STATISTICAL ANALYSIS OF PHARMACOKINETIC DATA--- BIOEQUIVALENCE STUDY By Qingmin Guo A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of BiostatisticsŠMaster of Science 2015 ABSTRACT STATISTICAL ANALYSIS OF PHARMACOKINETIC DATA-- BIOEQUIVALENCE STUDY By Qingmin Guo Bioequivalence (BE) studies are widely carried out in the pharmaceutical industry. The assessment of BE adopted by the Food and Drugs Adminis tration (FDA) is a moment-based criterion evaluating log-transformed pharm acokinetic responses such as Area Under the Curve (AUC), Maximum Concentration ( max C), which are usually estimated from drug plasma time profiles. Average BE (ABE) is based solely on the co mparison of population averages but not on the variances, while Population BE (P BE) and individual BE (IBE) approaches include comparisons of both averages and variances. The objective of this thesis is to review the standard approaches to statistical analyses of pharmacokinetic data. It also covers estimation of AUC, max Cand other pharmacokinetic (PK) pa rameters as introduced in a Non-Compartmental Analysis (NCA) approach and Compartmental Models Analysis approach. Widely c ited data sets from the published literature are used to illustrate these two approaches. They show the benefits of parameter estimation and subsequent statistical inference with an appropriate compartmental model, even though the model fitting coul d be a little complicated. iii ACKNOWLEDGEMENTS I would never have been able to finish my th esis without the guidance of my committee members Dr. Joseph Gardiner, Dr. Zhehui Luo and Dr. Do rothy Pathak, help from friends, and support from my family. iv TABLE OF CONTENTS LIST OF TABLES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vi LIST OF FIGURES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii CHAPTER 1 INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 1.1 Pharmacokinetics (PK) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 1.2 Area Under the Curve (AUC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 1.3 Peak Concentration ( max C). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 1.4 Bioequivalence (BE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 1.5 Crossover Design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 1.5.1 Treatment Effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 1.5.2 Period Effect . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . 4 1.5.3 Carryover Effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 1.6 Statistical Model . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . 5 1.7 Average Bioequivalence (ABE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7 1.8 Population Bioequivalence (PBE) . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 1.9 Individual Bioequivalence (IBE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 CHAPTER 2 BIOEQUIVALENCE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 2.1 ABE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 2.2 Sample Size Calc ulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 2.2.1 Formula for sample size. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 CHAPTER 3 AUC ESTIMATION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 3.1 Definition of PK Parameters. . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . 20 3.1.1 Elimination Rate Constant. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 3.1.2 Absorption Rate Constant. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 3.1.3. Half-life . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . 21 3.1.4. Bioavailability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 3.1.5. Volume of Distribution. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 3.1.6. Clearance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22 3.2 Non-Compartmental Analysis (NCA) . . . . . . . . . . . . . . . . . . . . . . . . . . 22 3.3 One Compartment Model. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24 3.3.1 AUC estimation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 3.3.2 Tmax and Cmax . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . 26 3.3.3 T 1/2 (half life) estimation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27 3.3.4 Application. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28 3.3.5. Model for C(t) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28 CHAPTER 4 DISCUSSION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 v BIBLIOGRAPHY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37 vi LIST OF TABLES Table 1 The standard 22 crossover design . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 Table 2 Expected Cell Means for Model (1) . . . . . . . . . . . . . . . . . . . . . . . . . . 6 Table 3 Expected Means and Effects for Model (1) . . . . . . . . . . . . . . . . . . . . . . 6 Table 4 Bioequivalence Types and Evaluation Criteria . . . . . . . . . . . . . . . . . . . 9 Table 5 Expected Cell Means for log(AUC) . . . . . . . . . . . . . . . . . . . . . . . . . . 13 Table 6 Expected Means and Effects for log(AUC ) . . . . . . . . . . . . . . . . . . . . . . 13 Table 7 TTEST output for log(AUC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 Table 8 GLIMMIX output for log (AUC) . . . . . . . . . . . . . . . . . . . . . . . . . . 15 Table 9 Expected Cell Means for log (Cmax ) . . . . . . . . . . . . . . . . . . . . . . . . . . 15 Table 10 Expected Means and Effects for log (Cmax ) . . . . . . . . . . . . . . . . . . . . . . 15 Table 11 TTEST output for log (Cmax ) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 Table 12 GLIMMIX output for log (Cmax ) . . . . . . . . . . . . . . . . . . . . . . . . . . 17 Table 13 Comparison of AUC by NCA and compartment model based estimates . .. . . . . 30 Table 14 Comparison of Cmax by NCA and compartment model based estimates . . . . . . . 31 Table 15 Comparison of Tmax by NCA and compartment model based estimates . . . . . . . 31 vii LIST OF FIGURES Figure 1 Schema of plasma concentration-vs.-time curve . . . . . . . . . . . . . . . . 2 Figure 2 Profiles over Treatment A and B for log(AUC) in two periods . . . . . . . . 13 Figure 3 Treatment A vs. B Agreement of log(AUC) in two periods . . . . . . . . . . . . 14 Figure 4 Profiles over Treatment A and B for log(Cmax ) in two periods . . . . . . . . 16 Figure 5 Treatment A vs. B Agreement of log(Cmax ) in two periods . . . . . . . . . . . . 16 Figure 6 One compartmental model with i.v. administration . . . . . . . . . . . . . . . . . 24 Figure 7 One compartmental model with extravascular administration . . . . . . . . 25 Figure 8 Individual concentration profiles by NCA . . . . . . . . . . . . . . . . . . . . 30 1 CHAPTER 1 INTRODUCTION 1.1 Pharmacokinetics (PK) PK is the study of the movement of drugs in the body, involving the processes of absorption, distribution, metabolism, and excretion (ADME), incl uding the rate and extent of each of these processes. The science of PK concerns on how th e body converts an active drug molecule into metabolites, the time course of drugs in the body, and what the body does to the drug. PK data are often derived from blood (serum or plasma) sa mples in small to medium-size datasets of individuals over time. From a PK profile, pharmacokinetic parameters can be estimated such as Area Under the Curve (AUC), Maximum Concentration ( max C), Time to Maximum Concentration ( max T), Half Life (1/2 T), Bioavailability, Clearance, Volume of Dist ribution, etc. These PK parameters are very useful in optimization of the dosage form and dose interval. 