Sunteți pe pagina 1din 11

A Comparison of the Prediction Accuracy of Two

IVIVC Modelling Techniques

CLARE GAYNOR,1 ADRIAN DUNNE,1 JOHN DAVIS2


1
UCD School of Mathematical Sciences, University College Dublin, Belfield, Dublin 4, Ireland
2
Clinical Pharmacology, Pfizer Global Research and Development, Sandwich, England

Received 30 April 2007; revised 20 July 2007; accepted 3 September 2007


Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.21220

ABSTRACT: The goal when developing an in vitro–in vivo correlation (IVIVC) model is
the ability to accurately predict the in vivo plasma concentration profile of a drug
formulation using only its in vitro dissolution data. The prediction accuracy of any model
depends on the reliability of the method used to develop it. Some statistical concerns
regarding methods based on deconvolution have been highlighted and a convolution
based technique has been proposed as an alternative. This comparison shows, by means
of a simulation study, that the modelling approach which uses convolution produces far
more accurate results, accurately predicting the observed plasma concentration–time
profile and, therefore, comfortably meeting the FDA validation criteria. The fact that the
model developed using the deconvolution based technique fails to describe the simulated
data and thus fails the FDA validation test when it ought to pass should be of great
concern to those currently implementing this method. ß 2007 Wiley-Liss, Inc. and the
American Pharmacists Association J Pharm Sci 97:3422–3432, 2008
Keywords: in vitro–in vivo correlations (IVIVC); mathematical model; deconvolution;
convolution

INTRODUCTION importance and the methods used to establish


them should be carefully scrutinised.
Establishing an in vitro–in vivo correlation The procedure routinely employed when
(IVIVC) model has become an integral part of attempting to establish an IVIVC for an extended
the drug development process. The ability to release (ER) drug product involves the study of
accurately predict an in vivo response from a number of formulations of the dosage form of
in vitro observations has far-reaching conse- interest, each with a different release rate. In
quences in several areas including quality control order to develop the IVIVC model, three kinds of
and formulation development. Such a model can data (in vitro, in vivo ER and in vivo reference) are
also act as a surrogate for human bioequivalence frequently collected. A sample of the ER dosage
studies and be used in the setting of in vitro units of interest from each formulation are
dissolution specifications.1 Given that these dissolved in vitro and the fraction which has
IVIVC models are in such widespread use, their dissolved is recorded at a series of time points for
accuracy of prediction and reliability are of key each dosage unit. Further ER dosage units from
each formulation are administered to a number of
human subjects, usually as part of a crossover
study, and their plasma drug concentrations are
Correspondence to: Adrian Dunne
(E-mail: adrian.dunne@ucd.ie) repeatedly measured for a predetermined period.
Journal of Pharmaceutical Sciences, Vol. 97, 3422–3432 (2008) A unit reference dose, which may take the form
ß 2007 Wiley-Liss, Inc. and the American Pharmacists Association of a solution, IV injection or dosage unit which

3422 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008


PREDICTION ACCURACY OF TWO IVIVC MODELLING TECHNIQUES 3423

instantly dissolves, is administered to each of a deconvolution and a convolution based method


the subjects and, once again, their plasma drug and quantify any difference in accuracy of
concentrations are measured over time. The prediction by means of a simulation study.
resulting data are used to develop a model which
can be employed to predict the in vivo plasma drug
concentration profile using in vitro dissolution
data only. DECONVOLUTION BASED APPROACH
There are a number of levels of IVIVC1 but, for
the purposes of this study, only the ‘level A A fundamental element of this method involves
correlation’, which predicts the entire in vivo relating the fraction of a dosage unit which has
plasma drug concentration profile from the dissolved in vitro to the fraction dissolved in vivo
in vitro dissolution data, will be examined. The at corresponding times. The fraction which has
less informative correlations (levels B, C and dissolved at each observation time in vitro is
multiple level C) which relate summary para- recorded as part of the dissolution study. The
meters of the in vitro and in vivo profiles are not fraction dissolved in vivo, however, is not directly
included in this study. observable and must be derived from the available
In order to establish an IVIVC model’s validity, data using the technique of deconvolution. There
its accuracy of prediction must be evaluated; ‘a are many methods of deconvolution4,5 which rely
correlation should predict in vivo performance on the same principles and assume linearity and
accurately and consistently’.1 The United States time invariance of the system being studied. This
Food and Drug Administration (FDA) recommend group of methods is based on the convolution
that formulations with three or more release rates integral, which is defined as
be used to develop the model. Assessment of the
model’s ability to describe these data is referred to Zt
as internal validation. The model must be able to Cp ðtÞ ¼ cd ðt  tÞRðtÞdt (1)
predict the area under the plasma concentration 0
curve (AUC) and the peak plasma concentration
(Cmax) to within acceptable limits as set out by the where Cp(t) is the plasma drug concentration at
FDA.1 Data collected using dosage units from a time t following administration of an ER dosage
fourth or subsequent formulation which was not unit, cd(t) the unit impulse response and R(t) the
used in the model development stage may also be in vivo dissolution rate. Deconvolution involves
used to similarly assess external predictibility. estimating the rate at which a drug dissolves
Many methods of developing IVIVC models in vivo using the observed in vivo ER data (which
have been proposed which, generally, fall into contains information on dissolution, absorption,
two categories: those based on deconvolution and distribution, metabolism and excretion of the
those based on convolution. A number of areas of drug) and observed in vivo reference data (which
concern regarding the deconvolution based meth- does not involve dissolution). This process may be
ods have been highlighted2 and it has become thought of as a kind of ‘subtraction’ technique
clear that, in principle, the use of these techniques used to isolate the information of interest, that is,
is not advisable. A non-linear mixed effects the rate at which the dosage unit dissolves in vivo.
modelling approach, which is not subject to the This rate is used to calculate the fraction dissolved
same criticisms, has been developed,3 this con- in vivo at each of the observed time points. In
volution based procedure should, in theory, order to establish an IVIVC, the relationship
produce more accurate results. In spite of this, between the fraction dissolved in vivo and that
the deconvolution based methods are routinely dissolved in vitro must be examined. These
used. There are perhaps many reasons for this fractions are plotted against one another at
practice, among which may be the fact that the common times and an appropriate line or curve
extent of any empirical difference between the two is fit to the data points. This is the core of the
approaches has never been demonstrated. Since IVIVC model and can be used to predict the
methods involving deconvolution are regularly fraction of the dosage unit dissolved in vivo using
used, it is of vital importance that their theoretical the in vitro data. In order to predict plasma drug
drawbacks be carefully examined and any prac- concentration, a further step is necessary. The
tical significance substantiated. The aim of this predicted in vivo fractions dissolved must be ‘re-
research is to investigate the performance of both convolved’ with estimates of the pharmacokinetic

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008
3424 GAYNOR, DUNNE, AND DAVIS

