throbber
Chapter 18
`Bioavailability and
`Bioequivalence Testing
`
`Steven B. Johnson, PharmD
`
`INTRODUCTION 349
`DISSOLUTION Jc19
`ABSORPTION FACTORS 350
`BIOEQUIVALENCE TESTING 351
`METHODS FOR DETERMINING BIOEQUIVALENCE 351
`
`MINIMIZING THE NEED FOR BIOEQUIVALENCE
`STUDIES 352
`
`EVALUATION OF BIOEQUIVALENCE DATA 352
`BIOEQUIVALENCE ASSESSMENT AND DATA
`EVALUATION 352
`
`CRITERIA FOR BIOEQUIVALENCE 35,~
`
`STUDY DESIGN 356
`
`THERAPEUTIC EQUIVALENCE EVALUATIONS 358
`
`INTRODUCTION
`Understanding the concepts of bioavailability and bioequiva-
`lence testing is essential in the drug development process be-
`cause they create the foundation for regulatory decision making
`when evaluating formulation changes and lot-to-lot consistency
`in innovator products. They also serve as the primary compo-
`nents to demonstrate therapeutic equivalence between generic

`products and the reference innovator product.
`Changes in bioavailabiJity can be thought of in terms of chang-
`es in exposure to the drug. If these changes are substantial, then
`they can alter the safety and efficacy profile of the compound in
`question. The bioavailability of orally administered drugs can be
`affected by numerous factors. These include food or fed state,
`differences in drug metabolism, drug-drug interactions, gastro-
`intestinal transit time, and changes in dosage form release char-
`acteristics (especially for modified release products).
`Bioequivalence is an important consideration in ensuring lot-
`to-lot consistency, including whenever evaluating changes in a
`marketed product's formulation, manufacturing process, and dos-
`age strength. Bioequivalence is also critical in regulatory author-
`ity decision malting when determining whether a generic product
`is therapeutically equivalent to the original innovator product.
`In addition, chemical equivalence, lot-to-lot uniformity of
`physicochemical characteristics, and stability equivalence are
`other factors that are important, as they too can affect product
`quality. In this chapter, bioavailability and bioequivalence top-
`ics are emphasized for solid oral dosage forms. However, many
`of the general concepts can also be applied to other dosage
`forms, including biologics.
`
`GENERAL CONCEPTS
`The terms used in this chapter require careful definition, since,
`as in any area, some terms have been used in different contexts
`by different authors.
`Bioa'Vailability is a term that indicates measurement of both
`the rate of drug absorption and the total amount (extent) of
`drug that reaches the systemic circulation from an adminis-
`tered dosage form. It is specific to the parent or active drug
`substance as contrasted to metabolites.
`Equi'Valence is more a general and relative term that indi-
`cates a comparison of one drug product with another. Equiva-
`lence may be defined in several ways:
`
`• Bioequi'Valence indicates that a drug in two or more
`similar dosage forms reaches the systemic circulation at
`the same relative rate and extent (i.e., the plasma level
`
`profiles of the drug obtained using the two dosage forms
`are the same).
`• Chemical equi'Valence considers that two or more dosage
`forms contain the same labeled quantities (within speci-
`fied limits) of the drug.
`• Clinical equi'Valence occurs when the same drug from
`two or more dosage forms gives identical in 'Vi'Vo effects as
`measured by a pharmacological response or by control of
`a symptom or disease.
`• Pharmaceutical equi'Valence refers to two drug products
`with the same dosage form and same strength.
`• Therapeutic equi'Valence implies that two brands of a
`drug product are expected to yield the same clinical
`result. The FDA specifically uses the term "therapeutic
`equivalence" in the evaluation of multi-source prescrip-
`tion drug products (generic drugs).
