Vous êtes sur la page 1sur 8

The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

Themed Issue: Bioanalytical Method Validation and Implementation: Best Practices for Chromatographic and Ligand Binding Assays
Guest Editors - Mario L. Rocci Jr., Vinod P. Shah, Mark J. Rose, and Jeffrey M. Sailstad

Key Elements of Bioanalytical Method Validation for Macromolecules


Submitted: February 7, 2007; Accepted: April 19, 2007; Published: May 18, 2007
Marian Kelley1 and Binodh DeSilva2
1Centocor R&D Inc, Radnor, PA
2Amgen Inc, Thousand Oaks, CA

ABSTRACT preclinical and clinical studies. Most of these methodologies


are used in quantitative assays supporting pharmacokinetic
The Third American Association of Pharmaceutical Scien-
and toxicokinetic parameters of the therapeutic agents.
tists/US Food and Drug Administration (FDA) Bioanalyti-
cal Workshop, which was held May 1 and 2, 2006, in The methods that are primarily used in these evaluations are
Arlington, VA, addressed bioanalytical assays that are being ligand binding assays (LBAs [or, for this publication’s pur-
used for the quantification of therapeutic candidates in sup- poses, immunoassays]), where the specificity and selectivity
port of pharmacokinetic evaluations. One of the main goals of the assays depend on the interactions of other biological
of this workshop was to discuss best practices used in bio- molecules, such as receptors, antibodies against the thera-
analysis regardless of the size of the therapeutic candidates. peutic candidates, and aptamers. The response observed in
Since the last bioanalytical workshop, technological these methods is indirectly related to the concentration of
advancements in the field and in the statistical understand- the therapeutic, that is, the basis of the detection is an enzy-
ing of the validation issues have generated a variety of inter- matic or radiochemical response tied to a variety of binding
pretations to clarify and understand the practicality of using interactions. There is no direct physicochemical property of
the current FDA guidance for assaying macromolecular a macromolecule that can be used in this determination
therapeutics. This article addresses some of the key ele- (unlike for a small-molecule drug candidate). Because of the
ments that are essential to the validation of macromolecular nature of these binding interactions, the dynamic range of
therapeutics using ligand binding assays. Because of the the standard curves is narrow as well as nonlinear/sigmoidal.
nature of ligand binding assays, attempts have been made There are several publications that discuss the validation
within the scientific community to use statistical approaches aspects of LBAs in detail.1,2 The purpose of this article is to
to interpret the acceptance criteria that are aligned with the highlight the key elements of validation of bioanalytical
prestudy validation and in-study validation (sample analy- methods that support the pharmacokinetic and toxicokinetic
sis) processes. We discuss, among other topics, using the assessments of macromolecules that were discussed at the
total error criterion or confidence interval approaches for Third American Association of Pharmaceutical Scientists
acceptance of assays and using anchor calibrators to fit the (AAPS)/US Food and Drug Administration (FDA) Bioana-
nonlinear regression models. lytical Workshop in 2006 and that warrant further consider-
ation. These key elements include the selection of reagents
KEYWORDS: Bioanalytical validation, ligand binding for these methods, the format of these assays, the determi-
nation of the accuracy and precision of these methods
assays, macromolecules, biological matrices, immunoassay
(where there is no extraction procedure that is usually used),
the importance of the reference material that is available for
INTRODUCTION use as a standard, and the selectivity of the matrix.
The increased number of biological agents used as thera-
peutics (in the form of recombinant proteins, monoclonal
SELECTION OF REAGENTS AND ASSAY FORMATS
antibodies, vaccines, etc) has prompted the pharmaceutical
industry to review and refine aspects of the development One of the most important aspects of developing and vali-
and validation of bioanalytical methods for the quantification dating an LBA is the availability of reagents. For most
of these therapeutics in biological matrices in support of novel and innovative macromolecular therapeutic mole-
cules, there are no commercially available reagents. As a
Corresponding Author: Binodh DeSilva, Amgen Inc, result, unique reagents must be developed within the inno-
One Amgen Center Drive, Department of Pharmacokinetics vator organizations.
and Drug Metabolism, Mail Stop 30E-3-C, Thousand The critical building blocks of LBAs are the ligand reagents,
Oaks, CA 91320. Tel: (805) 447-5562, (805) 630-5346; which typically are an antibody or a pair of antibodies for
Fax: (805) 499-9027; E-mail: bdesilva@amgen.com immunoassay-based assays. Other reagents may include
E156
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

binding proteins, receptors, oligonucleotides, and peptide ing activity/epitope for the capture or detection molecule,
fragments. These reagents must be selected in a manner that making the method unsuitable for the intended purpose. At
allows for suitable specificity and selectivity for the intended the least, an assessment of the appropriateness of the new
use and should have binding characteristics that allow for lot should be conducted.
durable and stable antibody/antigen complex formation. When an endogenous counterpart exists in the matrix
Two additional aspects that are overlooked in many situa- (eg, erythropoietin), it is critical to have the appropriate ref-
tions are the availability of sufficient quantities of these erence material for the quantification of therapeutics. The
reagents and the stability of these reagents. Some reagents assay specificity and the accuracy of the measurements must
are subject to lot-to-lot variation (eg, conjugated antibodies, be evaluated carefully under these conditions. Depending
radiolabeled ligands). Therefore, it is imperative that suffi- on the binding properties of the ligands used in the assay,
cient quantities be available to support long-term studies. there could be an underestimation or an overestimation of
The reagents that are commonly used in LBAs are macro- the concentrations measured when an endogenous protein is
molecules themselves, and it is essential that the handling present in the biological matrix. Table 1 compares the major
and storage be accomplished without destroying the integ- characteristics of small-molecule and macromolecule
rity of these reagents. The LBAs are only as good as the compounds.
reagents that are used; hence, the assay sensitivity and the
robustness may be drastically affected if the reagents are
unstable.3 It is important that the quality and characteristics SPECIFICITY AND SELECTIVITY
of these critical reagents be fully documented.
Specificity is the ability to measure the macromolecular
In addition to considering the critical reagents used in the therapeutic unequivocally in the presence of other compo-
LBAs, one must carefully consider the assay diluents, the nents in the assay matrix. The specificity of the ligands
specific characteristics of the analyte, the intended matrix, (antibodies, receptors, etc) determines the applicability,
and the binding entities (eg, antibodies or receptors). For sensitivity, and robustness of an LBA. LBAs have to be spe-
example, the addition of heavy metals or chelating agents cific for the macromolecular therapeutic of interest, espe-
may be required to enable the necessary confirmation for cially because they measure the therapeutic without extraction
optimal binding. Additionally, the need for detergents (eg, procedures. There are 2 types of nonspecificities: specific
Tween 20 or Triton X-100) or bulking proteins (eg, albu- nonspecificity and nonspecific nonspecificity.4 Specific
min, casein, or gelatin) must be considered to optimize nonspecificity results from the interferences of compounds
assay performance. that have physicochemical properties similar to those of the
There are a multitude of assay formats for LBAs, depending
on their intended use. Assay formats can include sandwich, Table 1. Comparison of the Characteristics of Small-Molecule
and Macromolecule Compounds
competition, direct or indirect binding, inhibition, and solid
phase or solution phase assays. In recent years, there have Characteristic Small Molecules Macromolecules
been advances in the detection platforms in addition to the
standard colorimetric assays (eg, luminescence, electroche- Size Small (<1000 Da) Large (>5000 Da)
miluminescence), providing a wide array of assay formats Structure Organic molecules Amino acid
to choose from. biopolymers; could
be multimeric
Purity Homogeneous Heterogeneous
REFERENCE STANDARD Solubility Often hydrophobic Often hydrophilic
Stability Chemical Chemical, physical,
Macromolecule therapeutic agents are produced in cell cul-
and biological
ture; hence, they are not characterized as rigorously as
Presence in matrix Xenobiotic (foreign) Endogenous
small-molecule drug candidates. There is a greater potential
Synthesis Organic synthesis Biological production
for lot-to-lot variability in purity and potency in these prep-
Metabolism Defined Not well defined;
arations. In many instances, a true “reference standard” may
could be
not be available; rather, a well-characterized material may
biotransformed
be the only choice. It is critical to develop and validate the
depending on the
methods for macromolecules with the appropriate reference
environment as well
material used in the relevant study (ie, the lot of material
as in vivo
used in the validation may not be the same as the adminis-
conditions
tered material in the clinical study). The reference material
Serum binding Albumin Specific carrier
used in the clinical study may have different posttransla-
proteins
tional modifications, which could result in the loss of bind-
E157
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

analyte (eg, endogenous compounds, isoforms, and vari- quantification of macromolecules. Small-molecule assays
ants with different posttranslational modifications that may often include a preassay extraction, which is often helpful to
have similar epitopes). Specific nonspecificity is sometimes alleviate problems from individual matrix variability. In
referred to as cross-reactivity. Theoretically, an anti-idiotypic addition, the use of either analog or stable isotope-labeled
antibody reagent is considered to have high specificities internal standards in liquid chromatography/mass spectrom-
toward the analyte and, therefore, low cross-reactivity. For etry assays for small molecules normalizes the influence of
a given critical reagent, data that describe the binding char- matrix effects and system fluctuations. The inherent charac-
acteristics (eg, cross-reactivity with related compounds) teristics of macromolecular therapeutics make it difficult
must be taken into consideration before the reagent’s use and often impossible to extract samples before analysis.
in an LBA. During method development, the specificity LBAs used to quantify macromolecules, therefore, are often
of the ligands should be evaluated using compounds that developed to measure analyte in complex matrices without
are variant forms of the therapeutic as well as other physi- extraction. Many macromolecular therapeutics are recom-
cochemically similar compounds, and anticipated con- binant or modified variants of endogenous proteins. It is
comitant medication. highly unlikely that most of the LBA reagents used will be
Nonspecific nonspecificity arises from interferences from able to distinguish between the therapeutic and the endoge-
unrelated compounds, especially matrix components (eg, nous counterpart, which could affect the accuracy of mea-
heterophilic antibodies, rheumatoid factor, proteases), in surement of the assay. In these cases, special considerations
the LBA. Nonspecific nonspecificity is sometimes referred must be made for matrix selection and for analysis of data.
to as the matrix effect. This matrix effect is one of the main The matrices collected for bioanalysis include plasma,
reasons that LBAs need more method development or vali- serum, urine, cerebrospinal fluid, synovial fluid, and homog-
dation work to be conducted during the switch from one enized tissue. The characteristics of the macromolecule can
matrix to another across animal species or even within the be affected by the methods used for sample preparation, the
same species. It is strongly recommended during clinical need for additives (anticoagulants, protease inhibitors, etc)
study support that the matrix from the relevant disease at the time of collection, the stability of the macromolecule
populations be tested for matrix effects as soon as it becomes during collection procedures (whole blood before separa-
available. The matrix effect should be evaluated by compar- tion of plasma or serum), and the postcollection processing
ing the concentration-response relationship of both spiked and storage conditions (temperature, vial type, shipping,
and unspiked samples of the biological matrix (the recom- freeze-thaw cycles, etc), so these characteristics must be
mendation is 10 lots from individual sources) to a compa- evaluated during the method development phase. Assay for-
rable buffer solution. It is recommended that the spiked mat, sample collection conditions, and other factors may
sample concentrations be at the low and high end of the influence the choice of matrix in the assay (eg, plasma is the
dynamic range. preferred matrix for labile analytes because of the extended
Nonspecific nonspecificity can usually be reduced or elimi- time needed for the preparation of serum and because of the
nated by dilution of the matrix with a buffer containing presence of proteolytic enzymes).
chaotropic or chelating agents. This is referred to as the min- Spiked samples (ideally at the low and high concentrations)
imal required dilution (MRD). Other sample cleanup proce- should be prepared in the same matrix as the anticipated
dures such as liquid-liquid, solid phase, or immunoaffinity matrix of the unknown study samples to evaluate the accu-
extractions are also applicable where the nonspecific inter- racy (recovery) of the method. In the absence of an endoge-
ferences are stronger. In either situation, the sensitivity (the nous component, simple spiked recovery studies using the
lower limit of quantification [LLOQ]) should be reported as nominal concentrations will be sufficient to qualify a
the concentration of the therapeutic in the 100% matrix. matrix.1,2 The use of a stripped matrix (eg, charcoal, immu-
Specificity and selectivity evaluations verify that the assay noaffinity) or an alternative matrix (eg, protein buffers, dia-
is specific for the intended analyte and can select the ana- lyzed serum) is not recommended but is necessary when no
lyte from a complex matrix without positive or negative other strategy for quantification can be designed for mea-
interference. suring endogenous analytes.
Regardless of the source of the matrix interference, validation
samples (ie, quality control [QC] samples used during the
MATRIX SELECTION, SAMPLE PREPARATION, prestudy validation phase) must be prepared using the same
AND MRD type of neat, unaltered matrix as was used for the study samples
The considerations that pertain to matrix selection are one for the determination of the assay’s precision and accuracy.
of the key differences between the assays developed for The MRD for an assay is the minimum magnitude of
small-molecule analysis and the LBAs developed for the dilution to which a sample must be subjected to optimize
E158
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

accuracy and precision in an assay run. When the standard is truncated for that particular run and the samples out of the
prepared in 100% matrix, no MRD exists, and samples can be range must be repeated.
assayed undiluted or neat. In other cases, where the endoge-
nous material does not generate a linear signal or a back-
ground signal is observed due to matrix effects, dilution of PRECISION AND ACCURACY
the sample may be required to establish acceptable linearity. Method precision (random error, variation) and accuracy
If there needs to be a matrix lot change during the course of (systematic error, mean bias) for LBAs should be evaluated
study sample analysis, appropriate QC samples must be by analyzing validation samples (QC samples) that are pre-
prepared to evaluate the comparability of the data obtained pared in the same biological matrix as the anticipated study
during the prestudy validation. samples. For analytes with endogenous components, the
reader is referred to the section Matrix Selection, Sample
Preparation and MRD, for further details about preparing
NONLINEAR STANDARD CURVES AND MODEL validation samples using an altered matrix. At least 5 con-
SELECTION centration levels (anticipated LLOQ, less than 3 times
LBAs measure the signal of a series of interactions that fol- LLOQ, mid, high, and anticipated ULOQ), with at least 2
low the law of mass action, resulting in a nonlinear and independent determinations per assay, should be run during
often sigmoidal standard curve. The response error relation- the prestudy validation phase. The interbatch variance com-
ship is not constant (heteroscedastic); therefore, the highest ponent is usually higher in LBAs than the intrabatch vari-
precision does not necessarily coincide with the highest ance component is. Therefore, it is strongly recommended
sensitivity. In general, it is highly recommended that results that at least 6 batches be run during prestudy validation to
from multiple runs be used to estimate the response error assess the accuracy, precision, and total error of the method.
relationship. Because of the heteroscedastic nature of the For each validation sample, the repeated measurements
response variance, a weighted, nonlinear, least-squares from all runs should be analyzed together using an appro-
method is generally recommended for fitting concentration priate statistical method.2
response data from LBAs.5,6 Four- and 5-parameter logistic Based on our current understanding and the knowledge
calibration models are often used to fit the LBA standard from the available data discussed in the literature and at
curves. Standard points outside of the range of quantifica- recent meetings (3rd AAPS/FDA Bioanalytical Workshop,
tion (anchor calibrators) are often used to assist in fitting Round Table discussion at the AAPS Annual Meeting and
these nonlinear regression models.7 The details of standard Exposition, AAPS Bioanalytical Method Validation of
curve calibration point selection to be used during method Ligand Binding Assays to Support Pharmacokinetic Assess-
validation are described in DeSilva et al.2 In summary, it is ments of Macromolecules: A Post-Crystal City III Perspec-
recommended that at least 3 runs be used to establish the tive 2006), in general an LBA method can be regarded as
calibration model, with at least 8 non-anchor standard points being acceptable for generating pharmacokinetic and toxi-
run in duplicate.2 The acceptance of the model must be verified cokinetic data if the interbatch precision % coefficient of
by evaluating the relative bias between the back-calculated variation (CV) and the absolute mean bias %RE are both
and nominal concentrations of the calibration standards.8 The ≤20% (25% at the LLOQ) and the method total error (sum
use of the correlation coefficient is not recommended for of the %CV and absolute %RE) is ≤30% (40% at the LLOQ).
confirmation of the regression model.9 The term “total error” describes the combination of system-
Following prestudy validation, the standard curves for sam- atic error (the deviation of the calculated value from the
ple analysis (in-study validation) should be monitored using nominal value) and random error (deviation of the calcu-
the criteria established during prestudy validation. The gen- lated value from the analytical mean).1,2 However, it is
eral recommendation for acceptance is that at least 75% of important to note that in situations where more stringent cri-
the standard points should be within 20% of the nominal teria are needed to support a clinical (ie, bioequivalence)
concentration (% relative error [RE]), except at the LLOQ study or a preclinical study, an effort is made to develop and
and the upper limit of quantification (ULOQ), where the validate LBAs for this purpose.
%RE should be 25%. The editing of the standard curve is Once the prestudy validation is completed and the method’s
permitted with only a priori documented criteria and should accuracy, precision, and total error have been established,
be independent of and completed before the assessment of this information will be used to set the acceptance criteria
the QC acceptance. If edited, the standard curve must be re- for the sample analysis phase. The in-study acceptance cri-
regressed and reassessed for acceptance. The final standard teria must be consistent with the data obtained during the
curve should have at least 6 nonzero standards besides the prestudy assessment. If these criteria are inconsistent, there
anchor points. In situations where the lowest or the highest may be higher assay failures than expected. Run acceptance
standard point has been edited, the assay range should be criteria that have been embraced for both chromatographic
E159
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

assays and LBAs require at least two thirds of all QC results pared in undiluted sample matrix. Therefore, they may be
for a run to be within a specific percentage (eg, 15%, 20%, subjected to MRD before analysis. In cases where an MRD
25%, 30%) of the corresponding nominal reference values, is used, it is acceptable to define the range of quantification
with at least 50% of the results within the specified limit for as the standard concentration values in either neat matrix or
each QC sample. Assays of conventional small-molecule as the range of standard concentration values obtained after
drugs have adopted a 4-6-15 rule.10 In contrast, a 4-6-30 applying the MRD. As an example, a standard curve of 100
rule was proposed for LBAs of macromolecules at the to 1000 ng/mL in neat matrix is equivalent to a standard
March 2000 AAPS workshop.7 This was challenged at the 3rd curve range of 10 to 100 ng/mL with an MRD of 10 (ie,
AAPS/FDA Bioanalytical Workshop in 2006, and the sur- 10% matrix).
vey results indicated that most responders did not use the A plot of the precision profile can be helpful in assessing the
total error criterion during the assessment of validation data prospective limits of quantification (Figure 1). It is neces-
and that the commonly used run acceptance criteria for sary to obtain the data for the precision profile from multiple
LBAs was 20% to 25%. Although there was much discus- runs over time rather than using one set of data, because of
sion at this workshop on the use of point estimates for run the high interassay variance over the intra-assay variance.
acceptance criteria, we support the adoption of relevant sta- The range of quantification established during prestudy val-
tistical approaches (eg, total error, confidence intervals, tol- idation is the range into which samples must be diluted if
erance intervals) that describe the data from the prestudy necessary during in-study validation. Samples that fall
validation in assigning the run acceptance criteria during above the ULOQ must be reassayed at a greater dilution.
in-study validation primarily based on the intended use of Samples already at the MRD and below the LLOQ must
these results.1,8,9,11 be reported as <LLOQ (below the limit of quantification).
During sample analysis, the LLOQ for a run must be revised
RANGE OF QUANTIFICATION upward if editing of the standard curve results in no cali-
brator at or below the validated LLOQ. In this case the
The range of quantification for LBAs is based on the lowest LLOQ is increased to the lowest remaining standard
(LLOQ) and highest (ULOQ) validation samples that meet concentration.
the target precision and accuracy criteria. Because of the
nonlinear nature of the standard curves in LBAs, it is neces-
sary to define both ends of the standard curve range to obtain OTHER VALIDATION PARAMETERS
the range of quantification. Another difference in the chro- The key elements of method validation of the LBAs were
matographic assays is that because anchor calibrators are discussed in detail above. There are other validation param-
frequently used in LBAs, the LLOQ and the ULOQ concen- eters that should be evaluated and acceptance criteria that
trations may not be exactly the concentrations of the lowest should be set up prior to the initiation of study sample anal-
and the highest calibration standards. LBA standard curves ysis (in-study validation). Other publications have offered
have a narrow dynamic range, so it is necessary to reassay detailed discussions about evaluating these parameters,1,2
samples that span beyond the ULOQ to be within the range so only some key points will be reported here.
for quantification using appropriate dilutions. Validation
samples used to define the range of quantification are pre- The stability of the macromolecular therapeutic in the antic-
ipated matrix and in conditions the sample will be subjected
to should be demonstrated. In situations where an altered
matrix is used for standard curve and QC preparation, sta-
bility samples must be prepared in the unaltered matrix. The
experiments must mimic the conditions under which the
study samples will be collected, stored, and processed. Sta-
bility types that need to be assessed include the stability of
the analyte in blood when processed into plasma or serum;
storage stability such as benchtop, short-term, and long-
term storage at –20°C and –70°C; and freeze-thaw stability.
It is important to understand the physicochemical properties
of the macromolecule during the stability evaluation. For
instance, does a protease inhibitor cocktail need to be added
during collection? Is the molecule more hydrophobic than
Figure 1. Example of a typical precision profile. %CV indicates others, which may warrant the use of highly proteinaceous
percent coefficient of variation; LLOQ, lower limit of buffers for storage? Formal stability experiments must be
quantification; ULOQ, upper limit of quantification. conducted with an established method during prestudy
E160
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

validation, but long-term stability experiments may extend study samples. The degree of nonparallelism that is accept-
into the in-study validation phase. It is important to note able for a method depends on its intended application.14 It is
that a freshly prepared standard calibrator curve and QC recommended that the precision (%CV) between samples in
samples or those that are within the acceptable expiration a dilution series be ≤30%. The procedure for reporting a
should be used as the reference for comparison of the stabil- result when a sample does not dilute linearly (ie, dilutes in a
ity samples. The acceptance criteria applicable to the QC nonparallel manner) should be defined a priori.
samples may be used for stability evaluations, or other sta- Robustness and ruggedness are closely related parameters
tistically appropriate methods may be used.12 that should be tested during the prestudy validation phase to
Many immunoassays have narrow standard curve ranges ensure the performance of the assay during the in-study
(<1 order of magnitude), which necessitates the dilution of phase (during sample analysis for study support). The
samples that are above the ULOQ to within the range of the robustness of the assay is determined by its consistency
assay. Dilutional linearity should be evaluated on spiked when changes are implemented that may affect the assay
samples that have been made into the sample matrix (typi- (these should be clearly documented in the method; exam-
cally 100- to 1000-fold greater than the ULOQ) and then ples are incubation temperature, light exposure, matrix).15
should be diluted into the assay matrix. The back-calculated The ruggedness of an assay is determined by its consistency
concentration for each diluted sample should be within 20% when routine changes are implemented, resulting in differ-
of the nominal value, and the cumulative precision should ent operational conditions (eg, analysts, instruments, batch
be ≤20%. It is also necessary to conduct dilutional experi- size, environmental factors).16
ments for the identification of a possible prozone or “hook Validations can fall into 3 broad categories: full, partial, and
effect” (Figure 2 contains an example of signal suppression cross. A full validation, which is done for any new method,
caused by high concentrations of analyte).13 involves method development, prestudy validation, and in-
Dilutional linearity should not be confused with parallel- study validation. Full validations are required for a change
ism. Parallelism is a performance characteristic that is typi- in species (eg, rat to mouse) or a change in matrix within a
cally evaluated during in-study validation using incurred species (eg, rat serum to rat urine). A partial validation is
samples (actual study samples). Parallelism can be assessed conducted where method changes are considered minor; it
using Cmax samples from a given study, pooling the samples can range from a single intra-assay accuracy and precision
to prepare the parallelism validation samples. This approach run to a full validation. Some examples include method
eliminates the generation of multiple values for individual transfer, changes to anticoagulant (eg, EDTA, heparin,
citrate), changes to critical reagents in a method, sample
processing changes (how fast a clot needs to be spun, col-
lection vessels, storage condition), sample volumes, extension
of the concentration range, selectivity issues (concomitant
medication), conversion of a manual to an automated method,
and qualification of an analyst.
Method transfer occurs when a method established in one
laboratory (the transfer laboratory) is transferred to another
laboratory (the receiving laboratory); it requires at least a
partial validation.
Cross-validation is conducted when 2 validated bioanalyti-
cal methods are used within the same study or submission
(eg, enzyme-linked immunosorbent assay [ELISA] to meso-
scale delivery, ELISA to liquid chromatography/mass spec-
trometry). The general practice is to test samples (spiked
Figure 2. A typical sigmoidal dose-response curve for a 2-site and/or pooled incurred samples) with the 2 bioanalytical
Enzyme Immune Assay (EIA) (solid line), including the high- methods. Data should be evaluated using appropriate pre-
dose hook effect (dotted line) demonstrating when the higher defined acceptance criteria or an appropriate statistical
concentrations of analyte result in a lower than expected method.17,18
response. If there were no hook effect, as shown by the graph
with a solid line, higher concentrations of analyte would result in
a >ULOQ response. Without a hook effect, the quantification DOCUMENTATION
range of the curve is between the LLOQ and the ULOQ. OD
indicates optical density; LLOQ, lower limit of quantification; During the discussions on the best practices in validating
ULOQ, upper limit of quantification. LBAs, it has been concluded that changes occur in the selection
E161
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).

of critical reagents, the assay format, and the understanding CONCLUSIONS


of the drug product during the method development phase. This article has addressed the key elements in validating an
These factors, which would be part of the final selection crite- LBA to support pharmacokinetic studies. Most of the param-
ria, require some form of documentation, typically in a lab eters that are essential for the successful validation of an
notebook or binder. As the assay matures during development LBA must be initiated during the method development
and the expected format is defined, this too should be docu- phase, confirmed during the prestudy validation phase, and
mented. With the final format in place, these parameters should closely monitored during the in-study (sample analysis)
be captured in a draft method standard operating procedure phase. The quality of the reagents used in LBAs is pivotal to
(SOP) or an analytical procedure.
the development of a robust method; therefore, applying
The prestudy validation plan guides the validation process criteria for reagents’ lot-to-lot acceptance can be critical.
and relies on an analytical procedure or method SOP, which The availability of a well-characterized therapeutic is
may be still in final draft or may be approved. All validation another important factor to be considered. Most LBAs are
experiments should be documented in a laboratory note- quantified in matrices without an extraction procedure,
book (or as per the individual company policies). As the making the assays more susceptible to matrix interferences.
validation comes to completion, the details of the experi- For this reason, it is essential that both specificity and selec-
ments and the data that contribute to the validation must be tivity be assessed critically.
compiled in a validation report. It is important to summarize
Given the complexity and heterogeneity of these macromo-
all the runs to accurately illustrate the assay performance.
lecular therapeutics as well as the methods routinely used to
The prestudy validation data should, to some extent, predict
quantify these molecules, proper controls must be used and
what to expect during sample analysis (in-study validation).
appropriate acceptance criteria established to support these
Since stability data are frequently obtained over time, the
studies. It is essential that appropriate statistical methodol-
validation report should be updated periodically as new data
ogies be used in the assessment of these parameters so that
are generated.
the in-study validation criteria are consistent with the data
The final validation report can include the following: obtained from the prestudy validation. Furthermore, apply-
ing rigorous statistical methods like the concept of total
1. A description of the assay, including reference error during the prestudy validation can help ensure that the
standard, critical reagents, and the regression used assay used during sample analysis will not experience sig-
for analysis nificant failures.
2. The dates that the validation was conducted and
last updated
3. The location of stored data REFERENCES
4. The names of analysts who conducted the validation
1. Findlay JWA, Smith WC, Lee JW, et al. Validation of immunoassays
5. A description of the parameters tested and the
for bioanalysis: a pharmaceutical industry perspective. J Pharm Biomed
methods used to perform the experiments Anal. 2000;21:1249-1273.
6. A summary of the standard curve back-calculated 2. DeSilva B, Smith W, Weiner R, et al. Recommendations for the
values, including overall statistics of accuracy and bioanalytical method validation of ligand-binding assays to support
precision pharmacokinetic assessments of macromolecules. Pharm Res.
7. A summary of all the validation samples (QC 2003;20:1885-1900.
samples run during validation) used in the conduct 3. Crowther JR. The ELISA Guidebook. Methods in Molecular Biology.
of the validation, including overall statistics on ac- Totowa, NJ: Humana Press; 2001.
curacy and precision (intra-assay and interassay) 4. Chard T. An introduction to radioimmunoassay and related
8. Summary tables for each of the parameters in- techniques. In: Burdon RH, van Knippenberg PH, eds. Laboratory
Techniques in Biochemistry and Molecular Biology. vol. 6. 3rd ed. New
cluded in the validation (eg, dilutional linearity,
York, NY: Elsevier Press; 1987:Part 2, 175-193.
stability, specificity, selectivity)
5. Rodbard D, Frazier GR. Statistical analysis of radioligand assay data.
9. Deviations from the validation plan Methods Enzymol. 1975;37:3-22.
10. A conclusion, including that the validation passed/
6. Dudley RA, Edwards P, Ekins RP, et al. Guidelines for immunoassay
failed and the final acceptance criteria to be ap- data processing. Clin Chem. 1985;31:1264-1271.
plied during sample analysis 7. Miller KJ, Bowsher RR, Celniker A, et al. Workshop on Bioanalytical
Methods Validation for Macromolecules: summary report. Pharm Res.
Also, a final summary page of all that constituted the valida- 2001;18:1373-1383.
tion is advantageous since this summary page can be associ- 8. Karnes HT, March C. Calibration and validation of linearity in
ated with sample analysis conducted for studies to document chromatographic biopharmaceutical analysis. J Pharm Biomed Anal.
the validity of the sample analysis data. 1991;9:911-918.

E162
The AAPS Journal 2007; 9 (2) Article 17 (http://www.aapsj.org).
9. Hartmann C, Smeyers-Verbeke J, Massart DL, et al. Validation of 14. Plikaytis BD, Holder PF, Pais LB, Maslanka SE, Gheesling LL,
bioanalytical chromatographic methods. J Pharm Biomed Anal. Carlone GM. Determination of parallelism and nonparallelism in
1998;17:193-218. bioassay dilution curves. J Clin Microbiol. 1994;32:2441-2447.
10. Shah VP, Midha KK, Findlay JWA, et al. Bioanalytical method 15. ICH. Guideline on validation of analytical procedures: definitions
validation. A revisit with a decade of progress. Pharm Res. and terminology. Paper presented at: International Conference of
2000;17:1551-1557. Harmonization (ICH) of Technical Requirements for the Registration of
11. Hubert H, Chiap P, Crommen J, et al. The SFSTP guide on the Pharmaceuticals for Human Use; November 6, 1995; Geneva. 1995
validation of chromatographic methods for drug analysis: from the 16. Riley CM, Rosanke TW. Development of Validation of Analytical
Washington Conference to the laboratory. Anal Chim Acta. Methods: Progress in Pharmaceutical and Biomedical Analysis. New
1999;391:135-148. York, NY: Elsevier; 1996.
12. Timm U, Wall M, Dell D. A new approach for dealing 17. Bland JM, Altman DG. Measuring agreement in method comparison
with the stability of drugs in biological fluids. J Pharm Sci. studies. Stat Methods Med Res. 1999;8:135-160.
1985;74:972-977. 18. Hartmann C, Smeyers-Verbeke J, Penninckx W, et al. Reappraisal of
13. Rodbard D, Feldman Y, Jaffe ML, Miles LEM. Kinetics of two-site hypothesis testing for method validation: detection of systematic error
immunoradiometric (‘sandwich’) assays, II: studies on the nature of the by comparing the means of two methods or two laboratories. Anal
‘high-dose hook effect’. Immunochemistry. 1978;15:77-82. Chem. 1995;67:4491-4499.

E163

Vous aimerez peut-être aussi