1.2 Area Under the Curve (AUC) AUC has units of concentration ×time (e.g., /mghrL ×), is a measure of the total systemic exposure of a drug integrated over time. AUC is usually estimated from concentration-time data. There are two major approaches to estimation of AUC: one is Non-Compartmental Analysis (NCA), which calculates the AUC following the tr apezoidal rule by adding up the area under the curve between consecutive time points, the requi rement of the Food and Drugs Administration (FDA) [1][2] for AUC estimation; the other is compartmental modeling analysis, which will be discussed in chapter 3. 2 1.3 Peak Concentration ( maxC) max Crefers to the maximum (or peak) serum concen tration that a drug achieves in a specified compartment or test area of the body after the drug has been administrated and prior to the administration of a second dose, i.e., in brief, max Cis the maximum concentration observed. Figure 1 Schema of plasma concentration-vs.-time curve. In bioequivalence studies, the key pharmacoki netic parameters are log-transformed AUC and max C.[3] Figure 1 shows the schema of AUC, max Cand max T in the plasma concentration -vs.- time curve after a single oral drug dose, cited from Atkinson AJ, et al. 2007 [4] with a little modification. max Tis Time to Peak Concentration, the term used in pharmacokinetics to describe the time at which the max C is observed. 1.4 Bioequivalence (BE) BE studies are widely carried out in the pha rmaceutical industry. The US Food and Drugs Administration - Code of Federal Regulations ( FDA- CFR) definition [5] of BE is that the absence 3 of a significant difference in the rate and extent to which the active ingredient in pharmaceutical equivalents or pharmaceutical alte rnatives becomes available at the site of drug action when administered at the same molar dose under similar conditions in an appropriately designed study. Although it is seen sometimes that measures used in BE study are pharmacological or clinical end-points, the most sensitive measures used in BE studies [6] are drug concentrations in the blood. From subject-level time and concentration data, subject-level AUC and max Ccan be estimated, which form the BE outcome data for statistical analyses. If the drugs' plasma concentration curves are superimposable, then th ese drugs are considered as bioequivalent in extent and rate of absorption. The FDA guidelines for BE studies recommend a minimum of 12 samples collected over time following drug administration with an additional sample prior to dosing. [2] These investigations on BE are best made through randomized clinical trials, and BE of tw o drugs is assessed by analysis of logarithmic transformed AUC and max C typically obtained from a crossover design. 1.5 Crossover Design Crossover Design is probably the most comm only used statistical design for comparing bioequivalence between two formulations of a drug. We shall refer to a two-sequence, two- period, crossover design as the standard 2 2 crossover design, also called AB|BA design. Table 1 The standard 22 crossover design Crossover Designs for Two Fo rmulations Period 1 Period 2 Sequence AB = 1 A B Sequence BA = 2 B A A standard 2 2×crossover study will generate paired outcomes 12(,) YY in two sequences of subjects: (a) In sequence AB subj ects receive drug A in period 1 and are crossed over to drug B 4 in period 2; (b) In sequence BA subjects receive drug B in period 1 and are crossed over to drug A in period 2, as shown in Table 1. The dosing periods are separated by a washout period of sufficient length for the drug received in the first period to be completely metabolized or excreted from the body. Now, let's discuss here treatment effect, period effect and carry-over effect. 1.5.1 Treatment Effect. The objective of a cross-over trial is to focus attention on within- patient treatment differences, the difference betwee n different measurements in the same subject, also called within-subject difference. The difference between these measurements removes any component that is related to the differences betw een the subjects, which is called ‚subject-effect™, from the comparison. 1.5.2 Period Effect. The within-subject difference could al so be thought of as a comparison between the two treatment periods, which is the reason why usually one group of subjects received the treatments in the order AB and the other group received the treatments in the order BA. 1.5.3 Carryover Effect. A carryover effect is defined as effect of the treatment from the previous time period on the response at the curren t time period. The presence of carryover is an empirical matter. [7] It depends on the design, the setting, the treatment, and the response. The washout periods are usually included in the desi gn, to allow the active effects of a treatment given in one period to be washed out of the body before each subject begins the next period of treatment. The disadvantage of the 22×crossover trial is that several important effects, such as 5 carryover effect and interaction effect are aliased with each other. Therefore, the carryover effect cannot be removed just by randomization alone in 2 2× crossover design. Washout period has to be clarified, usually 5 Half-life time ( 1/2 T) is necessary. The objective of crossover design is to estimate the treatment effect, the expected difference in mean of logarithm transformed AUC and max C of drug A versus drug B obtained from each subject. In each sequence, Treatment A and Tr eatment B produce a difference measure, provided that the period effect can be assumed to be cons tant, the information from both sequences can be combined to obtain the estimate of expected difference. Since the difference measure is within one subject, the difference removes any ‚subject-effect™ from the comparison. 1.6 Statistical Model Consider a statistical model without considering carryover effect used by Byron Jones and Michael G. Kenward. [7] Let ijk Y=response in k-th patient, in j-th period for i-th treatment. There is implied ‚nesting™ of treatment in period. The model is: ijkijkijk Yµ =++++ (1) where µ is effect of an overall mean; iis effect of i-th treatment effect, i=1, 2, ..., t; jis effect of the j-th period effect, k=1, 2, ..., p; k is random effect associated with the k-th subject. 