You are on page 1of 9

Journal of Pharmaceutical& Biomedical Analysis 0731-7085/90 $3.00 + 0.

00
Vol. 8, Nos 8-12, pp. 629-637, 1990 (~) 1990 Pergamon Press plc
Printed in Great Britain

Method validation in the bioanalytical laboratory*


A.R. BUICK, M.V. D O I G , S.C. J E A L , G.S. L A N D and R.D. M c D O W A L L t

Department of Bioanalytical Sciences, The Wellcome Research Laboratories, Langley Court, Beckenham,
Kent BR3 3BS, UK
Abstract: Bioanalytical methods, based on a variety of physico-chemical and biological techniques such as
chromatography, immunoassay and mass spectrometry, must be validated prior to and during use to engender confidence
in the results generated. The fundamental criteria for assessing the reliability and overall performance of a bioanalytical
method are: the evaluation of drug and analyte stability, selectivity, limits of quantification and detection, accuracy,
precision, linearity and recovery. The extent to which a method is validated is dependent on its prospective use, the
number of samples to be assayed and the use to which the data are put.
Specific analytical techniques may require additional validation such as antibody-binding characteristics, peak purity
determination, evaluation of matrix effects or structural confirmation of the analyte. Ideally each assay should be cross-
validated with a method utilizing a highly specific detector such as a mass spectrometer. Once in use, the performance of
the method should be monitored using quality control standards. If a method is set up in another laboratory, the
performance of that assay should be monitored with quality control standards sent from the originating laboratory.

Keywords: Bioanalytical method validation; immunoassay; chromatography; mass spectrometry; precision; accuracy;
selectivity.

Introduction undertaken is dependent on the analyst's


experience and judgement. Validation of bio-
Why validate bioanalytical methods? analytical methods will be discussed mainly in
The reason for validating a bioanalytical the context of the drug discovery and develop-
procedure is to demonstrate the performance ment process within the pharmaceutical in-
and reliability of a method and hence the dustry; however the principles outlined are, in
confidence that can be placed on the results. In the opinion of the authors, applicable to all
addition, Shah has stated that all bioanalytical bioanalytical methods.
methods must be validated if the results are
used to support registration of a new drug or The responsibilities of the bioanalyst
the reformulation of an existing one [1]. It The r61e of the bioanalyst is a very important
should be noted that the initial validation is one; should analytical data be incorrect, then
only a beginning, as a method should be the efforts of scientists or physicians could be
monitored continually during its application to in vain. Dubious analytical data could waste
ensure that it performs as originally validated. valuable resources, cost money through delays
in registration of new compounds or even
Aims and objectives worse be a cause for wrong diagnosis and
The aim of this paper is to discuss method treatment. Therefore, the bioanalyst has a vital
validation and to provide the basis for a contribution to make. It is against this back-
comprehensive framework for validating bio- ground of responsibility that a bioanalyst
analytical methods. There are very few articles validates a method to demonstrate to himself,
in the literature that address this topic [1-5] as well as to other scientists, that the data
and there is little guidance on method valid- produced are reliable.
ation for the submission of bioanalytical
methods to journals; this can result in a International G L P requirements
variable quality of such manuscripts. There is Directives for bioanalytical method valid-
no standard plan to follow for the validation of ation are not clearly addressed in the Good
a bioanalytical method and the work that is Laboratory Practice (GLP) regulations [6-9];

* Presented at the "Second International Symposium on Pharmaceutical and Biomedical Analysis", April 1990, York,
UK.
t Author to whom correspondence should be addressed.

