Bioequivalence and Bioequivalency Testing

Similar documents
BIOAVAILABILITY & BIOEQUIVALENCE TRIALS

Robert L. Talbert, Pharm. D. College of Pharmacy UT Austin. UT Health Science Center San Antonio

Overview of Dissolution for BA/BE

Guidance for Industry

Bioavailability / Bioequivalence

Statistics and Pharmacokinetics in Clinical Pharmacology Studies

Clinical Study Synopsis

Bioequivalence Study Design Considerations. Dr. John Gordon

PHAR 7633 Chapter 19 Multi-Compartment Pharmacokinetic Models

IV solutions may be given either as a bolus dose or infused slowly through a vein into the plasma at a constant or zero-order rate.

Generic drugs are copies of innovator drug products

Bioequivalence: An overview of statistical concepts

Letter Date March 27, 2007 Stamp Date March 28, 2007 PDUFA Goal Date January 28, 2008

A Peak at PK An Introduction to Pharmacokinetics

5.5 Pharmacokinetics - Sublingual

Bundesinstitut für Arzneimittel und Medizinprodukte. Dissolution Testing. Analytik,Methodenentwicklung, Bioäquivalenz SAQ. Olten, 25.

Humulin (LY041001) Page 1 of 1

Clinical Study Synopsis for Public Disclosure

Introduction to Enteris BioPharma

Statistical estimation using confidence intervals

Guidance for Industry

NIMULID MD. 1. Introduction. 2. Nimulid MD Drug delivery system

PHAR 7633 Chapter 21 Non-Linear Pharmacokinetic Models

Guidance for Industry

Public Assessment Report Scientific discussion. Tenofovir disoproxil Teva (tenofovir disoproxil) SE/H/1432/01/DC

PUBLIC ASSESSMENT REPORT Scientific Discussion. Perindopril arginine Amlodipine FR/H/ /01-04/DC. Applicant: Servier

Compilation of individual product-specific guidance on demonstration of bioequivalence

Guidance for Industry

Post-Approval Change Management: Challenges and Opportunities An FDA Perspective

HYDROCORTISONE 10 MG TABLETS

Guidance for Industry

Revision of The Dissolution Procedure: Development and Validation 1092

CTC Technology Readiness Levels

Disclosure. This presentation contains forward-looking statements.

Longitudinal Modeling of Lung Function in Respiratory Drug Development

Guidance for Industry

The Japan Generic Market Drivers and Obstacles for Change. Matt Heimerdinger Anterio Inc.

Nursing 113. Pharmacology Principles

Sponsor. Novartis Generic Drug Name. Vildagliptin. Therapeutic Area of Trial. Type 2 diabetes. Approved Indication. Investigational.

PHARMACEUTICAL MANAGEMENT PROCEDURES

Sample Size and Power in Clinical Trials

A MULTIVARIATE TEST FOR SIMILARITY OF TWO DISSOLUTION PROFILES

Introduction to pharmaceutical technology

New anticoagulants: Monitoring or not Monitoring? Not Monitoring

The 505(b)(2) Drug Development Pathway:

MULTISOURCE (GENERIC) PHARMACEUTICAL PRODUCTS: GUIDELINES ON REGISTRATION REQUIREMENTS TO ESTABLISH INTERCHANGEABILITY DRAFT REVISION

Human ADME and Studies with Radiolabeled Compounds: Phase I-IIa

U.S. Scientific Update Aricept 23 mg Tablets. Dr. Lynn Kramer President NeuroScience Product Creation Unit Eisai Inc.

Teriflunomide is the active metabolite of Leflunomide, a drug employed since 1994 for the treatment of rheumatoid arthritis (Baselt, 2011).

Guidance for Industry

PCNE Classification for Drug related problems

Study (s) Degree Center Acad. Period Grado de Farmacia FACULTY OF PHARMACY 3 Annual PDG Farmacia-Nutrición Humana y Dietética

THE LOG TRANSFORMATION IS SPECIAL

Questions and Answers for Health Care Providers: Renal Dosing and Administration Recommendations for Peramivir IV

February page 1 / 9

Food, Medicine and Health Care Administration and Control Authority

M4E(R2): The CTD Efficacy

GDUFA Regulatory Science Update

DESCRIPTIVE STATISTICS. The purpose of statistics is to condense raw data to make it easier to answer specific questions; test hypotheses.

Standard Deviation Estimator

The Patient-Centered Medical Home How Does Managed Care Pharmacy Add Value?

Pharmacy Operating Guidelines & Information

Research on Tablet Splitting

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE

Guidance for Industry

Guideline on Generic Substitution of Immunosuppressive Drugs

Not All Clinical Trials Are Created Equal Understanding the Different Phases

GENERAL CONSIDERATIONS FOR CLINICAL TRIALS E8

IN VITRO BINDING BIOEQUIVALENCE STUDY SUMMARY TABLES AND SAS TRANSPORT FORMATTED TABLES FOR DATASET SUBMISSION

EXIGENCIA DE ESTUDIOS DE BIOEQUIVALENCIA A TRAVÉS DE METODOS IN VITRO

Two-Sample T-Tests Allowing Unequal Variance (Enter Difference)

Biological importance of metabolites. Safety and efficacy aspects

Generating Randomization Schedules Using SAS Programming Chunqin Deng and Julia Graz, PPD, Inc., Research Triangle Park, North Carolina

Absorption of Drugs. Transport of a drug from the GI tract

PharmaPendium. The definitive source of best-in-class drug information

ICH Topic E 8 General Considerations for Clinical Trials. Step 5 NOTE FOR GUIDANCE ON GENERAL CONSIDERATIONS FOR CLINICAL TRIALS (CPMP/ICH/291/95)

Guidance for Industry

MULTISOURCE (GENERIC) PHARMACEUTICAL PRODUCTS: GUIDELINES ON REGISTRATION REQUIREMENTS TO ESTABLISH INTERCHANGEABILITY. REVISION (JULY 2014)

VITAMIN C AND INFECTIOUS DISEASE: A REVIEW OF THE LITERATURE AND THE RESULTS OF A RANDOMIZED, DOUBLE-BLIND, PROSPECTIVE STUDY OVER 8 YEARS

NONCLINICAL EVALUATION FOR ANTICANCER PHARMACEUTICALS

Public Assessment Report. Decentralised Procedure

Frequently Asked Questions

Fexinidazole a new oral treatment for sleeping sickness update of development

Applying Statistics Recommended by Regulatory Documents

Veterinary Compounding

Comparative Bioavailability Study of a Generic Sustained Release Diclofenac Sodium Tablet

Ibuprofen 200mg Soft Capsules PL 03105/0105 UKPAR

Methods for Measuring Dose Escalation in TNF Antagonists for Rheumatoid Arthritis Patients Treated in Routine Clinical Practice

Medication Utilization. Understanding Potential Medication Problems of the Elderly

Two-Sample T-Tests Assuming Equal Variance (Enter Means)

ICH Topic Q 1 A Stability Testing Guidelines: Stability Testing of New Drug Substances and Products

Proof-of-Concept Studies and the End of Phase IIa Meeting with the FDA

Pharmaceutical Sciences

For personal use only

ANDA CHECKLIST FOR CTD or ectd FORMAT FOR COMPLETENESS and ACCEPTABILITY of an APPLICATION FOR FILING

Annex 7 Guidelines on pre-approval inspections

Hybrid or Mixed Marketing Authorization Application in the European Union: Not a Trivial Decision in New Development Programs for Established Drugs

TRAMADOL HYDROCHLORIDE AND PARACETAMOL 37.5 MG/325 MG FILM-COATED TABLETS (tramadol hydrochloride and paracetamol ) PL 30684/0222 UKPAR

West Midlands Centre for ADRs. Jeffrey Aronson. Robin Ferner. Side Effects of Drugs Annuals. Editor Meyler s Side Effects of Drugs

Comparisons of Retail Prices of Generic Prescription Drugs in Canada vs. United States: A Comprehensive Study

Transcription:

Bioequivalence and Bioequivalency Testing Joseph P. Balthasar 1 Department of Pharmaceutics and Pharmaceutical Chemistry, 421 Wakara Way, Suite 318, University of Utah, Salt Lake City UT 84108 PROLOGUE This lecture material is covered in one and one-half fiftyminute lecture periods. The primary objectives of the lecture are to: (i) review interpatient and intrapatient pharmacokinetic variability; (ii) introduce the concepts of therapeutic equivalence and bioequivalence; (iii) introduce the current FDA standards on bioequivalence; (iv) introduce the basic approach of bioequivalency testing; (v) provide an example of a clinical study assessing bioequivalence, showing calculations and conclusion regarding bioequivalency; and (vi) discuss the current FDA standards of bioequivalence as they relate to the approval of generic formulations of highly variable drugs and to the interchangeability of formulations of drugs with low therapeutic indices. The overall goal of the lecture is to introduce pharmacy students to bioequivalence and to bioequivalency testing, such that they may appropriately address the concerns of patients regarding generic substitution and appropriately advise physicians regarding the interchangeability of approved drug formulations. INTRODUCTION Generic drug formulations are often substituted for brand name (i.e., innovator) formulations by pharmacists in an effort to reduce the cost of prescription-drug therapy. In most states, generic substitution is allowed and encouraged, provided that the generic formulation is deemed to be therapeutically equivalent to the innovator formulation by the FDA. The FDA publishes a list of drug products and equivalents, which is entitled, Approved Drug Products with Therapeutic Equivalence Evaluations, and is commonly known as the Orange Book. The FDA s designation of therapeutic equivalence indicates that the generic formulation is (among other things) bioequivalent to the innovator 1 Assistant Professor. 194 American Journal of Pharmaceutical Education Vol. 63, Summer 1999

Fig. 1. Interpatient variability in propranolol pharmacokinetics following a single oral dose of 80mg to 5 healthy volunteers. Redrawn from Shand, D.G., Nuckolls, E.M., and Oates, J.A.: Plasma propranolol levels in adults, with observations in four children, Clin. Pharmacol. Then, 11, 112-120(1970). Reproduced with permission of Mosby, Inc. formulation and signifies the FDA s expectation that the formulations are likely to have equivalent clinical effect and no difference in their potential for adverse effects. (1) The 1984 Amendments to the Drug Price Competition and Patent Term Restoration Act require that manufacturers seeking approval of generic formulations submit to the FDA data demonstrating bioequivalence to the innovator drug product (1). Currently, bioequivalence is determined by assessing the equivalence of the rate and extent of drug absorption. Typically, this investigation is made through a 12-36 subject, two-treatment crossover study, conducted with healthy normal adult subjects. Under current FDA standards, bioequivalence is concluded in cases where the innovator and test product differ in terms of their rate and extent of absorption by -20/+25 percent or less. The FDA guidance, Statistical Procedures for Bioequivalence Studies Using a Standard Two-treatment Crossover Design, outlines a statistical method for assessing data and provides a decision rule for concluding bioequivalence(2). In spite of the efforts of the FDA, controversy and disagreement exist regarding the therapeutic equivalence of drug products listed in the Orange Book. Additionally, there is a seemingly continual debate in the pharmaceutical literature over the appropriateness of the FDA s recommended approach of testing the rate and extent of drug absorption(3-6). Perhaps in part as a consequence of these debates, and perhaps in part due to the publicity of the recent generic drug scandals, pharmacists are often queried by patients, health care professionals, and HMO representatives regarding the therapeutic equivalence of drug products. This lecture is designed to strengthen the students understanding of bioequivalence and bioequivalency testing, in an attempt to increase students confidence in addressing these questions. LECTURE FORMAT AND CONTENT Interpatient and Intrapatient Pharmacokinetic Variability Interpatient variability in pharmacokinetics is recognized by pharmacists and other health care professionals as an important factor to consider in evaluating and planning drug therapy. Fig. 2. Intrapatient variability in verapamil pharmacokinetics following single oral doses of verapamil tablets from the same lot on two separate occasions in two healthy volunteers. Redrawn from: Tsang, Y.C., Pop, R., Gordon, P., Hems, J. and Spino, M., High variability in drug pharmacokinetics complicates determination of bioequivalence: experience with verapamil, Pharm. Res., 13, 846-50(1996). Reproduced with permission from Plenum Publishing Co. In fact, much of the work in the field of clinical pharmacokinetics is devoted to attempts to understand and overcome this form of pharmacokinetic variability. Interpatient variability is often best demonstrated by examining the concentration vs. time profiles of a drug following the administration of a fixed dose to several patients. Between-patient differences in pharmacokinetics (i.e., the processes of drug absorption, distribution, metabolism and excretion) are expressed as differences in resultant concentration vs. time profiles (Figure 1)(7). Intrapatient pharmacokinetic variability refers to variabilities in pharmacokinetics that occur within a patient from dose to dose during the course of drug therapy (Figure 2)(8). In contrast to the mindfulness of practitioners regarding interpatient variability, intrapatient variability is virtually ignored in discussions of pharmacokinetics, outside of those in the bioequivalence literature. The relative lack of attention paid to intrapatient pharmacokinetic variability may be partially explained by the argument that drugs exhibiting high intrapatient pharmacokinetic variability rarely are drugs with low therapeutic indices, as this combination would not be likely found in an approved drug(9). This argument, which likely has merit, implies that intrapatient pharmacokinetic variability is rarely a factor in maintaining the safety and efficacy of drug therapy. However, intrapatient variability appears as a key factor in designing and assessing bioequivalence studies. American Journal of Pharmaceutical Education Vol. 63, Summer 1999 195

Fig. 3. Schematic representation of the standard two-treatment crossover study design commonly employed in bioequivalence trials. Therapeutic Equivalence Bioequivalency is evaluated by the FDA in ascertaining the therapeutic equivalence of pharmaceutical formulations. The definition of therapeutic equivalents and the criteria for determining therapeutic equivalence are listed in the Orange Book as follows: Therapeutic Equivalents. Drug products are considered to be therapeutic equivalents only if they are pharmaceutical equivalents and if they can be expected to have the same clinical effect and safety profile when administered to patients under the conditions specified in the labeling. The FDA classifies as therapeutically equivalent those products that meet the following general criteria: (i) they are approved as safe and effective; (ii) they are pharmaceutical equivalents in that they, (a) contain identical amounts of the same active drug ingredient in the same dosage form and route of administration, and (b) meet compendial or other applicable standards of strength, quality, purity, and identity; (iii) they are bioequivalent in that, (a) they do not present a known or potential bioequivalence problem, and they meet an acceptable in vitro standard, or (b) if they do present such a known or potential problem, they are shown to meet an appropriate bioequivalence standard; (iv) they are adequately labeled; and (v) they are manufactured in compliance with Current Good Manufacturing Practice regulations(1). It is important to note that the FDA designation of therapeutic equivalence does not consider several formulation characteristics, including packaging, scoring configuration, shape, or pharmaceutical excipients (e.g., colors, preservatives, flavors). These formulation characteristics may be important to the care of particular patients (e.g., patients allergic to certain pharmaceutical excipients). With this limitation, however, the FDA has the expectation that therapeutically equivalent products will produce the same clinical effect and toxicity profile of the prescribed product(1). Therapeutic Equivalence Codes The FDA provides a code to indicate the level of bioequivalency demonstrated for therapeutic equivalents. The AB code signifies that the formulation has been submitted with a study demonstrating bioequivalence. The main focus of this lecture is to instruct students in process and requirements needed to attain the AB rating. Bioequivalence Bioequivalence is defined to the class using the terms provided in section 505(j)(7)(B) of the Federal food, Drug, and Cosmetic Act: Table I. AUC and Cmax data for example bioequivalence study based on cyclosporine pharmacokinetics Test AUC Subject ng/ml x h Reference AUC ng/ml x h Test Cmax ng/ml Reference Cmax ng/ml 1 2560 4020 140 226 2 118 211 13.6 20.1 3 905 572 78.8 51.8 4 764 815 88.0 105 5 729 715 54.7 40.6 6 2460 1940 76.4 52.6 7 3180 1750 310 175 8 2740 2720 110 135 9 3260 4670 182 337 10 1220 1930 192 326 11 2340 2240 364 346 12 1050 1010 112 126 the rate and extent of absorption of the test drug do not show a significant difference from the rate and extent of absorption of the reference drug when administered at the same molar dose of the therapeutic ingredient under similar experimental conditions in either a single dose or multiple doses; or the extent of absorption of the test drug does not show a significant difference from the extent of absorption of the reference drug when administered at the same molar dose of the therapeutic ingredient under similar experimental conditions in either a single dose or multiple doses and the difference from the reference drug in the rate of absorption of the drug is intentional, is reflected in its proposed labeling, is not essential to the attainment of effective body drug concentrations on chronic use, and is considered medically insignificant for the drug. Metrics of the Rate and Extent of Drug Absorption Bioequivalence studies attempt to gain insight on formulation switchability (i.e., the ability to substitute one formulation for another without concern of the potential for reduced effectiveness or increased probability of adverse effects). A key assumption is that switchability may be inferred from plasma concentration vs. time data and metrics reflecting the rate and extent of drug absorption. The area under the plasma concentration vs. time curve (AUC) is commonly employed as the metric describing the extent of drug absorption, while the maximal concentration observed following drug administration (Cmax) is the metric recommended by the FDA to evaluate the rate of drug absorption(2). The AUC resultant from a single dose of a drug formulation is commonly assessed with the linear trapezoidal method(10): = i n( Ci + Ci 1 ) Cn AUC = Cdt ( ti t i 1) + Eq. 1. 2 λ 0 i = 1 Where C refers to the concentration of drug in plasma, t refers to the time of sample collection, and (z refers to the magnitude of the terminal In-linear slope of the declining concentration vs. time data. Cmax is determined by observation from the collected concentration vs. time data. 196 American Journal of Pharmaceutical Education Vol. 63, Summer 1999