6 ijk is random error associated with the k-th subject who received the i-th treatment in the j-th period, k=1, 2, ..., ni. ()2~0, kN, 2~(0,) ijk N, k and ijk are independent. ()()222 ijkkijk VarYVar =+=+= , and 2,') ( ijkijk CovYY = for all jj. In this random subject-effects model, 2 is the inter-subject variability and 2 is the within- subject variability. In Table 2, there are only four sample means 111221 .,.,., yyy and 22.y. 11yrepresents the mean of samples of treatme nt 1 and period 1, ..., 21.yrepresents the mean of samples of treatment 2 and period 1; Table 3 lists the m ean of treatment and period, peri od effect and treatment effect with regard to this 22× crossover design. Table 2 Expected Cell Means for Model (1) Sequence Period 1 2 1 1111 .yµ=++ 2222 .yµ=++ 2 2121 .yµ=++ 1212 .yµ=++ Table 3 Expected Means and Effects for Model (1) Mean of Treatment A 1112112 ½(..)=½½ yyµ ++++ Mean of Treatment B 2122212 ½(..)=½½ yyµ ++++ Mean of Period 1 1121112 ½(..)½½ yyµ +=+++ Mean of Period 2 1222212 ½(..)½½ yyµ +=+++ Treatment (A-B) Effect 12 Period (1-2) Effect 12 7 It is known from empirical studies that after logarithmic tr ansformation, AUC and max C are normally distributed or may be assumed to be approximately no rmally distributed. The core modeling component of SAS proc GLIMMIX ca n be illustrated for both fixed and random subject-effects models. [7] Suppose there are variates including subject, period, direct treatment and response variables in the da taset, model fitting and inference for fixed subject-effect models follow conventional ordinary least squares (O LS) procedures and for random subject-effect models Restricted Maximum Like lihood (REML) analyzes are used. [7] The benefit of crossover design is that each subj ect serves as their own control, and statistical efficiencies are gained with re spect to power and precision. Alt hough crossover design has great advantages, it also brings a pot ential disadvantage: (1) Carryove r may be confounded with direct treatment effects. (2) There are at least 2 pe riods, patients may withdraw from the trial, or become "lost to follow-up". Special consideration is needed while doing statistical analysis for the crossover design. The typical method of Null Hypothesis Significance Testing (NHST) is designed to assess the evidence against the null hypothesis. The null hypoth esis is rejected if the observed p-value is less than the stated significance level ; if not rejected, the null hypothesis will be retained. However, equivalence is not concluded just b ecause we do not reject null. Here, the Two One- Sided Tests (TOST) is applied to assessing bioequivalence. 1.7 Average Bioequivalence (ABE) ABE is a conventional method for the BE study, which solely compares the population averages of a BE measure of interest but not the vari ances of the measures for the T and R products, 8 following the FDA 1992 guidance on Statistical Procedures for Bioequivalence Studies. [3][8] TOST has been used to determine whether the ratio of the logarithm transformed averages of the measures for the test and reference products were comparable. [9] The two null hypotheses of TOST: (1) the mean difference is larger than the upper value of the BE limit ; and (2) the mean difference is below the lower bound of the BE limit - , versus the alternative hypothesis of the difference falls within the range of the BE limit. where T is Test , and R is Reference . BE is established at significance level of if a t-interval of confidence ()12100% × is contained in the interval (,) , which is called Westlake™s Confidence Interval. [10] Therefore, to establish BE at significant level of 0.05 =, a 90% confidence interval should fall within the BE limit (,) . For PK measures after logarithmic tr ansformation, ln 1.25 =,ln 1.25 =,[3] while for PK measures without logarithmic transformation, the BE limit is a little different. For the case of no logari thmic transformation, let TR/Dµµ= be the ratio of the averages of the measures for the Test and Reference products. 01T02 : / , : / : / µµµµµµ<(Reference-scaled criterion) when220TRT (Constant-scaled criterion) 10 In the equation, TRµµ is still the mean difference between the test and reference. 2TT is the total variance of test, and 2TR is the total variance of reference. 20 T is the FDA specified threshold value, currently the values recommended by the FDA is 200.04 T=.[11] When the total variance of Reference 2TR is greater than the FDA specified threshold value 20 T, Reference-scaled criterion is to be applied. Otherwise, Constant-s caled criterion is to be used. Currently FDA recommended value for P is 1.7448,[11] if PBEp <, and ABE is concluded, then PBE is also concluded. 1.9 Individual Bioequivalence (IBE) IBE approach uses the means and variances of T and R, and the subject-by-formulation interaction to assess within-subject variability for the T and R products, as well as the subject-by- formulation interaction. when220WRW >( Reference-scaled criterion) when220WRW (Constant-scaled criterion) Here, 2WT is within-subject variance for test drug, and 2WR is within- subject variance for reference drug. 2BT is between-subject variance for test drug, 2BR is between-subject variance for reference drug. ()22 2 (1) =+DBTBRBTBR , assesses the subject-by- formulation interaction. is the correlation coefficient be tween individual average test and reference formulation, they both c ontribute to the IBE determination. [3][11] 2222 22222 20()()TRDWTWR WRIBE TRDWTWR Wµµ µµ ++=++11 20W is specified threshold value, currently the FDA recommended 200.04 W=.When the within variance of reference 220 WRW >, Reference-scaled crite rion is to be applied. Otherwise, Constant-scaled criterion is to be used. FDA recommended value for I is 2.4948[11], if IBEI <, and ABE is concluded, then IBE is also concluded. The two-period two-treatment crossover design is the simplest prototype, which is not enough for PBE and IBE. When additional periods a nd/or treatments are considered, the possible configurations would increase. Some examples of a three-period two-treatment crossover design are (i) Sequences ABA and BAB; (ii) Sequences AAB, ABA and BAA; (iii) Sequences ABB and BAA.[3] Just as in the AB|BA design, we note that the treatment difference can be estimated in each period from any of these three designs. The benefit of having add itional periods and/or treatments contributes to detecting if there is car ryover effect in crossover design. Moreover, the IBE can be estimated with high order crossover design in addition to ABE and PBE. 12 CHAPTER 2 BIOEQUIVALENCE A data set called "pkdata" from STATA documentation [12] is used here for our illustration. The data comprises two concentrations CONCA, C ONCB in the same n=16 subjects assesses at 13 time points, including t=0. At baseline the con centration is zero. Eight patients are randomly assigned to sequence 1, which means they take the drug A in the period 1, and then take the drug B in the period 2; the other 8 patients are assigned to sequence 2, take the drug B in the period 1, and then take the drug A in the period 2. Assume the Drug B is the reference drug (R), and Drug A is the test drug (T). We will calculate AUC and max C by Non Compartmental Analysis approach. Take the logarithmic transformation of AUC and max C as the variables for the ABE study. 