parameters obtained from the reference data to tion has a negative impact on the model’s
produce plasma concentration profiles. predictions.
Once these fractions are plotted against one
another, an appropriate line or curve is fit to the
data. There is an abundance of instances docu-
mented in the literature where straight-line
Some Statistical Concerns
models are fit to data where a curve would be
There are many theoretical arguments against more appropriate.9–11 This can lead to inaccurate
using deconvolution based methods which are (either over or under) predictions. It is apparent
described in detail elsewhere.2 Descriptions of that care must be taken when choosing the model
those weaknesses which are believed to have the which describes the relationship between the
most significant effect on the model’s ability to in vitro and in vivo fractions dissolved.
predict with accuracy are given below, and are The process of plotting in vivo fractions dissolved
also summarised in Table 1. at a particular time against their in vitro counter-
Very often, the observed data are averaged parts is based on the assumption that their
across subjects or dosage units at each time point relationship is time invariant. This procedure
before analysis. This leads to a significant loss of forces the assumption that the link between
information particularly if the between-subject in vitro and in vivo dissolution does not change
(or dosage unit) variation is high. There may also over time; this is a somewhat ingenuous approach.
be a marked difference between the curve which is In effect, the medium in which the dosage unit
being analysed (made up of data averaged at each dissolves in vitro does not change as time passes,
time point) and the original individual profiles. but it may significantly alter in vivo as the dosage
Deconvolution can, of course, be carried out at the unit’s location in the gastrointestinal tract, and
individual level6–8 but this is not as frequently consequently the pH, changes. Of course this is
seen as analysis based on averages. In order to not necessarily the case, there are many solid
ensure comparable conditions, the methods of systems that are inert and whose dissolution is not
analysis being compared in this study were both influenced by their location in the gastrointestinal
carried out at the individual subject level. tract. It may, however, be unwise to assume that
When the in vivo dissolution rate has been this is always true and that the resulting IVIVC
converted to give the fraction dissolved in vivo at a relationship is time-invariant.
given time, these fractions are plotted against Generally, the method of ordinary least squares
the in vitro fractions dissolved at the same time. is used to fit the chosen model but two of its
Clearly only in vitro and in vivo data collected at main assumptions are violated in this case. The
common times can be used. This leads to the first is that the independent variable (usually
discarding of data where the observation times in vitro fraction dissolved) is measured without
are not coincident. The resulting loss of informa- error, which, in terms of this application, is an

Table 1. A Summary of Statistical Concerns with Deconvolution Based Methods

Statistical Concern Effect


Only data at common times are used Loss of information leads to inefficiency
A straight line can be used to model in vitro–in vivo Produces biased predictions
relationship when it is inappropriate
Assumes in vitro–in vivo relationship is Model is somewhat naı̈ve and unrealistic
time invariant
Assumptions of ordinary least squares violated Error in independent variable leads to biased predictions
Correlation exists between observations made on a single
tablet or subject which leads to reduced efficiency
Averaged data are used Cannot identify inter-subject or inter-dosage
unit differences
Curve of averages is different to the average of the
individual curves
Use of deconvolution Deconvolution is inherently unreliable

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008 DOI 10.1002/jps
PREDICTION ACCURACY OF TWO IVIVC MODELLING TECHNIQUES 3425

unattainable expectation and is known as the function of time and incorporates random effects
Errors in Variables issue.12 Secondly, it is which can account for some of the correlation
required that all observations be uncorrelated between repeated measurements on the same
or independent while it is clear that measure- subject or dosage unit. Observations in vitro and
ments made on the same subject or dosage unit in vivo do not have to be made at corresponding
cannot be uncorrelated. Violating these two times, eliminating the loss of information caused
underlying assumptions of the least squares by discarding some of the data. The use of this
method results in a model which can produce method removes the need for a deconvolution step
inaccurate predictions. and predicts plasma concentrations rather than
A fundamental shortcoming of this entire group their deconvolved equivalents.
of methods is that the process of deconvolution
is itself unstable13 and may be thought of as a
‘roughening’ of the data. It, therefore, by nature, METHOD—SIMULATION STUDY
introduces errors and reduces the accuracy with
which the model can predict the plasma concen- A simulation study to compare these two appro-
tration profiles. aches to establishing an IVIVC was undertaken.
There are a number of ways of evaluating Four batches of a drug, each with a different
accuracy of prediction, two of the most commonly release rate, were simulated. The first three
used measures are bias and efficiency. A biased batches will be referred to as the fast, medium
estimate will tend to consistently over or under and slow formulations while the fourth was
predict the quantity of interest, for example, AUC included for the purposes of external validation.
or Cmax. A low bias is therefore usually preferable. The data were simulated such that a marked
An inefficient estimate will produce predictions relationship between in vitro and in vivo dissolu-
that are widely scattered around the true value. tion would exist. The three kinds of data: in vitro,
The combination of the issues mentioned above in vivo ER and reference were produced for 12 ER
is believed to have a substantial impact on the dosage units and 12 subjects with 22 observations
accuracy with which the deconvolution method made from time of administration (0 h) up to 34 h
can predict the plasma concentration curve, and it in order to simulate a study with a crossover
is expected that this method will produce biased design.
and inefficient predictions. The reference data were simulated according to
a one compartment pharmacokinetic model with
first order absorption as follows:
CONVOLUTION BASED APPROACH  
l3k
cdk ðtÞ ¼ ðel2k t  el3k t Þ (2)
Vk ðl3k  l2k Þ
An alternative modelling approach which incor-
porates the convolution integral (Eq. 1) has been where Vk, l2k and l3k represent the kth subject’s
developed. This method, like the deconvolution volume of distribution and rate constants of drug
based method described above, assumes the elimination and absorption, respectively. The
linearity and time invariance of the system being plasma drug concentration for the kth subject at
studied.4 The version of this convolution based time t following administration of a unit reference
method proposed by O’Hara et al.3 first fits a dose is given by
standard compartmental model to the unit
Y3k ðtÞ ¼ cdk ðtÞ þ "3k ðtÞ (3)
impulse response data in order to estimate each
subject’s pharmacokinetic parameters separately. where e3k is approximately normally distributed
The in vivo plasma concentrations resulting from ðNð0; s 22 ÞÞ with s 22 accounting for in vivo measure-
administration of the ER dosage unit to each ment error.
subject, and the in vitro fractions dissolved for The observed in vitro fraction of drug dissolved
each dosage unit are modelled directly (i.e. from the ith dosage unit at time t is described as
without averaging) in a one-step process. The follows:
resulting IVIVC model does not suffer from the
Yi1 ðtÞ ¼ f1 Fi1 ðtÞ þ "i1 ðtÞ; "i1 ðtÞ  Nð0; s 1 2 Þ (4)
same theoretical flaws as the deconvolution based
method previously described. It allows the rela- where Fi1(t) is the true fraction dissolved at time t,
tionship between the fraction dissolved in vitro s 21 represents the in vitro measurement error and
and that dissolved in vivo to be described as a the parameter f1 accounts for any difference

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008
3426 GAYNOR, DUNNE, AND DAVIS

Table 2. Values of Parameters Used to Simulate Dataset

Description of Parameter Value


Tablet to tablet standard deviation (v1) 0.034
Number of tablets in vitro 12
Number of batches 4
Standard deviation of in vitro error term (s1) 0.0005
Number of time points in vitro 22
Dissolution rate constant (l1) 0.799 (fast formulation)
0.299 (medium formulation)
0.100 (slow formulation)
0.200 (external formulation)
Scale parameter in vitro (f1) 0.996
Weibull parameter (a) 1
Subject to subject standard deviation (v2) 0.61
Number of subjects 12
Standard deviation of in vivo error term (s2) 0.62
Number of time points in vivo 22
Scale parameter in vivo (f2) 0.996
Elimination rate constant for typical subject (l2) 0.2
Absorption rate constant for typical subject (l3) 0.9
Standard deviation of l2k 0.04
Standard deviation of l3k 0.18
Theta1 (u1) 1.29
Theta2 (u2) 0.900
ER dose 100
IR dose 50
Volume of distribution of typical subject 1
Standard deviation of V 0.2

between actual dose and label claim. Variation in The in vivo plasma concentration measured
Fi1(t) between dosage units was modelled as: from the kth subject at time t following admin-
istration of the ith dosage unit is described as
logitðFi1 ðtÞÞ ¼ logitðF1 ðtÞÞ þ ui ; ui  Nð0; v1 2 Þ (5) Z
0
Yi2k ðtÞ ¼ Dose f2 cdk ðt  tÞFi2k ðtÞdt þ ei2k ðtÞ;
The term ui is a random effect which allows ei2k ðtÞ  Nð0; s 2 2 Þ ð7Þ
for differences between dosage units while also
accounting for the correlation between repeated where Fi2k0 (t) is the in vivo dissolution rate at time
observations made on the same dosage unit. The t, f2 allows for a difference in bioavailability
inter dosage unit variation is given by v21 . F1(t) is between the reference dose and the ER dose and
the fraction of the typical dosage unit (where ui is s 22 represents in vivo measurement error as
zero) dissolved at time t and is modelled according before.
to the Weibull function as follows: The error terms ei1, ei2k and e3k are all modelled
according to truncated normal distributions to
F1 ðtÞ ¼ 1  expðl1 ta Þ (6)
ensure that the values of Yi1, Yi2k and Y3k remain
positive.
where the rate of dissolution of the drug is
The relationship between in vitro and in vivo
determined by l1 and a. The use of the logit
dissolution is defined as follows:
transformation logit(x) ¼ log(x/(1x)) in equation
five ensures that the simulated fraction, even with logitðFi2k ðtÞÞ ¼ logitðF1 ðtÞÞ þ u1 þ u2 t þ ui þ sik ;
the addition of a random effect (ui), remains
within the interval [0,1]. with ui  Nð0; v1 2 Þ; and sik  Nð0; v2 2 Þ ð8Þ

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008 DOI 10.1002/jps
PREDICTION ACCURACY OF TWO IVIVC MODELLING TECHNIQUES 3427

where sik is a subject specific random effect which RESULTS


allows correlation between observations made on
a particular subject and accounts for differences While all analysis was carried out at the
between subjects so v22 gives the subject-to-subject individual subject (or dosage unit) level, for ease
variation and v21 represents inter dosage unit of presentation the in vitro dissolution data and
variability as before. The use of the logit link as in vivo ER plasma concentrations were averaged
seen above ensures that both Fi2k(t) and Fi1(t) are at each time point for each formulation and are
constrained to be between zero and one. The u2t shown in Figure 1a and b, respectively.
term allows for the possibility that the relation- The in vivo reference data were first analysed.
ship between in vitro and in vivo dissolution Each subject’s elimination and absorption rate
may vary with time as might be expected in some constants and volume of distribution were esti-
circumstances. mated separately. These parameter estimates
In order to ensure that the results of this were used to predict the immediate release
simulation could be meaningfully interpreted, the profiles which are shown in Figure 2 and were
values of all the above parameters were based on used as part of both methods of analysis.
those estimated using real datasets and are listed
in Table 2.
A dataset was simulated according to the
model described above and analysed twice: first
using a deconvolution based method and then an
alternative, convolution based technique. The
fast, medium and slow formulations were used
to develop the model in both cases, while the
fourth batch was excluded to be used later for
the purposes of external validation. First, each
subject’s reference data was analysed separately
using the one compartment pharmacokinetic
model provided in the ADVAN2 subroutine of
the NONMEM software developed by Beal and
Sheiner.14 The resulting PK parameters were
then used in both the deconvolution and con-
volution methods as required. The convolution
method used was a modified version of that
reported by O’Hara et al.,3 which fit the model
described by Eqs. (4–8) above by means of a
custom written PRED subroutine for NONMEM.
The method of deconvolution used was Con-
strained Deconvolution (CoDe) as proposed by
Hovorka et al.5,15 The required software was
kindly provided by Dr. Hovorka. The two IVIVC
models established were used to predict in vivo
plasma concentration profiles for each subject
and all four formulations as both methods of
analysis were carried out at the individual
subject level. The Cmax and AUC were computed
for each of the four batches using the in vivo ER
data and the profiles predicted by each method of
analysis. The AUC was calculated using the
trapezoidal rule, while the Cmax for each subject
was located by inspection and results were Figure 1. (a) Simulated in vitro dissolution data
averaged over all 12 subjects. The average averaged across dosage units for each of the four for-
values of AUC and Cmax obtained using each mulations. (b) Simulated in vivo plasma concentration
method were compared to the true values to data averaged across subjects for each of the four for-
calculate percentage prediction error (%PE). mulations.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008
3428 GAYNOR, DUNNE, AND DAVIS

Figure 2. In vivo reference data and fitted curves for each subject.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008 DOI 10.1002/jps
PREDICTION ACCURACY OF TWO IVIVC MODELLING TECHNIQUES 3429

The deconvolution based method was first to


be implemented. The fraction dissolved in vivo
for each subject at each observation time was
calculated and averaged across subjects to obtain
an in vivo dissolution profile for each formulation
as shown in Figure 3. These in vivo fractions were
plotted against the corresponding in vitro value
and the method of Ordinary Least Squares was
used to fit a polynomial curve of degree four to the
data as illustrated in Figure 4. This fitted curve
was used to predict the fraction dissolved in vivo
which corresponded with the fraction dissolved
in vitro at each time point. These in vivo fractions
were then reconvolved with each subject’s PK
parameters to produce predicted plasma concen-
trations for each subject and each formulation.
Figure 4. Average in vivo fraction dissolved against
The convolution-based method was used to average in vitro fraction dissolved at common times.
develop the IVIVC model in a one-step process, The fast formulation is represented by the þ sign, the
analysing the in vitro and in vivo data simulta- medium by  and the slow formulation by the symbol.
neously. Predicted plasma concentration curves The fitted polynomial curve is given by the solid line.
were produced for each subject directly.
To illustrate the differences between the two
modelling techniques, Figure 5 shows simulated
and predicted plasma concentration curves for each CONCLUSIONS AND DISCUSSION
of the four batches averaged over all 12 subjects.
The solid line represents predictions made when The summary in Tables 3a and 3b shows that
using the convolution method, while the dashed analysis using the deconvolution based method
line shows those of the deconvolution method. yields inaccurate predictions, while those of the
The corresponding percentage prediction errors convolution based technique are very precise.
for each formulation resulting from analysis using The United States FDA require, that for an
each method are shown in Tables 3a and 3b. IVIVC model to be validated, average absolute
percentage prediction error of both AUC and Cmax
must be 15% or less for each batch (and less
than 10% on average over the three batches) to
establish internal predictibability and 10% or less
for external predictability.1 For this sample
dataset, where an IVIVC relationship does exist,
the convolution method produces an IVIVC
model that comfortably meets the FDA criteria
while the deconvolution based method fails to
establish the in vitro–in vivo relationship.
There is a noticeable difference in the accuracy
with which the deconvolution-based method pre-
dicts the four different formulations. The reasons
for this can easily be seen by examining the core of
the IVIVC model which is illustrated in Figure 4.
The majority of points representing the fastest
formulation are in the upper right hand corner
of the plot and are quite close to the fitted curve.
Figure 3. Average in vivo fractions dissolved esti- The early time points, however, lie below the fitted
mated using the deconvolution method for each of the curve. This leads to overprediction of plasma
three formulations included in developing the model. concentrations at earlier times and quite a good fit
The fast formulation is represented by the þ sign, the at later times as can be seen in Figure 5a. The
medium by  and the slow formulation by the * symbol. batch with a medium rate of dissolution has points

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008
3430 GAYNOR, DUNNE, AND DAVIS

Figure 5. Observed and predicted plasma concentration profiles averaged across


subjects for each formulation. Panels (a–d) represent the fast, medium, slow and
external formulations, respectively. In each panel, the symbols denote the simulated
data, the solid line shows the convolution method’s predictions and the dashed line
represents the deconvolution method’s results.

which lie quite close to the fitted curve in Figure 4, can see the resulting underprediction of in vivo
resulting in good predictions in Figure 5b. Most of plasma concentration values in Figure 5c at early
the points in Figure 4 which belong to the slowest time points and, similarly, concentrations at later
formulation are above the fitted curve and one times are overpredicted. This lack of accuracy in

Table 3a. Percentage Prediction Errors (Used for Internal Validation)

Batch Method of Analysis %PE AUC %PE Cmax


Fast Deconvolution 0.4692 6.1409
Convolution 0.2427 0.0320
Medium Deconvolution 0.1988 2.2250
Convolution 0.0380 0.3745
Slow Deconvolution 0.5670 28.0391
Convolution 0.0791 0.0324
Average j%PEj Deconvolution 0.4116 12.135
(first three batches) Convolution 0.1199 0.1463

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008 DOI 10.1002/jps
PREDICTION ACCURACY OF TWO IVIVC MODELLING TECHNIQUES 3431

Table 3b. Percentage Prediction Errors (Used for based method may also have an impact on the
External Validation) results. This is a question which has previously
been studied, albeit not in this context. Zhang
Method of et al.17 point out that ‘conditioning on some of the
Batch Analysis %PE AUC %PE Cmax
data or on prior fits to them may yield inaccurate
External Deconvolution 0.2730 11.0301 standard errors as there is not a proper account-
Convolution 0.0025 0.0232 ing for the uncertainty in conditioning quantities’.
In general, it is agreed that simultaneous analysis
of data is, statistically, preferable to first estimat-
prediction leads to a very high %PE value for ing parameters, which are then fixed and used in
the Cmax of this formulation, resulting in the the second stage of the analysis.17,18
failure to satisfy the internal validation criteria. The findings of this study confirm the expecta-
The formulation used for external validation tion that the convolution based method would
has a release rate which lies between the medium outperform its counterpart. The concerns about
and slow formulations as seen in Figure 1a, the deconvolution method are supported by the
predictions made using deconvolution for this results and its lack of accuracy in prediction is a
formulation are not sufficiently accurate to meet result of the combined influence of its theoretical
the FDA external validation criteria. flaws. While these may have seemed like purely
The fact that the convolution method is, in statistical considerations, they have a very strik-
effect, fitting the correct model leads to results ing practical effect in terms of quality of predic-
that are, perhaps, over optimistic. The degree to tions and, therefore, ability to meet the FDA
which the predictions would deteriorate when validation criteria. These results substantiate the
using real data would depend on the extent to statistical theory, illustrating and quantifying the
which the model used is mis-specified. superiority of the convolution based approach and
The deconvolution method not only fails to providing support for its use in preference to the
produce a model which meets the FDA validation deconvolution-based group of methods.
criteria but, as can be seen in Figure 5 (particu-
larly in panel c), the predicted plasma concentra-
tion curve is entirely the wrong shape. This is
potentially very misleading if the IVIVC model is REFERENCES
to be used during product development.
The deconvolution method and software used in 1. Food and Drug Administration. 1997. Guidance for
this study (CoDe15) has been chosen from a large industry: Extended release oral dosage forms:
number of such techniques. The CoDe method was Development, evaluation, and application of in
selected on the basis of its good performance in vitro/in vivo correlations.
testing such as that conducted by Madden et al.5 2. Dunne A, Gaynor C, Davis J. 2005. Deconvolution
based approach for level A in vivo-in vitro correla-
There are also many variations possible at the
tion modelling: Statistical considerations. Clin Res
implementation stage of both deconvolution and
Regul Aff 22:1–14.
the convolution-based methods so a number of 3. O’Hara T, Hayes S, Davis J, Devane J, Smart T,
further decisions were necessary. The choice of Dunne A. 2001. In vivo–in vitro correlation (IVIVC)
whether or not to average the data before analysis modeling incorporating a convolution step. J Phar-
is believed to be a crucial one. Current advice on macokinet Pharmacodyn 28:277–298.
best practise16 suggests that deconvolution should 4. Dunne A. 2007. Approaches to developing in vitro-
take place on an individual subject (or dosage in vivo correlation models. In: Chilukuri DM,
unit) level. Consequently, all of the analysis Sunkara G, Young D, editors. Pharmaceutical pro-
conducted as part of this study was carried out duct development in vitro-in vivo correlation.
entirely at the individual subject level for both the pp 47–70.
5. Madden FN, Godfrey KR, Chappell MJ, Hovorka R,
deconvolution and convolution-based methods.
Bates RA. 1996. A comparison of six deconvolution
Averaging of the raw data is believed to be so
techniques. J Pharmacokinet Biopharm 24:283–
fundamental an issue that it warrants further 299.
exploration and, as such, this point will form the 6. Abuzarur-Aloul R, Gjellan K, Sjölund M, Löfqvist
basis of a separate study. The use of a one-stage M, Graffner C. 1997. Critical dissolution tests
rather than a two-stage approach to fitting the of oral systems based on statistically designed
in vitro and in vivo ER data in the convolution experiments. I. Screening of critical fluids and

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008
3432 GAYNOR, DUNNE, AND DAVIS

in vitro/in vivo modelling of extended release coated 12. Snedecor GW, Cochran WG. 1980. Statistical meth-
spheres. Drug Dev Ind Pharm 23:749–760. ods, 7th edition. Ames, Iowa: Iowa State University
7. Abuzarur-Aloul R, Gjellan K, Sjolund M, Graffner Press.
C. 1998. Critical dissolution tests of oral systems 13. Wing GM. 1991. A primer on integral equations of
based on statistically designed experiments. II. In the first kind—The problem of deconvolution and
vitro optimization of screened variables on ER- unfolding. Philadelphia: Society for Industrial and
coated spheres for the establishment of an in Applied Mathematics.
vitro/in vivo correlation. Drug Dev Ind Pharm 14. Beal SL, Sheiner LB. 1992. NONMEM user’s
24:203–212. guides, NONMEM Project Group. San Francisco:
8. Abuzarur-Aloul R, Gjellan K, Sjolund M, Graffner University of California.
C. 1998. Critical dissolution tests of oral systems 15. Hovorka R, Chappell MJ, Godfrey KR, Madden FN,
based on statistically designed experiments. III. In Rouse MK, Soons PA. 1998. CODE: A deconvolution
vitro/in vivo correlation for multiple-unit capsules program implementing a regularization method of
of paracetamol based on PLS modeling. Drug Dev deconvolution constrained to non-negative values.
Ind Pharm 24:371–383. Description and pilot evaluation. Biopharm Drug
9. Liu Y, Schwartz JB, Schnaare RL, Sugita ET. 2003. Dispos 19:39–53.
A multi-mechanistic drug release approach in a 16. Farrell C, Hayes S. 2007. IVIVC for oral drug
bead dosage form and in vitro/in vivo correlations. delivery: Immediate and extended release dosage
Pharm Dev Technol 8:409–417. forms. In: Chilukuri DM, Sunkara G, Young D,
10. Cardot JM, Beyssac E. 1993. In vitro/in vivo corre- editors. Pharmaceutical product development in
lations: Scientific implications and standardisation. vitro-in vivo correlation. pp 47–70.
Eur J Drug Metab Pharmacokinet 18:113–120. 17. Zhang L, Beal S, Sheiner L. 2003. Simultaneous vs.
11. Kortejarvi H, Mikkola J, Bacjman M, Antila S, sequential analysis for population PK/PD data I:
Marvola M. 2002. Development of level A, B Best-case performance. Journal of Pharmacoki-
and C in vitro-in vivo correlations for modified- netics and Pharmacodynamics 30:387–403.
release levosimendan capsules. Int J Pharm 241: 18. Verbeke G, Molenberghs G. 2000. Linear mixed
87–95. models for longitudinal data. New York: Springer.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 8, AUGUST 2008 DOI 10.1002/jps

S-ar putea să vă placă și