throbber

`
`Logarithmic Transformation in Bioequlvalence: Application with
`Two Formulations of Perphenazine
`
`K. K. MIDHA*", E. D. ORMSBY*, J. W. HUBBARD", G. MCKAY*, E. M. HAWES*, L. GAVALAS§,
`AND I. J. MGGILVERAY‘
`
`Received July 20, 1990, from the “Colleges of Pharmacy and Medicine, University of Saskatchewan, Saskatoon
`Saskatchewan, Canada S7N 0W0, *Health Protection Branch, Tunney‘s Pasture, Ottawa, Ontario, Canada K1A 0L2, and
`Accepted for publication August 5,1992.
`§Zenith Laboratories Inc. Northvale, NJ 07647.
`
`
`Abstract DThe rationale for using the logarithmic transformation on
`concentration-dependent pharmacokinefic parameters a priori is pre-
`sented. This rationale is based on theoretical pharmacokinetic and
`statistical grounds, but is also applicable to the practice of physicians in
`dealing with variations of drug treatment within and between patients.
`The implications of the transformation on data analysis, specificwa
`analysis of variance, and estimation and inference from the analysis as
`it pertains to bioequivalence decisions are explored. implementation of
`the transformation is shown, with an example of two perphenazine
`formulations in a single-dose crossover study. It is concluded that the
`transformation has to be accepted on theoretical grounds because
`sample sizes are too small in bioequivalence studies and too susceptible
`to extreme values to state with any certainty the actual distribution of
`pharmacokinetic parameters or their differences within a subject.
`
`Over the past two decades there has been much discus-
`sion1-4 on the use of the logarithmic (log) transformation of
`concentration-dependent pharmacokinetic parameters before
`examination by analysis of variance (ANOVA) in bioequiv-
`alence studies. In several important papers5-8 in which
`decision rules for acceptance of bioequivalence are discussed,
`application ofthis transformation is alluded to, but its routine
`use has not been recommended. Recommendations to the
`_ Canadian Health Protection Branch9 have specified that
`bioequivalence decisions must be made on the basis of log
`transformed data. By contrast, the United'States Food and
`Drug Administration (FDA)10 requires that ANOVA be car-
`ried out on the raw scale unless there is strong evidence that
`supports the transformation. However, the Generic Drugs
`Advisory Committee11 of the FDA voted recently in favor of
`using the log transformation a priori. A guideline12 produced
`by the European Committee for Proprietary Medical Products
`has also recommended the transformation.
`In light of the recent ruling of the FDA advisory committee
`and the obvious trend towards the log transformation, the
`authors felt it was necessary to present the rationale and
`statistics concerning the analysis of a bioequivalence study
`based on the log scale. At a major bioavailability conference13
`in 1989, the point was raised that there was a need to educate
`both regulatory reviewers and the pharmaceutical industry
`on application of and inference from the log transformation.
`An example of the analysis is provided.
`
`Theoretical Section
`
`The purpose of a bioequivalence study is to show that the
`plots of concentration of drug in plasma versus time (plasma
`concentration—time profiles) of two drug products containing
`the same dosage of the same drug can be superimposed
`Because profiles from drug products even from the same lot,
`let alone two different formulations, will never be identical
`within a subject, the degree to which the two curves super-
`
`138 / Journal of Pharmaceutical Sciences
`Vol. 82, No. 2. February 1993
`
`impose must take account of this ”intrasubject” variation
`while still ensuring that the test formulation does not deliver,
`on the average, a different profile. Although this variation
`plays a significant role in the precision of the estimated
`relative bioavailability, the main interest lies in comparing a
`measure of central location of the pharmacokinetic parame-
`ters derived from the administration of test and reference .
`formulations.
`For immediate-release formulations, superimposition of
`the profiles following single dosings can usually be assessed
`by comparing extent and shape. These two characteristics of
`the profiles are estimated with the values of area under the
`curve (AUC) and the observed maximum concentration
`(CI-n“)! respectively. A crossover study is executed, and the
`probability that the relative average difference of the two 4
`products for each parameter falls within a prespecified range
`is calculated. This probability must not be greater than the
`consumer risk of a% (conventionally 5%) for both tails of the
`distribution. This prespecified range is called the bioequiva—
`lence interval (BI) and is the only link between clinical and
`blood concentration significance. It is generally agreed that,
`for most drugs, up to a 20% difference in AUC or C1mm between
`two formulations would have no clinical significance. The
`implication of accepting bioequivalence is that the two for-
`mulations would have the same clinical effect.