2.1 ABE Consider the parameter AUC. Let BRµµ= be the reference group population mean, TAµµ= be the test group population mean. Let DTRµµµ= be the treatment difference, ln1.25 = be the lower bound, and ln1.25 =(=0.2231) be the upper bounds on DTRµµµ= that define the region of equivalence. ABE involve s the calculation of a 90% CI for DTRµµµ=, the difference in the means of log-transformed AUC. The ABE will be concluded based on the calculated 90% confidence limits falling within 0.2231 0.2231 TRµµ . First, the expected cell means for ()logAUC in Table 5 are listed following notations used in Table 2. 13 Table 5 Expected Cell Means for ()logAUC Sequence Period 1 2 1 11.5.0069 y= 22.4.9270 y= 2 21.5.0077 y= 12.4.8740 y= Similarly, the expected means and effects for ()logAUC in Table 6 are listed following notations used in Table 3. Table 6 Expected Means and Effects for ()logAUC Mean of Treatment T 1112 ½(..)=4.9404 +yy Mean of Treatment R 2122 ½(..)=4.9673 +yy Mean of Period 1 1121 ½(..)5.0073 yy+= Mean of Period 2 1222 ½(..)4.9005 yy+= Treatment (T-R) Effect -0.0269 Period (1-2) Effect 0.1067 Figure 2 Profiles over treatment A and B for ()logAUC in two periods. The objective of this study with cross-over design is to focus attention on within-subject treatment differences. Figure 2 s hows profiles over treatment for crossover designs. The subject- 14 profiles in Figure 2 are plotted for each sequenc e the change in each subject™s response over the two treatment periods, which show no strong treatment effect or period effect. Figure 3 Treatment A vs. B Agreement of ()logAUC in two periods. The treatment agreement in Figure 3 is plotte d for the response associated with the second treatment against the response associated with the first treatment. The figure indicates the strength of the treatment effect is small, and the treatment effect A-B is negative. The spread of points within sequence AB being wider indicates the bigger between-subject variability. Table 7 TTEST output for ()logAUC Treatment Method Mean Lower Bound 90% CL Mean Upper Bound Assessment Diff (1-2) Pooled -0.0269 -0.2231 < -0.1462 0.0924 < 0.2231 Equivalent Diff (1-2) Satterthwaite -0.0269 -0.2231 < -0.1503 0.0965 < 0.2231 Equivalent For crossover design, TOST option of PROC TTEST requests Schuirman ™s TOST equivalence test, with the option of specifying the equivale nce bounds. After log - tran sformation of PK data, given the BE limit is (-0.2231, 0.2231), the assessment of BE is finally shown in Table 7. Exactly the same calculations can be carried out in PROC GLIMMIX with LSMEANS statement (1) compute least squares (LS) means of fixed effects (2) compute the 90% CI for LS-mean 15 difference, and (3) see if 90% CI falls in the stated BE limits (- , ). The BE limits are (-0.2231, 0.2231) in ABE evaluation. Table 8 GLIMMIX output for ()logAUC Estimates Label Estimate Standard Error DF t Value Pr> |t| Lower Upper T-R -0.02690 0.06774 14 -0.40 0.6973 -0.1462 0.09241 In Table 8, the PROC GLIMMIX output shows that the 90% CI (-0.1462, 0.09241) falls within the range (-0.2231, 0.2231), the ABE is concluded for ()logAUC at significance level 0.05 = Table 9 Expected Cell Means for ()max logC Sequence Period 1 2 1 11.1.9763 y= 22.2.5126 y= 2 21.2.0101 y= 12.2.4469 y= Similarly, the expected cell means for ()max logC in Table 9 are listed following notations used in Table 2. ()max logC of Drug T and Drug R are es timated by PROC TTEST and PROC GLIMMIX procedures. Table 10 Expected Means and Effects for ()max logC Mean of Treatment T 1112 ½(..)=2.2116 +yy Mean of Treatment R 2122 ½(..)=2.2614 +yy Mean of Period 1 1121 ½(..)1.9932 yy+= Mean of Period 2 1222 ½(..)2.4798 yy+= Treatment (T-R) Effect -0.0498 Period (1-2) Effect -0.4866 Similarly, the expected means and effects for ()max logC in Table 10 are listed following notations used in Table 3. 16 Figure 4 shows profiles over treatment for crossove r designs. The subject-profiles in Figure 4 are plotted for each sequence the change in each subject™s response over the two treatment periods, which show no strong treatment effect but maybe a period effect. Figure 4 Profiles over treatment A and B for ()max logC in two periods. Figure 5 Treatment A vs. B Agreement of ()max logC in two periods. The treatment agreement in Figure 5 is plotte d for the response associated with the second treatment against the response associated with the first treatment. The figure indicates the 17 strength of the treatment effect is small, and the treatment effect of A-B is negative. Substantial location differences between the two sequences indicate a strong period effect. Table 11 TTEST output for ()max logC Treatment Method Mean Lower Bound 90% CL Mean Upper Bound Assessment Diff (1-2) Pooled -0.0498 -0.2231 < -0.1280 0.0285 < 0.2231 Equivalent Diff (1-2) Satterthwaite -0.0498 -0.2231 < -0.1285 0.0290 < 0.2231 Equivalent For crossover design, TOST option of PROC TTEST requests Schuirman ™s TOST equivalence test, with the option of specifying the equiva lence bounds. After logarithmic transformation of PK data, given the BE limit is (-0.2231, 0.2231), the assessment of bioequivalence is finally shown in Table 11. Table 12 GLIMMIX output for ()max logC Estimates Label Estimate Standard Error DF t Value Pr> |t| Lower Upper T-R -0.04976 0.04379 14 -1.14 0.2749 -0.1269 0.02737 Similarly use PROC GLIMMIX with LSMEANS statement: (1) compute LS means of fixed effects, (2) compute the 90% CI for LS-mean difference, and (3) see if 90% CI falls in the stated BE limits (- , ), here the BE limits are (-0.2231, 0.2231). In Table 12, PROC GLIMMIX output shows that the 90% CI (-0.1269, 0.02737) falls within the range (-0.2231, 0.2231), the ABE is concluded for ()max logC at significance level 0.05 =. Therefore, summarizing the two primary response variables for BE study, ()logAUC and ()max logC , the ABE is concluded for T and R at the significance level 0.05 =. 18 2.2 Sample Size Calculation Sample size calculation for crossover design in bioavailability and bioequivalence study is an essential question, which establishes bioequivalence within meaningful limits in the case of logarithm transformed AUC and max C.[13] A minimum number of 12 evaluable subjects should be included in any BE study according to FDA guidelines. [3] Based on Schuirmann™s TOST procedure fo r interval hypothesis, use the data DTRµµµ= as the expected mean differ ence after logarithmic tran sformation. If the 100(1-2 )% CI (),22,22 ‹‹‹‹ ,DnDn ttµµ+ of the mean µDis entirely within the BE limit (-ln1.25, ln1.25), then 0H is rejected at significance level and no drug-drug inter action is concluded; otherwise, 0Hfails to be rejected. The type-I error of the TOST procedure is often set as 5%.Total number of subjects should provide adequate power for BE demonstration, and the adequate power means at least 80% power to detect a 20% difference in products™ BE . In practice the power usually is about 80% - 90%. 2.2.1 Formula for sample size . Let n = number of subjects required per sequence. is the significant level, is the type error, CV= coefficient of variation, /100% eRCVµ=, = the BE limit, is the expected difference compared to expected mean of Reference , 100TRRµµµ=; ‹is the intra-subject standard deviati on. Assuming a normal distribution of logarithm transformed PK data (AUC, max C), for 0=, n can be estimated [13][14] by 19 22,22/2,22 ‹2[][/] nnntt + The approximate sample size calculation for the TOST tests for > 0, the equation is: 22,22,22 ‹2[][/()] +nnntt The n on the right hand side is unknown (in the degr ees of freedom). Start with an initial value n0, the n1 is calculated. The calculation is iterative until n is almost unchangeable. As an illustration, consider the STATA dataset de scribed at the beginning of this chapter. Let =0.05, =0.2; For ()logAUC , 0.2231 =, ‹=0.1355, use the initial value 08n=22/22222[(22)(22)] [1.7611.345] ‹220.13558 0.2231 tntn n++ So n per group = 8, and total n = 16. The n per group =8 is also estimated from PROC POWER. The example is for ABE study. The number of subjects for PBE or IBE studies can be estimated by simulation according to the FDA guideline, [3] which will not be discussed here. 20 CHAPTER 3 AUC ESTIMATION There are two representative examples of one compartmental pha rmacokinetic models: [15][16] (A) One compartmental model with i.v. adminis tration; (B) One compartmental model with extravascular administration. One compartmental model with i.v. dosing means administering a dose of drug over a very short time period, there is no absorption rate constant ( ak) considered. A one compartmental model with extravascular administration means absorpti on phase is involved in the whole process. As shown in Chapter 1, the max C and max T are simple measures for summarizing the absorption process. In one compartme nt model, assessment of max T depends on the value of both elimination rate constant ( ek) and absorption rate constant ( ak). ()maxlog/ aeaekkTkk=. Let us look into the concepts of ek and ak, and how they appear in a pharmacokinetic model. 3.1 Definition of PK Parameters 3.1.1 Elimination Rate Constant (ek, units are h-1) describes the rate of decrease in concentration per unit time, usua lly the time unit is hour. It is estimated from the log-linear terminal part of the concentration-time curve, ekslope =. 3.1.2 Absorption Rate Constant (ak, units are h-1) is the rate of absorption of a drug absorbed from its site of application according to assumption of first-order kinetics, which is for a drug 21 administered by a route (for exampl e, oral) other than the intrave nous. The first-order differential equation that governs the drug amount remained ()Xt:()()adXt kXt dt=. (0) DX= is the actual dose (mg) that is available to the body for kinetic s, whereas the oral dose is given in mg/kg. Next we define the four most useful PK parameters charact erizing the in vivo disposition of a drug. [15][16] 3.1.3 Half-life (1/2 T) is given by ()1/2 log2/k eT=, that is, the time from max T to reach one- half of the maximum concentration max C. 3.1.4 Bioavailability (F, has no unit) is described as the fraction of the extravascular dose of the administered drug that reaches the absorption de pot. If the drug is injected intravenously, it is assumed that bioavailability F =100%. B ioavailability generally decreases when a medication is administered via other routes (such as orally), such as oraliv ivoral AUCDose FAUCDose ×=×, F is often measured by quantifying the "AUC". 3.1.5 Volume of Distribution (V, the units of volume, e.g., L ) or apparent volume of distribution is a pharmacological, theoretical volume that the to tal amount of administered drug would have to occupy (if it were uniformly distri buted), to provide the same concentration as it currently is in blood plasma. There are two quantities: the concentration ()Ct in plasma and the amount of drug ()Xt in tissue. It is assumed that ()/() XtCtV = is constant. 22 3.1.6 Clearance (CL, the units are volume per time, e.g., L/hr) is called the drug clearance rate, can be defined as the volume of plasma which is completely cleared of drug per unit time. CL is calculated using the dose administered divi ded by the subsequent measured AUC, ()(0) eOraldoseFX CLVk AUCAUC ×===× , where F is the bioavailability. Unless we have information on F, the parameter CL/F is only identified, this is called the apparent clearance. With the drug plasma concentration-time profile, AUC can be estimated by NCA, and also by compartmental modeling analysis. 3.2 Non-Compartmental Analysis (NCA) Based on the theory of statistical moments, the mo ments of a function are used in the analysis of pharmacokinetic data. [17] Suppose drug concentration ()Ct is a real-valued function defined on the interval [0, ); the zero th moment of ()Ct is 0S: 00()SCtdtAUC ==, fithe area under the curve from time zero to infinityfl; and the first moment of ()Ct is 1S: 10()StCtdtAUMC ==, fithe area under the first moment curvefl, is the area under the curve of concentration-time versus time curve from time zero to infinity, AUMC can be used to estimate some other PK parameters. Non-compartmental Analysis estimate AUC using the trapezoidal rule without making any assumption concerning the number of compar tments. Following the trapezoidal rule, concentration-time curve is considered as a series of trapezoids and the AUC estimate is the total area of all the trapezoids. 23 For non-compartment model, (0) tAUC is AUC from 0 h to the last quantifiable concentration to be calculated; (0)(0)() ttAUCAUCAUC =+, represents the total drug exposure over time. (0) AUC requires extrapolation of th e elimination-phase curve beyond the last measurable plasma concentration. The ex trapolation of AUC from t to infinity requires several assumptions: (1) At low concentrations, drug usually declines in mono exponential fashion; (2) The terminal elimination rate constant does not change over time or with different concentrations of circulating drug; (3) other processes such as ab sorption and distribution do not play a significant role in the terminal phase of the pharmacokinetic profile. These assumptions usually are valid in almost all PK applications. Therefore, (0) AUC can be calculated as (0)(0) C/ tlaste AUCAUCk =+, where last Cis the last observed quantifiable concentration and ek is the terminal pha se rate constant, ekslope =(units: h -1). When a regression line is fitted to terminal phase data points on log-scale, then elimination half-life 1/2 log(2)/ eTk= can be estimated. With NCA, the observed max C and max T are obtained directly from the data without interpolation. Non-compartmental analysis allows a simple estimation of AUC. It basically summarizes the concentration-time pr ofile without modeling assumpti ons. However, non-compartmental methods are unable to visualize or predict plasma concentration time profile for other dosing regimens. It assumes the kinetics to be linear and stationary (i.e., time independent) for simple applications. In more sophisticated analyses of PK data, the one compartment model or multi- compartment analysis with nonlinear mixed effects models (NLMEM) are increasingly used in drug development. [18] 24 We will use the widely cited example of drug theophylline, [19][20][21] which has serum concentrations measured at 11 time points over a 25 hour period in 12 subjects to illustrate the NCA method and compare the results with that of a one compartment model. 3.3 One Compartment Model Compartmental modelin g in pharmacokinetics estimate the concentration- time curve using kinetic models that depend on the rate of drug distribution to the different parts of the body. [15] In a one compartment model, the drug is consid ered to be distributed instantaneously into all parts of the body . The simplest case, if i.v. drug is received, it instantaneously equilibrates within the compartment and is eliminated at a constant rate ek. For a concentration measure ()Ct, first- order kinetics is assumed: ()()edCt kCt dt= where 0ek> is the elimination rate constant. We get ()(0)exp() eCtCkt = where C(0) is the initial concentr ation. The elimination rate is h -1, per hour. (0)(0)/ CXV = is the initial amount of dose in mg per unit volume in L, where V is the volume distribution. Figure 6 One compartmental model with i.v. administration. i.v. ek Central Compartment 25 Figure 7 One compartmental model with extravascular administration. For extravascular (such as oral) administration, th e body receives the drug and is absorbed at constant rate ak proportional to the amount of drug available for absorption. The drug instantaneously equilibrates within the compartment and is eliminated at a constant rate ek. The first-order differential equations that govern the amounts 1()Xt:11()()adXt kXt dt=, and 2()Xt: 212()()() aedXt kXtkXt dt=.The initial amounts are 1(0) XD= and 2(0)0 X=; at time t, the drug amount in the absorption depot is: 11()(0)exp() aXtXkt =, as shown in Figure 6; and the drug amount in the central compartment is ()12(0) ()exp()exp() aeaaekXXtktkt kk= , as shown in Figure 7. Note that this equation makes sense only when aekk>. The focus is on the equation of central compartment, which can be ex pressed as the con centration equation: ()()exp()exp() ()aeaaekDCtktkt Vkk = . From the equations /ekCLV = and (0)() XOraldoseF =×, an operational expression of drug concentration in the central compartment at time t is ()()()exp()exp() ()aeeaaekkoraldoseF Ctktkt CLkk ×= . input ak ek Absorption depot, 1()Xt Central compartment, 2()Xt 26 The unknown parameters are ,ak ,ek CL that must be estimated from observed concentrations {():0,1,,} Cttm =–in individuals over a grid of time points. 3.3.1 AUC estimation. From the equation of the drug co ncentration, the AUC is defined as 0()AUCCtdt = and also denoted by (0) AUC . From the formula for ()Ct we get ()0()exp()exp() ()()11() .()aeeaaeaeaeea kkoraldoseF AUCktktdt CLkk kkoraldoseF oraldoseF CLkkkkCL ×= ××== Only if the underlying pharmacokinetic model is identified, can the parameters be accurately estimated, otherwise this method of estimation is not to be recommended.[22] 3.3.2 max T and max C estimation. . Since max T is the time to maximum concentration we obtain the maximum value of ()Ct by solving ()0dCt dt= and showing that the unique solution is indeed the maximum value. ()()()exp()exp() ()aeeeaa aekkoraldoseF dCt kktkkt dtCLkk ×=+. The derivative is a continuous function; at t=0 it is positive; as t +, the derivative approaches zero. When aekk>, the solution max T is given by ()maxlog/ aeaekkTkk=. To obtain max C: 27 ()maxmaxmax ()()exp()exp() ()aeeaaekkoraldoseF CTkTkT CLkk ×= . Although we defined 0()AUCCtdt =, another quantity of interest is max max()(0,) 0TAUCCtdt T=. Using the formula max max 01exp() exp() TaaakTktdt k= and repeating the previous calculation gives ()max (0,) max0maxmax max()exp()exp() ()()exp()exp() 11()()exp()exp ()()TaeTea aeaeea aeeaea aee aee kkoraldoseF AUCktktdt CLkk kkoraldoseFkTkT CLkkkkkk kkoraldoseFkT oraldoseF CLCLkkk ×= ×= ××=max ()aakTk The first term on the right hand side is (0,) AUC that was calculated previously. 3.3.3 1/2 T (half life) estimation . The half-life 1/2 T is the time from max T to reach one-half of the maximum concentration max C. Initially at t= 0 the concentration is zero. With the passage of time the concentration ()Ct increases to a peak max C at time max T and then ()Ct declines to zero asymptotically. The relationship between elimination rate constant ( ek) and half-life ( 1/2 T) is: 1/2 log2 ekt=. The half-life ( 1/2 T) is determined by clearance ( CL) and volume of distribution (V): 28 1/2 log2 VTCL×=. An objective of PK studies is to obtain estimates of parameters from observations of concentrations {():0,1,,} Cttm =– in individuals over a grid of time points. The parameterization may allow for some parameters to be individual-specific which makes them random effects instead of pure constants. 3.3.4 Application. Take the widely cited example of drug theophylline, [19][20][21] serum concentrations measured at 11 time points ove r a 25 hour period in 12 subjects. First the NCA method is used to estimate the PK parameters, comparing the re sults from the one compartment model. As described above, the one compartment model is used to estimate the PK parameters, and then estimates of AUC, max C and max T are derived from the form ulas. Among the benefits of the one compartment model analysis is that the PK parameters are estimated together with their standard errors and 95% confidence interv als. In addition indivi dual (subject-specific) prediction of drug conc entration can be made. 3.3.5 Model for ()Ct. Assume normal distribution for ()Ct given (,,) aekkCL . The parameters (,) akCL are subject-specific, i. e., random effects, but ek is a fixed parameter. All parameters are transformed to their logged form: ()alogk , ()elogk and ()logCL ;and the random effects are jointly normal and indepe ndent of the error term in ()Ct. Therefore, formally the model is desc ribed by the equation (for one subject) ()()()exp()exp()() ()×= +aeeaaekkoraldoseF Ctktktt CLkk 29 where (i) the error term(s) ()t are serially independent (within subject), normally distributed, mean zero and variance 2 . (ii) 11(), CLlgb o=+()22,ablogk =+()3,elogk =with 1, 2, 3 as fixed parameters, 1,b 2b as subject-specific ra ndom effects Š means 0, covariance matrix (3-parameters 221212 ,,). (iii) 12(,) bb independent of the error term. Across subjects independence is assumed. Hence we can construct a joint likelihood for the sample data {():0,1} iCttin for the n=12 subjects with 11 concentrations assessed at the same grid of time points from (0, 25hr). Maximum likelihood estimation (MLE) provides es timates of all model parameters and their covariance matrix. There are 7 parameters: 1, 2, 3, 21, 22, 12, 2.The formulas for AUC, max C and max T for the compartment model now give their estimates. Because (,) akCL is individual-specific, we will get individual- specific estimates for the PK parameters. The additional big advantage of the compartment model is the calculation of standard errors of these estimates. Table 13 shows the 0inf AUC calculation by NCA approach, and AUC estimates by compartment model. It shows the values estimated by these two approaches are very similar except subject 1 and subject 10. 30 Table 13 Comparison of AUC by NCA and compartment model based estimates NCA Compartment model based estimates Obs subject 0inf AUC AUC Stderr AUC Lower 95% CI Upper 95% CI 1 1 270.004 144.816 7.00102 129.217 160.416 2 2 95.050 110.005 5.87935 96.905 123.105 3 3 107.599 111.415 5.95405 98.148 124.681 4 4 121.926 118.535 6.25209 104.604 132.465 5 5 146.878 137.689 6.62215 122.934 152.444 6 6 87.877 87.023 5.54399 74.670 99.376 7 7 115.931 106.554 6.28655 92.547 120.561 8 8 104.732 102.774 5.87002 89.695 115.853 9 9 96.641 95.818 5.34297 83.914 107.723 10 10 207.536 154.629 7.40864 138.122 171.137 11 11 85.472 96.932 5.54048 84.587 109.277 12 12 126.815 141.073 6.77517 125.977 156.169 Figure 8 Individual concentration profiles by NCA. Figure 8 shows individual concentration profile s by NCA, AUC of subject 1 and subject10by NCA are higher than AUC of most subjects. Higher last Cof these 2 subjects explains the higher AUC by NCA, and furthermore, explains the discrepancy of AUC by NCA and compartment model analysis. AUC=270 AUC=207 31 Table 14 Comparison of max C by NCA and compartment model based estimates NCA Compartment model based estimates Obs subject max C max C Stderr max C Lower 95% CI Upper 95% CI 1 1 10.50 10.3420 0.33180 9.60273 11.0813 2 2 8.33 8.2205 0.32169 7.50370 8.9372 3 3 8.20 8.3918 0.32066 7.67732 9.1063 4 4 8.60 8.2855 0.31677 7.57966 8.9913 5 5 11.40 9.8981 0.32666 9.17026 10.6259 6 6 6.44 6.1226 0.30897 5.43414 6.8110 7 7 7.09 6.9453 0.31625 6.24068 7.6500 8 8 7.56 7.3470 0.31939 6.63534 8.0586 9 9 9.03 7.7379 0.31140 7.04408 8.4318 10 10 10.21 9.7244 0.31813 9.01554 10.4332 11 11 8.00 7.5903 0.32118 6.87468 8.3060 12 12 9.75 9.5540 0.32690 8.82562 10.2824 Table 15 Comparison of max T by NCA and compartment model based estimates NCA Compartment model based estimates Obs subject max T max T Stderr max T Lower 95% CI Upper 95% CI 1 1 1.12 2.10624 0.18124 1.70241 2.51008 2 2 1.92 1.57581 0.16126 1.21650 1.93511 3 3 1.02 1.48354 0.17806 1.08680 1.88028 4 4 1.07 2.35712 0.21209 1.88455 2.82969 5 5 1.00 2.02911 0.15872 1.67545 2.38277 6 6 1.15 2.28098 0.27775 1.66212 2.89984 7 7 3.48 3.17439 0.32442 2.45154 3.89723 8 8 2.02 2.09445 0.22878 1.58470 2.60420 9 9 0.63 0.66861 0.13729 0.36270 0.97452 10 10 3.55 3.59319 0.26621 3.00003 4.18634 11 11 0.98 1.02902 0.15033 0.69407 1.36397 12 12 3.52 2.72684 0.20907 2.26101 3.19268 Similarly, the max C and max Testimates by NCA approach and compartment model are shown in Table 14 and 15. It indicates the values estimate d by these two approaches are very similar. Our analysis shows the benefits of parameter esti mation and subsequent statistical inference with an appropriate compartmental model, even though the model fitting could be a little complicated. 32 This pharmacokinetic model is well identified; the parameters can be accurately estimated. If it failed AUC estimation is recommended by NCA. [22] Failure to fit a compartment model to a given data set could be due to ma ny factors. As seen the statistical model is highly non-linear, introduction of too many random effects can be problematic if the data set cannot support a complex structure. A good approach would start w ith a ‚fixed™ parameters model to obtain initial parameter values for building a more complex model. A few attempts might be needed before a stable model can be obtained. Extension beyond a one compartment model is possible. Two-compartment models view the body as a central compartment that receives the drug with transfer from the central compartment to a peripheral blood compartment that absorbs th e drug. Transfer in the opposite direction from peripheral to central is also possible. Elimination occurs from the central compartment. 33 CHAPTER 4 DISCUSSION The objective of this thesis is to review the st andard approaches to statistical analyses of pharmacokinetic (PK) data. It covers estimation of Area Under the Curve (AUC), Peak Concentration (max C) and other PK parameters and how a bioequivalence (BE) study can be conducted with crossover design. Parameters such as AUC and max C are the key parameters in a PK study and used for bioavailability and bioe quivalence. They are identified as population parameters and estimated from observed drug concentration-time profiles. The assessment of AUC adopted by the Food and Drugs Administration (FDA) is the Non- Compartmental Analysis (NCA) approach that es timates AUC using the trapezoidal rule without making any assumption concerning the number of compartments. Following the trapezoidal rule, concentration-time curve is considered as a series of trapezoids and the AUC estimate is the total area of all the trapezoids. The other appr oach is based on compartmental models. The one compartment model is used to estimate the PK parameters, such as Absorption Rate Constant (ak), Elimination Rate Constant ( ek) and Clearance ( CL). They can be estimated from observed concentrations {():0,1,,} Cttm =– in individuals over a grid of time points, and then estimates of AUC, max C and max T are derived from formulas. They show the benefits of parameter estimation and subsequent statistical inference with an appropriate compartmental model, even though the model fitting could be a little complicated. Among the benefits of the one compartment model analysis is that the PK parameters are estimated together with their 34 standard errors and 95% confidence intervals. In addition individual (subj ect-specific) prediction of drug concentration can be made. For Pharmacokinetic/P harmacodynamic modeling, the compartmental pharmacokinetic models are widely used, providing continuous description of the drug concentration that can serve as the input of pharmacodynamic models. [23] Fitting of compartmental models can be a complex and lengthy process. As seen the statistical model is highly non-linear, introduction of too many random effects can be problematic if the data set cannot support a comple x structure. A good approach would start with a ‚fixed™ parameters model to obtain initial parameter values for building a more complex model. A few attempts might be needed before a stable model can be obtained. If it failed AUC estimation is recommended by NCA. [22] The widely cited example of drug "theophylline" data [19][20][21] is used to illustrate these two approaches. Comparison of the PK paramete rs by NCA and one compartmental model shows the parameters estimated from these two methods ar e very close, the model is identified. Only when the underlying pharmacokinetic model parame ters are identified, can AUC be accurately estimated, otherwise AUC estimation is recommended by the NCA. BE studies are widely carried out in the pharmaceutical industry. For small molecule drug products, a bioavailability and bioequivalence study are required by FDA for approval of generic drug products, which contain the exact same activ e ingredient as the innovator drug. Biosimilars are large molecule biological drug products ma de via living systems. As generic forms of 35 biological products instead of the classical generic drugs, biosimilars are only similar to the reference product; with no exactly the same activ e ingredient as the innovator drug. The more stringent assessment include safety, purity, and pote ncy, to show that a follow-on biologic is not clinically different from the reference biological product. [24] Average bioequivalence (ABE) is based solely on the comparison of population averages but not on the variances, while population bioequivale nce (PBE) and individua l bioequivalence (IBE) approaches include comparisons of both averages and variances. For statistical analyses in a bioequivalence study, we used the "pkdata" example with AB|BA design to illustrate how the crossover design is applied and ABE is te sted. SAS procedures PROC TTEST and PROC GLIMMIX are applied to the logarithm-transformed AUC and max C to estimate ABE . Available from a public resource even though there are quality issues in this data, the "pkdata" example is the reasonable example of data with blood c oncentration time profile s, from which we can estimate AUC and max C, the two key parameters to compare in the BE study. The deficit of the data for BE study includes: (1) For a AB/BA design, since there are only 4 combinations of periods and treatme nts, the period effect in this particular parameterization is aliased with the carryover effect. [25] Our results show that there is period effect when comparing max log() Cfor A and B. There is no information available if there is carryover effect, therefore, it is not clear if the period effect is a real period effect. The purpose of this thesis is to illustrate how a bioequivalence (BE) study can be conducted with crossover design, so we claim there is no carryover effects. (2) For the AB/BA design, it is not well-suited for comparison of the 36 within-unit variance 2A and 2B[25] in the statistical model, we have only have one common variance 2 for treatment A and treatment B. Theref ore, total variance of A and B cannot be calculated. The "pkdata" example cannot be used for Population BE and individual BE study. We will need a replicated crossover design. [3] In recent years statisticians in the pharmaceuti cal industry have given a ttention to developing strategies for statistical analyses for Biosimilars and Biobetters. The FDA recently (April 2015) issued guidance on the scientific issues to be considered in demonstrating biosimilarity to a reference product. [26] The FDA™s definition states: Biosimilar or biosimilarity means that fithe biological product is highly similar to the refere nce product notwithstanding minor differences in clinically inactive components,fl and that fithere are no clinically meaningful differences between the biological product and the refe rence product in terms of the safety, purity, and potency of the product.fl Therefore, the role that crossover designs have in bioequivalence demonstration on a single endpoint or outcome measures must now be ex panded in ways to asse ss multiple endpoints and measures. It will bring biostatis ticians and methodologists in pharm acology together to craft the statistical designs and studies for clinical evaluations that can answer these questions. 37 BIBLIOGRAPHY 38 BIBLIOGRAPHY [1] Guidance for Industry--- Exposure-Response Rela tionships --- Study Design, Data Analysis, and Regulatory Applications. U.S. Department of Health a nd Human Services Food and Drug Administration, Center for Drug Evaluation a nd Research (CDER), Center for Biologics Evaluation and Research (CBER) April 2003. [2] Guidance for Industry --- Bioavailability and Bioe quivalence Studies Submitted in NDAs or INDs --- General Considerations. U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Ev aluation and Research (CDER) March 2014. [3] Guidance for Industry ---Statistical Approac hes to Establishing Bioequivalence. U.S. Department of Health and Hu man Services Food and Drug Ad ministration Center for Drug Evaluation and Research (CDER) January 2001. [4] Atkinson AJ, Abernethy DR, Daniels CE, et al. Principles of clinical pharmacology. Second edition. Academic Press, 2007. [5] FDA-CFR. Title 21--- Food and Drugs chapter 1--- Food and drug administration department of health and human services Subchapter D---Drugs and human use . Part 320 Bioavailability and Bi oequivalence, 2014. [6] Niazi SK. Handbook of Bioequivalence Testing. Second Edition.CRC Press 2014. [7] Jones B, Kenward MG. Design and Analysis of Cross-Over Trials. Third Edition. Chapman and Hall/CRC, 2014. [8] CENTER FOR DRUG EVALUATION AND RESEARCH --- Guidance for Industry. The FDA published Good Guidance Practices in February 1997. [9] Schuirmann DJ . A comparison of the two one-sided tests procedure and the power approach for assessing the equivalence of average bioavailability . J Pharmacokinet Biopharm. 1987; 15: 657-80. [10] Westlake WJ . Symmetrical confidence intervals for bioequivalence trials. Biometrics. 1976; 32: 741-4. [11] McNally RJ , Iyer H , Mathew T. Tests for individual and population bioequivalence based on generalized p-values. Stat Med. 2003; 22: 31-53. [12] StataCorp. Stata 14 Base Reference M anual. College Station, TX: Stata Press. Datasets for Stata Base Reference Manual, Release 14. 2015. 39 [13] Chow SC, Jen-pei Liu, JP. Design and Analysis of Bioavailability and Bioequivalence Studies. Third edition. Chapman and Hall/CRC, 2008. [14] Statistical Approaches To Establishing Bioequivalence . Appendix 1: statistical issues --- Food and Drug Administration. 2002. [15] Hedaya MA. Basic pharmacokinetics. Second edition .CRC Press, 2011. [16] Benet LZ, Zia-Amirhosseini P. Basic principles of pharmacokinetics. ToxicolPathol.1995; 23: 115-123. [17] Chow SC, Hsieh TC, Chi E, and Yang J. A comparison of moment-based and probability- based criteria for assessment of follow-on biologics. Journal of Biopharmaceutical Statistics. 2010; 20: 31-45. [18] Wang Z, Kim S, Quinney S, Zhou J, Li L . Non-compartment model to compartment model pharmacokinetics transformation meta-analysis Œ a multivariate nonlinear mixed model.BMC Systems Biology. 2010; 4: S8. [19] Pinheiro JC and Bates DM. Approximations to the Loglikelihood Function in the Nonlinear Mixed Effects Model. Journal of Computational and Graphical Statistics.1995; 4: 12-35 [20] Wolfinger RD. Fitting Nonlinear Mixed Models with the New NLMIXED Procedure . SUGI Proceedings, 1999. [21] Russek-Cohen E, Ma rtinez MN, Nevius AB. A SAS/IML program for simulating pharmacokinetic data. Computer Methods and Programs in Biomedicine.2005; 78: 39-60. [22] Buncher CR. Statistics in the Pharmaceutical Industry. Second edition. Marcel Dekker Inc, 2006. [23] Derendorf H, Meibohm B. Modeling of pharmacokinetic/pharmacodynamic (PK/PD) relationships: concepts and perspectives. Pharm Res. 1999; 16: 176-85. [24] Chow SC, Ju C. Assessing biosimilarity and interchange ability of biosimilar products under the Biologics Price Competition and Innovation Act. Generics and Biosimilars Initiative Journal.2013; 2: 20-5. [25] Vonesh, EF and Chinchilli, VM. Linear and Nonlinear Models for the Analysis of Repeated Measurements. First edition. Marcel Dekker Inc, 1997. [26] Scientific Considerations in Demonstra ting Biosimilarity to a Reference Product Guidance for Industry. U.S. Department of Health a nd Human Services Food and Drug Administration Center for Drug Evaluation a nd Research (CDER), Center for Biologics Evaluation and Research (C BER).April 2015 Biosimilarity.