Table II. Data transformations performed in analysis of the simulated cyclosporine bioequivalence study Subject lnauc test lnauc reference Difference test-reference lncmax test lncmax reference Difference test-reference 1 7.85 8.30-0.45 4.94 5.42-0.48 2 4.77 5.35-0.58 2.61 3.00-0.39 3 6.81 6.35 0.46 4.37 3.95 0.42 4 6.64 6.70-0.06 4.48 4.65-0.18 5 6.59 6.57 0.02 4.00 3.70 0.30 6 7.81 6.57 0.24 4.34 3.96 0.37 7 8.07 7.47 0.60 5.74 5.16 0.57 8 7.92 7.91 0.01 4.70 4.90-0.20 9 8.09 8.45-0.36 5.21 5.82-0.62 10 7.11 7.57-0.46 5.26 5.79-0.53 11 7.76 7.72 0.04 5.90 5.85 0.05 12 6.95 6.91 0.04 4.71 4.84-0.12 Mean -0.0427-0.0668 SD 0.368 0.405 Two-Treatment Crossover Study The typical study design employed in bioequivalence studies is the two-treatment, two-period, crossover design (Figure 3). In this study design, subjects are randomly separated into two groups of equal number. The innovator formulation is administered to group A in the first study period, and the test formulation is administered to group B in the first period. During the second study period, group A receives the test formulation and group B receives the innovator formulation. The first and second study periods are separated by a washout period, which is designed to be of sufficient duration to allow elimination of the drug administered in the first period. Subjects are separated into two groups to allow identification of period or sequence effects in the study results. Bioequivalence Study Example Data (AUC and Cmax) are presented from a simulated cyclosporine bioequivalence study with 12 subjects. Briefly, data were simulated with ADAPTII software(11) using a one compartment model and assuming the following values for pharmacokinetic parameters (mean ± interpatient SD): bioavailability 0.3±0.4, absorption rate constant 0.28h-l±0.13, clearance 53.8L/h±21.4, volume of distribution 301L±217. Intrapatient variances in model parameters were assumed as follows: bioavailability (0.2 x mean value) 2, absorption rate constant (0.23 x mean value) 2, volume of distribution and clearance (0.1 x mean values) 2. Data were simulated for a two-treatment, crossover study of 300mg of cyclosporine administered orally. These data were simulated assuming the same parameter values and variances for both study periods; consequently, there was no true difference between the formulations in terms of their rate and extent of absorption. However, these parameter values lead to moderately high values of intrapatient variability in AUC (percent coefficient of variation: 19.3 percent) and in Cmax (percent coefficient of variation: 23 percent). These intrapatient variabilities agree well with the experimental results of Kovarik et al. in their investigation of intrapatient variability in cyclosporine pharmacokinetics following administration of the Sandimmune oral formulation(12). Simulated AUC and Cmax data are presented in Table I. Analysis of Data for Bioequivalence Determination A step-by step approach is used to evaluate data for the determination of bioequivalence. 1. Calculation of the natural log of the AUC and Cmax data The FDA advocates logarithmically transforming AUC and Cmax data prior to analysis(2). Either log 10 or the natural logarithms of the data may be used. Natural logarithms are used in this example (Table II). 2. Calculation of the difference between the transformed data for each metric for each subject The transformed value of the reference AUC is subtracted from the transformed value of the test AUC for each patient. This procedure is repeated for the transformed Cmax values (Table II). 3. Calculation of the mean difference between the transformed data The mean difference exhibited in the transformed data are determined by summing the individual differences and dividing by the number of subjects in the study (e.g., ((lnauctest-lnaucreference)/number of subjects). The mean differences for the example data set are provided in Table II. 4. Calculation of the standard deviation of the difference between the transformed data Standard deviation calculation is performed through the use of a hand-held calculator by students in class. Results are shown in Table II. 5. Selection of the appropriate t-value T-values appropriate for the construction of confidence interval limits are dependent on the a-value of the statistical test and on the on the degrees of freedom of the test. For bioequivalence studies, α is always 0.05 and the degrees of freedom are determined as n-1, where n is the number of patients in the study. Common subject sample sizes and t- values for bioequivalence studies are as follows: 12-subjects: 1.7959, 18-subjects: 1.7396, 24-subjects: 1.7139. 6. Calculation of the high and low bounds of confidence intervals about the mean difference of the transformed data High and low confidence bounds are calculated as: HB = e ( MD + [ t-value x SD / n]) LB = e ( MD [ t-value x SD / n]) Where HB and LB refer to the high and low bound of the confidence interval; MD and SD refer to the mean difference and standard deviation of the transformed metric. For example, for the AUC data, the HB of the confidence interval may be calculated as: HB = e ( MD + [ t-value x SD / n]) = e (- 0.0427[1.7959 0.368/ 12]) = 1.16 The confidence intervals for AUC and Cmax are 0.79-1.16 and 0.76-1.15, respectively. American Journal of Pharmaceutical Education Vol. 63, Summer 1999 197

6. Decision rule Bioequivalence is concluded if the confidence intervals about the ratio of AUC and Cmax both fall within the range of 0.8-1.25. That is the lower bounds of the confidence intervals must be greater than or equal to 0.8 and the upper bounds of the confidence intervals must be less than or equal to 1.25. In the present example, bioequivalence is not concluded as the lower bound of each confidence interval is less than 0.8. Limitations of the Present Approach of Determining Bioequivalence Formulations of Highly Variable Drugs. Highly variable drugs have been defined as those demonstrating intrapatient variabilities greater than 30 percent in metrics of the extent and rate of absorption (e.g., AUC and Cmax)(9). Unfortunately, present FDA bioequivalence standards tend to penalize against generic formulations of drugs exhibiting high intrapatient variability, as this high variability often translates to wide confidence intervals about the mean difference in the bioavailability metrics (as demonstrated in the example above). For example, Tsang et al. have shown a failed bioequivalence determination from a small study assessing verapamil absorption from the same lot of sustained release tablets, administered on two occasions in a crossover study(8). In practical terms, large numbers of subjects are often required to produce confidence interval limits within the current guidelines. It has been argued that the current standards present an unfair obstacle to companies developing formulations of these drugs; however, the FDA is currently considering a draft guidance to address this concern(13). Additional Concerns. The intent of the FDA designation of therapeutic equivalence is to ensure the switchability of drug formulations. Unfortunately, the typical approach of assessing bioequivalence is through small, tightly controlled studies with normal healthy subjects. Commonly, subjects are not permitted to take other medications during the study period; additionally, several factors are often controlled. For example, subjects are often selected based on age, weight, and smoker-status. Moreover, variables such as the timing and content of meals, liquid intake, and the amount of physical activity allowed during the study period are often regulated. These controlled factors may differentially influence the rate and extent of absorption of compared formulations; consequently, the results of these studies may not be indicative of results obtained in less controlled situations (e.g., typical outpatient therapy). These concerns are particularly significant when one considers the switchability of formulations of drugs with low therapeutic indices. HOMEWORK ASSIGNMENT Ten 12-patient bioequivalence trials are simulated for five different levels of intrapatient variability (i.e., 10 trials with 0, 10, 20, 30, and 40 percent intrapatient variability in bioavailability and absorption rate) with a one-compartment population model, using ADAPT II software(11). The simulations are performed such that no true difference exists between the test and innovator formulation in the extent or rate of absorption. Each student is provided with data from a unique clinical trial and is required to determine if the results are indicative of bioequivalence. Results of the homework are reviewed in the next class. Students report the results of their study, which allows determination of the fraction of studies yielding the conclusion of bioequivalence under each level of intrapatient variability. The results (invariably) demonstrate that the fraction of studies showing bioequivalence decreases with increasing intrapatient variability; however, equally impressive is the intrapatient variability in AUC and Cmax shown in studies yielding the conclusion of bioequivalence. Overall, the homework exercise strengthens the students understanding of bioequivalence, and particularly the students understanding of the influence of intrapatient variability in bioequivalence studies. AUTHOR S NOTES It is the author s belief that the above homework assignment greatly enhances student understanding of the process and limitations of bioequivalency testing. In turn, the educational value of the homework assignment is believed to be enhanced through the distribution of individual data sets with specific values of intrapatient variability (and potentially specific true differences in the rate and extent of drug absorption). Data of this nature are unavailable in the literature, but may be generated with clinical trial simulation software (as described above). Please contact the author if you wish to obtain copies of recently used data sets or copies of executable files capable of performing the clinical trial simulations described. Am. J. Pharm. Educ., 63, 194-198(1999); received 2/1/99, accepted 4/15/99. References (1) Food and Drug Administration - Center for Drug Evaluation and Research, Approved drug products with therapeutic equivalence evaluations, [may be accessed from: http://www.fda.gov/cder/orange/adp.htm], (1998). (2) Food and Drug Administration - Office of Generic Drugs - Division of Bioequivalence, Guidance on statistical procedures for bioequivalence studies using a standard two-treatment crossover design [may be accessed from: http://www.fda.gov/cder/guidance/index.htm], (1992). (3) Schall, R. and Williams, R.L., Towards a practical strategy for assessing individual bioequivalence. Food and Drug Administration Individual Bioequivalence Working Group, J. Pharmacokinet. Biopharm., 24, 133-149(1996). (4) Patnaik, R.N., Lesko, L.J., Chen, M.L. and Williams, R.L., Individual bioequivalence. New concepts in the statistical assessment of bioequivalence metrics. FDA Individual Bioequivalence Working Group, Clin. Pharmacokinet., 33, 1-6(1997). (5) Basson, R.P., Cerimele, B.J., DeSante, K.A. and Howey, D.C., Tmax: an unconfounded metric for rate of absorption in single dose bioequivalence studies, Pharm. Res., 13, 324-8(1996). (6) Lacey, L.F., Keene, O.N., Duquesnoy, C. and Bye, A., Evaluation of different indirect measures of rate of drug absorption in comparative pharmacokinetic studies [see comments], J. Pharm. Sci., 83, 212-215(1994). (7) Shand, D.G., Nuckolls, E.M. and Oates, J.A., Plasma propranolol levels in adults with observations in four children, Clin. Pharmacol. Ther, 11, 112-120(1970). (8) Tsang, Y.C., Pop, R., Gordon, P., Hems, J. and Spino, M., High variability in drug pharmacokinetics complicates determination of bioequivalence: experience with verapamil, Pharm Res, 13, 846-850(1996). (9) Shah, V.P., Yacobi, A., Barr, W.H., Benet, L.Z., Breimer, D., Dobrinska, M.R., Endrenyi, L., Fairweather, W., Gillespie, W., Gonzalez, M.A., Hooper, J., Jackson, A., Lesko, L.J., Midha, K.K., Noonan, P.K., Patnaik, R. and Williams, R.L., Evaluation of orally administered highly variable drugs and drug formulations, Pharm Res, 13, 1590-1594(1996). (10) Gibaldi, M. and Perrier, D., Pharmacokinetics, 2nd ed., Marcel Dekker, New York NY (1982) (11) D Argenio, D.Z. and Schumitzky, A., ADAPT II User s Guide: Pharmacokinetic / Pharmacodynamic Systems Analysis Software, Release 4, Biomedical Simulations Resource, Los Angeles (1997). (12) Kovarik, J.M., Mueller, E.A., van Bree, J.B., Tetzloff, W. and Kutz, K., Reduced inter- and intraindividual variability in cyclosporine pharmacokinetics from a microemulsion formulation, J. Pharm. Sci., 83, 444-446(1994). (12) Food and Drug Administration - Center for Drug Evaluation and Research, In vivo bioequivalence studies based on population and individual bioequivalence approaches - Draft guidance [may be accessed from: http://www.fda.gov/cder/guidance/index.htm], (1997). 198 American Journal of Pharmaceutical Education Vol. 63, Summer 1999