`Consideration of the Log Transformation—It is com-
`monly believed that the distributions of many biological
`parameters have much longer right tails than would be
`expected had the parameters come from a normal distribu-
`tion. Kenny and Keeping“ state that if an outcome random
`variable is affected by many random causes, each of which
`produces a small proportional effect, the resulting distribu-
`tion can be represented by the log normal distribution.
`Applying this random process to pharmacokinetics, one can
`expect that a concentration measured at any time is a
`function of many random processes (absorption, metabolism,
`elimination) that act proportionally to the amount of drug
`present. Therefore, it can be envisaged that AUC or CmaXma)’
`take on a log normal distribution due to environmental and
`genetic influences on the many random processes from which
`the parameters arise. The values from this distribution may
`differ by a factor of < 10 to a factor as large as 100. Many drugs
`exhibit polymorphic metabolism, which can account for the
`wider ranges seen for some drugs. The distribution of the!)8
`parameters is difficult to ascertain because of the small
`number of subjects used in a bioequivalence study. However.
`the distribution of the concentration-dependent parameters
`for many drugs does show an extended right tail.15-17
`Although inter-subject or between-subject variation, 85 .
`described above, may have large clinical implications, it.is not
`of major importance in the crossover study because each.
`subject receives the drug from each formulation. The mot!t‘
`
`0022-3549/93/0200—0138302.5
`
`© 1993, American Pharmaceutical Associ
`
`
`ENDO - Ex. 2048
`
`Amneal v. Endo
`
`lPR2014-00360
`
`ENDO - Ex. 2048
`Amneal v. Endo
`IPR2014-00360
`
`

`

`x is
`
`'1
`
`
`
`t variation is the intrasubject or within-subject
`;
`oe» which is the variation exhibited by a single person
`'ven the same dose of drug (e.g., solution) over
`
`administrations (i.e., independent of formulation).
`
`;
`‘tude of the intrasubject variance depends greatly
`
`harmacokinetics of the drug. For instance, drugs with
`kinetics (e.g., little metabolism, well absorbed) will
`
`y have intrasubject coefficients of variation (CVs)
`
`‘
`- .
`to be <20%, whereas drugs with complicated
`
`cs (e.g., high first-pass effect, variable absorption) can
`estimated intrasubject CVs as large as 40% or more.
`
`in the above paragraph that we have switched from
`bject variance to intrasubject CV. This is the common
`
`‘7 professionals make when they wish to describe indi-
`in the population. The CV is used because it describes
`
`ation expected for individual patients from the pop-
`r. ., whether they exhibit large or small values. Implicit
`
`" use of the CV is the concept of relative variation. That
`jects with larger values will have larger variation in
`
`licated values than subjects with smaller values, but,
`' : tion relative to their individual means is the same.
`
`trast, ANOVA assumes constant variation, not relative
`
`on, and thus needs implimentation of the log transfor-
`n.
`
`ake18 suggested AUC and Cmax would have this
`
`e variation over subjects given the same drug. He
`ed, based on the common expression that AUC was
`
`to amount of drug absorbed over clearance, that vari-
`within a subject would be larger for subjects with larger
`
`(e.g., slower elimination) than for those with faster
`tion. Although this expression is satisfied for drugs
`
`,
`ery general conditions, we feel the same subject
`phcative effect holds true for most drugs. Further,
`
`'
`most analytical methods also show an increase in
`oe (but somewhat constant relative variance) in re-
`
`- as the response increases, we can expect
`larger
`
`on in larger AUCs, especially with higher Cmax values.
`
`main use of the log transformation is, therefore; to
`' e or make equal the intrasubject variation and not to
`
`ze the intersubject parameters, although the latter is
`also accomplished. Because ANOVA assumes that
`
`the model act in an additive way, the transformation
`
`the data from a proportional or relative scale into an
`ve one. The basic premise of the transformation is that
`
`.
`') = log (A) — log (B).