`
`Area under the Concentration-Time Curve (AUG) is the inte-
`gral of the concentration- time curve after administration of a single
`dose of drug or after achieving a steady state. The calculated area
`under the serum, blood, or plasma concentration-time curve is re-
`ported in amount/volume multiplied by time (e.g., IW'mL x h or
`!1/100 mL x h) and can be considered representative of the amount
`of drug absorbed. Several variants of AUC exist, including AU Co.,;
`AUC0_; and AU~, SS• corresponding to the calculated area from
`time zero to a truncated time point (e.g., AUC048), the total area
`under the curve, and the area when steady state has been achieved.
`Peak-height Concentration (Cmax) is the peal' of the blood
`level-time curve and represents the highest drug concentration
`achieved after drug administration.
`Time of Peak Concentration (Tmax) is the measured length of
`time necessary to achieve the maximum concentration (~")
`after drug administration.
`
`DISSOLUTION
`For a drug to be absorbed, it must first go into solution. In Figure
`18-1 the steps in the dissolution and subsequent absorption
`process of a tablet or capsule dosage form are outlined. Simi-
`lar profiles could be obtained for any solid or semisolid dosage
`form, including oral suspensions, parenteral suspensions, and
`suppositories. The theory and mechanics of drug dissolution
`rate are described in detail in Chapter 22. The physical charac-
`teristics of the drug and the composition of the tablet (dosage
`form) can have an effect on the rates of disintegration, deaggre-
`gation, and dissolution of the drug. As such, these can affect the
`rate of absorption and resultant blood levels of the drug.
`
`349
`
`AMN1067
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`350
`
`Solid dosage form
`Dissoluti~n
`of
`drug
`
`°• IARMfiCEUTICAl_ ANALYSIS AND QUALITY CONTROL
`Disintegration
`Deaggregation
`Granules
`
`Small particles
`
`drug
`
`j Diss~~ution/issolution
`Drug in solution ! Drug absorption
`
`of
`drug
`
`Drug in blood
`Figure 18-1. Sequence of events involved in the dissolution and
`absorption of a drug from a solid oral dosage form.
`
`An important aspect of product quality for marl<eted oral solid
`dosage forms relates to dissolution testing. The dosage forms ac-
`tually used by patients will be from lots that have not directly
`undergone human bioavailability testing. Once adequate product
`quality has been established by bioavailability testing, subsequent
`batches manufactured using the same formulation, equipment,
`and process are likely to be bioequivalent to the original batch
`tested in humans. This is an important concept in the regulatory
`control of product quality and is where in vitro testing such as
`assay, content unifom1ity, tablet hardness, and dissolution are
`important. Among these several in vitro tests, dissolution test-
`ing is probably the most important, related to bioavailability. As
`part of the drug approval process, a dissolution test procedure is
`established for all oral solid dosage forms. These dissolution tests
`are incorporated into the United States Pharmacopeia (USP) and
`apply both to innovator and generic drug products. All marketed
`batches of these drug products must meet the Abbreviated New
`Drug Application (ANDA)/New Drug Application (NDA)IUSP dis-
`solution tests throughout the shelf-life of tl1e product. Products
`failing their approved dissolution test and/or a USP dissolution
`test must be removed from the market.
`PROPERTIES OF THE DRUG
`The physical characteristics of the drug that can alter bioavail-
`ability are discussed in Chapters 9 and 54 and consist of the
`polymorphic crystal form, choice of the salt form, particle size,
`hydrated or anhydrous form, wettability, and solubility of the
`drug. Chapter 9 also discusses several other properties that can
`adversely affect drug product quality. These factors should be
`investigated during product development and should not, there-
`fore, affect the bioavailability of the drug product.
`PROPERTIES OF THE DOSAGE FORM
`The various components of the solid or semisolid dosage form,
`other than the active ingredient, are discussed in Chapter 45.
`Only an overview, for tablet dosage forms, is given here. In ad-
`dition to the active ingredient, a tablet product usually will con-
`tain the following types of inactive ingredients:
`• Glidants are used to provide a free-flowing powder from
`the mix of tablet ingredients so that the material will flow
`when used on a tablet machine.
