You are on page 1of 12

Journal of Pharmaceutical Sciences xxx (2018) 1-12

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


journal homepage: www.jpharmsci.org

General Commentary

Past, Present, and Future of Bioequivalence: Improving Assessment


and Extrapolation of Therapeutic Equivalence for Oral Drug
Products
Rodrigo Cristofoletti 1, 2, Malcolm Rowland 3, Lawrence J. Lesko 4, Henning Blume 5,
Amin Rostami-Hodjegan 3, 6, Jennifer B. Dressman 2, *
1
Brazilian Health Surveillance Agency (ANVISA), Division of Therapeutic Equivalence, Brasilia, Brazil
2
Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
3
Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
4
Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, Florida 32827
5
SocraTec C&S, Oberursel, Germany
6
Certara, 1 Concourse Way, Sheffield, UK

a r t i c l e i n f o a b s t r a c t

Article history: The growth in the utilization of systems thinking principles has created a paradigm shift in the regulatory
Received 2 March 2018 sciences and drug product development. Instead of relying extensively on end product testing and one-
Revised 3 May 2018 size-fits-all regulatory criteria, this new paradigm has focused on building quality into the product by
Accepted 12 June 2018
design and fostering the development of product-specific, clinically relevant specifications. In this
context, this commentary describes the evolution of bioequivalence regulations up to the current day and
discusses the potential of applying a Bayesian-like approach, considering all relevant prior knowledge, to
Keywords:
bioequivalence guide regulatory bioequivalence decisions in a patient-centric environment.
pharmacokinetics © 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
regulatory science
dissolution
mechanistic modeling
pharmacokinetic/pharmacodynamic
(PK/PD) correlation

Introduction agencies across the globe to develop systematic toxicity testing


protocols.2 In the United States, for example, the Federal Food,
Historically, the development of the regulatory sciences has Drug, and Cosmetic Act was amended in 1962 by the so-called
been stimulated by public health catastrophes, as illustrated by the Kefauver-Harris Amendments, which required not only more
chain of events in the United States. Before 1938, one could intro- stringent toxicological testing but also evidence of efficacy for
duce a drug product in the United States market without any the product's intended use to support a new drug marketing
toxicity testing or demonstration of effectiveness. The deaths of 107 application (NDA), that is, premarket clinical trials.3,4 Because of a
children in September and October of 1937, who were poisoned by grandfather clause, no further studies were required for drugs that
an elixir of sulfonamide which contained ethylene glycol intended were already on the market before the 1938 law; however, efficacy
to solubilize the drug, symbolized the catastrophic flaw in the data for drugs that entered the U.S. market after 1938 became
caveat emptor approach.1 Almost immediately, in 1938, amend- mandatory.3,4 Some manufacturers contested the legality of the
ments to the Food and Drugs Act of 1906 requiring toxicity testing Kefauver-Harris Amendments, but the U.S. Supreme Court upheld
and safety studies before the introduction of any medicine into the the retroactive efficacy requirement.5 Owing to the lack of
marketplace were enacted.1,2 resources to evaluate the efficacy of the several thousand drug
The thalidomide tragedy, which originated in Europe in the products that had been approved between 1938 and 1962, the U.S.
early 1960s, marked a further turning point in regulatory supervi- Food and Drug Administration (FDA) recognized that a transitional
sion of the pharmaceutical market, as it prompted regulatory period would be necessary and elaborated an abbreviated pro-
cedure to handle generic drugs. According to this procedure, if the
* Correspondence to: Jennifer B. Dressman (Telephone: þ49 (0) 69 798 29680). regulatory agency already had evidence supporting the safety and
E-mail address: dressman@em.uni-frankfurt.de (J.B. Dressman). efficacy of a certain drug substance in the dossier of the respective

https://doi.org/10.1016/j.xphs.2018.06.013
0022-3549/© 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
2 R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

innovator drug product, generic manufacturers would not have to approval in general were made before the formation of the EU, with
submit clinical trials. Instead, generics were required to be proven the passage of EC Directive 65/65/EEC in 1965.22 After the forma-
equivalent to the respective brand name products to maintain their tion of the EU in 1993, there were significant changes, eventually
marketing authorizations.4-7 For the first time, the focus shifted leading to a centralized procedure for new drug applications at the
from the drug substance to the formulation. Nevertheless, at that European Medicines Agency (EMA) but continuing to allow
time, bioequivalence (BE) standards were in a state of flux and had approval of generic versions at a national level. Today, generic drug
not yet been established.4,5,8 approvals within the EU can be obtained in one of the 3 ways: a
The post-1962 scenario is even more complex. New clinical trials central approval which applies to the whole European market,
had to be conducted even for generics and, thus, the cost of stepwise approval at the national level (which can lead to approval
developing generics of post-1962 drugs increased dramatically.7 throughout the EU once 3 countries have approved the drug
This period was marked by intense legal battles among the regu- product, provided a certain mix of countries has been involved), or
latory authority, the innovative pharmaceutical companies, and the approval only at the national level, which can be instigated for
generic manufacturers. Worth mentioning is the Roche Products Inc. example when the sponsor decides that the product will only be
v. Bolar Pharmaceutical Co. case, in which the innovative company marketed in that given country.23
questioned whether the generic manufacturers could conduct Generics have played a role in many European countries for
testing on patented products solely for obtaining approval of many years, as in the United States. The first generic drug company
competitor products, and the United States v. Articles of Drug, appeal in Germany, Ratiopharm, was founded in 1973, and in that year,
of the Lannett Company, Inc. case, in which the generic manufacturer generic products, such as acetylsalicylic acid and paracetamol
contended that because the drug substances had already been tablets, were introduced into the market. Other companies soon
proven to be safe and effective by the innovative companies, clinical followed (e.g., Stada in 1975), and by 1997, generics already
trials should be superfluous for the amended new drug application. accounted for around 40% of prescriptions.24 This percentage has
Federal courts reached contrary decisions on these matters, continued to climb to over 80% over the ensuing 20 years.
increasing the uncertainty around what was required to bring a Again taking Germany as an example, the regulation of generics
generic product to market.5,7 Consequently, as many as 150 brand in Europe provides a variety of ways in which generic drug products
name drug products lacked generic versions, despite being can be introduced into the market. In Germany, there are still
off-patent, in the following years.7,9 so-called “Standard Rezeptur” products on the booksdand some of
Paralleling the judicial disputes, academic concerns about the these are on the market. Standard Rezeptur products must be
in vivo equivalence of generics arose after some clinical results were manufactured according to a predefined composition and, if this is
published showing that different formulations containing digoxin the case, no BE testing is required to achieve a marketing authori-
led to different pharmacokinetic (PK) profiles.10-12 For instance, zation.25 Furthermore, when a drug product has established itself
Lindenbaum et al.11 investigated the systemic exposure of digoxin over a number of years in the market and scientific material addi-
released from 4 different formulations in healthy volunteers. The tional to the approval package is available, a generic manufacturer
authors found pronounced differences in the bioavailability (BA) of is permitted to rely on this database. For example, glucocorticoids
different commercially available digoxin tablets. These differences and endocrine hormones were introduced into the market in the
in BA results, in combination with the well-known narrow thera- 1950s and 1960s, and since then, many scientific papers and clinical
peutic index of digoxin, caught the attention of the regulators at the studies have been published regarding their use. Against this
FDA. Subsequently, Wagner et al.,13 under contract with the FDA,4,8 background, it was deemed to be no longer necessary to perform
confirmed Lindenbaum's findings of lack of equivalence between clinical studies for the approval of generic versions. However, this
the plasma levels of digoxin released from multisource tablets. In type of waiver is only granted when the drug, excipients, and
fact, Wagner et al.13 reported nonequivalence in the systemic dosage form are all identical with products already on the market.25
exposure of digoxin even with tablets that met the pharmacopoeial Another interesting modality for generic drug approval in Germany
standards for both potency and disintegration time. Similar obser- is the literature-based approval, known as the “bibliographische
vations were also reported for other drug products, including those Zulassung” procedure. In this case, regulatory relief in the form of a
containing tetracycline,14,15 chloramphenicol,16 phenylbuta- waiver of preclinical and clinical data is entertained. Requirements
zone,17,18 and oxytetracycline.19 An editorial written by Levy and are that the drug must have already been on the market for 10 years
Gibaldi20 in 1974 reinforced the urgency of developing an adequate and that the sponsor must produce thorough literature evidence of
means of assuring the BA of oral drug products. Recognizing the efficacy combined with an acceptable side-effect profile. Both
existence of BA problems in marketed products, the FDA Office of positive and negative literature must be cited and discussed, with
Technology Assessment convened a panel of 10 senior medical peer-reviewed publication being accorded more weight than in-
consultants who concluded that the then current compendial ternal documents or nonepeer-reviewed articles.26 It should be
reference standards and regulatory practices did not assure uni- noted, however, that the usual procedure for approval of generic
form BA.21 products in Germany (and elsewhere in Europe) is to demonstrate
Finally, in 1984, almost 20 years after the Kefauver-Harris BE using PK studies in healthy human volunteers.
Amendments, the U.S. Congress passed the Drug Price Competi- Outside the United States and Europe, there are many different
tion and Patent Term Restoration Act (commonly known as the approaches to approval of generic drugs, ranging from very strin-
Hatch-Waxman Act), which was intended to make low-cost ge- gent requirements and a low % of prescriptions filled with generics
nerics more widely available while simultaneously maintaining in Japan (around 18% in 2007) to the situation in Iran, where,
adequate incentives for innovation, by, for example, extending because of a government decree shortly after the revolution, only
brand name market exclusivity. The Act authorized the FDA to generic versions were permitted to be marketed. Still today,
approve generic drug products through BE studies, fostering the approximately 96% of drugs available on the market are manufac-
development of multisource drug products and generic-based tured by local generic companies.27,28 Interestingly though, 86% of
public health policies worldwide.6,9 Iranians hold the perception that bioequivalent generics are
In Europe, concepts for generic drugs and BE evolved at the not therapeutically equivalent to the respective reference drug
national level, with each country forming its own rules. The first products,29 a statistic which has driven both the government and
efforts to standardize European regulations regarding drug generic manufacturers to sponsor prospective, blinded efficacy, and
R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 3

safety studies for head-to-head comparisons between approved Table 1


generics and branded versions.30,31 Historical Turning Points and Scientific Evolution of Regulations Concerned With BE