`in el and Assumptions—The general linear model for a
`
`riod,
`two-treatment crossover design,
`ignoring the
`, over efi‘ect,19 is:
`
`
`1“ Wijm1=Xim=u+n+Bjm+Wk+¢1+eijkl
`
`(1)
`
`'”
`
`and is used to describe the data after taking the natural logs
`of the individual parameters. The (SJ-(i) is the random effect of
`subject nested within sequence and is assumed to be normal
`(0, 023). The residual term is also assumed to be normal (0, 0%,)
`and independent of the random subject effects. The other
`terms are the fixed effects of sequence (7,), of period (ark), and
`of formulation (4),). As defined, the variance of a single
`observation is equal to 02 = 0% + 0%,, and the two observa—
`tions from the same subject have a covariance 0% or a
`correlation of 02,3/(0f3 + 0%,).
`The residual is the estimate of the intrasubject variation,
`0%,. As the name implies, it is the variation remaining that
`the model cannot explain. For this design, it is sometimes
`called the subject-by-formulation interaction term because it
`is mainly made up of variation due to a day-to-day variation
`in drug pharmacokinetics in a subject, the “true” intrasubject
`variation, and formulation variation. However, clean esti—
`mates of intrasubject and formulation variances can never be
`obtained because a tablet can only be given once and the
`pharmacokinetics of the drug in the subjects are unique for .75
`each dosing. The best that can be done is to estimate the
`subject-by-formulation interaction for each formulation when
`each formulation is repeated.20 Therefore, one assumption is
`that both the inter- and intra-variances from each formula-
`tion are equal. Also, contributing to the residual is analytical
`error and sample placement error. The analytical error is
`allowed to be as high as 15%21 but is more commonly ~5%.
`This error is more evident in the variation of a single
`concentration such as Cmax than with AUC, because AUC is
`an integral over many points with the same relative error.
`Recently, Gafl‘ney22 has studied the contributions of these
`variance components in bioequivalence studies. The last
`component due to placement of sampling times can have a
`great effect when input is fast and the frequency of sampling
`does not reflect the rate or the variation in time to reach C'max
`(than).
`The degrees of freedom, expected mean squares, and the
`appropriate error term to test for each source are given in
`Table I for the two-period, two-treatment, crossover model.
`Estimation and Inference from the Log Transforma-
`tion—We must consider the purpose of performing a bioequiv-
`alence study to decide what statistical parameters have to be
`estimated, but also the study design to account for design
`effects. What we need is an estimator of average relative
`bioavailability and its standard error such that a probability
`
`i—Expected Mean Squares for a Two-Period, Two-Treatment, Crossover Design
`
`roe
`
`d1a
`
`Expected Mean Squares
`
`02 + 202 +
`w
`B
`
`2nmnm(rm — Tm)2
`"Tn + "RT
`
`nTR + ”RT — 2
`
`05,, + 20‘?3
`
`2nrnnm'(77'm - Whrlz
`a5, +——"TR + nm
`
`02
`W
`
`+ .—.—_——__
`2nmnm'(¢m — <I>RT)2
`"TR + "RT
`
`”TR + nRT — 2
`
`03"
`
`F-Ratio
`Denominator
`
`Subject (SE0)
`
`Residual
`
`Residual
`
`Residual
`
`_b
`
`ass of freedom. " Not determined.
`
`
`
`Journal of Pharmaceutical Sciences/ 139
`Vol. 82, No. 2, February 1993
`
`

`

`statement or inference can be made. If the parent distribution
`of a parameter onr each formulation is log normal, then the
`resulting distributions after transformation of Y to X are
`normal (ubaf). There are two competing estimates of central
`tendency for Y, the median and the mean. The medians of the
`distributions can be estimated with eq 2:
`
`MEDIAN; = exp?!
`
`(2)
`
`For 1 = T,R (for the test and reference formulations, respec-
`tively), the least square means are X: (XTR, + XRT,)/2. A
`least squares mean is simply the mean1ofthe means from each
`of the sequence TR and RT. If the number of subjects in
`sequence TR (nTR) equals the number in sequence RT (nRT),
`then the least squares mean equals the overall mean for each
`formulation. Equation 2 also defines the geometric mean.
`The mean is obtained from eq 3:
`
`MEAN, = expi’l’sm
`
`(3)
`
`In eq 3, $123 is the estimated variance of the Xs. Note that the
`median will always be smaller than the mean for a skewed
`distribution. Land23 has given tables to calculate the exact
`confidence interval (CI) for the mean and they differ from the
`usual CI for the median because the exact CI must be wider
`to account for the estimation of 0123.
