

E. Data Analysis Methods

This section should contain a description of the treatment of outliers and missing data (where

applicable), and a detailed description of the criteria and procedures for model building and

reduction incorporating exploratory data analysis. A flow diagram(s) of the analysis performed

and representative control/command files for each significant model building/reduction step

should be provided

F. Results

The key results of the analysis should be compiled into comprehensible tables and plots. To aid

interpretation and application, a thorough description of the results should be provided. Complete

output of results obtained for the final population model and key intermediate steps should be

included.

G. Discussion

The report should include a comprehensive statement of the rationale for the model building and

reduction procedures, interpretation of the results, protocol violations, and any other relevant

information.

H. Application of Results

A discussion of how the results of the analysis will be used (e.g., to support labeling,

individualize dosage, support safety, or define additional studies) should be provided. In

addition, the use of graphics to communicate the application of a population model (e.g., for

dosage adjustment) is recommended.

I. Appendix

Page 32: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

The appendix should contain the data set(s) used in population analysis. The output table from

the final model should be in this section, as well as any additional plots that are deemed

important. Where the analysis was performed as a result of a clinical study or a population

pharmacokinetic study, the study protocol should be included in the appendix.

J. Electronic Format Files

Data set and representative command files used for population analyses may be submitted as

ASCII files and/or PDF files with the filing of a new drug application. It is understood that data

format may be software specific. The Agency may, on some occasions, request that the data be

formatted in a manner that is compatible with another type of software. An electronic copy of the

report may also be a part of the submission. However, the submission of these data and reports in

electronic form does not eliminate the need to submit a paper copy.

X. LABEL

Where population model parameter estimates are included in the label, the total number of

subjects used for the analysis and the precision with which the parameters were estimated should

be included in the report. Where the results of the population analysis provide descriptive

information for the label, it should be stated that the information was obtained from a population

analysis. Information from population analyses used to characterize subpopulations should

include the total sample size used for the analysis and the proportion of subjects belonging to the

subpopulation.

ASTHMA:

DEFINITION:

An expert panel of the National Institutes of Health National Asthma Education and

Prevention Program (NAEPP) has defined asthma as a chronic inflammatory disorder of the

airways in which many cells and cellular elements play a role. In susceptible individuals,

Page 33: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

inflammation causes recurrent episodes of wheezing, breathlessness, chest tightness and

coughing. Air flow obstruction often reversible either spontaneously or with treatment.

CHRONIC OBSTRUCTIVE PULMONARY DISEASE (COPD):

The American Thoracic Society (ATS) defines COPD as a disease process involving

progressive chronic airflow obstruction because of chronic bronchitis,

emphysema (or) both.

Chronic bronchitis is defined clinically as excessive cough and sputum production on most days

for at least three months during at least two consecutive years.

Emphysema is characterized by chronic dyspnea resulting from the destruction of lung tissue

and the enlargement of air spaces.

Asthma and COPD Treatment with Theophylline:

Theophylline has been used in the treatment of asthma and chronic obstructive pulmonary

disease (COPD) for over 60 years and remains one of the most widely prescribed drugs for the

treatment of airway disease.

ASTHMA: Theophylline is used as bronchodilator in the symptomatic treatment of asthma. The

drug relieves the primary manifestation of asthma, including shortness of breath, wheezing and

dyspnea, and improves pulmonary function as measured by increased flow rates and vital

capacity. Theophylline also suppresses exercise- induced bronchospasm.

CHRONIC OBSTRUCTIVE PULMONARY DISEASE:

Page 34: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

In the stepped–care approach to drug therapy for chronic obstructive pulmonary disease

(COPD), theophylline may be administered orally (as an extended- release preparation) as

alternative therapy in patients with mild COPD when inhaled β2-agonist bronchodilator are

unavailable (or) as an adjunct to short (or) long –β2 acting bronchodilator (e.g. Ipratropium, a

selective inhaled β2 agonist in patients with moderate to severe COPD who require additional

therapy because of in adequate response.

THEOPHYLLINE

Theophylline is a methyl xanthine used to treat bronchospasm including asthma and obstructive

airway disease.

Theophylline occurs as a white, odourless crystalline powder. The theophylline is slightly

soluble in water and in chloroform, very slightly soluble in ether.

PHARMACOLOGY

BRONCHODILATOR ACTION

Theophylline is a weak and non-selective inhibitor of Phosphodiesterase enzymes that break

down cyclic nucleotides in the cell, thereby leading to an increase intracellular

cAMP and cyclic guanosine 3¹, 5¹- monophosphate concentrations. It relaxes airway smooth

muscle by inhibition of PDE activity (PDE3, PDE4, and PDE5), but relatively high

concentrations are needed for maximal relaxation15. It is a potent inhibitor of adenosine receptors

in the bronchi resulting in relaxation of the smooth muscle.

ANTIINFLAMMATORY EFFECT

Theophylline has anti-inflammatory effects in asthma. In patients with nocturnal asthma, low-

dose theophylline inhibits the influx of neutrophils and to a lesser extent, eosinophils in the early

Page 35: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

morning. In patients with asthma, low dose of theophylline reduce the number of eosinophils in

bronchial biopsies, bronchoalveolar lavage and induced sputum. Interleukin-10 has a broad

spectrum of anti-inflammatory effects, and its secretion is reduced in asthma. interleukin-10

release is increased by theophylline and this effect may be mediated via PDE inhibition.

OTHERS EFFECTS

Theophylline also increases sensitivity of the medullary respiratory centre to carbon dioxide, to

reduce apneic episodes. It prevents muscle fatigue, especially that of the diaphragm. It also

causes diuresis and cardiac and CNS stimulant16.

PHARMACOKINETICS

ABSORPTION

Theophylline is well absorbed from oral liquids and uncoated plain tablets, maximal plasma

concentrations are reached in 2 hrs for immediate release oral preparations and between 4 and 12

hrs after ingestion of sustained release preparations. Food may alter rate of absorption, especially

of some extended release preparations.

DISTRIBUTION17

Distributed throughout the extracellular fluids. Equilibrium between fluid and tissues occurs

with in an hour of an I.V loading dose. Average volume of distribution is 0.45L/kg

(Range 0.3 to 0.7 L/kg). Theophylline does not distribute in to fatty tissue but readily crosses the

placenta and is excreted in to breast milk. Approximately 40% is bound to plasma protein.

Therapeutic serum levels generally range from 10 to 20 mcg /ml.

METABOLISM

Metabolized in the liver to inactive compounds. Half-life is 7 to 9 hours in adults,

Page 36: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

4 to 5 hours in smokers, 20 to 30hours in premature infants, and 3 to 5 hours in children.

EXCRETION

Theophylline is primarily eliminated by metabolism via the hepatic cytochrome P-450 mixed

function oxidase microsomal enzymes primarily the CYP1A2 Isoenzyme, with 10% of dose is

excreted in the urine unchanged. The other metabolites include 1.3-dimethyl uric acid, 1-

methyluricacid and 3-methylxanthine. But in neonates around 50% is excreted un changed, and a

large proportion is excreted as caffeine.

CONTRAINDICATIONS AND PRECAUTION

Contraindicated in patients with hypersensitivity to xanthine compounds, such as caffeine and

theobromine, and in those with active peptic ulcer and seizure disorders.

Use cautiously in the elderly; in neonates, infants under age 1,and young children; and in

patients with COPD, cardiac failure, corpulmonale, renal or hepatic disease, peptic ulcer,

hyperthyroidism, diabetes mellitus, glaucoma, severe hypoxemia, hypertension, compromised

cardiac or circulatory function, angina, acute MI, or sulfite sensitivity.

INTERACTIONS

DRUG-DRUG:

Allopurinol (high dose), calcium channel blockers, cimetidine, corticosteroids, erythromycin,

interferon, mexiletine, oral contraceptives, propranolol, quinolones, troleandomycin – May

increase serum levels of theophylline. Dose adjustment may be needed if use together can’t be

avoided.

Activated charcoal, barbiturates, ketoconazole, phenytoin, rifampicin – Decreased plasma

theophylline levels. Dose adjustment may be needed if use together can’t be avoided.

Beta blockers: Exert an antagonist pharmacological effect. Avoid use together.

Page 37: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides

carbamazepine, isoniazid, loop diuretics: May increase or decrease theophylline levels.

Dose adjustment may be needed if use together can’t be avoided.

Lithium: Increased excretion of lithium. Lithium dose may require dose adjustment. Monitor

patients carefully.

Drug- Herb- Cocoa tree: Possible inhibition of theophylline metabolism. Patient should avoid

ingesting large amounts of cocoa when using theophylline.

Guarana, caffeine: Additive CNS and CV effects. Avoid use together.

Drug–Food- Any food: Accelerates absorption. Advise patient to take drug on an empty

stomach.

Drug-Lifestyle: Smoking (cigarettes, marijuana): Increases elimination of theophylline. Monitor

theophylline response and serum levels. Dose adjustment may be required.

Page 38: INTRODUCTION the Measurement of Drug Concentrations in Blood Samples Provides