Year Major Events

1906 Food and Drugs Act


Initiation and Evolution of the BE Concept
1937 Deaths of 107 children who were poisoned by an elixir of
sulfonamide
The first U.S. legal definitions of BA and BE were stated in the 1938 Federal Food, Drug, and Cosmetic Act (FD&C)
Code of Federal Regulation 21CFR320.1. For systemically acting 1960s Thalidomide tragedy in Europe and further countries
drugs, BA was defined in the Act as “the rate and extent to which 1962 Drug Efficacy Amendment (also known as Kefauver-Harris
Amendments)
the active ingredient or active moiety is absorbed from a drug
1973 First generic company in Germany
product and becomes available at the site of action” and BE as “the 1974 Editorial on digoxin written by Gerhard Levy and Milo Gibaldi
absence of a significant difference in the rate and extent to which 1977 75/75 decision rule
the active ingredient or active moiety in pharmaceutical equiva- 1980s 80/20 rule or power approach
lents or pharmaceutical alternatives becomes available at the site of 1984 Drug Price Competition and Patent Term Restoration Act (also
known as Hatch-Waxman Act)
drug action when administered at the same molar dose under 1987 Two one-sided tests
similar conditions in an appropriately designed study.” This statu- 1990s Bio-International conferences on Bioavailability, Bioequivalence,
tory definition of BA is purposely different from the academic view, and Pharmacokinetic Studies, held biennially
which defined BA as the percentage of an administered dose that 1999 Regulatory initiatives on individual BE and population BE were
stopped
enters the systemic circulation in an unchanged form, being a
2000 BCS-based biowaiver introduced for generics
function of the fraction of the dose that is effectively absorbed into e Narrowing acceptance interval for NTIs
the enterocytes (Fabs) and of that, the fraction escaping first-pass e Reference-scaled average BE
elimination on passage through the gut wall (Fg) and liver (Fh) e Partial AUCs
before reaching the systemic blood supply32: e Flexible BE criteria for NDAsdfocus on the clinical aspect of BE
e Prioritizing the biopharmaceutical aspect of BE in case of ANDAs
2017 Applying model-informed drug development approaches for
BA ¼ Fabs  Fg  Fh generics
As defined, for example, in the Biowaiver Guidances,33,34 oral ANDA, abbreviated new drug application.
absorption refers to the movement of drugs across the mucosal
membranes of the gastrointestinal (GI) tract, and Fabs represents
the fraction of the administered drug that reaches the systemic Average BE
circulation as either a parent or metabolite. Thus, BA would equal
Fabs only in the absence of presystemic metabolism.32 Nevertheless, The first acceptance limits for declaring equivalence were
by using measures of both the rate and extent of absorption to established in the early 1970s.35 A group of medical experts in
define BA, the regulatory authorities emphasize the importance of consultation with the FDA reached a consensus that only differ-
assessing the drug product performance in vivo. ences of greater than 20% for the PK metrics mean peak (Cmax) and
By focusing on the “availability at the site of drug action,” reg- extent of exposure (AUC0-t) would be clinically significant for drug
ulators pointed out their concern about ensuring therapeutic products,36 and this criterion has since then been applied across the
equivalence. From a clinical perspective, the metrics used to assess board. At the time, some forceful arguments against using the same
BA and BE should be surrogates of drug efficacy and safety. In other PK metrics and setting the same, essentially arbitrary, acceptance
words, the BE metrics should bridge efficacy and safety data be- limits for every drug were voiced.12,37 Nevertheless, these voices
tween compared formulations by showing that variations in BA, did not prevaildlikely because there was limited knowledge of PK/
which fall within an acceptable range, do not alter the clinical pharmacodynamic (PD) relationships for many drugs, making it
outcome. difficult for regulatory authorities to define drug-specific BE re-
Indeed, the careful choice of the words used in the statutory quirements and because there was pressure on the regulatory au-
definitions of BA and BE highlights the 2 fundamental regulatory thorities to increase the number of approved generics to support
aspects of the BE concept: biopharmaceutical and clinical. Inter- the new policy. A consensus around the new paradigm for the BE
estingly, the legislators did not specify which metrics should be field, that is, the “± 20%” rule, was achieved within a shorter time
used to assess the rate and extent of drug absorption or what is frame than would be usual in other scientific fields.38 The concerns
meant by “significant differences,” delegating this responsibility to expressed by, for example, Levy12 about setting the same arbitrary
the regulatory authorities. The statutory definitions, thus, afford a acceptance limit for every drug, regardless of where its usual dosing
higher level of flexibility than has generally been practiced by lies on the sigmoidal exposure-response (E-R) relationship, were
regulatory authorities, not only in the United States but also in unable to convince either the FDA or the majority of the scientific
other jurisdictions. community at the time. Some years later, even though he had
Retrospectively, with a view to the delineation of drug/formu- demonstrated that the delay between the time course of plasma
lation parameters from systems parameters, it is apparent that the concentration and drug effect should be taken into account to guide
regulatory definitions of BA and BE do not stipulate the conditions the decision about which exposure metrics would be clinically
of the system under which BA and BE are to be assessed. This relevant, Sheiner37 also failed to succeed in reopening the debate
omission implies that once BA or BE is established in one group of on this field.
individuals or under one set of conditions, it will be valid regardless In contrast to the steadfast adhesion to the equivalence criteria
of any changes to the systems parameters. Based on a literal (“± 20%” rule), the statistical methods used to establish BE after
interpretation of the aforementioned rules, one would postulate administering different formulations have evolved substantially
that BA and BE may be extrapolated from healthy young adults over the last decades. In 1977, the 75/75 decision rule was proposed
(who are typically used in such studies) to, for example, pediatric as an addition to the requirement that there should be a difference
and elderly patients without any adjustments. of no more than 20% in the mean AUC and Cmax between the test (T)
The scientific evolution of regulations concerned with BE is and reference (R) formulations. This rule was proposed with the
summarized in the following text and in Table 1. aim of accounting for individual variability in BE. According to the
4 R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

75/75 rule, 2 formulations were to be deemed bioequivalent pro- Population and Individual BE
vided that the individual subject ratios (i.e., T/R for AUC0-t and Cmax)
exceeded 0.75 in at least 75% of the subjects studied, in addition to Already in the late 1970s, some authors had been expressing
the average comparison. Recognizing that this decision rule would concerns about the suitability of average BE to address
not ensure similarity in the safety profiles between the compared genericebrand name drug product interchangeability, especially in
formulations, it was further amended with the addition of an upper patients who had already been started on treatments using the R
bound on the ratios as well, that is, 0.75-1.25.39 However, this rule formulation. In particular, they were concerned that the average BE
garnered widespread criticism in the literature because of its high approach focuses only on the comparison of population averages of
sensitivity for drugs showing large intrasubject or intersubject a BE measure of interest and not on the variances of the measure for
variability, and it was later abandoned.40,41 In the early 1980s, a T and R products.49
statistical method for assessing BE between formulations based on To address the issue of interchangeability, 2 new concepts were
conventional hypothesis testing (i.e., null hypothesis of no differ- proposed, population BE and individual BE. Extensive reviews of
ence between the 2 compared averages; H00 : mT  mR ¼ 0) was individual and population BE, including the respective statistical
proposed. If the null hypothesis was not rejected, it was still methods, have been provided elsewhere.50-53 The population and
necessary to demonstrate that the study was large enough to individual BE approaches aim at addressing 2 different aspects of
provide at least 80% chance of correctly detecting a 20% difference the interchangeability concept: prescribability and switchability.
of the R average; the so-called “80/20” rule or “power” approach.42 Drug prescribability, which is assessed by population BE studies,
However, it became apparent that simple conventional null hy- refers to the clinical setting in which a physician prescribes a drug
pothesis testing the equality of 2 formulations was inadequate for product to a drug-naive patient. In this setting, the prescriber relies
BE purposes.43 First, because the main goal of BE studies is not to on an understanding that the average performance of the generic
assess whether there is a statistically significant difference between has been well characterized and a comparative PK study has
T and R formulations but whether the difference is clinically rele- bridged the safety and efficacy information from innovator's clin-
vant or not. Second, because using the renowned “lady tasting tea ical trials, that is, generic or brand name formulation can be chosen
experiment,” Ronald Fisher had already demonstrated that “the to start the treatment. Besides comparing population averages, the
null hypothesis is never proved or established, but is possibly dis- population BE approach assesses the total variability of the measure
proved, in the course of experimentation. Every experiment may be in the population. Although population BE is an improvement over
said to exist only in order to give the facts a chance of disproving average BE, it is still not sufficient to ensure that a given individual
the null hypothesis.”44 Consequently, if one aims to demonstrate will respond similarly to the 2 formulations.54-56
equivalence between T and R formulations, the “equivalence Drug switchability refers to the setting in which a practitioner
hypothesis” should be the alternative rather than the null transfers a patient from a brand name drug product to a generic
hypothesis.42,44,45 formulation. Individual BE testing, that is, establishing BE on an
In 1987, Schuirmann attempted to resolve the issues of hy- individual subject basis is needed for switchability, especially in
pothesis testing by recommending that statistical analysis of Cmax those instances in which the within-subject variability of the
and AUC0-t be based on a 2 one-sided test procedures.42 Briefly, the generic product is greater than the brand name product55 or in
alternative hypothesis was redefined as a range of values with an which the patient is switched from one generic product to another.
equivalent effect after assuming a clinically significant difference of The advantages and challenges of using the individual BE approach
±20% and a ¼ 0.05. The first condition tests whether the mean peak were intensely debated throughout the 1990s.36 Finally, in 1999,
and extent of drug exposure after administering the T are signifi- the FDA decided to stop the regulatory initiative related to indi-
cantly lower than the mean peak and extent of drug exposure after vidual BE testing because of the practical, economic, and statistical
the R formulation, while the second condition tests the opposite challenges in its measurement and interpretation.51,57,58 Moreover,
scenario, that is, whether the mean Cmax and AUC0-t after admin- refuting the claim that approved generics might exhibit non-BE
istering the R are significantly lower than the Cmax and AUC0-t after results when retested in special patient subgroups, the FDA Indi-
the T formulation. This analysis has the benefit of yielding the same vidual Bioequivalence Expert Panel concluded that “at this time,
outcome regardless of which product is designated as T and which individual bioequivalence still remains a theoretical solution to
as R. Because we conventionally note BE intervals as T/R, the second solve a theoretical clinical problem. We have no evidence that we
BE limit is 1.25 (i.e., reciprocal of 0.8), illustrating the rationale have a clinical problem, either a safety or an efficacy issue, and we
behind the 80%-125% confidence interval criteria. This approach is have no evidence that if we have the problem that individual bio-
termed average BE.42 equivalence will solve the problem” and “switchability is not a
problem for approved generics.”58 Thus, the average BE approach
remained the key method for the evaluation of therapeutic equiv-
Development of BE Requirements During the 1990s alence and interchangeability. The conclusions reached by the
expert panel are examined in detail in the section on group-by-
While the United States was the driving force for the devel- formulation interactions. Hereinafter, the term BE will refer to
opment of the BE concept in the 1980s and early 1990s, Europe average BE unless otherwise stated.
and Japan started contributing significantly from the mid-1990s.
In fact, many developments in the BE field described in the Measures of Exposure Versus Measures of Rate and Extent of
following sections of this article (e.g., measures of rate and extent Absorption
of absorption vs. measures of exposure, individual and population
BE, optimizing study design to increase sensitivity to detect Because assessing the active moiety kinetics at the site(s) of
formulation differences, and so forth) were initiated and debated action is generally not possible and direct measurements in the GI
during the Bio-International conferences that were held bienni- tract to quantify the rate and extent of drug absorption are rather
ally in Europe, Japan, or North America and co-sponsored by the laborious,59 exposure metrics derived from the plasma
FIP (International Pharmaceutical Federation), the FDA, and concentration-time curve are generally used, under the assumption
several other regulatory authorities and pharmaceutical organi- that the substance in the general circulation is in exchange with the
zations.46-48 substance at the effect site.60-62 In this context, peak (Cmax) and
R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 5

extent of exposure (AUC) are by far the most common metrics used intraindividual and interindividual variability in gastric emptying,
to assess BE. Although AUC is also a reliable metric for evaluating transporters, and presystemic metabolism, as well as in post-
extent of absorption,63 finding a unanimously acceptable measure absorptive processes such as distribution and clearance.74
or parameter for describing the absorption rate has been the clay With respect to PK-based determination of BE, both the FDA and
foot of BE. Cmax is not a pure measure of absorption rate as it (a) is the EMA, along with several other industrial countries, have pub-
partially dependent on the extent of drug absorption; (b) is lished guidelines containing recommendations to carry out BE
generally insensitive to changes in ka, especially when ka > kel; (c) studies for specific products. While deviations from the standard
contains little mechanistic information about the drug absorption one-size-fits-all approach within the European jurisdiction are
process, and (d) cannot discern differences in Tmax or lag-time limited to formulations containing narrow therapeutic index (NTI)
between formulations.61,63-70 For these reasons, some authors drugs (by narrowing the acceptance interval) and highly variable
have argued that the regulatory authorities should concentrate on drugs (HVDs; by widening the acceptance interval), requirements
the clinical relevance of the plasma PK profile by focusing on the to additionally assess partial AUC (e.g., various modified-release
systemic exposure concept. In this context, Cmax is deemed a useful formulations), compare within-subject variability (e.g., warfarin,
metric, not because it is an accurate measure of absorption rate but rivaroxaban, and dabigatran) and even assess subject-by-
because it is a useful measure of the peak of systemic exposure, a formulation interaction variance (e.g., methylphenidate) are not
metric which has clinical relevance in terms of safety and efficacy of uncommon within the US jurisdiction. Moreover, PD end points
the drug product with respect to, for example, maximum thera- have been introduced for specific drug products along with tailored
peutic response, adverse events, and dose-dumping.61,71 This line of acceptance criteria, for example, evaluation of the forced expiratory
thinking has subsequently prevailed, and modern BE guidelines volume in one second in the case of metered dose inhalers. These
have incorporated exposure concepts, to better focus on the clinical product-specific amendments to the standard BE approach aim at
aspects of BE.33,34 increasing the sensitivity of the in vivo test to detect formulation
differences and ensure therapeutic equivalence. In the following
Optimizing Study Design to Increase Sensitivity to Detect sections, some detailed examples of additions/amendments to the
Formulation Differences usual BE requirements are provided.

The FDA and the EMA, working together with prominent Partial AUC Approaches When ABE Metrics Are Insufficient to
pharmacokineticists, took the lead toward setting the most sensi- Compare Drug Products
tive study conditions to detect formulation differences. For
example, it was recognized that single-dose rather than multiple- The regulatory approach of using systemic exposure metrics to
dose studies are generally better able to detect formulation ef- indirectly assess the luminal behavior of products is not straight-
fects. For the same reason, it was contended that BE studies should forward because Cmax is not a pure measure of absorption rate and
be conducted under fasting conditions and that the parent drug, systemic variability (i.e., drug related rather than formulation
rather than metabolites, should be quantified whenever possible. related) also plays a major role in systemic drug exposure.61,71,74,75
Furthermore, to reduce variability that does not arise from product- In fact, concomitant plasma and GI fluid sampling in healthy adults
related differences, it was decided that BE studies should be per- has revealed a lack of correlation between intraluminal and sys-
formed in healthy volunteers unless the drug is associated with temic Cmax and AUC for immediate-release formulations containing
safety concerns that render studies in healthy volunteers unethical. spironolactone, fosamprenavir, and simvastatin.76-78 Furthermore,
According to this paradigm, if the T and R drug products were able modified-release formulations containing budesonide, mesal-
to show similar in vivo dissolution and absorption in healthy adults, amine, zolpidem, nifedipine, and methylphenidate illustrate that
assessed indirectly by means of comparing systemic exposure satisfying the current BE criteria in terms of Cmax and AUC0-t does
surrogates, it can be considered highly likely that both formulations not necessarily ensure similarity in the drug release characteristics;
would behave similarly in all target patient groups and clinical these are better addressed by using partial AUC metrics derived
scenarios. The aforementioned requirements for the study design from the initial or later parts of the respective plasma profiles.79-86
have been integrated in modern BE guidelines, focusing on the Similarly, consideration of early exposure may be useful when a
biopharmaceutical aspect of BE.33,34 rapid input is important to achieve an optimal efficacy profile, for
example, an analgesic effect.61,67,87
BE in the New Millenium
Group-by-Formulation InteractionsdRisks Associated With
A major innovation in BE that was introduced in the early 2000s Extrapolating BE Results From Healthy Adults to Special
is the possibility of waiving in vivo BE studies for certain generic Populations
drug/drug product combinations. This is known as the BCS-based
biowaiver procedure, where BCS is the Biopharmaceutics Classifi- Group-by-formulation effects have been reported for several
cation System originated from the study by Amidon et al.72 The drugs in the literature. While on the one hand recent research
underlying assumption of the BCS-based BE procedure is that if 2 sponsored by the FDA demonstrated BE between a previously
drug products dissolve similarly under in vitro conditions repre- approved generic lamotrigine and the brand name product, Lam-
sentative of the GI tract, they should result in indistinguishable ictal®, in “generic-brittle” patients with epilepsy who were already
plasma profiles and thus be bioequivalent with each other. This taking lamotrigine,88 several clinical examples of subgroup-by-
approach has been published in guidances from the FDA, EMA, and formulation interactions have also been reported, for example, for
WHO and is currently undergoing discussions at the International products containing methylphenidate, verapamil, or prasugrel.
Committee on Harmonization.33,34,73 Importantly, in vitro studies Meyer et al.89 found that a T product containing methylpheni-
assess product performance more directly than do conventional date was more variable than the respective R formulation and their
PK- or PD-based BE studies because in vitro studies focus on results further indicated a borderline subject-by-formulation
comparative drug release from T and R formulations, while in vivo interaction, in line with the clinical observation that some
BE testing can be influenced by confounding factors because of its patients manifested a more rapid onset of action and a shorter
indirect approach. Examples of confounding factors include duration of drug response after taking the T formulation. As a
6 R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

result, the FDA recommended that BE between formulations surgery patients and so forth). Indeed, virtual BE has already been
containing methylphenidate be based on Cmax, partial AUCs used by the Office of Generic Drugs at the FDA to set clinically
and AUC0-∞ under fasting and fed state conditions. Similarly, relevant specifications on the maximum amount of prasugrel free
to ensure the switchability between Concerta® and generic base (i.e., 20%) in hydrochloride salt products to ensure BE of ge-
products, a subject-by-formulation test for each PK metric is now nerics in subjects that may be taking PPIs.101 By taking the lead in
recommended. The specific instructions in the Concerta® guideline this area, regulatory bodies, such as the FDA, will no doubt generate
are illustrative of the trend to deviations from the traditional even more interest in the use of modeling and simulation for
one-size-fits-all criteria in applying average BE,86 noting that an assessing potential group-by-formulation interactions.
evaluation of the robustness of this particular guidance is outside
the scope of this commentary. Prescribable but Not Switchable?dBX Drug Products and
Carter et al.90 reported an age-based subject-by-formulation Hybrid Applications
interaction for an already approved generic verapamil immediate-
release dosage form. For one of the T formulations, the authors Theoretically, a new drug product that has failed to demonstrate
observed average AUC0-t and Cmax values that were 43% and 77% BE to the innovator drug product can still be approved in the United
higher, respectively, in elderly than in young subjects, while these States (i.e., when coded as BX, meaning that the data reviewed by
values remained similar between the R and another T formulation in the FDA are insufficient to determine that there is therapeutic
both subgroups. In yet another case, the systemic exposure after the equivalence to the R formulation) or in Europe (i.e., hybrid appli-
administration of 2 different products containing a calcium channel cation based on article 10(3) of Directive 2001/83/EC), provided
blocking agent was superimposable in male subjects but divergent claims of noninferior safety and efficacy profiles can be adequately
with respect to peak and extent of exposure in female subjects, supported. Examples include the applications of Absorica® (iso-
suggesting a gender-based subject-by-formulation interaction.91 tretinoin, NDA 021951), Triglide® (fenofibrate, NDA 021350), and
Mechanistically, subject- or group-by-formulation interactions Sumavel® DosePro® (sumatriptan succinate, NDA 022239). All 3
can be viewed as a manifestation of a unique interaction among drug products failed to show BE to the respective R formulation
characteristics of the anatomy, physiology, or biochemistry of the GI under at least one of the required conditions: for Absorica®, it was
tract (e.g., related to GI pH, gastric emptying rate, residence time in the fasted state PK; for Triglide®, it was the fed state PK; and for
the GI tract, and presystemic drug metabolism), or even lifestyle of Sumavel® DosePro®, it was when the injection site was the deltoid
a specific patient/subpopulation, and the product dissolution muscle. Marketing authorizations in the US were granted for these
in vivo. In fact, some intrinsic or extrinsic factors that affect gastric products, albeit coded as BX, based on favorable results in clinical
emptying and gastric pH have been reported to cause formulation- trials, or, subsequent to an adjustment to the SmPC (“a suboptimal
dependent absorption, an effect that may not be observed in dose may be delivered when Sumavel® DosePro® is administered to
traditional BE studies enrolling healthy young adults.92-98 For the arm, and therefore, the arm is not recommended as a site of
example, Seiler et al.99 compared the systemic exposure of 2 administration”).102
different formulations containing prasugrel, a poorly soluble free In the United States, some BX-rated products can be converted
base (F1), and its hydrochloride salt (F2) in healthy adults with and in AB-rated drug products (e.g., DiaBeta, glyburide 5 mg, NDA
without proton pump inhibitor (PPI) pretreatment. Under normal 017532) and then become new R drug products (i.e., ABX, where
gastric acid secretion, the Cmax and AUC0-t ratios (F2/F1) were 1.2 X ¼ 1, 2, 3…), to avoid shielding the former BX drug products from
and 1.1, respectively, whereas in PPI-pretreated volunteers, the direct generic competition.103,104 By contrast, in the EU, the current
ratios were 2.2 and 1.4, respectively. The clinical relevance of these legislation forbids the utilization of “hybrid” drug products as the R
findings persuaded the FDA to sponsor a project on the evaluation product in subsequent BE studies to bridge safety and efficacy to
of formulation dependence of drug interaction with PPIs, awarded subsequent generic formulations.105 Examples include supra-
to Biopharma Services USA, Inc. in 2016.100 bioavailable products which have been approved and whose
These examples suggest that the more heterogeneous the dosage recommendations have been supported by clinical studies
sampled population of volunteers enrolled in fully replicated BE or, alternatively, “hybrid” drug products which show a reduced food
studies, the higher the chances of detecting subject- or group-by- effect.106 In summary, some drug products have been shown to be
formulation interactions. Bringing matters to a further level of safe and efficacious even though they were not pharmacokineti-
complexity, one should consider the possibility that effects from a cally equivalent to the respective innovator's product under all
combination of factors could lead to different BE results. For circumstances.
instance, while 2 formulations may be BE in healthy adults and in
elderly patients of Caucasian origin, this may not be the case in Can PD Information and Intrasubject Variability Be Used to
Japanese subjects, where older patients are known to have a higher Set Clinically Relevant Acceptance Intervals for PK End Points?
incidence of achlorhydria.98
The statement made by the panel of experts in 1999 that During a meeting in 2004 of the Advisory Committee for Phar-
switchability is not a problem for approved drug products (see maceutical Science at the FDA, Prof. Les Benet declared the then
section on population and individual BE) is clearly challenged by current BE guideline to be Procrustean.107 In Greek mythology,
the aforementioned examples. However, as permutations of Procrustes offered hospitality to travelers, who were invited to rest
various conditions where a “formulation-by-condition” effect on his special bed. Nevertheless, if the travelers were too tall or too
might manifest itself are almost endless, it would be highly short to perfectly fit the bed, Procrustes would cut off their feet or
impractical to recruit the whole range of possible patient subsets to stretch them out. This is indeed a very good picture for the one-
assess product performance over the entire range of physiologically size-fits-all criteria of BE guidelines. Benet emphasized that “the
relevant conditions. An attractive and efficient alternative would be BE requirement for each drug should be established based on
to assess these permutations in relevant in vitro environments,57 known measures of intrasubject variability and the PKPD rela-
with subsequent incorporation of the data into in silico models tionship.”107,108 With the latter, it was highlighted that, in contrast
(i.e., in vitro to in vivo extrapolation using physiologically relevant to the prevailing assumption that if 2 drug products have the same
PK models to investigate drug absorption in different virtual pop- PK this will result in the same PD, products with different PK can
ulations (e.g., children, elderly, hypochlorhydric patients, bariatric also result in the same PD (depending where the administered dose
R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 7

is on the E-R curve), or alternatively, products with the same sys- scaled as a function of the R drug product variability, to keep the
temic exposure can produce different PD behavior (e.g., because of sample size needed for in vivo studies within a range that would be
formulation-dependent target site distribution). feasible for the sponsors of the generic application to conduct.118
Indeed, using PKPD information to set clinically relevant PK Under the assumption that an approved HVD must have a wide
limits would work analogously to replacing the classical toxico- therapeutic index, as otherwise there would have been significant
logical approach to estimate the “no observed adverse effect level” safety issues and lack of efficacy during phase III trials, the FDA
by the benchmark dose approach, which involves fitting a mathe- focuses only on the within-subject variability of the R formulation
matical model to all the dose-response data within a study, and to apply the RSABE approach to Cmax and AUC.120
thus enabling more biological information to be incorporated in the On the other hand, the EMA only allows the use of the RSABE
resulting estimates of guidance values (e.g., acceptable daily method to widen Cmax acceptance range for HVD if there are no
intakes).109 clinically relevant implications, that is, decisions are made on a
Perhaps, the most emblematic initiative in the field of BE is case-by-case basis.33,106 For example, even though the EMA Phar-
the recognition of the necessity to integrate predictive dissolution, macokinetics Working Party has acknowledged there are “some
physiologically relevant PK and PKPD models for decision-making data suggesting the existence of a plateau response in the inhibition
about generic drug BE standards, chosen as a priority for the fiscal of platelet aggregation” for clopidogrel, which is a HVD, they
year 2018 based on inputs from the FDA, industry, and other concluded that “it is currently not entirely clear what would be the
stakeholders.110 Indeed, this integrative approach has already been influence of variable clopidogrel concentrations on PD” and thus
applied to demonstrate that 2 pharmacokinetically inequivalent “the widening of 90% confidence intervals for Cmax is not recom-
formulations containing ibuprofen can nevertheless be therapeu- mended.”106 Extensive reviews on the statistical methods behind
tically equivalent.97,111 On the flip side, given the steep E-R rela- different RSABE approaches are provided elsewhere.118,121 It is also
tionship for efficacy end points of dabigatran and rivaroxaban, the noted that the specifics of the RSABE method applied vary between
FDA took a more pragmatic approach and denied widening the regulatory agencies and are in need of harmonization.
acceptance interval, although both are HVDs.112-115
What Approach Should Be Taken When the Drug Is Dosed High on
Narrowing the Acceptance Interval Because of Considerations of the E-R Relationship?
Safety
Some authors have reported that it is not uncommon to have
As the overall intrasubject variability is usually small for NTIs, phase III trials carried out using doses near to the maximum
generic formulations of them can be approved in BE studies even if tolerated doses derived from phase I trials to maximize the chances
the mean differences between T and R formulations are close to the of demonstrating drug effectiveness.122,123 Consequently, it is not
acceptance limits, leading to concerns about the transitivity of BE surprising to have approved therapeutic doses at or near the
testing (i.e., potential to drift to nonequivalence among generic plateau phase of E-RE-R curves, where the law of “diminishing
productsdthe NTI paradox).116 An additional concern arises from returns” plays a major role, for example, if the exposure is already
the overlap between the exposure needed for efficacy and that greater than the concentration producing 80% of drug effect (EC80),
leading to unacceptable toxicity, so that a minor change in exposure a 90-fold change in PK will result in only a 9% change in effect.124 In
could lead to the patient experiencing unacceptable adverse drug this scenario, using the traditional PK-based BE limits (80%-125%) to
reactions. To address these issues, regulators have amended the 80/ indirectly assess therapeutic equivalence isdat first glanceda
125 rule by setting product-specific criteria. Mostly, this has rather conservative approach. However, widening the PK-based BE
resulted in a tightening of the BE acceptance interval: for NTIs to limits using prior knowledge on E-R relationship may come with a
90%-111% (or similar limits) in Europe (for AUC and also for Cmax if caveat: focusing on E-R curve for just one biomarker or surrogate
the latter is of importance for safety, efficacy, or drug level moni- may not ensure similar efficacy and safety in all indications. For
toring); in Canada (only for AUC); and in Japan (for AUC and Cmax). example, fluctuations in PK which do not change drug efficacy may
According to the deductivism of Popper,117 the BE paradigm should still trigger safety concerns. Two different formulations, immedi-
have been challenged in a more fundamental way by the NTI case; ate- and modified-release products containing tacrolimus, an NTI
instead, a “quick fix” approach has been applied to maintain the BE drug, were equivalent in terms of effectiveness in Caucasians but
status quo. turned out to be inequivalent with regard to safety in African-
In contrast to other regulatory authorities, the FDA uses a American population.125 Therefore, care should be exercised,
reference-scaled average BE (RSABE) approach to scale down the BE especially in case of drugs prone to unacceptable adverse effects.
limits for Cmax and AUC to the within-subject variability of the R On the other hand, for well-characterized, wide therapeutic
product (but only if it is <20%); in other cases, the 80/125 rule index drugs, the available PKPD information may enable identifi-
should be applied.118 Even though such initiatives are welcome, cation of the most sensitive biomarker or surrogate end point to PK
there are still some remaining drawbacks: (a) the lack of worldwide changes (e.g., steepest E-R curve). In this case, the E-R curve could
harmonization,119 (b) the lack of a precise and broadly applicable be used to calibrate the PK-based BE limits (i.e., widening or nar-
definition of NTI; and (c) the lack of consistent utilization of rowing the acceptance limits), allowing generalization of PK-based
available drug-specific PD/toxicity information, including PD vari- decisions on therapeutic equivalence and thus covering all in-
ability, to set clinically relevant PK limits. dications. Such a scenario could be applied to some nonsteroidal
anti-inflammatory drugs whose EC50 for different clinical outcomes
Widening the Acceptance Interval Because of Consideration of is well established, for example, if the EC50 for pain relief > EC50 for
Variability antipyresis, when a wider PK fluctuation is not clinically relevant in
terms of pain relief it will not be relevant in terms of antipyresis
In the other direction, the original BE paradigm has also been either.
extended to accommodate HVDs, that is, those with an intrasubject In this context, the language to modernize the BE approach, at
coefficient of variation >30% (a cutoff value which was set by the least for new drug applications (NDAs), is already in place within
participants during the Bio-International '92 conference held in Bad the U.S. jurisdiction. For example, the FDA emphasizes that
Homburg, Germany).47 The amplitude of the BE goalposts was “exposure-response data can also support a new formulation that is
8 R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

unintentionally pharmacokinetically different from the formula- tissue distribution of methicillin in rabbits.134-136 In other words,
tion used in the clinical trials… Rather than reformulating the in case of a saturable cellular uptake process, nonequivalent
product or repeating the bioequivalence study, a sponsor may be systemic exposure may still be translated into similar target site
able to support the view that use of a wider confidence interval or distribution and hence therapeutic effectiveness.
accepting a real difference in bioavailability or exposure would not Stepping away from oral dosage forms for a moment,
lead to a therapeutic difference.”126 Recently, the FDA reinforced its formulation-dependent target site distribution has already been
position in a specific draft guidance on BA and BE for NDAs, reported in both preclinical and clinical situations for parenteral
encouraging sponsors to “demonstrate that the differences in rate products. For example, 2 pegylated liposomal doxorubicin formu-
and extent of absorption do not significantly affect the safety and lations showed similar plasma PK profiles (in terms of total drug
efficacy based on available dose-response or concentration- concentration) but distinctly different tissue distribution in mice.137
response data.”120 Similarly, the liver-to-plasma AUC ratio was about 2-fold higher in
An example illustrating the application of this policy by the FDA mice after administering a paclitaxel formulation made without
is aliskiren. Quoting from the Web site ipsis litteris: “In the bio- polyoxyethylated castor oil (Abraxane®) than with poly-
equivalence study (CSAH100A2102), the 90% CI for Cmax of aliskiren oxyethylated castor oil (Taxol®).138 Translating these results to
(0.76,0.93) was not contained within the predetermined BE limits humans, it has been shown that polyoxyethylated castor oil causes
of 0.8 e 1.25. However, this is not clinically significant, because of a a profound alteration of paclitaxel accumulation in erythrocytes by
shallow exposure-response relationship.” Also noteworthy is that, reducing the free drug fraction available for cellular partitioning,
despite the massive negative food effect observed for aliskiren (i.e., due to drug trapping in micelle-like structures. The authors
mean AUC and Cmax are decreased by 71% and 85%, respectively), concluded that changing the primary drug carrier in the systemic
the drug can be taken regardless of food intake, clearly suggesting circulation from the erythrocytes to micellar structures has a
that the therapeutic dose is on the plateau of the E-R curve.127,128 It substantial impact on how paclitaxel PK in humans should be
is interesting to contemplate what ramifications there would have interpreted.139 Consequently, quantification of total drug concen-
been, had the food effect been to increase Cmax and AUC to the same tration in plasma was deemed to be inadequate for assessing
extent. In such a case, one would also have to consider the safety therapeutic equivalence. Instead, in cases where paclitaxel is
aspects. Even if efficacy were to remain the same (i.e., dosing on the administered parenterally as a complex drug delivery system, the
plateau of the exposure-efficacy response curve), this may not standard proof for therapeutic equivalence requires not only that T
necessarily also be the case for the exposureetoxicity response and R formulations have compositions that are qualitatively (Q1)
relationship. In summary, given that BE is meant to cover both ef- and quantitatively (Q2) the same, but also a demonstration of
ficacy and safety, analysis based on E-R relationships should focus in vivo BE for both unbound and total paclitaxel, as well as
on whichever PD or clinical end point, including adverse events, is demonstrating in vitro similarity in terms of particle size distribu-
most sensitive to PK changes. tion using a population BE approach.140 An even more complex
Exceptions to the “golden rule” of average BE can also be found product-specific BE criteria has been set for pegylated liposomal
in the realm of generics. For example, the United Kingdom Medi- doxorubicin hydrochloride, involving demonstration of in vivo BE
cines and Healthcare products Regulatory Agency approved the for both free and liposome encapsulated doxorubicin, in vitro
registration of 3 generic products containing losartan even though population BE for liposome size distribution, and further, stringent
the 90% CIs for Cmax of the parent compound fell outside the BE demonstration of equivalent liposome characteristics.141
acceptance limits. In this case, the regulators took a pragmatic
approach and based their decisions on available evidence which
indicated that losartan Cmax has only a minor influence on its  -vis
Best Practice Evaluation of BEdClinical Aspects vis-a
clinical efficacy.129-131 Whether this represents a one-time initiative Biopharmaceutical Aspects of BE
of the British regulators or heralds a new regulatory trend is still
unclear, but such flexibility might be seen in the light of modern, Overall, the regulatory authorities tend to prioritize one of the
patient-centric approaches such as the Quality by Design and the BE aspects in lieu of the other depending on the type of the sub-
Biopharmaceutics Risk Assessment Roadmap,132,133 both of which mission. BE in the world of NDAs focuses on the clinical aspect of
advocate departure from a theoretical, oversimplified one-size-fits- the concept, and regulators tend to be flexible with regard to
all BE regulatory criterion in favor of product-specific, clinically acceptance criteria, whereas in the realm of generics, the bio-
relevant specifications. pharmaceutical aspects are prioritized, and these tend to be fol-
lowed more rigorously.142 Nevertheless, a new regulatory paradigm
What If the Link Between Systemic Exposure and Response Is envisioned by the FDA, focusing on applying model-informed drug
Indirect?dSlow or Saturable Target Site Distribution Kinetics development approaches to both brand and generic drug products,
may be useful to extract the best from both BE aspects in a product-
Translating PK changes/differences under nonsteady state con- specific manner. In this context, the workshop entitled “Leveraging
ditions (e.g., single-dose BE studies) into clinical response during Quantitative Methods and Modeling to Modernize Generic Drug
chronic therapy may not be straightforward if the measured Development and Review,” held by the FDA on October 2-3, 2017, is
concentration in plasma is not directly linked to the effect site an important step forward. While the classical one-size-fits-all
concentration. Indeed, 26 years ago, Lewis Sheiner had already approach may well prove to be most suitable for many generics,
raised the question of “what should be measured in BE studies?,” for others, including some of the examples discussed herein, such
pointing out that the delay between the time course of plasma as complex active ingredients, complex formulations, complex
concentration and drug effect should be taken into account to guide routes of delivery, or complex drug device combinations, using a
the decision.37 Bayesian-like approach (in a conceptual sense rather than insisting
Moreover, for drugs acting at intracellular targets protected by on its mathematical formalism), considering all relevant prior
biological barriers, the permeability into the cell can be the rate- knowledge to guide the definition of the test system (in vitro, on a
limiting step to the effect. In this context, saturable transport to stand-alone basis or in combination with in silico tools, in vivo or
the site of action has been reported to cause concentration- even a combination of both systems), including optimized experi-
dependent hysteresis for alphaxalone in rats and capacity-limited mental design, metrics, acceptance limits, and so forth, in a
R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 9

product-specific manner, should lead to better, more clinically Proceedings of the Indo-US Symposium. New Delhi: University College of
Medical Sciences; 1990:7-14.
relevant BE decisions.
2. Kelsey FO. Thalidomide update: regulatory aspects. Teratology. 1988;38:221-
226.
Forecasting the Future of the BE Paradigm Through a Pair of 3. Skelly JP. Bioavailability policies and guidelines. In: Martin A, Doluisio JT, eds.
Industrial bioavailability and pharmacokinetics: guidelines, regulations and
Kuhnian Glasses controls. College of pharmacy, drug dynamics institute. Austin, TX: University of
Texas; 1977:2-41.
It has been demonstrated that the seminal work by Thomas 4. Skelly JP. A history of biopharmaceutics in the Food and Drug Administration
1968-1993. AAPS J. 2010;12(1):44-50.
Kuhn on scientific revolutions and paradigm shifts can be applied
5. Abood RR. Pharmacy Practice and Law. 7 ed. Burlington, MA: Jones & Bartlett
to various branches of natural sciences: physics, chemistry, and Learning; 2014.
biology.38,143-145 In this section, we scrutinize the evolution of BE 6. Carpenter D, Tobbell DA. Bioequivalence: the regulatory career of a pharma-
ceutical concept. Bull Hist Med. 2011;85(1):93-131.
criteria in the light of Thomas Kuhn's theory of scientific revolution.
7. Kesselheim AS, Darrow JJ. Hatch-Waxman Turns 30: Do We Need a Re-
Importantly, one should always keep in mind that demonstration of Designed Approach for the Modern Era? Yale J Health Pol Law Ethics.
equivalence, in either the realm of NDAs or amended new drug 2015;15(2). Available at: http://digitalcommons.law.yale.edu/yjhple/vol15/
applications, is not a scientific fact per se but rather a judgement, iss2/2. Accessed January 10, 2017.
8. Skelly JP. Bioavailability and bioequivalence. J Clin Pharmacol. 1976;16:539-
which (a) is based on facts (e.g., geometric mean ratios for Cmax and 545.
AUC as well as intrasubject variability); (b) also relies on assump- 9. Mossinghoff GJ. Overview of the Hatch-Waxman Act and its impact on the
tions (e.g., setting of the acceptance limits at 80/125 on the drug development process. Food Drug Law J. 1999;54(2):187-194.
10. Manninen V, Melin J, Ha €rtel G. Serum-digoxin concentrations during treat-
assumption that this would encompass the range of clinical simi- ment with different preparations. Lancet. 1971;298:934-935.
larity); and (c) is prone to bias (e.g., status quo bias, heuristic bias, 11. Lindenbaum J, Mellow MH, Blackstone MO, Butler Jr VP. Variation in
hindsight bias, and so forth). Thus, decision-makers should always biologic availability of digoxin from four preparations. N Engl J Med. 1971;285:
1344-1347.
ask “on what basis do we make these judgments?” 12. Levy G. Bioavailability, clinical effectiveness, and the public interest. Phar-
Since the early 1980s, when a consensus about using the one- macology. 1972;8(1):33-43.
size-fits-all approach to compare product performances and 13. Wagner JG, Christensen M, Sakmar E, et al. Equivalence lack in digoxin plasma
levels. JAMA. 1973;224:199-204.
decide about therapeutic equivalence was reached, deviations from
14. Barnett DB, Smith RN, Greenwood ND, Hetherington C. Bioavailability of
the standard paradigm (i.e., the so-called “anomalies” using Kuh- commercial tetracycline products. Br J Clin Pharmacol. 1974;1:319-323.
nian vocabulary) have been accumulating, as demonstrated by the 15. Barr WH, Gerbracht LM, Letcher K, Plaut M, Strahl N. Assessment of the
biologic availability of tetracycline products in man. Clin Pharmacol Ther.
increasing numbers of drug products for which there are product-
1972;13:97-108.
specific recommendations for BE testing. The heretofore presented 16. Glazko AJ, Kinkel AW, Alegnani WC, Holmes EL. An evaluation of the
risks of incurring false-positive results, for example, when failing to absorption characteristics of different chloramphenicol preparations in
extrapolate BE results from healthy adults to special populations, or normal human subjects. Clin Pharmacol Ther. 1968;9:472-483.
17. Chiou WL. Determination of physiologic availability of commercial phenyl-
when Cmax and AUC do not necessarily ensure similarity in the butazone preparations. J Clin Pharmacol New Drugs. 1972;12:296-300.
in vivo drug release characteristics, as well as the risks of false- 18. Van Petten GR, Feng H, Withey RJ, Lettau HF. The physiologic availability
negative results by rejecting pharmacokinetically inequivalent of solid dosage forms of phenylbutazone. I. In vivo physiologic availability
and pharmacologic considerations. J Clin Pharmacol New Drugs. 1971;11:
(but still having therapeutically equivalence) formulations, suggest 177-186.
that at least some anomalies critically impact the main assump- 19. Barber HE, Calvey TN, Muir K, Hart A. Biological availability and in vitro
tions behind the biopharmaceutical and clinical aspects of the one- dissolution of oxytetracycline dihydrate tablets. Br J Clin Pharmacol. 1974;1:
405-408.
size-fits-all BE paradigm. In the context of the Kuhnian lens, the 20. Levy G, Gibaldi M. Editorial: bioavailability of drugs: focus on digoxin. Cir-
paradigm of average BE appears to be embedded in a crisis, and it culation. 1974;49(3):391-394.
will be necessary to propose alternative pathways for ensuring 21. Office of Technology Assessment (OTA). Drug bioequivalence. Recommenda-
tions from the Drug Bioequivalence Study Panel to the Office of Technology
therapeutic equivalence across the entire range of generic products.
Assessment, Congress of the United States. J Pharmacokinet Biopharm. 1974;2:
Indeed, there is already a movement toward applying systems 433-466.
thinking in drug discovery, moving away from a reductionist, target- 22. European Union Law. Council Directive 65/65/EEC on the Approximation of
Provisions Laid Down by Law, Regulation or Administrative Action Relating to
centric approach of drug development into a new holistic systems
Proprietary Medicinal Products. EUR-Lex; 1965. Available at: http://eur-lex.
approach, for example, systems pharmacology.146 Under this europa.eu/legal-content/EN/TXT/?uri¼CELEX:31965L0065. Accessed January
context, integrative approaches such as the model-informed drug 30, 2018.
development may be useful to explore new clinically relevant, 23. The European Parliament and the Council of the European Union. Regulation (EC)
No 726/2004. EUR-Lex; 2004. Available at: http://eur-lex.europa.eu/LexUriServ/
product-specific BE recommendations in lieu of using the standard LexUriServ.do?uri¼OJ:L:2004:136:0001:0033:en:PDF. Accessed February 7,
one-size-fits-all approach, in cases where the latter is not adequate. 2018.
An important part of this process will be close communication by 24. World Health Organization. Drugs and MoneyePrices, Affordability and Cost
Containment. Geneva, Switzerland: WHO; 2003. Available at: http://apps.
regulators internationally to ensure that drug (product)-specific BE who.int/medicinedocs/en/d/Js4912e/3.6.html. Accessed February 7, 2018.
requirements are harmonized across the globe. Efforts such as the 25. Eckstein N, Haas B, Ro €per L. Arzneimittelzulassung in besonderen Fa €llen.
European Federation for Pharmaceutical Sciences (EUFEPS) Global Deutsche Apotheker Zeitung; 2013. Available at: https://www.deutsche-
apotheker-zeitung.de/daz-az/2013/daz-22-2013/arzneimittelzulassung-in-
Bioequivalence Harmonization Initiative are already making besonderen-faellen. Accessed January 30, 2018.
important contributions in this area.147 26. Bundesinstitut für arzneimittel und medizinprodukte (BfArM). Bibliografische
Zulassung. BfArM; 2011. Available at: https://www.bfarm.de/DE/Arzneimittel/
Arzneimittelzulassung/Zulassungsarten/BibliografischeZulassung/_node.html.
Acknowledgments Accessed January 31, 2017.
27. Kuribayashi R, Matsuhama M, Mikami K. Regulation of generic drugs in Japan:
This article reflects the scientific opinion of the authors and not the current situation and future prospects. AAPS J. 2015;17(5):1312-1316.
28. Zargarzadeh AH, Emami MH, Hosseini F. Drug-related hospital admissions in
necessarily the policies of the Brazilian Health Regulatory Agency a generic pharmaceutical system. Clin Exp Pharmacol Physiol. 2007;34(5-6):
(Anvisa). 494-498.
29. Yousefi N, Mehralian G, Peiravian F, NourMohammadi S. Consumers'
perception of generic substitution in Iran. Int J Clin Pharm. 2015;37(3):497-
References 503.
30. Khoonsari H, Oghazian MB, Kargar M, et al. Comparing the efficacy of 8 weeks
1. Seife M. Evolution of the principal US food & drug laws. In: Sharma KN, treatment of Cipram® and its generic citalopram in patients with mixed
Sharma KK, Sen P, eds. Generic drugs, bioequivalence and pharmacokinetics: anxiety-depressive disorder. Iran J Psychiatry Behav Sci. 2015;9(2):e230.
10 R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

31. Tabatabaei O, Heshmat R, Taheri F, Larijani B. Comparison of a generic and a 63. Bois FY, Tozer TN, Hauck WW, et al. Bioequivalence: performance of several
brand metformin products in type II diabetes: a double blind randomized measures of rate of absorption. Pharm Res. 1994;11:966-974.
clinical trial study. Daru J. 2007;15(2):113-117. 64. Aarons L. Assessment of rate of absorption in bioequivalence studies. J Pharm
32. Gibaldi M, Levy G. Pharmacokinetics in clinical practice. I. Concepts. JAMA. Sci. 1987;76:853-855.
1976;235(17):1864-1867. 65. Endrenyi L, Fritsch S, Yan W. Cmax/AUC is a clearer measure than Cmax for
33. European Medicines Agency. Guideline on the Investigation of Bioequivalence. absorption rates in investigations of bioequivalence. Int J Clin Pharmacol Ther
London, UK: EMA; 2010. Available at: http://www.ema.europa.eu/docs/en_GB/ Toxicol. 1991;29:394-399.
document_library/Scientific_guideline/2010/01/WC500070039.pdf. Accessed 66. Schall R, Luus HE. Comparison of absorption rates in bioequivalence studies of
February 15, 2018. immediate release drug formulations. Int J Clin Pharmacol Ther Toxicol.
34. U.S. Food and Drug Administration. Waiver of In Vivo Bioavailability and 1992;30:153-159.
Bioequivalence Studies for Immediate-Release Solid Oral Dosage Forms Based 67. Chen ML. An alternative approach for assessment of rate of absorption in
on a Biopharmaceutics Classification System. Silver Spring, MD: FDA; 2015. bioequivalence studies. Pharm Res. 1992;9(11):1380-1385.
Available at: https://www.fda.gov/downloads/Drugs/Guidances/ucm070246. 68. Lacey LF, Keene ON, Duquesnoy C, et al. Evaluation of different indirect
pdf. Accessed February 15, 2018. measures of rate of drug absorption in comparative pharmacokinetic studies.
35. Brodie BB, Heller WM. Bioavailability of Drugs: Proceedings. Basel, Switzerland: J Pharm Sci. 1994;83:212-215.
S. Karger; 1971. 69. Reppas C, Lacey LF, Keene ON, et al. Evaluation of different metrics as indirect
36. Yu A, Sun D, Li BV, Yu LX. Bioequivalence history. In: Li BV, Yu LX, eds. FDA measures of rate of drug absorption from extended release dosage forms at
Bioequivalence standards. New York, NY: Springer; 2014:1-27. steady-state. Pharm Res. 1995;1995(12):103-107.
37. Sheiner LB. Bioequivalence revisited. Stat Med. 1992;11(13):1777-1788. 70. Endrenyi L, Al-Shaikh P. Sensitive and specific determination of the equiva-
38. Kuhn TS. The Structure of Scientific Revolutions. 3rd ed. Chicago, IL: University lence of absorption rates. Pharm Res. 1995;12:1856-1864.
of Chicago Press; 1996. 71. Rostami-Hodjegan A, Jackson PR, Tucker GT. Sensitivity of indirect metrics for
39. Dobbins TW, Thiyagarajan B. A retrospective assessment of the 75/75 rule in assessing ‘rate’ in bioequivalence studiesemoving the ‘goalposts’ or changing
bioequivalence. Stat Med. 1992;11(10):1333-1342. the ‘game.’ J Pharm Sci. 1994;83:1554-1557.
40. Haynes JD. Statistical simulation study of new proposed uniformity require- 72. Amidon GL, Lennern€ as H, Shah VP, Crison JR. A theoretical basis for a bio-
ment for bioequivalency studies. J Pharm Sci. 1981;70(6):673-675. pharmaceutic drug classification: the correlation of in vitro drug product
41. Haynes JD. FDA 75/75 rule: a response. J Pharm Sci. 1983;72(1):99-100. dissolution and in vivo bioavailability. Pharm Res. 1995;12(3):413-420.
42. Schuirmann DJ. A comparison of the two one-sided tests procedure and the 73. World Health Organization (WHO). Multisource (generic) pharmaceutical
power approach for assessing the equivalence of average bioavailability. products: guidelines on registration requirements to establish interchange-
J Pharmacokinet Biopharm. 1987;15(6):657-680. ability. Geneva, Switzerland: WHO; 2017. Available at: http://apps.who.int/
43. Kirkwood BL, Westlake WJ. Bioequivalence testingea need to rethink. Bio- medicinedocs/documents/s23245en/s23245en.pdf. Accessed January 30,
metrics. 1981;37(3):589-594. 2018.
44. Fisher RA. The Design of Experiments. 9th ed. New York, NY: Macmillan Pub- 74. Polli JE. In vitro studies are sometimes better than conventional human
lishing Co.; 1971. pharmacokinetic in vivo studies in assessing bioequivalence of immediate-
45. Anderson S, Hauck WW. A new procedure for testing equivalence in release solid oral dosage forms. AAPS J. 2008;10(2):289-299.
comparative bioavailability and other clinical trials. Comm Stat. 1983;12(23): 75. Davit BM, Conner DP, Fabian-Fritsch B, et al. Highly variable drugs: observa-
2663-2692. tions from bioequivalence data submitted to the FDA for new generic drug
46. McGilveray IJ, Midha KK, Skelly JP, et al. Consensus report from “Bio Inter- applications. AAPS J. 2008;10(1):148-156.
national '89”: issues in the evaluation of bioavailability data. J Pharm Sci. 76. Bønløkke L, Hovgaard L, Kristensen HG, Knutson L, Lennern€ as H. Direct esti-
1990;79(10):945-946. mation of the in vivo dissolution of spironolactone, in two particle size ranges,
47. Blume HH, Midha KK. Bio-International 92, conference on bioavailability, using the single-pass perfusion technique (Loc-I-Gut) in humans. Eur J Pharm
bioequivalence, and pharmacokinetic studies. J Pharm Sci. 1993;82(11):1186- Sci. 2001;12(3):239-250.
1189. 77. Brouwers J, Augustijns P. Resolving intraluminal drug and formulation
48. Blume HH, McGilveray IJ, Midha KK. BIO-international '94 Conference on behavior: gastrointestinal concentration profiling in humans. Eur J Pharm Sci.
Bioavailability, Bioequivalence and Pharmacokinetic Studies and Pre- 2014;30(61):2-10.
Conference Satellite on ‘In Vivo/In Vitro Correlation.’ Munich, Germany, 78. Geboers S, Stappaerts J, Tack J, Annaert P, Augustijns P. In vitro and in vivo
June 14-17, 1994. Eur J Drug Metab Pharmacokinet. 1995;20(1):3-13. investigation of the gastrointestinal behavior of simvastatin. Int J Pharm.
49. Hwang S, Huber PB, Hesney M, Kwan KC. Bioequivalence and interchange- 2016;510(1):296-303.
ability. J Pharm Sci. 1978;67:IV. 79. Endrenyi L, Tothfalusi L. Do regulatory bioequivalence requirements
50. U.S. Food and Drug Administration. Average, Population, and Individual adequately reflect the therapeutic equivalence of modified-release drug
Approaches to Establishing Bioequivalence. Silver Spring, MD: FDA; 1999. products? J Pharm Pharm Sci. 2010;13(1):107-113.
Available at: http://www.fda.gov/OHRMS/DOCKETS/98fr/3657gd1.pdf. 80. Lionberger RA, Raw AS, Kim SH, Zhang X, Yu LX. Use of partial AUC to
Accessed January 10, 2017. demonstrate bioequivalence of Zolpidem Tartrate Extended Release formu-
51. Hauck WW, Hyslop T, Chen ML, Patnaik R, Williams RL. Subject-by-formula- lations. Pharm Res. 2012;29(4):1110-1120.
tion interaction in bioequivalence: conceptual and statistical issues. FDA 81. Nicholls A, Harris-Collazo R, Huang M, Hardiman Y, Jones R, Moro L.
Population/Individual Bioequivalence Working Group. Food and Drug Bioavailability profile of Uceris MMX extended-release tablets compared with
Administration. Pharm Res. 2000;17(4):375-380. Entocort EC capsules in healthy volunteers. J Int Med Res. 2013;41(2):386-394.
52. Chen ML, Lesko LJ. Individual bioequivalence revisited. Clin Pharmacokinet. 82. Yu A, Baker JR, Fioritto AF, et al. Measurement of in vivo gastrointestinal
2001;40(10):701-706. release and dissolution of three locally acting mesalamine formulations in
53. Chow SC. Bioavailability and bioequivalence in drug development. Rev Com- regions of the human gastrointestinal tract. Mol Pharm. 2017;14(2):345-358.
put Stat. 2014;6(4):304-312. 83. U.S. Food and Drug Administration. Draft Guidance on Budesonide. Silver
54. Anderson S, Hauck WW. Consideration of individual bioequivalence. Spring, MD: FDA; 2017. Available at: http://www.fda.gov/downloads/drugs/
J Pharmacokinet Biopharm. 1990;18(3):259-273. guidancecomplianceregulatoryinformation/guidances/ucm426317.pdf. Accessed
55. Hauck WW, Anderson S. Types of bioequivalence and related statistical con- January 10, 2017.
siderations. Int J Clin Pharmacol Ther Toxicol. 1992;30(5):181-187. 84. U.S. Food and Drug Administration. Draft Guidance on Nifedipine. Silver
56. Hauck WW, Anderson S. Measuring switchability and prescribability: when is Spring, MD: FDA; 2017. Available at: http://www.fda.gov/downloads/Drugs/
average bioequivalence sufficient? J Pharmacokinet Biopharm. 1994;22(6): GuidanceComplianceRegulatoryInformation/Guidances/ucm118368.pdf.
551-564. Accessed January 10, 2017.
57. Levy G. The clay feet of bioequivalence testing. J Pharm Pharmacol. 85. U.S. Food and Drug Administration. Draft Guidance on Zolpidem. Silver
1995;47(12A):975-977. Spring, MD: FDA; 2017. Available at: http://www.fda.gov/downloads/drugs/
58. Benet LZ. Individual bioequivalence: have the opinions of the scientific guidancecomplianceregulatoryinformation/guidances/ucm175029.pdf.
community changed? Silver Spring, MD: 1999. Available at: http://www.fda. Accessed January 10, 2017.
gov/ohrms/dockets/ac/01/slides/3804s2_09_benet.ppt. Accessed January 10, 86. U.S. Food and Drug Administration. Draft Guidance on Methylphenidate.
2017. Silver Spring, MD: FDA; 2017. Available at: http://www.fda.gov/downloads/
59. Lennerna €s H. Human jejunal effective permeability and its correlation with Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM320007.
preclinical drug absorption models. J Pharm Pharmacol. 1997;49(7):627-638. pdf. Accessed January 10, 2017.
60. Chen ML. Rate of absorption in bioequivalence studies: replace with the 87. Cristofoletti R, Dressman JB. Bridging the gap between in vitro dissolution and
concept of exposure? In: Midha K, Nagai T, eds. Bioavailability, Bioequivalence the time course of ibuprofen-mediating pain relief. J Pharm Sci. 2016;105(12):
and Pharmacokinetic Studies. Tokyo, Japan: Bio-International; 1996:191-195. 3658-3667.
61. Chen ML, Lesko L, Williams RL. Measures of exposure versus measures of rate 88. Ting TY, Jiang W, Lionberger R, et al. Generic lamotrigine versus brand-name
and extent of absorption. Clin Pharmacokinet. 2001;40(8):565-572. Lamictal bioequivalence in patients with epilepsy: a field test of the FDA
62. European Medicines Agency. Note for guidance on investigational bioavailability bioequivalence standard. Epilepsia. 2015;56(9):1415-1424.
and bioequivalence. London, UK: EMA; 2000. Available at: http://www.ema. 89. Meyer MC, Straughn AB, Jarvi EJ, et al. Bioequivalence of methylphenidate
europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC5 immediate-release tablets using a replicated study design to characterize
00003519.pdf. Accessed January 30, 2018. intrasubject variability. Pharm Res. 2000;17(4):381-384.
R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 11

90. Carter BL, Noyes MA, Demmler RW. Differences in serum concentration of factor Xa inhibitorein healthy subjects. Int J Clin Pharmacol Ther. 2007;45(6):
and responses to generic verapamil in the elderly. Pharmacotherapy. 1993;13: 335-344.
359-368. 114. U.S. Food and Drug Administration. Draft Guidance on Dabigatran etexilate
91. Lesko LJ. Mechanistic aspects: the subject-by-formulation interaction. Amer- mesylate. Silver Spring, MD: FDA; 2017. Available at: http://www.fda.gov/
ican Association of Pharmaceutical Scientists (AAPS) Workshop, ‘Scientific and downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/
regulatory issues in product quality: narrow therapeutic index drugs and UCM308030.pdf. Accessed February 13, 2017.
individual bioequivalence’; 1998 Mar 16-18; Arlington, VA. 115. U.S. United States Food and Drug Administration. Draft Guidance on Rivar-
92. Aghazadeh-Habashi A, Jamali F. Pharmacokinetics of meloxicam administered oxaban. Silver Spring, MD: FDA; 2017. Available at: http://www.fda.gov/ucm/
as regular and fast dissolving formulations to the rat: influence of gastroin- groups/fdagov-public/@fdagov-drugs-gen/documents/document/ucm461150.
testinal dysfunction on the relative bioavailability of two formulations. Eur J pdf. Accessed February 13, 2017.
Pharm Biopharm. 2008;70(3):889-894. 116. Lionberger R, Jiang W, Huang SM, Geba G. Confidence in generic drug sub-
93. Almukainzi M, Jamali F, Aghazadeh-Habashi A, Lo € benberg R. Disease specific stitution. Clin Pharmacol Ther. 2013;94(4):438-440.
modeling: simulation of the pharmacokinetics of meloxicam and ibuprofen in 117. Popper KR. The Logic of Scientific Discovery. 15th ed. London: Routledge; 2002.
disease state vs. healthy conditions. Eur J Pharm Biopharm. 2016;100:77-84. 118. Davit BM, Chen ML, Conner DP, et al. Implementation of a reference-scaled
94. Mitra A, Kesisoglou F, Beauchamp M, Zhu W, Chiti F, Wu Y. Using absorption average bioequivalence approach for highly variable generic drug products
simulation and gastric pH modulated dog model for formulation development by the U.S. Food and Drug Administration. AAPS J. 2012;14(4):915-924.
to overcome achlorhydria effect. Mol Pharm. 2011;8(6):2216-2223. 119. Davit B, Braddy AC, Conner DP, Yu LX. International guidelines for bioequiv-
95. Dreyfuss D, Shader R, Harmatz JS, Greenblatt DJ. Kinetics and dynamics of alence of systemically available orally administered generic drug products: a
single doses of oxazepam in the elderly: implications of absorption rate. J Clin survey of similarities and differences. AAPS J. 2013;15(4):974-990.
Psychiatry. 1986;47:511-514. 120. U.S. Food and Drug Administration. Guidance for Industry Bioavailability and
96. Badawy SI, Gray DB, Zhao F, Sun D, Schuster AE, Hussain MA. Formulation of Bioequivalence Studies Submitted in NDAs or INDsdGeneral Considerations.
solid dosage forms to overcome gastric pH interaction of the factor Xa in- Silver Spring, MD: FDA; 2014. Available at: http://www.fda.gov/downloads/
hibitor, BMS-561389. Pharm Res. 2006;23(5):989-996. drugs/guidancecomplianceregulatoryinformation/guidances/ucm389370.pdf.
97. Cristofoletti R, Patel N, Dressman JB. Assessment of bioequivalence of weak Accessed August 11, 2016.
base formulations under various dosing conditions using PBPK simulations in 121. Tothfalusi L, Endrenyi L. An exact procedure for the evaluation of reference-
virtual populations. Case Examples: ketoconazole and posaconazole. J Pharm scaled average bioequivalence. AAPS J. 2016;18(2):476-489.
Sci. 2017;106(2):560-569. 122. Cross J, Lee H, Westelinck A, Nelson J, Grudzinskas C, Peck C. Postmarketing
98. Doki K, Darwich AS, Patel N, Rostami-Hodjegan A. Virtual bioequivalence for drug dosage changes of 499 FDA-approved new molecular entities, 1980-
achlorhydric subjects: the use of PBPK modelling to assess the formulation- 1999. Pharmacoepidemiol Drug Saf. 2002;11(6):439-446.
dependent effect of achlorhydria. Eur J Pharm Sci. 2017;109:111-120. 123. Heerdink ER, Urquhart J, Leufkens HG. Changes in prescribed drug doses after
99. Seiler D, Doser K, Salem I. Relative bioavailability of prasugrel free base in market introduction. Pharmacoepidemiol Drug Saf. 2002;11(6):447-453.
comparison to prasugrel hydrochloride in the presence and in the absence of 124. Holford NH, Sheiner LB. Understanding the dose-effect relationship: clinical
a proton pump inhibitor. Arzneimittelforschung. 2011;61(4):247-251. application of pharmacokinetic-pharmacodynamic models. Clin Pharmacoki-
100. U.S. Food and Drug Administration. FY 2016 Awarded GDUFA Regulatory net. 1981;6(6):429-453.
Research Contracts and Grants. Silver Spring, MD: FDA; 2016. Available at: 125. Kuypers DRJ. Tacrolimus formulations and African American Kidney Transplant
https://www.fda.gov/downloads/ForIndustry/UserFees/GenericDrugUserFees/ Recipients: when do details matter? Am J Kidney Dis. 2018;71(3):302-305.
UCM526210.pdf. Accessed January 31, 2018. 126. U.S. Food and Drug Administration. Guidance for Industry. Exposure-
101. Fan J, Zhang X, Zhao L. Utility of physiologically based pharmacokinetic ab- Response RelationshipsdStudy Design, Data Analysis, and Regulatory Appli-
sorption modeling to predict the impact of salt-to-base conversion on Pra- cations. Silver Spring, MD: FDA; 2003. Available at: http://www.fda.gov/
sugrel HCl product bioequivalence in the presence of proton pump inhibitors. downloads/drugs/guidancecomplianceregulatoryinformation/guidances/
AAPS J. 2017;19(5):1479-1486. ucm072109.pdf. Accessed February 13, 2017.
102. U.S. Food and Drug Administration. Drugs@FDA: FDA Approved Drug Prod- 127. U.S. Food and Drug Administration. Clinical Pharmacology and Bio-
ucts. Silver Spring, MD: FDA; 2017. Available at: http://www.accessdata.fda. pharmaceutic Review. Application number:200045Orig1s000. Silver Spring,
gov/scripts/cder/daf/index.cfm. Accessed January 31, 2017. MD: FDA; 2010. Available at: http://www.accessdata.fda.gov/drugsatfda_
103. Lachman Consultant Services, Inc. Citizen Petition to the US-FDA Docket No. docs/nda/2010/200045Orig1s000OtherR.pdf. Accessed February 13, 2018.
2004P-0504/CP1 requesting that the Commissioner of Food and Drugs desig- 128. U.S. Food and Drug Administration. Tekturna label. Silver Spring, MD: FDA;
nate DiaBeta Tablets, 5 mg, as a second RLD for the purposes of submitting an 2017. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2
ANDA for a duplicate version of that RLD product. Silver Spring, MD: FDA; 2004. 012/021985s023lbl.pdf. Accessed February 13, 2018.
Available at: http://www.fda.gov/ohrms/dockets/dailys/04/nov04/111204/ 129. Medicines and Healthcare Products Regulatory Agency. Public assessment
04p-0504-cp00001-vol1.pdf. Accessed January 31, 2017. report of the Medicines Evaluation Board in UK. Losartan potassium 25, 50
104. U.S. Food and Drug Administration. Response to the Citizen Petition to the US- and 100 mg tablets, PL 16002/0077e79, PL 16002/0081e83. London, UK:
FDA Docket No. 2004P-0504/CP1. Silver Spring, MD: FDA; 2005. Available at: MHRA; 2008. Available at: http://www.mhra.gov.uk/home/groups/l-unit1/
http://www.fda.gov/ohrms/dockets/dockets/04p0504/04p-0504-pav0001- documents/websiteresources/con014176.pdf. Accessed January 12, 2017.
vol1.pdf. Accessed January 31, 2017. 130. Medicines and Healthcare Products Regulatory Agency. Public assessment
105. Vogel AM. Hybrid or mixed marketing authorization application in the report of the Medicines Evaluation Board in UK. Losartan potassium/hydro-
European Union: not a trivial decision in new development programs for chlorothiazide 50/12.5 mg tablets, PL 24701/0001/PL 24701/0002. London,
established drugs. Drug Information Journal. 2012;46(4):479-484. UK: MHRA; 2008. Available at: http://www.mhra.gov.uk/home/groups/l-
106. European Medicines Agency. Questions & Answers: positions on specific unit1/documents/websiteresources/con014706.pdf. Accessed January 12,
questions addressed to the Pharmacokinetics Working Party (PKWP). London, 2017.
UK: EMA; 2015. Available at: http://www.ema.europa.eu/docs/en_GB/ 131. Medicines and Healthcare Products Regulatory Agency (MHRA). Public
document_library/Scientific_guideline/2009/09/WC500002963.pdf. Accessed assessment report of the Medicines Evaluation Board in UK. Losartan
February 1, 2017. potassium/hydrochlorothiazide 100/12.5 mg tablets, PL04416/1197e1199,
107. Benet L. Why highly variable drugs are safer, FDA Advisory Committee for Sandoz Limited. London, UK: MHRA; 2011. Available at: http://www.mhra.
Pharmaceutical Sciences and Clinical Pharmacology meeting transcript. Silver gov.uk/home/groups/par/documents/websiteresources/con132112.pdf. Accessed
Spring, MD: FDA; 2004. Available at: http://www.fda.gov/ohrms/dockets/ac/ January 12, 2017.
04/transcripts/4034T2.htm. Accessed February 1, 2017. 132. Selen A, Dickinson PA, Müllertz A, et al. The biopharmaceutics risk assessment
108. Benet LZ, Goyan JE. Bioequivalence and narrow therapeutic index drugs. roadmap for optimizing clinical drug product performance. J Pharm Sci.
Pharmacotherapy. 1995;15(4):433-440. 2014;103(11):3377-3397.
109. Filipsson AF, Sand S, Nilsson J, Victorin K. The benchmark dose 133. Yu LX, Amidon G, Khan MA, et al. Understanding pharmaceutical quality by
methodereview of available models, and recommendations for application in design. AAPS J. 2014;16(4):771-783.
health risk assessment. Crit Rev Toxicol. 2003;33(5):505-542. 134. Visser SA, Smulders CJ, Reijers BP, Van der Graaf PH, Peletier LA, Danhof M.
110. U.S. Food and Drug Administration. GDUFA Regulatory Science Priorities for Fiscal Mechanism-based pharmacokinetic-pharmacodynamic modeling of
Year 2018. Silver Spring, MD: FDA; 2018. Available at:https://www.fda.gov/ concentration-dependent hysteresis and biphasic electroencephalogram ef-
downloads/Drugs/ResourcesForYou/Consumers/BuyingUsingMedicineSafely/ fects of alphaxalone in rats. J Pharmacol Exp Ther. 2002;302(3):1158-1167.
GenericDrugs/UCM582777.pdf. Accessed January 31, 2018. 135. Gengo FM, Schentag JJ, Jusko WJ. Pharmacokinetics of capacity-limited tissue
111. Cristofoletti R, Dressman JB. Use of physiologically based pharmacokinetic distribution of methicillin in rabbits. J Pharm Sci. 1984;73(7):867-873.
models coupled with pharmacodynamic models to assess the clinical rele- 136. Danhof M. Kinetics of drug action in disease states: towards physiology-based
vance of current bioequivalence criteria for generic drug products containing pharmacodynamic (PBPD) models. J Pharmacokinet Pharmacodyn. 2015;42(5):
Ibuprofen. J Pharm Sci. 2014;103(10):3263-3275. 447-462.
112. Stangier J, Rathgen K, St€ ahle H, Gansser D, Roth W. The pharmacokinetics, 137. Cui J, Li C, Guo W, et al. Direct comparison of two pegylated liposomal
pharmacodynamics and tolerability of dabigatran etexilate, a new oral direct doxorubicin formulations: is AUC predictive for toxicity and efficacy? J Control
thrombin inhibitor, in healthy male subjects. Br J Clin Pharmacol. 2007;64(3): Release. 2007;118(2):204-215.
292-303. 138. Smith NF, Acharya MR, Desai N, Figg WD, Sparreboom A. Identification of
113. Mueck W, Becka M, Kubitza D, Voith B, Zuehlsdorf M. Population model of the OATP1B3 as a high-affinity hepatocellular transporter of paclitaxel. Cancer
pharmacokinetics and pharmacodynamics of rivaroxabanean oral, direct Biol Ther. 2005;4(8):815-818.
12 R. Cristofoletti et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

139. Sparreboom A, van Zuylen L, Brouwer E, et al. Cremophor EL-mediated 143. Hoyningen-Huene P. Thomas Kuhn and the chemical revolution. Found Chem.
alteration of paclitaxel distribution in human blood: clinical pharmacoki- 2008;10(2):101-115.
netic implications. Cancer Res. 1999;59(7):1454-1457. 144. Portin P. The development of genetics in the light of Thomas Kuhn's theory of
140. U.S. Food and Drug Administration. Draft Guidance on Paclitaxel. Silver scientific revolutions. Recent Adv DNA Gene Seq. 2015;9(1):14-25.
Spring, MD: FDA; 2017. Available at: http://www.fda.gov/downloads/ 145. Strohman RC. The coming Kuhnian revolution in biology. Nat Biotechnol.
drugs/guidancecomplianceregulatoryinformation/guidances/ucm320015. 1997;15(3):194-200.
pdf. Accessed January 10, 2017. 146. van der Greef J, McBurney RN. Innovation: rescuing drug doscovery: in vivo
141. U.S. Food and Drug Administration. Draft Guidance on Doxirrubicin hydro- systems pathology and systems pharmacology. Nat Rev Drug Discov. 2005;4:
chlorhide. Silver Spring, MD: FDA; 2017. Available at: http://www.fda.gov/ 961-967.
downloads/Drugs/.../Guidances/UCM199635.pdf. Accessed January 10, 2017. 147. Chen ML, Blume H, Beuerle G, et al. The global bioequivalence harmonization
142. Endrenyi L, Blume HH, Tothfalusi L. The two main goals of bioequivalence initiative: summary report for EUFEPS international conference. Eur J Pharm
studies. AAPS J. 2017;19(4):885-890. Sci. 2018;111:153-157.

You might also like