629
630 A.R. BUICKetal.

however, comments that have been made concentrations and requires a large dynamic
during inspections show that validation of range. In contrast, immunoassays often require
analytical methods is audited, especially by the sample dilution in order to cope with such
Food and Drug Administration (FDA). More- dynamic ranges of analyte concentrations.
over, a publication by Shah [1] stated the
viewpoint that certain criteria must be defined Matrix effects. There are many different
and tested prior to a method being used to biological matrices that can be encountered in
support regulatory studies. All GLP guidelines bioanalysis, a selection is presented in Table 1.
rely on the use of standard operating pro-
cedures (SOP) related to the tests conducted Table 1
during the course of a study to ensure the Some biologicalmatricesencounteredin bioanalysis
quality and integrity of the data generated, and
Fluids Tissues
a SOP on validation of an analytical method is
clearly required and therefore inspected. Plasma/serum Liver
A Quality Assurance Group (QAG) work- Whole blood Brain
Urine Lung
ing party [10] made the important point that Bile Spleen
different groups of analysts will have different Stomach content Kidney
opinions about which criteria to select for Cerebrospinal fluid (CSF) Skin
Milk Faeces
validation but it is important that a system for Saliva Adipose tissue
validating methods is agreed and defined in a Lachrymalfluid Heart
SOP. Vesicle fluid Muscle
Broncoscopyfluid
Semen
Differences between pharmaceutical analysis
and bioanalysis
It is appropriate first to discuss the differ- Each presents a slightly different challenge to
ences between pharmaceutical analysis and the analyst as each biological matrix can
bioanalysis to show the problems facing a contain many components which can influence
bioanalyst when considering validating a or interfere with the method.
method. Pharmaceutical analysis will be de-
fined as the detection or measurement of an Cellular material or debris - - varying from
analyte in a pharmaceutical formulation, whilst erythrocytes in whole blood to disrupted cells
bioanalysis is the detection or quantification of in tissue homogenates. These can release
an analyte in a biological matrix. There are the enzymes capable of metabolizing an analyte or
following differences to consider: can sequester analytes with similar physico-
chemical properties within membranes present
Concentration. In pharmaceutical analysis in the matrix.
the analyte concentration is fixed within known
manufacturing limits. In bioanalysis, the con- Macromolecules - - such as enzymes or other
centration of the analyte is dynamic, being proteins, can bind to or degrade the analyte or
determined by the absorption, distribution, interfere with binding in immunoassays. This
metabolism, excretion and sometimes chemical may require special treatment to the matrix to
degradation of the compound. Other factors overcome or inhibit these phenomena.
that can affect analyte concentration are the
dose and its route of administration, drug Metabolites or precursors - - of the analyte or
formulation, whether food was taken prior to compounds in the matrix which can interfere
ingestion of a dose, species and sex. with the detection of the analyte may have to
be removed or separated.
Concentration range. The above effects may
require the bioanalytical method to have a Concomitant drug therapy - - whereby co-
dynamic range of three or more orders of administered drugs or their metabolites can
magnitude in order to monitor the analyte interfere with the determination of an analyte.
concentrations effectively. This means that
analytical equipment, particularly a chromato- Species differences. Different species and
graphic detector, is often used at the lower sexes can exhibit differences in absorption,
limit of its specification to measure small metabolism and elimination of the analyte. In
BIOANALYTICAL METHOD VALIDATION 631

addition, there are differences in the com- repeatability (which is an assessment of pre-
position of the same matrix from different cision during a single analytical run) and
species which leads to the problem that an between-day precision, inter-assay precision or
assay for a compound in human plasma may reproducibility (which shows the variation of
not be valid for measuring the same analyte in precision with time and may also include
dog plasma. different analytical staff, equipment, and re-
Biomedical analysis places special emphasis agents).
on parts of the analytical method such as the
sample preparation and detection stages to Accuracy
ensure the specificity of an assay. This is the closeness of the determined
concentration to the true value. Accuracy is
measured as within-day or intra-assay (assess-
ment of a single assay) and between-day or
Validation Parameters
inter-assay (over several analytical runs)
What analytical parameters could be tested values; the latter figure is usually more rep-
and monitored? Ideally, all bioanalytical resentative and should be quoted. Accuracy is
methods should be evaluated against the an absolute measurement and an accurate
parameters listed in Table 2 and defined method depends on several factors such as
below. specificity and precision.