`• Binders provide cohesiveness to the tablet. Too little
`binder will produce tablets that do not maintain ilieir
`integrity; too much may affect adversely the release (dis-
`solution rate) of the drug from the tablet.
`• Fillers are used to give the powder bulk so that an accept-
`ably sized tablet is produced. Most commercial tablets
`weigh from 100 mg to 500 mg, so it is obvious that for
`many potent drugs the filler constitutes a large portion
`of the tablet. Binding of drug to the fillers may occur and
`affect bioavailability.
`• Disintegrants are used to cause the tablets to disintegrate
`when exposed to an aqueous environment. Too much
`will produce tablets that may disintegrate in the bottle
`because of atmospheric moisture, and too little may be
`insufficient for disintegration to occur and may therefore
`
`alter the rate and extent of release of the drug from the
`dosage form.
`• Lubricants are used to enhance the flow of the powder
`through the tablet machine and to prevent sticking of the
`tablet in the die of the tablet machine after the tablet is
`compressed. Lubricants are usually hydrophobic materi-
`als such as stearic acid, magnesium, or calcium stearate.
`Too little lubricant will not permit satisfactory tablets to
`be made; too much may produce a tablet with a water-
`impervious hydrophobic coat, which can inhibit the
`disintegration of the tablet and dissolution of the drug.
`
`ABSORPTION FACTORS
`A significant factor related to drug bioavailability is the fact
`that many drugs are administered, not as a solution, but as a
`solid dosage form. Optimal bioavailability might be expected
`from a solution, since a solid drug must first dissolve to be ab-
`sorbed, but considerations such as drug stability, unpalatable
`taste, and the desired duration of action (for controlled-release
`drug products) may prevent tl1e development of solution-based
`dosage form.
`In the dose titration of any patiem the objective is, in con-
`ceptual terms, to attain and maintain a blood level that exceeds
`the minimum effective level required for response but does not
`exceed the minimum toxic (side-effect) level. This is shown
`graphically in Figure 18-2. There are several absorption factors
`that can affect the general shape of this blood-level curve and
`thus drug response.
`DOSE ADMINISTERED
`The blood levels wi!J rise and fall in proportion to the dose
`administered.
`AMOUNT OF DRUG ABSORBED
`Blood levels achieved are also dependent on the amount of drug
`absorbed. For example, the effect of having only one-half of the
`drug absorbed from a dosage form is equivalent to lowering the
`dose (Figure 18-3).
`RATE OF ABSORPTION
`If absorption from tile dosage form is more rapid than the rate
`of absorption that gave profi.le C in Figure 18-4, minimum toxic
`(side-effect) levels may be exceeded. If absorption from the
`dosage form is sufficiently slow, minimum effective levels may
`never be attained, as in profi.le B in Figure 18-4.
`In either instance, the time course and extent of clinical re-
`sponse to the drug may be altered because of changes in dose or
`rate and extent of absorption.
`Both factors, rate and extent of drug absorption, can be
`affected by the dosage form in which the drug is contained. The
`
`Minimum toxic level
`
`c
`0
`:;::>
`~ c Q)
`0 c
`0
`0
`«l E 1/)
`«l a:
`
`Time
`Figure 18-2. Typical plasma-level curve of a drug with effective and
`toxic (side-effect) profile levels defined.
`
`AMN1067
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`Minimum toxic level
`---------------------------------------------------·
`
`Minimum effective level
`
`c
`.2
`~
`'E Q) u
`c
`0 u
`ro
`~
`ro a:
`
`nme
`Figure 18-3. Effect of the extent of drug absorption from a dosage
`form on drug-plasma levels and efficacy. The extent of absorption
`from dosage form B is 50% of that from dosage form A.
`
`Minimum toxic level
`
`c
`
`c
`0
`~
`'E
`Q) u c
`8
`ro
`E (/) ro a:
`
`Time
`Figure 18-4. Effect of the rate of drug absorption from a dosage form
`on the plasma-level profile and efficacy. The rates of absorption from
`dosage forms Band Care 1/10 and 10 times those from dosage form A.
`
`effect of rate of absorption may be intentional, as in controlled-
`release products, or unintentional, for example, as brought
`about by a change in the composition and/or method of manu-
`facture of the dosage form.
`The choice of the inactive ingredients (excipients) used to
`prepare a dosage form is up to the individual manufacturer. It is
`through these changes in composition and manufacturing tech-
`nique that unintended changes in bioavailability and bioequiva-
`lence may occur. Revalidation of bioequivalence may be needed
`for major changes in the manufacturing process, whereas small
`changes may not raise significant bioavailability concerns.
`In situations involving minor changes in the manufacturing
`process, comparative dissolution testing of the original and
`reformulated product provides adequate documentation of con-
`tinued product quality, if the resulting dissolution profiles are
`similar. These considerations apply to all drug manufacturers,
`both innovator and generic companies. A description of the for-
`mulation of dosage forms and the factors that must be consid-
`ered is given in Chapter 9.
`
`BIOEQUIVALENCE TESTING
`The awareness of
`the potential for clinical differences
`between otherwise chemically equivalent drug products has
`been brought about by a multiplicity of factors that include,
`among others:
`
`• better methods for clinical efficacy evaluation
`• development of techniques to measure microgram or
`nanogram quantities of drugs in biological fluids
`
`351
`BIOAVAILABILITY AND BIOEOUIVALENCE TESTING
`• improvements in the technology of dosage form formula-
`tion and physical testing
`• awareness of reported clinical differences from the litera-
`ture in otherwise similar products
`• increased cost of classical clinical evaluation
`• objective and quantitative nature of bioavailability tests
`• an increase in the number of chemically equivalent
`products on the market because of patent expirations
`and the Drug Price Competition and Patent Term
`Restoration Act of 1984 (1-Iatch-Waxman Act), which
`established the generic drug approval procedures that
`are in place today.
`
`The increase in the number of drugs that are available from
`multiple sources frequently has placed people involved in the
`delivery of healthcare in the position of having to select one
`from among several marketed products. As with any decision,
`the more data available, the more comfortable one is in arriv-
`ing at the final decision. The need to make these choices, in
`light of the potential failure to demonstrate in vivo equivalence
`between products or different batches of the same product, has
`increased the demand for quantitative data. Bioequivalence
`testing represents a bridging alternative to clinical testing for
`efficacy and safety in such cases and is the means by which ge-
`neric drugs are approved for marketing. In addition, this is also
`the means by which the quality of all drug products is main-
`tained in situations involving major changes in formulation or
`manufacturing process.
`Requirements for bioequivalence data on drug products
`should be applied reasonably. The reason for bioequivalence
`testing should not be overlooked (i.e., it is used as a surrogate,
`in certain situations, for the clinical evaluation of drug prod-
`ucts). In some instances, bioequivalence data cannot reliably
`be obtained if the bioanalytical methodology is not available.
`However, in such cases pharmacodynamic data may provide
`a more sensitive, objective evaluation of a product's thera-
`peutic equivalence and may be explored as an alternative
`evaluation method in the absence of relevant bioanalytical
`methodology.
`Basic pharmacokinetic evaluation of bioavailability data is
`not necessary to show bioequivalence of two drug products.
`Pharmacokinetics has its major utility in the prediction or pro-
`jection of dosage regimens and/or in providing a better under-
`standing of observed drug reactions or interactions that result
`from the accumulation of drug in some specific site, tissue, or
`compartment of the body. The basis for the conclusion that two
`drug products are bioequivalent must be that the drug concen-
`trations measured in a biological matrix, or alternatively the
`pharmacological response, for one drug product are essentially
`the same for the second drug product. The more straightfor-
`ward decisions in the evaluation of bioequivalence between two
`drug products are those in which the two products are exactly
`superimposable (definitely bioequivalent). Those in which the
`two products differ in their bioequivalence parameters by a
`large amount, such as SO% or more, are most definitely not bio-
`equivalent. Statistical evaluation of the data is necessary for all
`situations, particularly for data that exist between these two
`extremes.
`
`METHODS FOR DETERMINING BIOEQUIVALENCE
`Bioequivalence usually involves human testing but sometimes
`may be demonstrated using an in vitro bioequivalence stan-
`dard, especially when such an in vitro test has been correlated
`with human in vivo bioavailability data. In other situations,
`bioequivalence may be demonstrated through comparative
`clinical trials or pharmacodynamic studies.
`The FDA has categorized (21CFR320.24) various in vivo and
`in vitro approaches that may be utilized to establish bioequiva-
`lence. These are, in descending order of accuracy, sensitivity
`and reproducibility,
`
`AMN1067
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`352
`PHARMACEUTICAL ANALYSIS AND QUALITY CONTROL
`1. An in 'Vivo test in humans in which the active drug sub-
`stance, as well as active metabolites when appropriate, is
`measured in plasma.
`2. An in vitro test that has been correlated with human
`in vivo bioavailability data. This approach is most likely
`for oral modified release products and is described in
`detail in FDA Guidance.
`3. An in vivo test in animals that has been correlated with
`human bioavailability data.
`4. An in vivo test in humans, where urinary excretion of
`the active drug substance, as well as active metabolites
`when appropriate, is measured.
`5. An in vioo test in humans in which an appropriate acute
`pharmacological effect is measured.
`6. Well-controlled clinical trials in humans that establish
`the safety and efficacy of the drug product, for estab-
`lishing bioavailability. For bioequivalence, comparative
`clinical trials may be considered. This approach is the
`least accurate, sensitive, and reproducible approach and
`should be considered only if other approaches are not
`feasible.
`7. A currently available in 'Vitro test, acceptable to FDA,
`that ensures bioavailability. This approach is intended
`only when in 'Vitro testing is deemed adequate, but no
`in vitro-in v ivo correlation (IVIVC) has been estab-
`lished. It also can relate to considerations involving the
`Biopharmaceutics Classification System (BCS).
`
`Most bioequivalence studies involve the direct measurement
`of the parent drug, as described in item 1 above. Bioequiva-
`lence testing in animals is not a recommended approach due to
`possible differences in metabolism, gastrointestinal physiology,
`weight, and diet.
`
`MINIMIZING THE NEED FOR BIOEQUIVALENCE
`STUDIES
`If a drug product has been adequ ately tested and approved for
`marketing, and if no changes in the manufacturing of the prod-
`uct are made, it is reasonable to assume that all subsequent
`batches of the product would be expected to be bioequivalent
`to the original product. If subsequently manufactured batches
`meet all tests of quality, including the dissolution test, no fur-
`ther human bioequivalence testing is needed.
`Depending on the degree of change, bioequivalence may
`sometimes need to be reconfirmed. Although it is somewhat
`difficult to categorize such major changes, this issue has been
`described in a series of FDA guidance documents related to
`Scale-Up and Post-Approval Changes (SUPAC).
`Drug characteristics related to solubility and permeability
`may allow a reasonable expectation that the drug is unlikely
`to be subject to significant bioavailability problems. For such
`drugs, in 'Vitro dissolution testing may be adequate, in lieu of
`in 'Vivo testing. These concepts are described in the Biophar-
`maceutics Classification System (BCS). This classification sys-
`tem provides a scientific framework for classifying drugs based
`on aqueous solubility and intestinal permeability. In addition,
`criteria for rapid dissolution are described (not less than 85%
`dissolved in 30 minutes, using mild agitation and physiologi-
`cal media). The BCS permits waivers of in 'V'ivo bioequivalence
`testing for high solubility, high permeability drugs (Class I),
`which are formulated into immediate release dosage forms hav-
`ing rapid dissolution. The basic ten et behind the BCS is that
`solutions of drugs are thought to have few bioavailability or bio-
`equivalence issues. Dosage forms containing drugs that are of
`high solubility and exhibit rapid dissolution behave similarly
`to a solution. Particularly for such drugs that are, in addition,
`highly permeable (well absorbed), the likelihood of bioavailabil-
`ity problems is quite small, and consequently, bioequivalence
`testing for such drugs is thought to be unnecessary. Similarly
`for oral solutions, bioequivalence testing is not necessary.
`
`EVALUATION OF BIOEQUIVALENCE DATA
`The following sections highlight some considerations when
`evaluating data from bioequivalence studies. The topics dis-
`cussed are directed specifically toward plasma level evaluations.
`With minor modifications, the approaches outlined can be used
`for urinary excretion measurements or for suitable, quantita-
`tive, pharmacological response measurements.
`Bioequivalence studies are usually conducted in healthy
`adults under standardized conditions. Most often, single doses
`of the test and reference product will be evaluated. However,
`in selected cases, multiple-dose regimens may be used (e.g. ,
`when patients are used and they cann ot be discontinued from
`a medication). The goal of the study is to evaluate the in 'V'i'VO
`performance, as measured by rate and extent of absorption, of
`the dosage forms under standardized conditions to minimize
`patient-related and other variability.
`The protocol should define the acceptable age and weight
`range for the subjects to be included in the study, as well as the
`clinical parameters that will be used to characterize a healthy
`adult (e.g., physical examination observations , clinical chemis-
`try, and hematological evaluations). The subjects should have
`been drug-free for at least two weeks prior to testing to elimi-
`nate possible drug-induced influences on liver enzyme systems.
`Normally, the subjects will fast overnight for at least ten hours
`prior to dosing and will not eat until a standard meal is provided
`four hours post-dosing. The dosage forms should be given to
`subjects in a randomized manner, using a suitable crossover
`design, so that possible daily variations are distributed equally
`between the dosage forms tested. The protocol should define
`sample collection times and techniques to collect the biologi-
`cal fluid. The method of sample storage should also be defined.
`
`BIOEQUIVALENCE ASSESSMENT
`AND DATA EVALUATION
`Several parameters are used to provide a general evaluation of
`the overall rate and extent of absorption of a drug. An analy-
`sis of all characteristics is required before one can determine
`bioequivalence or lack of bioequivalence. It is implicit that the
`analytical methodology used for analysis of drug in the samples
`is specific, sensitive, and precise.
`In assessing the bioequivalence of drug products, one must
`quantitate the rate an d extent of absorption, which can be
`determined by evaluating parameters derived from the blood-
`level concentration-time profile. Three parameters describing
`a blood-level curve are considered important in evaluating the
`bioequivalence of two or more formulations of the same drug.
`These are the peak-height concentration (Cmax), the time of the
`peak concentration (Tm:uJ, and the area under the blood (se-
`rum or plasma) concentration-time curve (AUC).
`
`PEAK-HEIGHT CONCENTRATION (Cmax)
`The peak of the blood-level-time curve represents the highest
`drug concentration achieved after oral administration. It is re-
`ported as an amount per volume measurement (e.g., microgram/
`milliliter (pgtmL) , unit/mL, or gram/100 mL). The importance
`of this parameter is illustrated in Figure 18-5, wh ere tl1e blood
`concentration-time curves of two different formulations of a
`drug are represented. A line has been drawn across the curve
`at 4 pg.'mL. Suppose that the drug is an analgesic, and 4 pg.'mL
`is the minimum effective concentration (!vlEC) of the drug in
`blood. If the blood concentration curves in Figure 18-5 rep-
`resent the blood levels obtained after administration of equal
`doses of two formulations of the drug and it is known that an-
`algesia would not be produced unless the MEC was achieved
`or exceeded, it becomes clear that formulation A would be ex-
`pected to provide pain relief, while formulation B, even though
`it is well absorbed regarding extent of absorption, might be inef-
`fective in producing analgesia.
`
`AMN1067
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`/Peak
`
`..
`
`::::;
`-®,
`.=;,6.0
`c:
`0
`:;:>
`
`~ 13 4.0
`0 c:
`0
`0
`u g 2.0
`:0
`Q)
`Ol
`~
`Q)
`
`~ 0
`
`8
`6
`16
`14
`12
`10
`2 3 4
`Time after drug administraion (hours)
`Figure 18-5. Blood concentration-time curves obtained for two dif-
`ferent formulations of the same drug, demonstrating relationship of
`the profiles to the minimum effective concentration (MEC).
`
`20
`
`-
`
`-
`
`AREA
`(0-20 hours)
`.
`Jlg
`A= 34.4 ml x hours
`Jlg
`A-·-t:> A = 34.2 ml x hours
`Formulation A
`·- ·- ·- ·-·-·-·-·-·-·-·- ·- ·- ·- ·- ·- ·MTC
`-----.:.
`........ --~:>
`-,-Formulation B
`:" ·':c...._
`--- -- ~~-- - - · - · -· -· - ·- ·----MEC
`
`·-6.--
`
`·-e... _ ___ _
`.
`
`::::;
`.E
`E:6.0
`c: g
`~ 13 4.0
`0 c:
`0
`0
`u g 2.0
`:0
`Q)
`Ol
`~
`Q)
`~
`
`0 1 2 3 4
`8
`6
`16
`14
`12
`10
`Time after drug administraion (hours)
`Figure 18-6. Blood concentration-time curves obtained for two dif-
`ferent formulations of the same drug, demonstrating relationship of
`the profiles to the minimum toxic concentration (MTC) and the mini-
`mum effective concentration (MEC).
`
`20
`
`On the other hand, if the two curves represent blood con-
`centrations following equal doses of two different formulations
`of the same cardiac glycoside, and 4 j.ig/mL now represents the
`minimum toxic concentration (MTC) and 2 jlg/mL represents
`the MEC (Figure 18-6), formulation A, although effective, may
`also present safety concerns, while formulation B produces con-
`centrations well above the MEC but never reaches toxic levels.
`Time of Peak Concentration (T max)
`The second parameter of importance is the measurement of the
`length of time necessary to achieve the maximum concentration
`
`353
`BIOAVAILABILITY AND BIOEOUIVALENCE TESTING
`after drug administration. This parameter is called the time of
`peak blood concentration (T013J. In Figure 18-S, for formula-
`tion A the time necessary to achieve peak blood concentration
`is 1 h. For formulation B, Tmax is 4 h. This parameter is related
`closely to the rate of absorption of the drug from a formulation
`and may be used as a simple measure of rate of absorption bu t
`is normally not evaluated statistically.
`To illustrate the importance of Tmax> suppose that the two
`curves in Figure 18-6 now represent two formulations of an an-
`algesic and that in this case the MEC is 2.0gtmL. Formulation
`A will achieve the MEC in 30 minutes; formulation B does not
`achieve that concentration until 2 h. Formulation A would pro-
`duce analgesia much more rapidly than formulation B and would
`probably be preferable as an analgesic agent. On the other hand,
`if one were more interested in the duration of the analgesic ef-
`fect than on the tin1e of onset, formulation B would present more
`prolonged activity, maintaining serum concentrations above the
`MEC for a longer time (8 h) than formulation A (5.5 h).
`AREA UNDER THE CONCENTRATION-TIME
`CURVE (AUC)
`The third, and sometimes the most important parameter for
`evaluation, is the area under the serum, blood, or plasma con-
`centration-time curve (AUC). This area is reported in amount/
`volume multiplied by time (e.g., jlg/mL x h or g/100 mL x h)
`and can be considered representative of the amount of drug
`absorbed following administration of a single dose of the drug.
`Although several methods exist for calculating the AUC, the
`trapezoidal rule method is most often used. This method as-
`sumes a linear function, y = bt + a, and its accuracy increases
`as the number of appropriate sampling intervals are increased.
`Table 18-1 and Figu re 18-7 describe the process for calculating
`the AUC using the trapezoidal rule.
`Returning to Figure 18-6, the curves, although much differ-
`ent in shape, have approximately the same areas (A = 34.4 jlg/
`mL x h ; B = 34.2 j.ig/mL x h), and both formulations can be
`considered to deliver the same amount of drug to the systemic
`circulation. Thus, one can see that AUC should not represent
`the only criterion on which bioequivalence is judged. All there-
`sults, as a composite, must be considered in reaching a decision
`about bioequivalence since no single parameter is adequate to
`serve this purpose.
`The plasma concentration- time curve is the focal point of
`bioequivalence assessment and is obtained when serial blood
`samples are analyzed for drug concentration. The concentra-
`tions are plotted on the ordinate (y-axis), and the times after
`drug administration that the samples were obtained are plotted
`on the abscissa (x-axis).
`A drug product is administered orally at time zero, and the
`plasma drug concentration at this time clearly should be zero.
`As a product passes through the gastrointestinal (GI) tract, it
`must u ndergo a sequence of events depicted in Figure 18-1. As
`
`AUC(o-E;) is used for bioequivalence analyses when the AUC(O·I) makes up ~ 80% of the AUC(o- £;)· AUC(O-I) is used when the AUC(O·I)
`makes up < 80% of the AUC(o-E;)· When drugs with long half-lives are evaluated, AUC(o-r) may sometimes be used with a truncated
`time point.
`Area under the concentration-time curve from time zero to time t (AUCo.t)
`1. Plot the concentration-time data for each subject
`2. Divide the curve into trapezoids by drawing vertical lines from each datum point to the x -axis. Calculate the area of the trapezoids
`using the following formula:
`3. AUC(t2-t1l = ((C2 + C1)(t2 - t1)] I 2
`AUC(o-t) is then calculated by summing the individual area.s to the time of the last concentration:
`4. AUC(o-t) = AUC(t2·l1) + AUC(t3-t2) + ... + AUC(In-(ln-1))
`Area under the concentration-time curve from time zero to infinity (AUCo-E)
`5. To calculate AU~(O-E;)• the tail region of the c urve must be added to AUC(O-I): AUC(o-E;) = AUC(O·Il + AUC "tail"
`6. AUC "tail" = C1!"-z_, where: C1 is the last detectable c oncentration, and "-._ is the terminal elimination rate const ant (see Figure 18-9).
`
`AMN1067
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`354
`
`PHARMACEUTICAL ANALYSIS AND QUALITY CONTROL
`AUC0
`
`_
`
`c
`.Q
`~ <:
`~ c
`
`0
`(.)
`
`Sampling time
`Figure 18-7. Graphical depiction of use of the trapezoidal rule for
`calculating the area under the concentration-time curve.
`
`the drug is absorbed, increasing concentrations of the drug are
`observed in successive samples until the maximum concentra-
`tion is achieved. This point of maximum concentration (C,wx)
`is the peak of the concentration-time curve. If a simple model
`describes the pharmacokinetics of the drug tested, the peak
`concentration represents approximately the point in time when
`absorption and elimination of the drug have equalized.
`The section of the curve to the left of the peak represents
`the absorption phase (or absorption and distribution), during
`which absorption predominates over elimination. The section
`of the curve to the right of the peak is called the elimination
`phase, during which el

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket