throbber
REVIEW
`
`IV-IVC Considerations in the Development of
`Immediate-Release Oral Dosage Form
`
`SHOUFENG LI,1 HANDAN HE,2 LAKSHMAN J. PARTHIBAN,1 HEQUN YIN,3 ABU T.M. SERAJUDDIN4
`
`1Pharmaceutical Development Section, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover,
`New Jersey 07936
`
`2Absorption, Distribution, Metabolism and Excretion (ADME) Section, Novartis Pharmaceuticals Corporation,
`One Health Plaza, East Hanover, New Jersey 07936
`
`3Exploratory Clinical Development Section, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover,
`New Jersey 07936
`
`4Science, Technology and Outsourcing Section, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover,
`New Jersey 07936
`
`Received 23 July 2004; revised 25 November 2004; accepted 28 January 2005
`
`Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.20378
`
`ABSTRACT: Predictive scientific principles and methods to assess in vivo performance of
`pharmaceutical dosage forms based on in vitro studies are important in order to minimize
`costly animal and human experiments during drug development. Because of issues
`related to poor solubility and low permeability of newer drug candidates, there has in
`recent years been a special focus on in vitro–in vivo correlation (IV-IVC) of drug products,
`particularly those used orally. Various physicochemical, biopharmaceutical, and physio-
`logical factors that need to be considered in successful IV-IVC of immediate-release oral
`dosage forms are reviewed in this article. The physicochemical factors include drug
`solubility in water and physiologically relevant aqueous media, pKa and drug ionization
`characteristics, salt formation, drug diffusion-layer pH, particle size, polymorphism of
`drug substance, and so forth. The biopharmaceutical factors that need to be considered
`include effects of drug ionization, partition coefficient, polar surface area, etc., on drug
`permeability, and some of the physiological factors are gastrointestinal (GI) content, GI
`pH, GI transit time, etc. Various in silico, in vitro, and in vivo methods of estimating drug
`permeability and absorption are discussed. Additionally, how IV-IVC may be applied to
`immediate-release oral dosage form design are presented. ß 2005 Wiley-Liss, Inc. and the
`American Pharmacists Association J Pharm Sci 94:1396–1417, 2005
`Keywords:
`in vitro in vivo correlation; formulation; bioavailability; oral absorption;
`permeability; solubility; dissolution; particle size; in silico modeling; in vitro models;
`animal models
`
`INTRODUCTION
`
`Correspondence to: Abu T.M. Serajuddin (Telephone: (862)
`778-3995, Fax: (973) 781-8487;
`E-mail: abu.serajuddin@novartis.com)
`
`Journal of Pharmaceutical Sciences, Vol. 94, 1396–1417 (2005)
`ß 2005 Wiley-Liss, Inc. and the American Pharmacists Association
`
`The rate and extent of drug absorption after oral
`administration of a dosage form are dependent
`on various physicochemical and physiological
`factors.1– 3 The physicochemical factors include
`ionization constant, partition coefficient, solubi-
`
`1396
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0001
`
`

`

`lity, dissolution rate, crystal form, surface area,
`etc., of drug substances as well as the nature and
`properties of dosage forms. Some relevant phy-
`siological
`factors are: solubility of
`the drug
`substance in gastrointestinal (GI) environment,
`drug permeability through GI membrane, GI pH
`profile, GI transit time, presence of bile salts and
`other physiological surfactants, effect of food, and
`so forth. Since the pioneering works by Edwards4
`and Nelson5,6 in the 1950s in correlating dissolu-
`tion rates of aspirin and theophylline, respec-
`tively, with their in vivo appearance after oral
`administration, there have been a gradual in-
`crease in publications related to the influence of
`in vitro characteristics of drug substances and
`drug products on their in vivo performance. Early
`studies on in vitro–in vivo correlation (IV-IVC)
`were reviewed by Dakkuri and Shah7 and by
`Abdou.8
`IV-IVC has attained a much greater signifi-
`cance in pharmaceutical dosage form development
`and especially in immediate-release formula-
`tions during the past decade. As indicated by
`Serajuddin9 in a commentary in 2002, widespread
`application of combinatorial chemistry and high
`throughput screening in recent years has drama-
`tically increased the lipophilicity of new drug
`candidates and, as a consequence, the aqueous
`solubility decreased substantially. A look at drugs
`that were the subjects of intense scientific and
`regulatory scrutiny in 1970s and 1980s because of
`bioavailability issues show that most of them had
`aqueous solubility in the range of 20–100 mg/mL.
`In the present days, the situation has changed so
`much that drug candidates with intrinsic solubi-
`lity of less than 1 mg/mL are very common. Special
`IV-IVC considerations are needed to cope with this
`situation during drug development. To address
`issues related to low drug solubility and drug
`permeability, there have also been various scien-
`tific and regulatory initiatives over the past few
`years to classify drug molecules in different
`categories based on their physicochemical proper-
`ties10 and then address their in vivo performance
`accordingly.11,12
`The objective of the present review is to take a
`fresher look into various IV-IVC considerations
`necessary for the development of
`immediate
`release oral dosage forms. In particular, issues
`related to physicochemical properties and perme-
`ability of candidates will be discussed, and how
`such issues may be addressed by in silico model-
`ing and in vivo experimentations will also be
`considered.
`
`IV-IVC CONSIDERATIONS
`
`1397
`
`PHYSICOCHEMICAL PROPERTIES
`
`Most of the models or tools utilized in predicting
`oral absorption during dosage form development
`are heavily dependent on physicochemical proper-
`ties of drug candidates. Some of these models are:
`the pH-partition hypothesis13 utilizing ionization
`and partition coefficient of drugs as a function of
`pH; absorption potential (AP)14 based on physico-
`chemical properties; mass balance15,16 based on
`solubility and assuming that the small intestine
`as a tube; and compartmental absorption, such as
`mixing tank and compartmental absorption and
`transit (CAT) models.17– 19 The Rule of Five,20
`which provides experimental and computational
`approaches to estimate solubility and perme-
`ability in drug discovery and early development
`settings,
`is also dependent on chemical and
`physical properties of new drug molecules. Some
`of the physicochemical properties that influence
`rate and extent of drug absorption and relevant
`to oral dosage form development are discussed
`below.
`
`Solubility
`
`Solid drug substances must dissolve before
`absorption, and the rate of dissolution depends
`on drug solubility. However, the solubility of a
`compound must be considered together with its
`dose for an evaluation of its AP. Johnson and
`Swindell21 proposed a relatively simple approach
`for estimating maximum absorbable dose (MAD):
`MAD ¼ S  Ka  SIWV  SITT
`where S is solubility (mg/mL) at pH 6.5, Ka is
`intestinal absorption rate constant (min1) ob-
`tained from rat intestinal perfusion experiment
`that has been considered to be similar to human
`Ka, SIWV is small intestinal water volume (mL),
`which is considered to be 250 mL, and SITT is the
`residence time of drug in small intestine, gener-
`ally assumed to be 3 h. The MAD concept serves
`as an initial guide to ascertain whether a
`compound might have potential dissolution and
`absorption issues and whether considerations for
`special dosage forms to overcome or minimize
`such issues have to be made.
`In calculating MAD and, practically, for any
`other estimation of drug absorption, questions
`usually arise with respect to what drug solubility
`to use. In drug discovery settings, kinetic solubility
`based on the kinetics-driven turbidimetric light
`scattering method is generally used,20 while
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0002
`
`

`

`1398
`
`LI ET AL.
`
`thermodynamic equilibrium solubility values are
`usually measured for dosage form development.
`Further, the ‘‘in vivo solubility’’ in the GI tract,
`which is a function of concentration of lipids,
`surfactants and mixed micelles present in intest-
`inal fluids, might be more relevant to in vivo
`settings and for IV-IVC.22 –24 For compounds with
`logP > 3, it has been recommended that solubility
`and dissolution rate in simulated gastric and
`intestinal fluids or in human GI aspirates should
`be used for IV-IVC, since regular in vitro dissolu-
`tion media used for quality assurance purposes do
`not contain bile salts, lecithin, lipid digestion
`products, etc.25 For instance, when solubilities of
`danazol (logP 4.5) were compared in simulated
`gastric fluid (SGF), simulated intestinal fluid
`(SIF), and human aspirates, the solubility in
`human aspirates was found to be much higher
`than that in buffers alone.26 However, when dif-
`ferent concentrations of bile salts, lecithin, and
`sodium lauryl sulfate were added to buffers, the
`solubility varied widely depending on nature and
`concentration of the added components. Therefore,
`in the determination of solubility for in vivo
`relevance, consideration must be given to thecom-
`position of solvents used.
`
`pKa
`
`Solubility of a weak acid or a weak base is de-
`pendent on its ionization constant and intrinsic
`solubility (S0; solubility of unionized or non-
`protonated species) and, therefore, a function of
`pH of the dissolution medium. There are some
`excellent reports on the pH-dependence of drug
`solubility.27 –29 Both pKa and S0 are important in
`dictating what would be the drug solubility under
`gastrointestinal (GI) pH conditions; a relatively
`low pKa of an acidic drug or a relatively high pKa
`of a basic drug do not necessarily assure high drug
`solubility in the GI pH range unless the intrinsic
`solubility is also high. Figure 1A shows simulated
`pH-solubility profiles of four basic drugs where
`pKa is kept constant at 8.0 and S0 ranges from 0.1
`to 100 mg/mL. For the compound with S0 of 0.1 mg/
`mL, the solubility in the typical intestinal pH
`condition of 6 to 7 would be in the range of 1–
`10 mg/mL only, and the solubility must be lowered
`below 4 to obtain a solubility of 1 mg/mL and
`higher. One aspect of the pH-solubility profile
`that is not shown in Figure 1A is the solubility
`of the salt form. The pKa does not keep on
`increasing with a decrease in pH, as shown in
`Figure 1A,
`indefinitely; at a certain pH, the
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`Figure 1.
`(A) Theoretical pH-solubility profile of a
`weak base with pKa of 8.0 and different intrinsic
`solubility. (B) Theoretical pH-solubility profile of a weak
`base with So of 0.1 mg/mL and different pKa values.
`
`total solubility exceeds the solubility of its salt
`form, preventing any further increase.29 As
`expected, the solubility is also dependent on the
`pKa value; Figure 1B shows the simulated pH-
`solubility profiles of four free bases as a function
`of pKa values, where the S0 value has been kept
`constant.
`The pH gradient inherent to the GI tract in-
`fluences and gives rise to areas of preferred
`absorption. Drugs with pKa values in the range of
`1 to 8 undergo considerable changes in the
`equilibrium between their ionized and unionized
`species as they transit through different regions
`of GI tract having different pH conditions. This
`effect, coupled with the solubility differences
`shown in Figure 1, gives rise to pH-dependent
`absorption. For example, the dissolution of a basic
`drug, ketoconazole, at pH 6 after 1 h is only 10%,
`while at pH < 3 more than 85% dissolves in less
`than 5 min.30 This pH-dependence was evident
`in vivo when ranitidine was coadministered with
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0003
`
`

`

`ketoconazole to raise the gastric pH to 6; the Cmax
`of ketoconazole decreased from 8.2 to 0.6 mg/mL
`and the AUC decreased from 37.1 to 1.6 mg/h/mL.31
`Similarly, a decrease of almost 85% in Cmax and
`AUC values of cinnarizine was observed in
`subjects with elevated gastric pH as compared to
`those exhibiting low gastric acidity.32 Although it
`is often assumed that a basic drug dissolves well
`under the gastric environment, these examples
`illustrate that the actual amount dissolved could
`indeed be variable and depends on such factors as
`the pH and the gastric residence time. One way of
`improving the situation, as discussed under the
`next heading in this article, is to administer a
`base in its salt form, which usually has a higher
`dissolution rate.
`Potential effects of pKa on drug solubility,
`dissolution rate, and absorption need to be inves-
`tigated at the early stage of drug development.
`When dissolution is the issue, appropriate for-
`mulation strategy to ensure the desired disso-
`lution rate at the absorption site is necessary. Ho
`et al.33 demonstrated the interplay of pH, pKa,
`partition coefficient, concentrations of
`ionized
`versus unionized species, and permeability coeffi-
`cients of aqueous boundary layers for a series of
`beta blockers by using cell culture monolayers.
`Structural modification of drug molecule can
`ensure optimal drug absorption. Derivatization
`into prodrugs or substitution of isosteres may also
`improve oral absorption of ionizable lipophilic
`molecules.34
`
`Salt Formation
`Since the early studies by Nelson,5,6 there have
`been numerous reports on the effect of salt forma-
`tion on drug dissolution35– 40 and its potential
`effect on bioavailability. A salt usually provides a
`higher dissolution rate than that of its free acid or
`base form by modifying pH and solubility in the
`diffusion layer at the surface of dissolving solid.
`Earlier work on physicochemical and biopharma-
`ceutical aspects of pharmaceutical salts were
`reviewed by Berge et al.35 It may also be pointed
`out here that, under certain GI pH conditions, a
`free base may have higher dissolution rate than
`that of its salt form. For example, Serajuddin and
`Jarowski39 reported that the dissolution rate of
`the free base form of phenazopyridine at pH 1 is
`higher than that of the hydrochloride salt; how-
`ever, if the full GI pH range (1–7) is considered,
`the salt still provides a superior pH-dissolution
`profile.
`
`IV-IVC CONSIDERATIONS
`
`1399
`
`Because of pH-dependent solubility, there is a
`potential that free acid or base forms may pre-
`cipitate out during the dissolution of salts under
`certain GI pH conditions. Depending on pKa and
`solubilities of free forms, the precipitation of free
`acid occurs at a relatively lower pH, i.e., in the
`stomach, while the free base could precipitate at a
`relatively higher pH, i.e., in the intestine. This
`may be exemplified by the dissolution of phenytoin
`sodium under GI pH conditions. Phenytoin is
`an acidic compound with a pKa of 8.4 and the
`solubility of 37 mg/mL at 378C between pH 1 and
`6.41 Therefore,
`it is expected that after oral
`administration as a sodium salt at a dose of 100
`to 300 mg, it would precipitate out in its free acid
`form under the acidic environment of the stomach.
`It had been suggested that the precipitation of
`phenytoin acid in a very finely divided state would
`facilitate its rapid redissolution and faster absorp-
`tion,42 and later it was confirmed that it is indeed
`the case.41 Figure 2 shows that when a 100-mg
`phenytoin sodium capsule was dissolved in 500 mL
`of a pH 2 medium, an equilibrium was established
`(20% dissolved) in <10 min and the excess drug
`precipitated out as free acid. However, when an
`additional 500 mL of medium was added to the
`dissolution vessel, a new equilibrium was estab-
`lished in <5 min. Figure 2 also shows that there
`
`Figure 2. Dissolution profiles of fast-release pheny-
`toin sodium formulation (100 mg) in (&) water and (~)
`pH 2.1 medium at 378C. Each data point represents the
`average of three determinations. (Reproduced from Ref.
`41 with permission of copyright owner.)
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0004
`
`

`

`1400
`
`LI ET AL.
`
`was no precipitation of phenytoin when water by
`itself was used as the dissolution medium, because
`the pH of the medium increased gradually with the
`dissolution of salt. This is, however, not the case in
`GI fluids where acidity, buffering action, etc.,
`would keep pH relatively lower that could cause
`drug precipitation.
`Despite the high dissolution rate of phenytoin
`sodium and the rapid redissolution rate of pheny-
`toin acid that precipitated out in the stomach,
`phenytoin sodium dosage forms have been the
`subject of extensive IV-IVC studies. It was noted
`that there was a greater incidence of side effects if
`the innovator’s product was substituted with an
`equivalent dose of products from other manufac-
`turers,43 which was due to 2–3 mg/mL higher
`steady state plasma level from the non-innovators’
`products.44 To address this issue, the FDA classi-
`fied the innovator’s product as an ‘‘extended’’ or
`slow-release dosage form and the others as
`‘‘prompt’’ or fast-release dosage form.45,46 How-
`ever, considering the long half life (22  9 h)
`and Michaelis–Menten parameters of phenytoin,
`Sawchuk and Rector47 determined that any varia-
`tion in the release rate of phenytoin from dosage
`forms should not influence its steady state plasma
`level unless the extent of absorption is changed.
`They did not see any difference in plasma level
`when a situation assuming instantaneous absorp-
`tion of phenytoin was compared with a situation in
`which a continuous constant rate of absorption
`occurred. Indeed, Arnold et al.48 and, later Ser-
`ajuddin and Jarowski,41 demonstrated that there
`is a potential for incomplete release and, as a
`result, lower bioavailability of phenytoin from
`certain phenytoin sodium products. This example
`of phenytoin sodium highlights the importance
`of selecting not only a salt form but also of an
`appropriate dosage form for a drug to attain
`optimal plasma concentration and bioavailability.
`A similar situation may also arise for the salts of
`basic drugs, especially under intestinal pH condi-
`tions. If higher dissolution rate for a salt form cannot
`be ensured, a free base form may even be preferred.49
`If other factors remain constant, the dissolution
`rate of a compound should determine the rate of
`build-up of blood levels with time and the max-
`imum levels obtained. Nelson has reported that
`the rank order of dissolution rates correlated well
`with clinically determined blood levels.5 Correla-
`tion of urinary excretion rates and dissolution
`rates of tetracycline and some of its acid salts was
`also demonstrated by Nelson.6 Relative bioavail-
`ability of the vasodilator naftidrofuryl in oxalate
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`and citrate salt forms has shown that the rate of
`absorption is higher for the citrate than for the
`clinically used oxalate form of the drug.50 Lin
`et al.,51 on the other hand, reported no enhance-
`ment in bioavailability when salts of a basic
`antihypertensive agent, having significantly dif-
`ferent intrinsic dissolution rates, were compared.
`
`Diffusion Layer pH
`
`The effect of diffusion layer pH has been men-
`tioned above in relation to the dissolution of free
`acids and bases versus their salts. Such an effect
`should also be considered in assessing the dis-
`solution of one particular form of a compound (for
`example, free acid) under different pH conditions.
`It was reported that the diffusion layer pH on the
`dissolving surface of benzoic acid (pHh ¼ 0) would
`remain practically constant around 3 while the
`pH of unbuffered bulk media may range from 3
`to 11,36,37 and, as a result, the dissolution rate
`within such a wide pH range would also remain
`practically unchanged. A self-buffering effect to
`maintain steady state pH in the diffusion layer
`may vary from compound to compound, depend-
`ing on solubility and/or pKa values. Figure 3
`shows the effect of the pH of dissolution medium
`on the dissolution of three compounds having
`similar pKa values, namely benzoic acid, 2-
`naphthoic acid, and indomethacin.36 In this figure,
`the increase in dissolution rate (flux) relative to
`that of the unionized species (N/N0) has been
`plotted. Despite pH-dependent solubility of all
`three compounds studied, pH of dissolution
`medium (bulk pH) had minimal effect on the
`
`Figure 3. Relative flux (N/N0) versus pHbulk for
`several carboxylic acids at 258C. N0 is the respective
`flux at pH 2. (Reproduced from Ref. 36 with permission of
`copyright owner.)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0005
`
`

`

`dissolution of benzoic acid and 2-naphthoic acid,
`while a more pronounced effect was noticed in the
`dissolution of indomethacin. Indomethacin has
`much lower solubility values at different pH
`than that of the other two acids and, as a result,
`its self-buffering effect in the diffusion layers is
`limited. The buffer capacity of a dissolution
`medium also plays a critical role in modulating
`microenvironmental pH of a drug substance and,
`therefore, its dissolution rate.
`pH and drug solubility in the diffusion layer of a
`particular compound may be calculated theoreti-
`cally.36,38 Serajuddin and Jarowski37 described a
`practical method of estimating diffusion layer pH
`of drug substance as the thickness of the layer
`approaches zero (pHh ¼ 0) and thereby predicting
`its dissolution rate under different pH conditions.
`The authors established that the pH of a saturated
`solution of a drug substance (acid, base, or salt) in a
`particular medium is equivalent to pHh ¼ 0, and,
`therefore, the drug solubility at this pH would
`represent Cs,h ¼ 0, i.e, solubility in the diffusion
`layer as the thickness of the layer approaches zero.
`The flux (J) in the same medium is then deter-
`mined experimentally by determining dissolution
`rate from a constant surface area. After the
`determination of J/Cs,h ¼ 0 from these two data,
`the pHh ¼ 0 in a second medium would be deter-
`mined. The measurement or theoretical calcula-
`tion of saturation solubility at this new pH and
`then solving for J as per the equation, J/Cs,h ¼ 0 ¼ k,
`under sink condition, would give the dissolution
`rate in the second medium. The authors suggested
`that this method may also be applicable to dissolu-
`tion media containing buffering agents because
`any change in buffer type, buffer capacity, etc.,
`would be reflected in pHh ¼ 0.
`As apparent from the above discussion, various
`acidic and alkaline components added to dissolu-
`tion media or dosage forms would influence drug
`dissolution because of their effects on diffusion
`layer pH. However, one area that has not been
`explored adequately is the effect of counterions
`added to dissolution media or present in GI fluid
`on dissolution of pharmaceutical salts, where a
`common ion effect is not apparent. Li et al.52
`recently reported that the dissolution rate of halo-
`peridol mesylate (methanesulfonate) is adversely
`influenced by NaCl added to dissolution media.
`The presence of chloride ion converts the mesylate
`salt to a hydrochloride salt, possibly in the dif-
`fusion layer at the surface of the solid, thereby
`making its solubility and dissolution rate suscep-
`tible to the common ion effect.
`
`IV-IVC CONSIDERATIONS
`
`1401
`
`Particle Size
`
`Amidon et al.10 described the concept of dissolu-
`tion number, Dn, in relation to drug absorption.
`Dn may be defined as:
`Dn ¼ tres
`tdiss
`
`where tres is the mean residence time of drug in
`the GI tract and tdiss is the time required for a
`particle of the drug to dissolve. The higher the Dn,
`the better the drug absorption, and a maximal
`drug absorption may be expected when Dn is
`greater than 1, i.e., tres > tdiss. However, for most
`poorly water-soluble drugs, Dn < 1 and, as a result
`a dissolution rate-limited absorption is expected.
`As will be discussed later in this paper, tres is a
`physiological factor that is difficult to change by
`dosage form design. Therefore, a more practical
`way of increasing Dn is by decreasing tdiss, that is,
`by increasing the dissolution rate. One commonly
`used method of increasing dissolution rate is by
`reducing the drug particle size.
`It may be of interest to examine how the particle
`size reduction would increase surface area and,
`consequently, the dissolution rate of drug sub-
`stances. If the size of one specific drug particle is
`reduced from a radius of r1 to a radius of r2, the
`total number of particles produced will be
`ð4=3Þpr3
`¼ r3
`1
`ð4=3Þpr3
`r3
`2
`
`1
`
`2
`
`According to this equation, if the size of a particle is
`reduced, say, by a factor of 5, the total number of
`new particles produces will increase by a factor of
`(r1)3/(r1/5)3 or 125. Despite 125 times increase in
`drug particles, the surface area will, however,
`increase by a factor of only (4pr22  125)/(4pr1
`
`2) or 5.
`Thus, it is apparent that the surface area and,
`therefore, the dissolution rate would increase only
`linearly with the decrease in particle size. For
`example, if the initial particle size of a drug sub-
`stance is 10 mm in diameter and the size is reduced
`to 3–4 mm by micronization, which might be the
`lowest limit of particle size reduction by conven-
`tional milling, only 2–3 times increase in dissolu-
`tion rate may be achieved. This is assuming that no
`conversion of drug substance to an amorphous
`form that could enhance dissolution or no agglom-
`eration of drug particles that could reduce dissolu-
`tion have occurred.
`Johnson and Swindell21 analyzed the effect of
`particle size on drug absorption as a function of
`dose as well as drug solubility (Fig. 4), where
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0006
`
`

`

`1402
`
`LI ET AL.
`
`Figure 4. Computed percent of dose absorbed at 6 h
`versus mean particle size, with an absorption rate
`constant of 0.001 min1: (A) at doses from 1 to 100 mg,
`with a solubility of 0.001 mg/mL; and (B) at solubility
`from 0.001 to 1.0 mg/mL, with a dose of 1 mg. (Replotted
`from data in ref. 21.)
`
`the absorption rate constant was assumed to be
`0.001 min1. For dissolution rate-limited absorp-
`tion (Dn < 1), Figure 4A shows that the fraction of
`drug absorbed will decrease with the increase in
`dose. On the other hand, if the dose is kept
`constant, Figure 4B shows that the fraction of
`drug absorbed will increase with an increase in
`drug solubility, and the particle size becomes
`practically irrelevant for drugs with a solubility
`of 1 mg/mL at a dose of 1 mg.
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 94, NO. 7, JULY 2005
`
`Polymorphism
`
`Since the publication of the effect of polymorph-
`ism on the absorption of chloramphenicol from
`chloramphenicol palmitate suspensions by Aguiar
`et al.,53 the interest in polymorphism in drug
`development has increased greatly. Indeed, in
`certain circles, polymorphism has become synon-
`ymous with bioavailability issues. One must,
`however, be cautious in generalizing the findings
`of Aguiar et al.53 In their study, the solubility of
`Polymorph B used was observed to be four times
`higher than that of Polymorph A, while its bio-
`availability in humans was over 20 times higher;
`however, no reason for
`the disproportional
`increase in bioavailability was given. Also, inter-
`estingly, when three different suspensions of the
`same polymorphic form (Polymorph B) having
`three different particle sizes (1, 5, and 25 mm)
`were given to humans, no difference in bioavail-
`ability was observed. The suspension with 1-mm
`particles was ‘‘well-dispersed’’ because of the
`presence of a surfactant, while those with 5 and
`25-mm particles were ‘‘highly aggregated’’ since no
`surfactant was added. Based on this consideration
`alone, the surface area of 1-mm suspension should,
`respectively, be over 5 and 25 times higher than
`that of 5 and 25-mm suspensions, and these
`differences should be reflected in dissolution rate
`and bioavailability. Since no difference in bioa-
`vailability was seen, the authors concluded that
`‘‘the polymorphic form is of much greater sig-
`nificance than the particle size.’’ An alternative
`explanation of the results obtained by Aguiar et al.
`might be necessary. Since chloramphenicol pal-
`mitate undergoes hydrolysis into chlorampheni-
`col prior to absorption, its in-situ hydrolysis in the
`gut may complicate the over-all picture of its
`intestinal absorption. Thus, the situation with
`chloramphenicol palmitate could be different
`from common cases where polymorphism influ-
`ences drug solubility and dissolution and, there-
`fore, it may not be generalized.
`Recently, Pudipeddi and Serajuddin54 reported
`that solubility values of the polymorphic forms of
`most drugs reported in the literature do not differ
`more than two times, and any two polymorphic
`forms hardly show greater than five times differ-
`ence in solubility. Such a difference may or may not
`have biopharmaceutical significance depending
`on, as discussed earlier, doses used, particle sizes,
`and individual solubility values. Therefore, any
`potential influence of polymorphism on bioavail-
`ability and the risks involved because of their
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1045-0007
`
`

`

`potential interconversion in a dosage form need
`to be carefully investigated during dosage form
`development. In addition to solubility, polymor-
`phism may influence physical stability and proces-
`sing parameters for a dosage form, which need to
`be addressed separately.
`Unlike polymorphism, a conversion from crys-
`talline to amorphous form or vice versa might
`have a more profound effect on drug dissolution.
`Hancock and Parks55 reported that solubility of
`amorphous drug substances could be higher by
`factors of tens or even hundreds than that of their
`crystalline forms.
`
`BIOPHARMACEUTICAL PROPERTIES
`
`Drug Permeability
`
`Besides solubility and dissolution, permeability
`of a compound plays a major role in its absorption
`after oral administration. Transcellular perme-
`ability (Pm) of lipophilic compounds depends on
`their solubility in GI membrane lipids relative to
`their aqueous solubility. This is defined as the
`ratio of the product of membrane diffusivity (Dm)
`and membrane–water partition coefficient (Kp) to
`the membrane thickness (Lm):
`Pm ¼ KpDm=Lm
`Therefore, a variation in membrane permeabi-
`lity for a particular compound will depend on the
`fluidity of membrane and partition coefficient of
`compound.56
`Models to estimate or measure permeability
`of a compound range from simple to complex. Drug
`ionization, logP, artificial membrane permeabil-
`ity, Caco-2 cell line permeability and other in vitro
`epithelial cell culture permeability, as well as
`in situ animal perfusion study and human jejunum
`permeability (Peff), have variously been used. They
`will be discussed in the following sections. In
`general, a compound is considered highly perme-
`able when its absorption is determined to be 90%
`or higher.
`
`Ionization
`
`Under the simplest scenario, considering that
`membrane uptake of unionized solute is favored
`over that of ionized solute by the membrane-to-
`water partition coefficient (Kp), membrane perme-
`ability would reflect the solute ionization curve
`at Kp ¼ 1. When Kp is higher (i.e., in the case of
`lipophilic drugs), membrane uptake of unionized
`
`IV-IVC CONSIDERATIONS
`
`1403
`
`Figure 5. Relative absorption rate for a weak acid
`(pKa ¼ 3) as a function of mucosal pH for increasing
`(membrane) permeability (Pb) with fixed
`barrier
`unstirred aqueous layer permeability (PUL). X ¼ pH
`inflection point. (Reproduced from reference 57 with
`permission of copyright owner.)
`
`drug would shift the equilibrium in order to
`replace the unionized solute removed by the
`membrane. This would shift the sigmoidal curves
`of membrane permeability versus pH towards
`basic pH for weakly acidic drugs and acidic pH for
`weakly basic drugs.57 The shifts may be described
`by changes in inflection or half-maximal pH
`points (pHmax/2) as demonstrated in the following
`equation and in Figure 5:
`pHmax=2 ¼ pKa  log½1 þ ðPm=Paqފ
`where, Paq ¼ aqueous boundary layer perme-
`ability, Pm ¼ membrane permeability. The above
`equation describes the absorption profiles of
`sufficiently lipophilic drugs. It simply states that
`half maximal rate of absorption occurs at pH
`values close to their pKa and that shifts in
`equilibrium can be accounted for based on an
`analysis of membrane permeability against aqu-
`eous boundary layer permeability. At pH values
`where the drug is fully ionized and the equation
`above has accounted for the permeability shifts, a
`non-zero permeability would suggest that per-
`meation occurs through routes other than simple
`lipid partitioning such as pore transport or
`paracellular transport. For drugs with half-max-
`imal pH points in the range

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