Analytical reference standard Limit of detection (LOD)


Before validation can be contemplated, an The lowest amount of analyte that can be
authenticated analytical reference standard detected but not quantified. The calculation of
must be available to prepare solutions of the LOD is open to misinterpretation as some
known concentrations. This standard should be bioanalytical laboratories just measure the
of a known form, e.g. free base or salt and of lowest amount of a reference solution that can
known purity, if used over a period of time, be detected and others the lowest concen-
should be monitored to ensure no decom- tration that can be detected in the biological
position or contamination has taken place. sample. The latter method is more meaningful
as there may be a signal due to the matrix
Precision itself. This parameter is dependent on the
This describes the closeness of replicate background signal, whether it be due to endo-
determinations of an analyte by an assay. genous substances or electronic noise, at the
Precision can be further sub-divided into time of measurement and a better approach
within-day precision, intra-assay precision or may be to use the limit of quantification.

Table 2
Bioanalytical method parameters that ideally should be examined for every assay and every analyte and every matrix
Calibration curve: Number of standards, concentrations, replicates and line fit
Precision: Intra-assay (within-day, repeatability)
Inter-assay (between-day, reproducibility)
Accuracy: Intra-assay (within-day bias)
Inter-assay (between-day bias)
Limit of detection (LOD)
Limit of quantification (LOQ)
Working concentration range of the assay What to do if the measured concentration is outside of this range:
extrapolate curve or dilute sample
Linearity
Selectivity (specificity)
Matrix effects
Recovery
Stability: Drug solutions
In the biological matrix under storage and room temperatures
Investigate stability in the matrix between sampling and storage
Extracts pending analysis
Cross validation with another analytical technique

This list assumes the availability of an authenticated reference standard with known purity and confirmed structure.
632 A.R. BUICKetal.

Limit of quantification (LOQ) half-lives, therefore to stabilize the compound


This is defined as the lowest concentration of an inhibitor should be added, the effectiveness
analyte that can be measured with acceptable of which will need to be assessed and validated.
precision and accuracy by the method. The
definition of acceptable is dependent on the Selectivity
aims and objectives of the work which the This is the power of the bioanalytical method
method is supporting and should be decided by to distinguish between the analyte and its
the analyst. metabolites, endogenous compounds and arti-
facts introduced by the analytical method.
Range and linearity Initial experiments must be set up to demon-
The range of a bioanalytical assay is the strate selectivity but it is an on-going exercise,
concentration interval over which an analyte throughout the application of the method.
can be measured with acceptable precision and
accuracy. Linearity of a method is where the
determined response is directly proportional to
Experimental Plans for the Initial Validation of
the analyte concentration. All bioanalytical
a Bioanalytical Method
methods should have a good working range but
not necessarily be linear. This section will cover the experimental
plans, which in the opinion of the authors,
Stability should ideally be applied to the validation of
The stability of the analyte must be investi- every bioanalytical method. The expected re-
gated under various conditions: in the standard suits and error resolution of the validation tests
solutions used to prepare calibration curves, in are dependent on the nature of work supported
any biological matrix stored at - 2 0 ° C and at by the method. A n example of this is shown in
room temperature prior to analysis and also in Table 3 which outlines the requirements of an
the final extract awaiting analysis. There may assay for the various stages of drug discovery
also be the need to investigate the stability of and development. However, there is no uni-
the analyte between the sample being taken versally accepted plan for validation, the test-
and stored: some compounds are metabolized ing will depend on the judgement of the
by esterases in the blood and have very short individual scientist.

Table 3
Requirements for bioanalytical methods used in drug discovery and development
Development stage Analyticalobjectives Practical validation

Discovery Comparative assessment of a number of Precision and accuracyto within +20% or less
related compoundsfor absorption, Basic assessment of stability, and selectivity
metabolism and elimination LOQ relatively high
Preliminary recoverydata

Preclinical To support safety and metabolism studies Precision and accuracyto within + 10% or less
in various animal species Selectivity: the method must be revalidated for each
animal species and matrix requiring analysis plus
on-goingevaluation
Recoverycalculated
Stability investigated: on-goingassessment
LOQ: goodsensitivity
Range and linearity defined

Clinical Method sensitive and specificfor the Precision and accuracyto within + 10% or less
determination of human Selectivity: cope with the lower concentrations
pharmacokinetics measured in man, known metabolites and
concomitant medication. Method validated for each
matrix to be analysed
Therapeutic monitoring Ideal stability: minimum3 months in samples at
known storage conditions
High sample throughput LOQ: at least 10% of the minimumeffective
concentration
Cross-validated against another analytical
technique
Range and linearity defined for each matrix
BIOANALYTICALMETHOD VALIDATION 633

Validate the method for each matrix be performed using pooled material and the
Each biological matrix from each species method of standard additions [12].
should be validated. A method that was devel-
oped for plasma from one species may not be Analyte stability
applicable to urine from the same species or As part of validation, analyte stability must
plasma from another species due to the be investigated, this is a continuing process
presence of different endogenous compounds that involves a number of components.
or analyte related material. Therefore ad-
ditional sample preparation or modification of Standard solution stability. The analyst needs
the analytical conditions may be necessary [4]. to know how long and under what conditions
An area of increasing concern, which may standard solutions of the analyte(s) can be
require thought when validating bioanalytical kept. In the absence of any information, these
methods in the future, is the supply of control solutions should be freshly prepared until
matrix. A relatively large volume is required evidence of stability is available. Failure to do
for standards, quality controls and blank this could result in standards that do not
samples during validation and routine oper- represent the true concentration of analyte,
ation. Ethical problems may be raised, and leading to erroneous determination of un-
methods may need to be cross-validated with known samples.
aqueous or artificial standard solutions for
routine use, possibly with the use of correction Stability in the biological matrix. Analyte
factors. stability should be determined in each matrix
and species likely to be studied.

Preparation of spiked samples. The samples (1) How long can the sample be stored at both
used for the validation experiments are usually ambient temperatures and deep frozen before
prepared from a control matrix with known the analyte decomposes beyond acceptable
additions of analyte. The preparation of spiked limits? This information is essential as it will
aqueous or semi-aqueous matrices such as determine the time that a sample can be left
plasma, serum, urine and cerebrospinal fluid between sampling and freezing and the time
are relatively easy, as the analyte is normally samples can be stored and the time undiluted
readily miscible in them. Ideally, the analyte sample can be left in autosamplers.
should be added to the matrix to make a stock To investigate ambient temperature stabil-
solution which is then diluted with the control ity, an experiment should cover 6 or 24 h for
matrix. However, many methods use the direct whole blood or plasma where replicate samples
addition of suitable analyte concentrations to are taken and assayed at hourly or 2-hourly
individual aliquots of the matrix. Inappropriate time points after spiking. Urine and faecal
choice of solvent or spiking volume can cause samples are usually collected over longer time
protein precipitation and/or variation in ex- periods (24 h or more) and stability tests
traction recoveries. If consistency of solvent should reflect this. If instability is found, the
additions is not maintained between standards analyst should investigate what precautions
and unknown samples then experimental bias should be taken to prevent degradation or
can occur. artifact formation. This may include the ad-
Matrices become more difficult to spike with dition of antioxidants or enzyme inhibitors to
analyte the less fluid they are and consequently sample collection vessels or processing im-
the analytical results may have more error mediately after sampling.
associated with them. Solid matrices may cause Once the sample has been deep frozen,
even greater problems [11]. storage stability is investigated; this is a con-
When measuring endogenous compounds in tinuous process and may cover a long and
biological samples, how does one obtain an indeterminate time period. Samples are spiked
analyte-free control sample? This may be with the analyte at two concentrations, separ-
achieved by either chemical degradation or ate aliquots are prepared, and replicate
enzymic conversion, e.g. histamine can be samples taken for analysis at increasing time
removed from plasma by using diamine points until signs of instability are observed.
oxidase, followed by deactivation of the The length of a stability study is usually
enzyme. If this is impossible, the analysis can determined by the initial storage requirements,
634 A.R. BUICK et al.

although it is always useful to know likely analytical procedures provided that a pure
maximum sample storage times. Once instabil- reference standard exists. During initial
ity is observed further experiments could be method development, a preliminary indication
conducted to determine the actual time limits of recovery can be obtained very quickly by
of storage. comparing the responses of extracted and
The normal storage temperature for the unextracted standards at the mid-point of the
majority of biological samples is -20°C. calibration curve. For a formal validation of a
Plasma is normally frozen solid at these tem- method, two calibration curves prepared in this
peratures but around macromolecules, such as way should be compared over the whole range
glycoproteins, water still has sufficient poten- of the assay to determine if recovery varies
tial energy to be mobile at temperatures below with concentration. The latter approach is
-20°C. Urine, depending on the salt content, limited by the availability of radio-labelled
may still contain micro droplets of water at analyte and the facilities to use and measure it.
temperatures as low as -25°C which may Labelled analyte is added to the control matrix
influence stability of the analyte. Ideally at physiological concentrations and then taken
laboratories should have facilities for storing through the method. The advantage is that
samples at -40°C and -80°C which may over- every fraction from the extraction scheme can
come any instability problems observed at be rapidly measured to ascertain where any
-20°C. Some instability problems may be losses occur.
overcome by various methods of sample pre-
treatment such as heat inactivation, alteration Linearity of detector response
of pH or freeze-drying. The linearity of chromatographic detector
response should be established. This is import-
(2) The effects of freeze-thaw cycles must be ant when an assay is required to quantify an
investigated. This can be undertaken by spiking analyte over a large concentration range. Sol-
a control matrix with known concentrations of utions of pure analyte are injected in duplicate
the analyte(s) and subjecting aliquots to freez- into the chromatograph and the response
ing and thawing cycles. This approach will recorded. The increments between successive
determine the stability of the analytes to analyte concentrations should not exceed an
freezing and thawing, but unless the stability of order of magnitude in order to determine the
all metabolites is known, extrapolation of the concentration at which the detector becomes
findings to in vivo samples may be erroneous as non-linear. This is not usually done if the
unknown compounds (e.g. labile glucuronides) dynamic range required of the method is one
may break down to one of the analytes. or two orders of magnitude but is encouraged
as the bioanalyst can never guarantee when the
(3) The final area to investigate is the stability range of an assay needs to be extended.
of sample extracts. This information could
determine the size of the batch run and the Range and linearity of the assay
maximum time between extraction and re- During the validation, procedures should be
analysis. This area concerns mainly chromato- developed to cope when a result is outside of
graphic analysis. In contrast, the final stage of the calibration range of the assay. There are a
a radioimmunoassay involves labelled material number of options to consider:
which is stable over several days. - -How far can a calibration line be extrapol-
ated outside of the concentration range of
Recovery the standards?
The recovery of an analyte from sample - -Should a sample be repeated using a smaller
pretreatment prior to analysis is important as it or larger aliquot to ensure that the result is
is a determining factor in the limit of quantifi- within the working range of the assay? If so,
cation of the whole analytical procedure. It can what is the effect of the matrix when a
be used to improve extraction efficiencies and larger sample volume is assayed? If a
is an indicator of the robustness. Recovery can smaller volume is used, should it be made
be calculated in one of two ways: either up to the nominal volume used in the assay
comparison of extracted and unextracted stan- by using the control matrix or a buffer?
dards or using radioactive analyte. The former - - S h o u l d the analysis be repeated after
method can be applied to virtually all bio- dilution of the sample? What are the effects
BIOANALYTICAL METHOD VALIDATION 635

of diluting the sample with either a buffer or another more selective method, where this is
control biological fluid? possible, can provide more information about
The option chosen will depend on the analyt- the accuracy of the method.
ical technique used, e.g. immunoassay or The limit of quantification (LOQ) should be
chromatography, and the linear range of the assessed by analysing replicate samples of low
detector used. Regardless of the approach, the analyte concentrations to determine at what
analyst should have this information available concentration acceptable limits of precision
before it is required rather than having to and accuracy have been exceeded. The limit of
investigate retrospectively. detection (LOD) is often taken as two or three
times the background response which may be
Precision and accuracy experiments due to electrical noise and/or minor chromato-
Initial validation of a method should give an graphic peaks but should be determined in
indication of the intra-assay and inter-assay samples derived from the biological matrix.
variation and accuracy; therefore two exper- Estimates of concentrations that fall between
iments should be set up. Each will have six the LOD and LOQ may be useful in some
replicates at three concentrations covering the cases for pharmacokinetic interpretation, but
top, middle and bottom of the calibration in this instance an indication of the precision
curve. After comparison with calibration stan- attainable at these concentrations is highly
dards, the analyte concentration in the samples desirable lest too much weight be put on such
can be measured and then the precision and results.
accuracy of the assay can be calculated.
Readers may question the approach of using Selectivity
separate samples for this work, but it is the Assay selectivity is an important part of
authors' opinion that the samples for precision bioanalytical method validation as the main
and accuracy should be determined against a techniques used for analysis are comparative
normal set of calibration standards. For more and not absolute. This means that standards
detailed validation, a third experiment should are used to calculate the analyte concentration
be performed to give more confidence in the in the samples. There are two sources of
robustness of the assay. interference in bioanalytical methods: endo-
Precision is expressed either as relative genous substances that are present in the
standard deviation (RSD) or coefficient of biological matrix and exogenous material that
variation (CV); the smaller the values the arises from the apparatus (e.g. plasticizer
better the assay performance. As precision can additives) and reagents used in the method. In
vary over the range of an assay, it is important addition, binding of the analyte to the glass or
to determine it at the top, mid point and plastic vessels used in the analysis is a common
bottom of the range as a minimum require- phenomenon [11].
ment. Usually the precision of an assay is The impact of interference on an assay can
constant over most of the range of an assay but be variable. This can range from a small but
it may decrease at the extremes of the calib- constant bias at very low concentrations to
ration curve, hence the importance of testing at intermittent problems caused by interference
the top and bottom of the range. arising from the use of different batches of
The accuracy of analytical methods is ex- reagent or materials, and such factors can
pressed as percentage bias or error which can make an assay impracticable. Therefore the
be a positive or negative figure; like precision specificity must be monitored continually to
the lower the value, the better the assay ensure that there is no interference.
performance. Accuracy can be measured The nature of the analytical techniques used
against an in-house, an inter-laboratory or an means that the basic assumptions of each must
international standard. In some respects accur- be investigated, for example peak purity in
acy is a difficult parameter to measure for two chromatography and antibody-binding in
reasons. Firstly, accuracy is an absolute immunoassay. Therefore work must be under-
measurement and most analytical methods taken to test these assumptions continually
used in a bioanalytical laboratory are compara- during the use of the assay.
tive, e.g. chromatography and immunoassay. The initial application of a chromatographic
Secondly, how closely does a spiked sample assay in a drug's discovery may involve the use
reflect the in vivo situation? Comparison with of predose samples only with reagent and
636 A.R. BUICK et al.

matrix blanks included with each analytical run outlined earlier, has been determined, then
to monitor assay specificity. However in drug analysing quality control (QC) samples is the
development, specificity investigations may most popular method for monitoring assay
also include the use of diode array or mass performance. The assay precision, defined
spectroscopic detectors to improve confidence during validation, sets the acceptable limits of
in the method. Immunoassay methods require performance, and the results can be displayed
an investigation of cross-reactivity with similar on a Shewart plot [12]. Alternatively, the same
structural compounds, non-specific binding QC results can be plotted as a cumulative
and matrix effects as a means of ensuring summation (cusum) [12].
specificity. As more information about the Brooks and Weinfeld [2] used two concen-
compound becomes known then any metabol- trations of QC samples near the ends of the
ites, co-administered drugs and their metabol- calibration curve to monitor assay perform-
ites should be tested for either co-elution or ance. This is a good approach but the accept-
cross-reactivity as appropriate. ance or rejection criteria of any QC results that
Often there are instances when one analyt- fall outside the predefined limits should be
ical method will be replaced by another; during established. Such criteria may be dependent on
the change-over period the results generated how much sample remains to repeat the
by the two methods on the same samples analysis, how far outside the limits the results
should be compared. were and the speed at which the results are
required. In the end it is the responsibility of a
Additional validation options bioanalyst to judge whether to accept or reject
Other factors to consider when validating a the results of an analytical run.
method may be turn-around time, cost per
analysis and data management costs. Turn- Method modification and revalidation
around time may be a required option depend- If changes are made to a validated method,
ing on the context of the result; with accident then it is the bioanalyst who must judge how
and emergency cases the physician may require much revalidation should be undertaken. This
to know if a patient has ingested a poison and can range from a change in the grade or
request measurement of its concentration in supplier of a chemical to a major modification
body fluids. Time in this context may be life- of the method: the greater the modification,
saving hence the need to validate the correct- the greater the need to revalidate the method.
ness of the analytical result when the analyst is In this latter context, precision and accuracy,
operating under pressure. Again, rapid turn- LOQ and LOD are the minimum parameters
around and high throughput may be required that should be redetermined. Changes in
to support early studies on new drugs in man stability, apart from that in different sample
when little pharmacokinetic information may extracts should not occur.
be available. Assay performance needs to be
assessed critically in these circumstances be- Acknowledgements - - The authors wish to thank Dr P.D.
cause of the possibility of unknown factors Whiteman, Department of Bioanalytical Sciences, Well-
come Research, and Dr B.V. Fisher, Wellcome Biotech
affecting results. for discussions and comments during the preparation of
In a cost conscious business environment, it this article.
may be essential to assess the method against
operating costs such as those for instrument
time, disposable apparatus required and References
analyst's time, and against the efficiency of the [1] V.P. Shah, Clin. Res. Prac. Drug Reg. Affairs 5,
data management procedures. 51-60 (1987).
[2] M.A. Brooks and R.E. Weinfeld, Drug Dev. Ind.
Pharm. 11, 1703-1728 (1985).
[3] D. Dadgar and M.R. Smyth, Trend Anal. Chem. 5,
Routine Operation of a Bioanalytical Method 115-117 (1986).
[4] E. Gelpi, Life Sci. 41,849-852 (1987).
Once validated, a method should be moni- [5] L. Aarons, S. Toon and M. Rowland, J. Pharmac.
tored by quality control procedures to show Meth. 17, 337-346 (1987).
how it performs routinely. [6] The United Kingdom Compliance Programme,
Department of Health and Social Security, London
1986 and subsequent amendments, Department of
In-house monitoring of assay performance Health (1989).
Once the initial stability of the analyte, as [7] United States Food and Drug Administration, Title
B1OANALYTICAL METHOD VALIDATION 637

21 Code of Federal Regulations Part 58, Federal Practice, Drug Metabolism Working Party (1987).
Register 22 December 1978, and subsequent Unpublished report.
ammendments. [11] M.V. Doig and A.E. Jones, in Analysis of Drugs and
[8] Organization for Economic Co-operation and Devel- Metabolites Including Anti-infective Agents (E. Reid
opment, ISBN 92-64-12367-9, Paris (1982). and I.D. Wilson, Eds), pp. 157-162. Royal Society of
[9] Japan Ministry of Health and Welfare Notification Chemistry, Cambridge (1990).
No. Yakuhatsu 313, Pharmaceutical Affairs Bureau, [12] J.N. Miller and J. Miller, Statistics for Analytical
31 March 1982 and subsequent ammendment Notifi- Chemists. Ellis Horwood, Chichester (1984).
cation No. Yakuhatsu 870, Pharmaceutical Affairs
Bureau, 5 October 1988. [Received for review 5 April 1990;
[10] Quality Assurance Group (UK), Good Laboratory revised manuscript received 1 May 1990]

You might also like