`However, in bioequivalence, the paramount estimation is
`for the relative medians or means. Estimates for the ratio of
`medians (RELBIOMED) are obtained with eq 4:
`
`RELBIOMED = eprrT 5‘11
`
`(4)
`
`The standard error of the difference (STEdifl) in the exponent
`is:
`
`1
`1
`STEdjfi‘: (_+—)Sw
`nTR
`nRT
`
`(5)
`
`In eq 4, S3,, is the residual from the crossover ANOVA.
`RELBIOMED is a consistent maximum likelihood estimator
`but also has a slight bias. Recently, Liu and Wen24 examined
`the properties of RELBIOMED and recommended a correction
`factor. Figure 1 gives a plot of the correction factors for
`intrasubject CVs from 10 to 50% and crossover sequence sizes
`from 6 to 50. Although the estimator consistently underesti-
`mates the true ratio of medians, this bias is not likely to be of
`any consequence (< 1%) given the required sample sizes to
`
`1.000
`
`0.975
`
`FACTOR
`CORRECTION
`
`0.050
`
`o
`
`'10
`
`20
`
`so
`
`40
`
`so
`
`SAMPLE SIZE
`
`Figure 1—Bias correction factors for balanced crossover studies. Key to
`CVW: (~--—) 10%; (- - - -) 20%; (—) 30%; (— — —) 40%; (— —) 50%.
`
`140 / Journal of Pharmaceutical Sciences
`Vol. 82, No. 2, February 1993
`
`achieve the power to make a bioequivalence decision for largei
`intrasubject CVs.
`;?
`If one is unwilling to assume the distributional structure:
`noted above, a nonparametric method8 can be used. However, .
`this method is not without distributional assumptions. It
`assumes that the differences calculated for each subject come
`from a distribution that is symmetric and that the differences
`are independent.25 Thus,
`the log transformation is also ,
`recommended to stabilize the variation.
`Two other important parameters that are used in summa.
`rizing results of a bioequivalence study are measures of
`variation. As mentioned earlier, two main sources of varia.
`tion can be estimated. From the “expected mean squares of
`subject (sequence)” term from Table I, one can estimate the
`intersubject variance component 0%. This is a measure of the
`variance of the distribution of subject means if subjects were {
`sampled an infinite number of times. The estimate 8% is.
`obtained by using the expected mean squares (EMS) for
`subject (sequence) and residual terms and solving for 0323. It is ‘
`usually more convenient to express this variation as a CV.
`The CVB, or the intersubject CV, is estimated from the log;
`scale from eq 6:
`
`-
`
`CVB = V expSi —1 x 100%
`
`(6):
`
`A very good approximation (ACVB) for CVBs, up to 40%, is
`obtained by simply taking the square root of Sf; and multi-
`plying by 100%.
`The estimate of the intrasubject variance is simply the
`residual mean square from the ANOVA, and its CVs can be
`obtained from eq 6 or the approximation by substituting $2
`for S2. CVW or intrasubject CV estimates the relative van- =1
`ationBone expects from an individual when given the drug on 7
`different occasions.
`,
`Another CV estimate could also be calculated. This is the‘
`CV of the observed Ys. The model assumes that the variance .
`of an individual observation is the sum of the inter- and
`intrasubject variances. Thus, an estimate of this pooled CV '
`(CVP) is CVP =
`expsg + Sl' — 1 x 100%. CVP or its approx- ‘
`imation can be used to compare CVs from parallel studies.
`What needs to be defined now is the criterion for accepting.
`bioequivalence. This has produced a profusion of papers from
`all corners of statistical thought from Baysianz“,27 to multi- ‘-
`variate28 to robust statistics,29 from nonparametrict3 to clas- :
`sical parametric techniques,56 and to the latest swing to- _-
`wards individual bioequivalence.30 Common to all these .
`methods are the a priori defining of limits that have clinical}
`significance. These are the bioequivalence limits introduced'
`earlier in the paper and they give meaning to the whole}?
`concept ofaccepting a new formulation on bioavailability data‘l
`alone, in lieu of clinical data It18 generally agreed that a safe
`limitis 20%; that1s, if the test formulation on the average
`produced a concentration—time profile within 20% of the
`reference formulation, the test formulation could assume all},
`clinical support from the reference formulation for its mar-5:
`keting. Therefore, the BI is commonly referred to as being;
`80—120% when an additive scale is used. However, a syma
`metric interval on an asymmetric distribution seems inap-i
`
`propriate and, ifthe log scale13 used, the BI should be set atj
`
`80—125% because the chance of passing the criterion is
`optimal when the formulations have identical means Note,
`‘i
`that the log of 0.801s equal, except for sign, to log 1.25. ThuS; '
`a BI symmetric on the log scale expresses itself asymmetfl'
`
`cally on the original scale [i.e., a “20%” increase of the 178513
`
`formulation on log (additive) scale1s a 25% increase on thQ
`original (relative) scale]. Of course, narrower or wider limit9
`may be chosen a priori for safety or efficacy concerns of clini
`.,
`
`
`
`

`

`
`cance with particular groups of drugs.
`alluded to earlier, there have been many papers written
`- manipulation of data to state bioequivalence. However,
`of them do not take into account the design in which the
`
`“in. eters are collected. Therefore, only three methods seem
`g:acceptable to the regulatory authorities. These are (1)
`
`the 90% CI for RELBIOMED must fall entirely within the
`1’) that both of the two one-sided hypothesis6 tests should
`
`'ficant at the 5% level, and (3) that the nonparametric
`
`018 of RELBIOMED should fall within the BI.
`r log-transformed data, the 90% CI for RELBIOMED is
`
`ted as follows:
`
`ifLOWER,UPPER = expo—(T - in) : tones“ + m — stEm (7)
`
`
`
`_ (ET—ink In (0.80)
`STE
`
`2 t0.05,nTR + nRT — 2
`
`(8)
`
`" convenient to multiply the estimate (eq 4) and its
`Hence limits (eq 7) by 100 to express them as percents.
`{‘e two one-sided hypothesis tests decisions are based on
`
`and 9:
`
`.
`
`'1'
`
`ln(1.25) — (ET — ER)
`tu= —-— 2 t
`STE
`
`0.05m“ + nRT — 2
`
`(9)
`
`
`
`E‘I-th tests produce p values of <0. 05, equivalence is
`‘Ja.ted
`
`vée nonparametric 90% CI is formed by taking all the
`,'u 1 minus period 2 differences for sequence TR and
`g acting each from all the period 1 minus period 2
`
`C nces from sequence RT. These differences are ranked
`
`'ieach divided by 2 and then exponentiated. There will be
`11 'r differences. The estimate of RELBIOMED is the
`
`difference, and the lower and upper limits are the
`. priate observed differences depending on the sample
`
`1 of the two sequences. Tables identifying the appropriate
`ed difference for each limit for crossovers up to 12
`3 per sequence are given in Hauschke et al.,7 and a
`1"» : equation is given in Conover.25
`
`
`
`..
`
`
`
`
`Experimental Section
`1 —Perphenazine is a potent antipsychotic drug that undergoes
`- .ve first-pass metabohsm.31 The drugin the population may
`
`at least a bimodal distribution for AUC and C1mm according to
`mine slow and rapid hydroxylators.32
`Methodology—A modification to a published radioimmu—
`
`(RIA) procedures-‘1 was used to assay perphenazine1n plasma.
`
`_ uodification was an extraction step that guarded against inter—
`i v from metabolites The lower limit of quantitation was 39
`
`'
`1
`, which allows for good measure of the elimination phase of the
`Sup to 48 h or longer after the administration of a single oral dose
`
`4 ). Standard curves were linear over the range of 39 to 2.5
`
`ng/mL, with an average accuracy and precision of 0.9 ng/mL and 8%,
`respectively. Samples were subject to single extraction, and aliquots
`(150 ML) of the extracts were reconstituted in 7% bovine serum
`albumin and analyzed in triplicate. The test samples were always
`analyzed in parallel with the standards and quality control standards
`that were made in blank plasma obtained from each subject before
`dosing. The highest mean difference from the nominal (added) value
`for the quality control samples was 13 (:12)% at the lowest concen-
`tration (0.18 ng/mL).
`Subjects—Twenty-seven subjects were selected from a cohort of
`healthy male volunteers, aged 18—45 years, and deviating by no more
`than 10% from the ideal weight for height according to the Metro-
`politan Life Insurance Company Bulletin, 1983. Each subject was
`required to be free of cardiovascular, hepatic, renal, or gastrointes-
`tinal disease, or drug abuse or alcohol dependence, as assessed by
`physical examination and review of medical history by the supervis‘
`ing physician. In addition, the blood pressure, electrocardiogram, and
`results of clinical laboratory tests (blood chemistry, hematology, and
`urinalysis) were required to be within normal ranges. Smokers
`(15/27) were permitted to continue smoking at their normal rate of
`consumption throughout the study. All subjects were required to
`abstain from the use of all other drugs (including nonprescription
`drugs) for at least 1 week prior to until after completion of the study.
`The subjects were also required to refrain from consuming alcoholic
`or cafieine—containing beverages from 48 h prior to dosing until after
`the collection of the last blood sample. Physical examination and
`laboratory tests were also carried out during and after the study to
`monitor the health of participating subjects.
`Study Implementation—Each volunteer gave his informed con-
`sent before participating in the study. The study protocol was
`approved by the local institutional review board. Of the 27 subjects
`who started the study, 24 subjects completed both legs ofthe crossover
`study. Three subjects dropped out for personal reasons. Each subject
`received each of the 16-mg reference formulations (Trilafon, Schering
`Corporation, lot #3ADM3) and the test formulation (Zenith Labora-
`tories, lot #107-183) according to their randomized sequence with a
`2-week washout period. After an overnight fast and drawing a
`zero-time blood sample, the dose was given and 20-mL blood samples
`were collected at 0.5, 1.0, 1.5, 2.0, 3.0, 4.0, 6.0, 8.0, 12.0, 16.0, 24.0,
`28.0, 36.0, and 48.0 h. Plasma was separated by centrifugation within
`30 min of harvesting and stored at —23 °C until analysis. Standard-
`ized meals were provided at 4.0 and 9.0 h post-dosing.
`Parameters—Pharmacokinetic parameters for perphenazine were
`determined by standard techniques. The Cmax and tmax values were
`obtained directly from the plasma concentration—time curve. The
`AUL up to the last time showing a measurable concentration of
`perphenazine (AU03) was determined by the linear trapezoidal rule
`for ascending and the log trapezoidal rule for descending portions of
`the curve.34 A rate constant for the disappearance of drug from
`plasma (k) was calculated by least squares regression of the natural
`logs of the last three plasma concentrations. AUCE,n values were
`determined by adding to the appropriate AUG}, the quotient of the
`estimated last plasma concentration and the appropriate value of k.
`A half-life value (t1,2) for the disappearance of the drug from plasma
`was calculated from the quotient of In 2 and k.
`
`Results and Discussion
`
`Table II lists estimates for the medians and, when appro-
`priate, means and CVs of some pharmacokinetic parameters
`for the perphenazine study. Each parameter listed in Table II
`
`I ll—Summary Statistics for Perphenazlne Pharmacoklnetlc Parameters for Test and Reference Formulations
`Reference Formulation
`.-'
`Test Formulation
`arameter
`‘
`‘
`
`Median
`12.92
`14.58
`1.17
`—"
`—
`—
`
`Mean
`14.32
`16.59
`1.29
`2.38
`0.076
`13.40
`
`CVP
`47.9
`54.3
`45.6
`43.6
`49.3
`91.3
`
`ACVP
`45.4
`50.8
`43.5
`——
`—
`—
`
`Median
`13.39
`15.81
`1.18
`—
`—
`—
`
`Mean
`14.79
`17.50
`1.28
`2.54
`0.078
`12.84
`
`CV...
`46.9
`47.5
`41.4
`29.5
`55.9
`76.4
`
`ACVP
`44.6
`45.1
`39.8
`—
`—
`—
`
`
`
`
`‘1‘" possible to calculate for all subjects (see text). b —: N°i determined.
`
`Journal of Pharmaceutical Sciences] 141
`Vol. 82, No. 2, February 1993
`
`

`

`shows CVs of >40%, which is characteristic of phenothiazine
`antip'sychotics.35—37 These estimates of relative variation are
`mixtures of intersubject and intrasubject variance compo-
`nents. Note that fluctuations in post-Cmax plasma concentra-
`tions prevented the calculation of a value for k in some
`individuals. It is important to recognize that variations in the
`time to the last concentration were characteristic of the
`subject, not the formulation. After reaching Cm“, plasma
`concentrations showed evidence of secondary peaking in
`many subjects, as has been reported with other phenothiazine
`antipsychotic drugs.16v35’36 This phenomenon may be a result
`of biliary recycling. In any event, the difficulty of obtaining a
`reasonable estimate of k (and therefore the AUCf,/AUCE,n
`ratio) should not invalidate studies on such drugs. Therefore,
`various mean values for k, tm, and AUC3 were calculated
`from 17/24 subjects for the reference product, and 18/24 values
`for the test product. The inability to estimate these parame-
`ters in so many subjects effectively ruled them out of conten-
`tion as bioequivalence-determining variables.
`Figure 2 presents the average profiles for both the reference
`and test formulations. Table III lists the estimates Of AUCE,
`and Cm” on the log scale for each subject and formulation by
`sequence. The means presented in this table are used in the
`calculation of the two least squares means. The similar
`magnitudes of the standard deviation for each formulation
`and period indicate similar sequence groups. It is recom-
`mended that transformed values be reported to at least three
`decimal places to maintain the accuracy when summary
`statistics are transformed back to the original scale.
`Interaction diagrams for AUCf, and Cmax are given in
`Figures 3 and 4, respectively. The spreads of the values for
`each formulation for both parameters are similar, indicating
`that the formulation variance is similar. The steepness and
`criss-crossing of the lines joining subjects is an indication of
`how variable the subject-by—formulation interaction is across
`subjects.
`Table IV presents some intersubject distribution statistics
`for AUCI', and C,mam and their logs. The Shapiro—Wilk test
`indicated a lack of normality, probably due to the positive
`skewness of the distributions (W test, p < 0.01) of both AUC
`and Cmax on the raw scale. The distributions of log-
`
`1.2
`
`—L O
`
`(ng/ml) 99:5ca Pn
`CONCENTRATION
`
`4 812162024283236
`
`48
`
`Figure 2—Mean plasma concentration—time profiles following test (0)
`reference (A) formulations of perphenazine.
`
`TIME (Hrs)
`
`142/ Journal of Pharmaceutical Sciences
`Vol. 82, No. 2, February 1993
`
`.-
`
`Table Ill—Estlmates of lndivldual Perphenazlne AUC}, and Cmax
`Values on Log Scale
`AUC‘
`C
`
`Sequence
`Subject
`0
`max
`Test
`Ref
`Test
`Ref
`
`
`TR
`
`A
`B
`G
`I
`K
`M
`O
`R
`U
`V
`W
`
`1.894
`2.980
`2.717
`2.319
`2.085
`2.756
`2.665
`2.661
`2.576
`2.446
`1.664
`
`2.129
`2.794
`2.929
`2.651
`2.001
`2.813
`2.747
`2.460
`2.498
`2.649
`1.635
`
`—0.333
`0.116
`0.258
`0.053
`—0.580
`0.397
`0.664
`0.481
`0.226
`—0.090
`—0.71 5
`
`—0.190
`0.545
`0.550
`0.126
`—0.523
`0.436
`0.490
`—0.051
`0.097
`0.083
`-—0.358
`
`
`
`4..A___-__..'
`
`_
`XTR
`STD
`RT
`
`0.963
`0.944
`3.551
`3.406
`X
`0.181
`0.118
`2.571
`2.514
`—3
`0.430
`0.491
`0.491
`0.476
`—
`—0.248
`0.137
`2.335
`2.386
`C
`—0.094
`0.100
`2.256
`2.306
`D
`0.105
`0.887
`2.790
`3.328
`E
`—0.31 5
`—0.383
`1.998
`1.945
`F
`0.128
`0.061
`2.669
`2.449
`H
`0.239
`0.083
`2.625
`2.428
`J
`0.061
`~0.020
`2.851
`2.747
`L
`—0.057
`—0.01 2
`2.682
`2.684
`N
`0.428
`0.095
`2.501
`2.339
`O
`1 .152
`0.951
`3.597
`3.345
`P
`0.241
`0.494
`2.898
`3.103
`S
`0.248
`0.007
`2.211
`2.201
`T
`_
`0.157
`0.200
`2.618
`2.603
`—
`Xm
`
`
`
`
`
`— 0.447 0.416 0.387STD 0.381
`
`‘ Not applicable.
`
`3.6
`
`3.1
`
`1.6
`
`
`
`LnAUCI(ng.hr/ml) !° 0)
`
`2.1
`
`Reference
`
`in
`FORMULATION
`5
`Flgure 3—Comparison of In AUG; of each subject by formulationl
`(formulation—subject interaction).
`"
`
`Test
`
`transformed data were less skewed and less platykurtic thall-
`the distributions of natural data. The log transformation not.
`only reduced skewness and kurtosis but, in each case, Ten‘}
`
`
`
`
`

`

`1.2
`
`0.8
`
`0.4
`
`0.0
`
`
`
`-——1———————I—
`Reference
`Test
`
`FORMU LATION
`_
`yre 4—Comparison of in Cmax values of each subject by formulation
`, ulation—subject interaction).
`
`; e W—Intersublect Distribution Characteristics
`
`Skewness
`
`1.44
`0.15
`1.52
`0.31
`
`Kurt sis
`o
`2.28
`0.34
`2.75
`0.60
`
`Shapiro-Wilk (W) Test
`p values
`0.003
`0.782
`0.003
`0.490
`
`
`
`
`
`" :
`. 2‘-
`
`eter
`
`f. t,
`" UC},
`'
`,
`'r max
`
`,
`
`
`4.:- the distribution approximately normal (W test, p >
`
`_
`). It is realized that any test on distributional assump-
`1: : with small sample sizes has very low power and is
`uenced greatly by extreme values. Therefore, it should not
`
`weft to the observed data set to decide which scale should be
`for analysis.
`
`examination of the intrasubject error for AUCE, was
`ed out by correlating the positive studentized residuals
`
`the ANOVA with the mean AUCf, values for each
`,
`
`ject. The studentized residuals are simply the residuals
`) from the model fit divided by their stande error. Each
`_'ect produces two residuals that are equal but opposite in
`
`. ; thus, only the positive ones were used. The usefulness of
`: standardization is that residuals from different scales can
`
`"compared. The correlation coefficient was significant for
`
`- raw data (p < 0.001) but was not significant in the log
`
`,1" ysis (p = 0.11). These data indicate that there was a
`rtionship between the size of the residual and the magni-
`
`fs e ofAUCf, that disappeared when the analysis was done on
`’3 log scale. Thus, in a theoretical intrasubject comparison,
`
`- given difference between test and reference has a greater
`‘ lute magnitude at higher parametric values. The impli-
`
`n of this result is that the use of the raw scale places
`I weight on the higher values and correspondingly
`
`r-weights the smaller values.
`,
`.' 81' results were obtained for analysis of Cmax data,
`Srough the correlation coefficient for the raw data (p =
`
`
`
`0.053) was slightly higher than the nominal 0.05. There was
`no significant correlation in the corresponding log trans-
`formed data (p = 0.291).
`The results of ANOVA on the log transformed AUCf,
`(Tables V and VI) or Cmax (Tables VII and VIII) revealed that
`neither pharmacokinetic parameter showed any effect of
`formulation, period, or sequence of administration. In each
`case, there was a significant effect of subject nested within
`sequence that showed the usefulness of the crossover design.
`The intrasubject CVW is estimated to be 14 and 20% for
`AUC and CW, respectively, which indicates reasonably
`consistent values were obtained within a subject. One feature
`of using the log transformation is that CVW is expected to be
`constant regardless of the magnitudes or units of the param-
`eters. Thus, a drug with a CVW of 20% would be expected to
`have a residual mean square of ~0.04 over many studies. For
`example, over three studies of chlorpromazine,3s the CVW
`varied by only 5% even when different analytical methods and
`subjects were used.
`The intersubject CVB was obtained by partitioning the
`subject (seq) mean square. This partitioning yields estimates
`of CVB of 46 and 39% for AUCE, and Cm”, respectively. Also
`calculated are the pooled CVP estimates, which agree well
`with the CVps for AUCB and C,mm for each formulation (Table
`II).
`Estimates of bioequivalence from the three methods are
`included in Tables V and VI. For these sequence sample sizes,
`the nonparametric estimate of RELBIOMED is the average of
`the 72n and 73rd difference and the lower and upper limits are
`the 43rd and 102nd difference, respectively. Note a

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket