throbber

`
`
`
`MARTINEZ AND AMIDONUNDERSTANDING THE FACTORS AFFECTING DRUG ABSORPTIONPHARMACOKINETICS AND PHARMACODYNAMICS
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`A Mechanistic Approach to Understanding
`the Factors Affecting Drug Absorption:
`A Review of Fundamentals
`
`Marilyn N. Martinez, PhD, and Gordon L. Amidon, PhD
`
`This article provides an overview of the patient-specific and
`drug-specific variables that can affect drug absorption fol-
`lowing oral product administration. The oral absorption of
`any chemical entity reflects a complex spectrum of events.
`Factors influencing product bioavailability include drug sol-
`ubility, permeability, and the rate of in vivo dissolution. In
`this regard, the Biopharmaceutics Classification System has
`proven to be an important tool for predicting compounds
`likely to be associated with bioavailability problems. It also
`helps in identifying those factors that may alter the rate and
`extent of drug absorption. Product bioavailability can also be
`markedly influenced by patient attributes such as the integ-
`
`rity of the gastrointestinal tract, physiological status, site of
`drug absorption, membrane transporters, presystemic drug
`metabolism (intrinsic variables), and extrinsic variables such
`as the effect of food or concomitant medication. Through an
`awareness of a drug’s physicochemical properties and the
`physiological processes affecting drug absorption, the skilled
`pharmaceutical scientist can develop formulations that will
`maximize product availability. By appreciating the potential
`impact of patient physiological status, phenotype, age, gen-
`der, and lifestyle, dosing regimens can be tailored to better
`meet the needs of the individual patient.
`Journal of Clinical Pharmacology, 2002;42:620-643
`©2002 the American College of Clinical Pharmacology
`
`A fundamental premise associated with the use of a
`
`therapeutic agent is that for any given patient, the
`clinical response can be predicted on the basis of the
`selected drug product, dose, and dosing regimen. This
`tenet provides the foundation for concepts of
`prescribability and switchability.1 Prescribability refers
`to an assumed relationship between a therapeutic out-
`come and the rate and extent of drug exposure. A physi-
`cian will prescribe a particular product in accordance
`with assumptions pertaining to this relationship.
`Generally, the process of drug movement from in-
`take (e.g., oral delivery systems) to its site of action can
`be schematically presented as follows (Figure 1).
`As depicted in Figure 1, the relationship between
`drug intake and a clinical response is highly complex,
`potentially affected by a host of intrinsic and extrinsic
`
`From the Office of New Animal Drug Evaluation, HFV-130, Center for Vet-
`erinary Medicine, Food and Drug Administration, Rockville, Maryland (Dr.
`Martinez) and the University of Michigan, College of Pharmacy, Ann Arbor,
`Michigan (Dr. Amidon). Submitted for publication August 15, 2001; re-
`vised version accepted February 15, 2002. Address for reprints: Marilyn N.
`Martinez, Office of New Animal Drug Evaluation, HFV-130, Center for
`Veterinary Medicine, Food and Drug Administration, 7500 Standish Place,
`Rockville, MD 20855.
`
`620 • J Clin Pharmacol 2002;42:620-643
`
`variables. Accordingly, deviations between drug re-
`sponse within or between individuals may be ascribed
`either to product bioavailability (i.e., the rate and extent
`of drug absorption), drug pharmacokinetics (which in-
`cludes the metabolism, distribution, and elimination of
`a compound), or the particular concentration-effect
`relationship.
`While product formulation can significantly affect
`processes leading to drug absorption, once in the circu-
`lation, the original formulation is generally considered
`to no longer affect the ultimate drug response. In other
`words, it is the concentration of the drug moiety, along
`with its corresponding effect, that will ultimately de-
`termine product safety and effectiveness. For this rea-
`son, once a patient is titrated to a particular product
`and dosing regimen, we assume that a comparable clin-
`ical response will be achieved if the patient elects to
`take a less expensive generic equivalent.
`The purpose of this article is to discuss basic princi-
`ples associated with the process of drug absorption.
`Special attention will be given to the use of the
`Biopharmaceutics Classification System (BCS) as a
`predictive tool for identifying compounds whose ab-
`sorption characteristics may be sensitive to intrinsic
`
`CELGENE 2141
`APOTEX v. CELGENE
`IPR2023-00512
`
`

`

`UNDERSTANDING THE FACTORS AFFECTING DRUG ABSORPTION
`
`ORAL DRUG INTAKE
`
`Drug Disintegration and Dissolution
`Loss via Luminal Degradation
`
`Diffusion through Gastrointestinal Fluids
`
`Membrane Permeation
`
`Uptake into Blood or Lymph
`
`Drug in Systemic Circulation
`
`Loss via Mucosal Metabolism
`
`Loss via Hepatic Metabolism
`
`Bound Drug
`
` Unbound Drug
`
` Clearance
`
`Bound Tissue Drug
`Concentrations
`
`Free Tissue Drug
`Concentrations
`
` Active Site Concentrations
`
`EFFECT
`
`Figure 1. Schematic diagram of the relationship between an oral
`dose of a drug product and its ultimate effect.
`
`(physiological) and extrinsic (e.g., food and formula-
`tion) variables. Accordingly, this review focuses on
`those factors that can affect drug dissolution, aqueous
`solubility, membrane permeability, and presystemic
`drug metabolism.
`
`WHAT IS THE BCS?
`
`One of the most significant prognostic tools created to
`facilitate product development in recent years has been
`the BCS.2 By knowing the solubility and permeability
`characteristics of specific compounds, we improve our
`ability to predict those variables (such as formulation,
`food, dosing regimen, and disease) that will alter oral
`drug absorption.
`Currently, all pharmaceutical compounds are
`grouped into one of the following categories:
`
`Class I—high solubility, high permeability: generally very
`well-absorbed compounds
`Class II—low solubility, high permeability: exhibit disso-
`lution rate-limited absorption
`Class III—high solubility, low permeability: exhibit per-
`meability rate-limited absorption
`
`Class IV—low solubility, low permeability: very poor oral
`bioavailability
`
`Solubility is calculated on the basis of the largest
`strength manufactured. It is defined as the minimum
`solubility of drug across a pH range of 1 to 8 and at a
`temperature of 37 ± 0.5°C. High-solubility drugs are
`those with a ratio of dose to solubility volume that is
`less than or equal to 250 ml. Permeability (Peff, ex-
`pressed in units of 104 cm per second) is defined as the
`effective human jejunal wall permeability of a drug.
`High-permeability drugs are generally those with an
`extent of absorption greater than or equal to 90% and
`are not generally associated with any documented in-
`stability in the gastrointestinal tract.
`It is interesting to note that for certain compounds,
`Peff is not necessarily constant. For example, nonlinear
`changes in Peff were observed with increasing doses of
`the surface-active molecule, fluvastatin. This nonlin-
`ear increase in Peff was attributed to its effects on mem-
`brane surface tension and to a possible decrease in
`poly-glycoprotein (P-gp) activity associated with an in-
`crease in intestinal membrane fluidity.3
`The application of this system to nonhuman species
`may require adjustment of these classification parame-
`ters based on physiological differences in gastric vol-
`ume and the pH of the gastrointestinal (GI) fluids. Ac-
`cordingly, at this time, we cannot be certain that the
`BCS classification of a compound remains constant
`across all species. This question is currently being ex-
`plored by the Food and Drug Administration’s (FDA’s)
`Center for Veterinary Medicine.
`By understanding the relationship between a drug’s
`absorption, solubility, and dissolution characteristics,
`it is possible to define situations when in vitro dissolu-
`tion data can provide a surrogate for in vivo
`bioequivalence assessments. The use of this surrogate
`relies on the validity of three fundamental assump-
`tions. First, it must be assumed that a comparison of
`product in vitro dissolution performance accurately re-
`flects relative differences in product in vivo dissolu-
`tion behavior. Second, we must assume that if two
`products present with equivalent in vivo dissolution
`profiles under all luminal conditions, they will present
`equivalent drug concentrations at absorptive mem-
`brane surfaces. Third, for comparable dissolution pro-
`files to ensure comparable in vivo absorption, the rate
`and extent of drug presented to absorptive membrane
`surfaces must determine the absorption characteristics
`of that drug product.
`Lobenberg and Amidon4 have summarized the rela-
`tionships between dose, dissolution characteristics,
`drug solubility, and drug absorption properties. These
`relationships can be described as follows:
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`621
`
`

`

`MARTINEZ AND AMIDON
`
`1. Absorption number (An) = (Peff/R) • <Tsi>,
`where R is the gut radius and <Tsi> the residence time
`of the drug within the intestine.
`2. Dissolution number (Dn) = (3D/r2) • (Cs/ρ) • <Tsi>,
`where D is the diffusivity of the dissolved drug, ρ is the
`density of the dissolved drug, Cs is the drug solubility,
`and r is the initial radius of the drug particle.
`M / V
`,
`3. Ratio of dose to dissolved drug (D0) =
`0
`C
`where M is the dose of the drug and V0 is the volume of
`fluid consumed with the dose.
`The fraction of drug absorbed is closely related to the
`drug’s effective permeability across mucosal cells.4 If
`the Peff of a drug is less than 2 • 10–4 cm/s, then drug ab-
`sorption will be incomplete, whereas complete absorp-
`tion can be expected for substances whose Peff exceeds
`this value. For poorly soluble drugs, critical variables
`include the volume of the intestinal fluids, GI pH, and
`GI transit time (where adequate time is needed to dis-
`solve poorly soluble materials). For these lipophilic
`compounds, food and bile salts may increase drug
`solubility.
`Class I compounds are highly permeable and readily
`go into solution (Dn > 1). In this case, the fraction ab-
`sorbed (F) can be expressed as follows:
`
`s
`
`F = 1 – exp(–2An).
`
`For these agents, as “An” increases, the fraction of
`drug absorbed increases, with 90% absorption (highly
`permeable compounds) occurring when An = 1.15. Re-
`ferring back to the equation for An, we see that F can be
`affected by a change in the compound’s membrane per-
`meability, the gut radius of the host, or the intestinal
`transit time. Based on these factors alone, it is evident
`that differences in GI physiology due to factors such as
`disease, age, or animal species can alter the value of An
`and, therefore, the fraction of drug absorbed.
`For Class II drugs (high permeability, low solubility),
`Dn < 1. In these cases, the relationship between D0 and
`Dn is critical for determining the fraction of drug ab-
`sorbed, and the rate of drug dissolution tends to be the
`rate-limiting step. Accordingly, anything that increases
`the rate and extent of in vivo dissolution will also in-
`crease the bioavailability of that compound.
`
`SOLUBILITY
`
`Aqueous solubility can be estimated by determining
`the ability of a drug to partition from lipid to aqueous
`environments. This partitioning behavior is often a
`function of solvent pH due to the latter’s effects on drug
`
`622 • J Clin Pharmacol 2002;42:620-643
`
`ionization. In general, ionized drugs tend to exhibit far
`greater aqueous solubility than the un-ionized counter-
`part. Consequently, the rate of solute dissolution in
`aqueous media can be markedly affected by the pH of
`that solvent.
`To examine the effect of pH on drug ionization, one
`can use a rearrangement of the Henderson-Hasselback
`equation:5
`
`Weak acid: % un-ionized = 100/(1 + antilog (pH-pKa)).
`Weak base: % un-ionized = 100/(1 + antilog (pKa-pH)).
`
`Weakly basic drugs tend to have a slower dissolution
`rate at higher pH (when more drug exists in its un-
`ionized form), whereas weakly acidic drugs dissolve
`faster at higher pH (when more drug exists in its ion-
`ized form). Examples of the relationship between the
`percentage of drug in its un-ionized form as a function
`of drug pKa and pH are found in Figures 2 and 3. For
`this reason, by increasing the proportion of drug exist-
`ing in its un-ionized state, meals that elevate gastric pH
`can decrease the dissolution of a weak base. For exam-
`ple, weak bases such as indinavir (with pKa of 3.7 and
`5.9) are expected to precipitate when gastric pH is ele-
`vated during a meal, resulting in a significant reduction
`in AUC and Cmax values in fed versus fasted human sub-
`jects.6 Conversely, the same meal can increase the dis-
`solution rate of a weak acid by increasing the propor-
`tion of drug existing in its ionized state, thereby making
`it more water soluble.7
`By definition, solubility is the extent to which mole-
`cules from a solid are removed from its surface by a sol-
`vent. While solubility may be expressed in many ways,
`some generalizations can be made:8
`
`Very soluble: Less than 1 part solvent needed to dissolve 1
`part solute
`Freely soluble: From 1 to 10 parts solvent needed to dis-
`solve 1 part solute
`Soluble: From 10 to 30 parts solvent needed to dissolve 1
`part solute
`Sparingly soluble: From 30 to 100 parts solvent needed to
`dissolve 1 part solute
`Slightly soluble: From 100 to 1000 parts solvent needed to
`dissolve 1 part solute
`Very slightly soluble: From 1000 to 10,000 parts solvent
`needed to dissolve 1 part solute
`Practically insoluble: More than 10,000 parts solvent
`needed to dissolve 1 part solute
`
`A compound’s aqueous solubility, as measured by its
`propensity to distribute between octanol and water, is a
`function of its ability to form hydrogen bonds with the
`water molecule. Generally, aqueous solubility is di-
`rectly proportional to the number of hydrogen bonds
`that can be formed with water.9 As discussed later,
`
`

`

`UNDERSTANDING THE FACTORS AFFECTING DRUG ABSORPTION
`
`percent drug
`unionized
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`pKa = 2
`pKa = 4
`pKa = 6
`pKa = 8
`
`0
`
`1
`
`2
`
`3
`
`4
`pH of solution
`
`5
`
`6
`
`7
`
`8
`
`Figure 2. Relationship between percentage of drug un-ionized, and
`pH and pKa of weak acids.
`
`percent drug
`unionized
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`pKa = 2
`pKa = 4
`pKa = 6
`pKa = 8
`
`0
`
`1
`
`2
`
`4
`3
`pH of solution
`
`5
`
`6
`
`7
`
`8
`
`Figure 3. Relationship between percentage of drug un-ionized, and
`pH and pKa of weak bases.
`
`while very high aqueous solubility is beneficial for
`drug dissolution in aqueous media, these same com-
`pounds often exhibit low permeability due to their
`high polarity and poor lipophilicity.
`Although lipid/water partitioning is often used to
`describe drug solubility, there is some evidence that
`solubility may better be described by the compound’s
`dynamic energy properties.10 The solubility parameter
`of any compound can be described in terms of the en-
`ergy required to fragment a molecule into its constitu-
`ent atoms (its cohesive energy). When described in
`terms of the square root of its cohesive energy density
`(energy of vaporization per unit volume), the solubility
`parameter will lie on a scale from 10 (nonpolar) to 48
`(water). For two materials to be miscible, their solubil-
`ity parameters must be similar.
`When comparing the percent absorbed versus log P
`(octanol/water partition coefficient), the percent ab-
`sorbed versus the solubility parameter, and the percent
`absorbed versus the number of hydrogen bond accep-
`tors, it was noted that a high level of negative correla-
`
`tion was observed for the latter two relationships. Con-
`versely, a large degree of scatter was observed in the re-
`lationship between permeability and log P. It was also
`noted that highly permeable compounds tend to have
`solubility parameter values almost identical to that of
`biological membranes (solubility parameter values of
`20-26 MPa1/2). Accordingly, thermodynamic consider-
`ations rather than physicochemical interactions10 may
`serve as the best predictors of a compound’s membrane
`permeability.
`
`IN VIVO DRUG ABSORPTION
`
`Efforts are currently under way to identify molecular
`quantitative structure-bioavailability relationships
`(QSBR) that predict drug bioavailability.11 Factors that
`negatively influence bioavailability include the num-
`ber of hydrogen bond donors, the presence of heavy at-
`oms, and the inclusion of fragments such as tetrazole,
`4-animopyridine, and benzoquinone. Factors that tend
`to enhance drug bioavailability include the presence of
`hydrogen acceptors, low molecular weight, and the
`presence of fragments such as azide, salicylic acid, and
`amides.
`To understand reasons for these structure-
`bioavailability relationships, it is important to recog-
`nize the complex series of events that occur during the
`process of drug absorption. Molecular movement
`across lipid bilayers, such as those existing within bio-
`logical membranes, is extremely complex due to the re-
`gional differences in membrane polarity, hydrophobicity,
`and density. Generally, the bilayer can be divided into
`four distinct regions.12 These include the following:
`
`1. The first (outermost) region contains a high proportion
`of water molecules and may be the region responsible
`for interactions with other membranes and proteins.
`2. The second region has the highest molecular density
`of all four regions (contains the polar headgroups),
`contains little or no water, and exerts the greatest
`barrier to solute diffusion (due to its density
`characteristics).
`3. The third region contains the highest density of
`nonpolar tails. This region serves as the primary bar-
`rier to membrane penetration and is primarily respon-
`sible for the limitations in molecular size and shape
`associated with membrane transport.
`4. The fourth region is the most hydrophobic region of
`the membrane, serving as a hydrophobic barrier in
`membrane transport.
`
`Owing to this membrane structure, it is evident that drug
`permeability is not a simply two-step process of
`solubilization and diffusion but rather represents a
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`623
`
`

`

`MARTINEZ AND AMIDON
`
`spectrum of complex molecular events. Accordingly,
`intestinal permeability reflects a multifunctional inter-
`action of factors such as molecular size (negatively cor-
`related), lipophilicity (positively correlated), polar van
`der Walls surface area (negatively correlated), and the
`molecular flexibility (intramolecular hydrogen bond
`formation).13
`In addition to cellular membrane barriers to drug
`diffusion, significant impedance is also effected by the
`components of the gastric and intestinal mucous
`layer.14 In examining the relative contribution of these
`various components, it was noted that lipid constitu-
`ents such as phosphatidyl choline, cholesterol, and
`linoleic acid significantly retard the diffusion of small
`lipophilic molecules such as propranolol and hydro-
`cortisone. Conversely, small hydrophilic molecules
`such as mannitol appear to freely diffuse through this
`lipoid barrier. Mucous gel-forming components, such
`as mucin and DNA, exert far less negative effects on the
`diffusion of lipophilic molecules. However, they may
`serve to block the diffusion of peptides and proteins.
`Molecular flexibility and the corresponding ability
`to undergo conformation changes can significantly af-
`fect the polar surface area of a molecule. The polar sur-
`face area and nonpolar surface area are powerful pre-
`dictors of intestinal permeability, being respectively
`inversely and directly related to membrane permeabil-
`ity.15 However, any particular set of descriptors may not
`adequately predict membrane permeability across
`nonhomologous compounds.9 Another important vari-
`able is the strength of the hydrogen bonds formed be-
`tween the molecules of water and solute.15 It is gener-
`ally assumed that these bonds must be broken
`(desolvation) before the solute can traverse the biologi-
`cal membrane. Which of these factors play the domi-
`nant role in determining drug permeability may vary
`across homologous drug series.
`Despite these complexities, certain generalizations
`can be made with regard to drug absorption processes.
`For example, the vast majority of orally administered
`drugs are absorbed via passive transcellular transport.15
`This necessitates that the drug traverse through a
`highly lipophilic membrane. Accordingly, diffusion
`processes are governed by Fick’s laws of diffusion and
`therefore influenced by the compound’s lipophilicity.
`This ability to diffuse through lipids has been found to
`be highly correlated with the ability of a drug to parti-
`tion between water and an organic solvent such as
`octanol. In fact, when expressed as log P0 (based on par-
`titioning between n-octanol and water), the optimal
`partition coefficient for a drug generally falls within the
`range of 2 to 7.5 Nevertheless, exceptions do occur, and
`while transcellular transport generally occurs when
`
`624 • J Clin Pharmacol 2002;42:620-643
`
`the compound is un-ionized, recently, several ionized
`molecules have been shown to be absorbed via
`transcellular processes.16,17 This finding reinforces ear-
`lier statements regarding the degree of scatter associ-
`ated with the relationship between log P and drug
`absorption.
`In addition to passive mechanisms, active transport
`is important to the absorption of several compounds.
`Both active and passive transport mechanisms may oc-
`cur simultaneously for the same molecule. Which of
`these mechanisms has the dominant role tends to be
`compound specific and may not be well predicted by
`in vitro systems.15 Nevertheless, it must be remem-
`bered that even active transport mechanisms require
`that the drug penetrate the intestinal cells via the
`transcellular route.
`The rate of passive diffusion of any molecule,
`whether it be absorbed via transport between mucosal
`cells or through the mucosal membrane, can be de-
`scribed by the following equation:18
`
`dM
`dt
`
`=
`
`A
`m
`
`•
`
`D
`m

`
`•
`
`C
`
`membrane
`
`•
`
`C
`
`lumen
`
`+
`
`•
`
`J
`
`fluid
`
`+
`
`K
`C
`
`m
`
`lumen
`
`J
`max
`+
`C
`lumen
`−
`•
`
`(1 α ,)
`
`+
`
`•
`
`A
`
`p
`
`D

`
`a
`q
`
`aq
`
`
`
`where
`dM/dt
`
`Daq
`
`aq
`

`Jfluid

`
`K
`
`Dm
`
`Jmax
`
`Km
`
`= the effective rate of passive drug
`absorption (concentration/time).
`= the diffusion coefficient of the
`compound in water.
`= the aqueous diffusion distance.
`= the fluid flow between epithelial cells.
`= the ratio of the water flow relative to the
`solute flux, both under the influence of
`the existing pressure gradient, and is
`dependent on molecular size, volume,
`charge, and hydration number. It may
`also be influenced by the dynamic
`width of the tight junction.
`= the partition coefficient describing the
`relative tendency of the substance to
`dissolve in the membrane phase (Cmembrane)
`as compared to the surrounding aqueous
`phase (Clumen).
`= the diffusion coefficient of the
`compound within the membrane, which
`is dependent on factors such as drug
`lipophilicity, hydrogen bonding
`capacity, polar surface area of the
`molecular, molecular volume, and
`shape.
`= the maximal transport capacity of the
`carrier-mediated process.
`= the substrate specificity of the
`membrane transporter (the Michaelis
`constant).
`
`

`

`UNDERSTANDING THE FACTORS AFFECTING DRUG ABSORPTION
`
`where dm/dt is the dissolution rate, expressed as the
`change in the amount of drug dissolved (m) per unit
`time (t); D is the diffusion coefficient; S is the surface
`area; h is the thickness of the diffusion film adjacent to
`the dissolving surface; Cs is the saturation solubility of
`the drug molecule; Ct is the concentration of the dis-
`solved solute; and V is the volume of the dissolution
`medium.
`Upon integration, this equation can be expressed as
`follows:25
`
`−
`
`exp(
`
`−
`
`Kt
`
`))
`
`C V
`
`
`
`s (1
`
`m =
`
`and
`
`K = S • D/h.
`
`The viscosity of the GI contents,23,26 as described by
`the following equation, can affect D, the diffusion
`coefficient:
`
`D = (KbT)/(6πR0η),
`
`
`where D is the diffusivity of a compound, Kb is the
`Bolzmann constant, T is the temperature, R0 is the sol-
`ute radius, and η is the viscosity of the diffusion
`medium.
`As seen in the latter equation, increasing the surface
`area of a particle (S) can enhance its dissolution rate. S
`can be increased by micronization, a process some-
`times applied to poorly water-soluble compounds.24
`Also to be considered are particle size and density,
`which both inversely affect the dissolution rate.27 Parti-
`cle shape is also important in determining the dissolu-
`tion behavior of a drug, and for many crystalline forms
`(particularly shapes of needles and platelets), shape
`and consequently dissolution behavior may change
`markedly as the particle dissolves.28,29
`Particle properties can also affect the rate of GI tran-
`sit, the latter being highly dependent on such proper-
`ties as size, shape, and density. For example, in swine,
`about 30% to 40% of ingested food materials pass into
`the duodenum within 15 minutes in an adult pig. How-
`ever, large particles (10 mm in diameter) are retained
`within the stomach of swine for several days, with resi-
`dence time increasing with particle density and
`length.30
`A problem with the Noynes-Whitney equation is
`that there is an inherent assumption that S remains
`constant over time. Unfortunately, this assumption is
`incorrect, and the S of powders and immediate-release
`preparations tends to decrease as dissolution pro-
`

`
`= the thickness of the rate-limiting
`diffusion barrier.
`Am and Ap = the available surface areas for
`transcellular and paracellular
`transport, respectively.
`
`Thus, for passive diffusion, whether a drug is absorbed
`via paracellular or transcellular mechanisms is deter-
`mined by both physicochemical and physiological
`factors.
`Paracellular diffusion involves both diffusion and a
`convective volume flow through water-filled
`intercellular channels whose diameter is approxi-
`mately 3 to 10 Å in humans.18 Accordingly, the size and
`number of paracellular spaces influence the intestinal
`absorption of most hydrophilic compounds. This, in
`turn, is affected by the mucosal surface area and by cel-
`lular density.19 Therefore, it is not surprising that the
`bioavailability of small hydrophilic compounds tends
`to be greater in species such as dogs, in which both pore
`diameter and surface area tend to exceed that in hu-
`mans.20 In humans, the small intestinal surface area for
`paracellular absorption is approximately 0.01% of the
`total membrane surface area. For this reason, unless the
`molecule is extremely small (e.g., < 200 Da),
`paracellular transport will have a minor role in drug ab-
`sorption.15
`With permeability-limited absorption, we can ex-
`pect that although the fraction of dose absorbed re-
`mains unchanged, the absolute amount of drug ab-
`sorbed will increase as dose increases (assuming linear
`kinetics). Conversely, for solubility-limited com-
`pounds, increasing the dose will have no effect on the
`absolute amount of drug absorbed. Consequently, in
`these cases, the fraction of dose absorbed will decrease
`as dose is increased.21 Moreover, permeability is not
`necessarily constant throughout the GI tract. While for
`some compounds, drug absorption appears to be site
`independent,22 for others, it is site dependent.23 When
`drug absorption is site dependent, the availability of
`dissolved drug at the absorption site can be the
`rate-limiting factor in product bioavailability.
`
`PRODUCT DISSOLUTION
`
`Drug absorption depends on delivery of the drug parti-
`cles to its site of absorption. The Noynes-Whitney
`equation describes the variables that can affect drug
`dissolution:24,25
`
`(C
`
`−
`Cs
`
`t
`
`),
`
`• •
`
`D S
`V h
`
`=
`
`dm
`dt
`
`PHARMACOKINETICS AND PHARMACODYNAMICS
`
`625
`
`

`

`MARTINEZ AND AMIDON
`
`drug absorbed. In this situation, fraction of drug ab-
`sorbed can only be improved by enhancing drug
`solubility (e.g., via the inclusion of surfactants in the
`product formulation). Conversely, particle size exerts
`its greatest effect when solubility is not a problem. In
`these cases, a significant improvement in the fraction of
`drug absorbed can be achieved by increasing surface
`area (i.e., decreasing particle size).
`The FDA’s Center for Drug Evaluation and Research
`(CDER) has written a guidance that provides for the
`waiver of in vivo bioequivalence study requirements
`for high-solubility/high-permeability drug products
`based on in vitro dissolution data. The scientific and
`regulatory considerations that must be applied to these
`procedures are described in the CDER guidance titled
`Waiver of In Vivo Bioavailability and Bioequivalence
`Studies for Immediate-Release Solid Oral Dosage
`Forms Based on a Biopharmaceutics Classification
`System (August 2000). For Class I compounds, the
`bioequivalence of generic or revised versions of mar-
`keted drug products can be confirmed if the sponsor can
`demonstrate that gastric emptying is the rate-limiting
`step in product absorption.
`To be granted a waiver of in vivo bioequivalence
`study requirements, the CDER recommends that in vi-
`tro dissolution tests be conducted under the following
`conditions:
`• The test apparatus is USP Apparatus I at 100 rpm or
`Apparatus II at 50 rpm. Testing is conducted in 900 ml
`of each of the following dissolution media: (1) 0.1 N
`HCl or Simulated Gastric Fluid USP without enzymes,
`(2) a pH 4.5 buffer, and (3) a pH 6.8 buffer or Simulated
`Intestinal Fluid USP without enzymes.
`• For each formulation, a minimum of 12 dosage units is
`evaluated to support a biowaiver request. Samples
`should be collected at time intervals adequate for char-
`acterizing the dissolution profile of the drug product.
`When comparing the test and reference formulations,
`the respective dissolution profiles should be compared
`using the similarity factor (f2), where
`
` 
`
`,
`
`•
`
`100
`
`−
`0 5
`.
`
` 
`
`−
`R T
`t
`t
`
`(
`
`2
`
`)
`
`t
`
`+
`
`
`
`1( /
`
`n
`
`)
`
` 
`
`1
`
` 
`
`=
`
`•
`
`50
`
`log
`
`f
`
`2
`
`n
`
`∑
`
`=
`
`1
`
`where Rt is the percent dissolved of the reference prod-
`uct, Tt is the percent dissolved of the test product, and n
`is the number of units tested.
`
`• Two dissolution profiles are considered similar when
`the f2 value is ≥ 50. To allow the use of mean data, the
`
`ceeds.25 In vivo, many of these parameters are also in-
`fluenced by the conditions of the GI tract, which will
`vary over time.31 For this reason, these equations, while
`providing insights into the parameters that can affect
`drug dissolution, should not be blindly applied to pre-
`dict in vivo dissolution rate.
`It should be noted that because the Noynes-Whitney
`equations are unable to adequately model either
`S-shaped data or data with a steep initial slope, the
`more general Weibull distribution has been used to de-
`scribe dissolution profiles.25 The Weibull distribution
`can be described as follows:
`M = 1 – exp(–αtβ),
`
`where M is the accumulated fraction of material in so-
`lution at time t, α is a scale parameter, and β is a shape
`parameter, where β = 1 indicates an exponential rela-
`tionship, β > 1 indicates an S-shaped relationship, and
`β < 1 indicates an exponential relationship with a steep
`slope.
`The rate of product dissolution may or may not in-
`fluence the resulting plasma concentration-time pro-
`file. For example, Class I compounds (highly soluble,
`highly permeable) may exhibit marked difference in
`the in vitro dissolution profiles without any resulting
`differences detected in product bioavailability.32-34 In
`these instances, gastric emptying is slower than prod-
`uct dissolution. Accordingly, it is the rate of gastric
`emptying rather than product performance that is the
`rate-limiting step in determining the bioavailability
`characteristics of that formulation. Similarly, highly
`soluble, poorly permeable compounds (Class III) dis-
`solve rapidly. However, in these cases, it is not the rate
`of drug dissolution that is usually rate limiting but
`rather the rate of permeation across biological mem-
`branes. Therefore, we can again assume that so long as
`dissolution is faster than the rate of gastric emptying,
`product dissolution will not determine product
`bioavailability. In the case of Class III compounds, so
`long as absorption occurs via linear processes, the ab-
`solute amount of drug absorbed may be increased by
`increasing the dose.21
`On the other hand, for high-permeability, low-
`solubility compounds (Class II), the rate and extent of
`product dissolution will have a significant role in de-
`fining the resulting blood concentration-time profile.27
`This may be attributable to problems associated with
`either particle size (termed dissolution-limited absorp-
`tion) or drug solubility (termed solubility-limited ab-
`sorption). In the case of solubility-limited absorption,
`particle size exerts minimal effect on the fraction of
`
`626 • J Clin Pharmacol 2002;42:620-643
`
`

`

`UNDERSTANDING THE FACTORS AFFECTING DRUG ABSORPTION
`
`coefficient of variation should not exceed 20% at the
`earlier time points (e.g., 10 minutes) or 10% at all other
`time points. If, under all dissolution conditions, both
`the test and reference products dissol

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