throbber
Encyclopediaof
`
`Pharmaceutical Technology
`
`Second Edition
`
`Volume 1
`A—D
`
`Pages 1-1032
`
`edited by
`
`James Swarbrick
`
`President
`
`PharmaceiiTech, Inc., Pinehurst, North Carolina
`and
`
`Vice President for Scientific Afiairs, aaiPharma, Inc.
`Wilmington, North Carolina, U.S.A.
`
`and
`
`James C. Boylan
`
`Pharmaceutical Consultant
`
`Gurnee, Illinois, U.S.A.
`
`
`
`MARCEL DEKKER, INC.
`
`NEW YORK ° BASEL
`
`Amgen Ex. 2006
`
`Complex Innovations v. Amgen
`
`|PR2016-00085
`
`Amgen Ex. 2006
`Complex Innovations v. Amgen
`IPR2016-00085
`
`

`
`Cover Art: Leigh A. Rondano, Boehringer angelheim Pharmaceuticals, Inc.
`
`ISBN: Volume 1:
`Volume 2:
`Volume 3:
`
`Prepack:
`
`0-8247-2822-X
`0-8247-2823-8
`0-8247-2824-6
`
`0-8247-2825-4
`
`ISBN: Onlinez
`
`0-8247-2820-3
`
`This book is printed on acid-free paper.
`
`Headquarters
`Marcel Dekker, Inc.
`270 Madison Avenue, New York, NY 10016
`tel: 212-696-9000; fax: 212-685-4540
`
`Eastern Hemisphere distribution
`Marcel Dekker AG
`
`Hutgasse 4, Postfach 812, CH-4001 Basel, Switzerland
`tel: 41-61-261-8482; fax: 41-61-261-8896
`
`World Wide Web
`http://www.dekker.com
`
`Copyright © 2002 by Marcel Dekker, Inc. except as occasionally noted on the opening page of each article. All Rights Reserved.
`
`Neither this book nor any part may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopying,
`microfilming, and recording, or by any information storage and retrieval system, without permission in writing from the publisher.
`
`Current printing (last digit)
`
`10
`
`9
`
`8
`
`7
`
`6
`
`5
`
`4
`
`3
`
`2
`
`1
`
`PRINTED IN THE UNITED STATES OF AMERICA
`
`

`
`colon cancers. Oral drug delivery systems (DD
`S) can ,_
`classified into three categories:
`immediate—re1
`preparations, control1ed—release (CR) preparations’ a
`F
`targeted-release preparations. This chapter describes “I
`recent technological advances in oral drug delivery an‘
`various physicochemical and biological barriers, and als
`provides some insight on future strategies to improve or 1-
`drug delivery.
`-
`
`
`
`ANATOMICAL AND PHYSIOLOGICAL
`CHARACTERISTICS OF THE GI TRACT
`
`Mechanisms and Pathways of Drug Absorption
`Orally administered drugs are mainly absorbed in th
`small intestine (duodenum, jejunum, and ileum) and int a‘.
`large intestine (colon); however, other regions, such ‘._
`buccal cavity, stomach, and rectum, also can be considered
`potential sites for drug absorption. The various anatomica
`and physiological characteristics of each segment at
`briefly described in Table 1.
`Once at the surface of intestinal epithelium, a drug c Ir_
`be absorbed across by one or a combination of th
`following mechanisms: passive transcellular, passiv
`paracellular, and carrier— and receptor—mediated transpol;
`systems. Paracellular transport involves the passage '|'
`drug molecules through aqueous pores created 0'
`epithelial tight junctions, and is the most likely route '1-
`polar, hydrophilic drugs since they exhibit poor rnembfl ‘I’
`partitioning. In the human small intestine, the averag‘3 5‘ ‘
`of these water—filled pores is approximately 7-9 A "'
`jejunum and 3-4
`for ileum (2).
`In the colon,
`f‘
`estimated pore radii are about 8-9 A, albeit this Value
`"
`obtained from rat colon (3). Nevertheless, the extent "
`paracellular transport is limited as tight junctions C0mP_ _‘
`only about 0.01% of the total absorptive surface area of
`intestine (villi) (4). Transcellular absorption inV0_IVeS I
`transport of drugs through the intestinal epithelial c :
`(enterocytes) and requires partitioning of drugs across I‘,
`the apical and the basolateral membranes. Obvi0lJS1Y’ :.'
`route is mainly limited to the transport of relatiV513f
`molecular weight lipophilic drugs. Furthermore, Stud”
`
`
`
`h 01 '
`.
`Encyclopedia ofPharn1aceutzcfll Tecesrgr
`,
`.
`Copyright © 2002 by Marcel Dekker, Inc. All rights r
`
`DRUG DELIVERY—ORAL ROUTE
`
`Kwon H. Kim
`
`Brahma N. Singh
`St. John's University, Jamaica, New York
`
`INTRODUCTION
`
`Oral drug delivery is the most desirable and preferred
`method of administering therapeutic agents for their
`systemic effects. In addition, the oral medication is generally
`considered as the first avenue investigated in the discovery
`and development of new drug entities and pharmaceutical
`formulations, mainly because of patient acceptance,
`convenience in administration, and cost—effective manu-
`facturing process. For many drug substances, conventional
`immediate—release formulations provide clinically effective
`therapy while maintaining the required balance of
`pharmacokinetic and pharmacodynamic profiles with an
`acceptable level of safety to the patient.
`However, the potential for oral dosage form develop-
`ment is sometimes limited for therapeutic agents that are
`poorly absorbed in the gastrointestinal
`(GI)
`tract and
`unstable to various enzymes, in particular, to proteolytic
`enzymes, such as peptide and protein drugs. The overall
`process of oral delivery is frequently impaired by several
`physiological and pharmaceutical challenges that are
`associated with the inherent physicochemical nature of the
`drugs and/or the variability in GI conditions, such as pH,
`presence of
`food,
`transit
`times,
`expression of
`P—Glycoprotein (P-Gp) and CYP3A, as well as enzymatic
`activity in the alimentary canal. Manipulation of these
`problems and challenges is considered an important
`strategy for improving oral drug delivery, and requires
`thorough understanding and appropriate integration of
`physicochemical principles, GI physiology and biochem-
`istry, polymer science, pharmacokinetics, and pharmaco-
`dynamics. Over the last 3 decades, much research effort
`has been made in this area to address various biological
`and technological issues. Research has opened many novel
`avenues for
`the more effective,
`sustained, or
`rate-
`controlled oral delivery of both existing and new
`therapeutic agents,
`including peptide and protein drugs
`emerging from the biotechnology arena.
`Furthermore,
`the oral
`route offers an attractive
`approach of drug targeting at the specific sites within GI
`tract for the treatment of certain pathological conditions,
`such as gastroesophageal reflux "disorder, gastroduodenal
`ulcers,
`inflammatory bowel disease, and stomach and
`
`886
`
`

`
`CW.
`
`O
`
`e
`
`788
`
`
`
`
`
`.a2>§dowmwumom550was
`
`
`
`dommfiofidmazmfi
`
`
`
`
`
`%uwombH058::MOmoumtouofimao~wo.~wo_o?¢.Enew~mo_E8m:<Hofimh
`
`Emmofiao
`
`
`
`xlra.Em_:nw..oo.5:Zina_m:n.._Hw..E§..<
`
`
`
`
`Rwiuaaoov®o:uEmo._gov:onE.5ma<no.8ounmuamfiwnoq
`
`
`w3:855muoficwasmefizuamcumuflmnrmEvmcacmno«E5932:3FEWAFCnflwom
`
`
`
`N3JOE.:E:2"woman:5.5mummu?miAS<.0dfieNofiaaem
`
`
`
`EmmmhoE~Eo-.omA«A:émmhu..mEommamcm3M“<OMdowe8:525
`
`mumQ35=25
`
`ocwmflfi
`
`
`
`mommflosa.323
`
`
`
`
`
`dwfiofimAme.8335dmflmfim,EmmoHm—omnwnooZfie.m.<,05ooo.m825?.
`
`
`
`.8835
`
`
`
`momwm:dummwumom
`
`
`EfiocanmA53Aommnflouuunovommcnmumouounmumqmnoozwinam.<"O.momo.vBaum
`
`.§&:fimommcumom
`
`
`
`
`
`
`
`:3.momSBmohmomfiosuomo.wm.0.n_madmm;dO—OU
`
`
`
`mumauuoofiosndomwflosc
`
`:9omfiaooz24we3uwbfi
`
`3m.<"U.mmodS.on8.o8:80
`
`onumuafi
`
`
`
`omfimbsm.mommEEm
`
`.m8nEmoobm
`
`asmfio858%
`
`
`
`
`
`émotmoouumHmEommadhham:3HHunommnmbwoufifiommHmEommnmbo>uo<H<wtommmabo>too>aoOHUuuommnmbwfimmamHm
`
`
`
`
`
`2.3.see
`
`

`
`Drllg Del1ve1.y\or
`
`
`
`phases (11, 12). Phase I (basal state) is a quiesce
`mp
`about 45-60 min without any contractions, exc
`occasions. Phase II (preburst state) is a periodept ‘u_-.
`duration (30-45 min)
`that consists of imof
`"
`peristaltic contractions that gradually increasfi ins‘ ._..
`and frequency. Phase III (burst state) is a Short
`“_l
`consists of large, intense peristaltic C0HtI‘aQtiQl:1:I‘1
`about 5 - 15 min. This phase serves to sweep the unadt,
`materials out of the stomach and for this reason
`:~.
`contractions are known as “housekeeper” waves’ p '
`As phase III of one cycle reaches the end Ofith
`ileum (ileocecal junction), phase I of the next cycle...
`in the stomach (proximal) or esophagus (lower es0p;,_
`sphincter). However, sometimes MMC may or-lgi In
`the duodenum or jejunum and some MMC may “__
`action potentials strong enough to traverse throu
`entire small intestine (12). Phase IV is a brief transi .1,
`phase (0-5 min) that occurs between phase III and p
`of two consecutive cycles.
`In the fed state, the onset of MMC is delayed. In .3.
`words, feeding results into delayed gastric emptying .
`duration of this delay is mainly dependent on the size (L.
`or heavy) and composition (fatty or fibrous) of the
`Consequently, the fate of pharmaceutical dosage in
`mainly subject to the pattern of GI motility in faste
`fed) state at the time of dosage administration.
`
`,
`
`PROBLEMS AND BARRIERS TO
`ORAL DRUG DELIVERY
`
`
`
`The biggest problem in oral drug delivery is low
`erratic bioavailability, which mainly results from on
`more factors
`such as poor aqueous
`solubility,
`s .-
`dissolution rate, low intestinal permeability, instabili-
`GI milieu, high first—pass metabolism through liver -Fr
`intestine variable GI transit, and P-gp mediated ill-
`This, in turn, may lead to unreproducible clinical resp I‘
`or a therapeutic failure in some cases due to subtherape
`plasma drug levels. Indeed, the incomplete and var -1“
`oral bioavailability will have its most serious imp
`‘
`drugs with a narrow “therapeutic window” (13) (
`theophylline, carbamazepine, quinidine, etc.) From '
`economic point of view, low oral bioavailability F6531
`the wasting of a large portion of an oral dose, and adfi
`the cost of drug therapy, especially when the drug ‘I
`expensive one (14). It is, therefore, extremely W190 L.
`that these issues be considered and a suitable techlllqu
`an animal model) be used while estimatlng
`[ll
`contributions from each factor responsible for 10W a
`variable bioavailability.
`
`888
`
`humans have demonstrated that absorption by the
`in the colon
`transcellular route decreases significantly
`(small intestine > ascending colon > transverse colon),
`which has implications for delayed or sustained release
`formulations, whereas no such gradient exists for the
`paracellular route (5).
`Carrier—mediated transport involves interaction of the
`drug with a specific transporter or carrier, in which drug is
`transferred across the cell membrane or entire cell and then
`released from the basal surface of the enterocyte into the
`circulation (6). The process is saturable and utilized by
`small hydrophilic molecules (7). Drugs that are shown to
`be transported by this mechanism include [3-Lactam
`antibiotics, cephalosporins, and ACE inhibitors (7).
`Receptor-mediated transport involves internalization of
`an external substance, which may be a ligand for which
`there is a surface bound receptor, or a receptor which binds
`to a surface located ligand (8). During this process, a small
`region of the cell invaginates and pinches off, forming a
`vesicle. This process, in general, is known as endocytosis
`and comprises phagocytosis, pinocytosis,
`receptor-
`mediated endocytosis (clathrin—mediated), and potocytosis
`(nonclathrin mediated) (9).
`After a drug is absorbed in the GI tract, it can gain
`access to the systemic circulation via two separate and
`functionally distinct absorption pathways—portal blood
`and the intestinal lymphatics. The relative proportion of
`drug absorbed Via these two pathways is largely dictated
`by physicochemical and metabolic features of the drug,
`and the characteristics of the formulation (10).
`The portal blood represents the major pathway for the
`majority of orally administered drugs as it has higher
`capacity to transport both water soluble and poorly water
`soluble compounds (10). During this process, hydrophilic
`molecules are carried to the liver via the hepatic portal
`vein, and then by the hepatic artery gain across to the
`systemic circulation for subsequent delivery to their sites
`of action. On the other hand, highly lipophilic drugs (log P
`> 5) that cross the same epithelial barrier are transported
`to the intestinal lymphatics, which directly delivers them
`to the vena cava, thereby bypassing the hepatic first—pass
`metabolism (10).
`
`Gastrointestinal Motility
`The process of GI motility occurs both during fasted and fed
`states; however, the pattern differs markedly in the two
`states. In the fasted (interdigestive) state, the pattern is
`characterized by a series of motor activities known as
`interdigestive myoelectric cycle or migrating motor
`complex
`(MMC), which
`usually
`occurs
`every
`80-120 min. Each cycle consists of four consecutive
`
`

`
`889
`
`dispersions, supersaturation is maintained for a long period
`due to better dispersion through particle size reduction.
`
`Lipophilicity
`
`The lipid solubility or lipophilicity of drugs has long been
`recognized as a prerequisite for transcellular diffusion
`across the intestinal membrane. Traditionally,
`the
`lipophilicity of drug substances is expressed as
`the
`apparent partition coefficient or distribution coefficient
`(log P) between n-octanol and an aqueous buffer (pH 7.4),
`which is pH-dependent in the case of ionizable compounds
`(29). In general, compounds with low log P are poorly
`absorbed, whereas compounds with log P > —1 offer
`satisfactory absorption (29). It is important, however, that
`the drug possess an optimum lipophilicity, as too low or
`too high lipophilicity may result in less than optimum oral
`bioavailability. For example, Mori et al.
`(30) did not
`observe any significant improvement in oral absorption of
`disodium cromoglycate (log P < -3) despite the fact that
`lipophilicity was increased by derivatization to a lipophilic
`(diethyl) ester prodrug (log P = 1.78). This observation
`was attributed to a marked decrease in the water solubility
`(from 195.3 mg/mL to 0.0052 mg/mL), which occurred
`simultaneously with increased lipophilicity. Similarly,
`Wils et al.
`(31) reported for the first
`time that high
`lipophilicity (log P > 3.5) decreases drug transport across
`the intestinal epithelial cells and could be accounted for
`loss of in vivo biological activity. The “cut—off” point of P
`value, that is, the P value corresponding to an optimal
`transepithelial passage of drugs,. was found to be around
`3000. Other consequences of high lipophilicity may
`include passive exsorption (32), and increased plasma
`protein binding and biliary excretion; the biliary excretion
`may limit the oral bioavailability of polypeptides.
`
`Aqueous boundary layer
`
`The aqueous boundary layer or the unstirred water layer
`(UWL) is a more or less stagnant layer, about 30— 100 am
`in thickness, composed of water, mucus, and glycocalyx
`adjacent to the intestinal wall that is created by incomplete
`mixing of the lumenal contents near the intestinal mucosal
`surface (33). The glycocalyx is made up of sulfated
`mucopolysaccharides, whereas mucus is composed of
`glycoproteins (mucin), enzymes, and electrolytes. Until
`recently, the resistance of the UWL to intestinal absorption
`was believed to be correlated to the effective intestinal
`
`permeability (Peff) values of the solutes; however,
`considerable evidence suggests instead that the available
`surface of the apical membrane of the intestinal mucosa is
`the main barrier
`for both actively and passively
`absorbed solutes (33). It is also interesting to note that
`coadministration of food and prokinetic (motility inducing)
`
`Defive,-y——Oral Route
`
`:1 S
`
`
`
`‘
`
`.
`
`,
`
`‘
`
`sicochemical Barriers to Oral Drug Delivery
`-V
`..
`eons solubility
`5
`long been recognized that before an orally
`hlflfliswred drug becomes available for absorption at
`fig sites within the GI tract, it must be dissolved in the
`id_ Since both the dissolution rate and the maximum
`01:1, of a drug that can be dissolved are dictated by the
`ability ofthe drug in the medium (15), aqueous solubility
`.
`.
`.
`8 drug could be regarded as a key factor responsible for
`‘ Oral bioavailability of poorly water—soluble drugs,
`‘Why 1imiting their therapeutic potential. Other issues
`"Med to low bioavailability for a sparingly soluble drug
`'
`, lack of dose proportionality, substantial food effect,
`I; intra. and intersubject variability, gastric irritancy, and
`I". onset of action (16). These problems are further
`.acerbated when attempts are made to develop CR dosage
`“ms (17). Unfortunately, many potent compounds,
`.cluding new chemical entities (NCES), possess very low
`ueous solubility at physiological pH, which could be
`fibuted to their high inherent lipophilicity incorporated
`.\ drug design in order to ensure good absorption (18).
`nsequently, various approaches are utilized to improve
`ueous solubility, which mainly include chemical
`odification (19), complexation (17, 20, 21), microniza—
`on (22), solubilization using surfactants, solid dispersion
`3, 24), and design of drug delivery systems. However,
`11 ere may be specific practical limitations to each of these
`pproaches. For instance, the salt formation approach is not
`feasible for neutral compounds, and in many cases also for
`_eakly acidic or weakly basic drugs because of the
`raconversion of salts into aggregates of their corresponding
`tee acid or base forms (23). This precipitation effect would
`:l'ivery is low and
`‘ us lead to slower dissolution rates and may cause failure
`esuns from one or
`—va clinical response or epigastric distress due to change in
`s
`solubility,
`.sl0.W _
`RH (24). Sometimes, the rate of renal excretion may also
`bility, instability Ill
`-
`-M9386, particularly when amine drugs are solubilized as
`;h1~ough liver and/07
`:“°‘d'Sa1.lS(25)-
`gp mediated effl1l>_i--
`_ Similarly, the commercial success of solid dispersion
`ale clinical respoll?
`btien limited due to use of higher temperatures
`tue to subthefapfwtii
`.1 10p C) or harmful organic solvents, such as chloroform
`mplete and Van“? r
`0? dichloromethane, which may result
`in chemical
`;t serious impact 0
`,_?“°0mposition of drugs and carriers or possible toxicity
`window” (13) (°~3"’.'
`{gm the residual solvent. These unacceptable problems
`fine, etc.) From
`flh Scientists to explore novel alternatives, particularly
`availability reslglgsfi
`-— e °°gI‘1nding method, which involves cogrinding of a
`ral dose, and 3 .5
`’when the drug 1531 mily Water-soluble drug with a water-soluble polymer,
`extremely irrtporw
`- £1 as Polyvinyl pyrrolidone, polyethylene glycol (PEG),
`suitable technlquemgl
`pol ml")/PTOPY1 cellulose, hY<1roxYlJroPY1m°thY1 0611111036,
`3'
`yvmyl alcohol (26, 27), or sugars such as D—Mannitol
`2
`)nsible for low 3l‘..._
`E0811)‘ These authors suggest that although the composi-
`°f the coground mixture is similar to that of solid
`
`v
`_
`nzvisf th
`_xtcyc1eebdl"‘
`Wet esoph
`13)’ Originate
`C may ROI .1;
`zrse through .'.
`brief t1‘aI1Sltl0.l
`
`;e III and phas II‘
`
`delayed. In oth_'
`ic emptying.
`i.'.
`L on the size (lig I:
`)us) of the meal
`.1 dosage forms. ’
`ility in fasted (d_ '
`tration.
`
`
`
`

`
`_
`therapeutic system (GITS®, Alza Corpor
`provide zero—order drug absorption, in
`hieh atti ..
`drug absorption from the GI tract is Constanha
`determined by the amount of drug available in tht
`However, none of the CR systems can control thee .
`drug absorption from the GI tract. This is
`to the fact that the extent of drug absorpti
`.0n fmm ii.
`regions of the GI tract is different, wh
`lch in fac .
`constitutes a basis for the biopharmaceutic C1aSS_ \.
`scheme for drugs administered as extended.relea :
`products (41). Thus, indirectly the extent of abs
`,
`determined by the GI transit time (GITT) of the _.
`form rather than its CR properties or deliver
`,_
`(42).
`In general,
`the GITT of most drug pr0I'_
`relatively short (8-12 h), which in tum impe
`'
`formulation of a once daily dosage form (43)_
`From the oral delivery standpoint, both .:.
`emptying time (GET) and small
`intestinal transi
`(SITT) are considered important since the majo
`drugs are preferentially absorbed in the upper pan 5-.
`GI tract (stomach, duodenum, and jejunum)_ ‘_
`the stomach and intestines have a limited site f .'
`absorption. This is known as an “absorption win
`The relatively short GET (1-3 h) and SITT (3-5
`provide limited time for drug absorption throu
`major absorption zone. These problems are
`"I"
`aggravated by the highly variable nature of the
`emptying process, which can vary depending on -...
`physiological factors, such as food, age, posture, -I-~
`body mass index, circadian rhythm, etc.; patholr
`factors, such as stress, diabetes, Crohn’s diseas
`motility disorders; and pharmaceutical factors, «'1
`size and density of formulation and coadministrat
`drugs like anticholinergic and prokinetic agents.
`ll
`apparent
`that GI transit
`is less likely to be a in
`determinant of bioavailability for drugs
`tha
`completely absorbed from the GI
`tract and if _"
`dosage forms reside in the GI tract for a consid
`period of time (~14-18 h).
`Since SITT has been demonstrated to be consta ~_
`
`'
`
`
`
`by gastric emptying. The variability in turn may 1""
`unpredictable plasma drug levels, and could 5° __
`impair the performance of pulsed— or time-releaS6 d°. '
`which are basically designed to deliver drug 3‘
`predetermined time to a specific site of the G1 tfac
`instance, if there is a large intrasubject variation In "Vi
`times,
`then the use of time-release S)/Stems ma
`precluded due to the unintended delivery Of drug
`inappropriate region of the GI tract (45).
`
`890
`
`agents such as cisapride tends to decrease the thickness of
`UWL by increasing segmental and propagative contrac-
`tions respectively, which may have implications for drug
`dissolution in the GI tract (34). The reverse is true for some
`viscous soluble dietary fibers, such as pectin, guar gum and
`sodium carboxymethylcellulose, which may increase the
`thickness of UWL by reducing intraluminal mixing
`(35-37) and could possibly decrease the intestinal exsorp—
`tion of lipophilic drugs like quinidine and thiopental (38).
`
`Biological Barriers to Oral Drug Delivery
`
`intestinal epithelial barrier
`
`lines the GI tract
`layer that
`The intestinal epithelial
`represents the major physical barrier
`to oral drug
`absorption. Structurally, it is made up of a single layer
`of columnar epithelial cells, primarily enterocytes and
`intercalated goblet cells (mucus secreting cells) joined at
`their apical
`surfaces by tight
`junctions or zonula
`occludens. These tight
`junctions are formed by the
`interaction of membrane proteins at the contact surfaces
`between cells and are responsible for restricting the
`passage of hydrophilic molecules during the paracellular
`transport.
`In fact, electrophysiological
`studies have
`suggested that epithelium gets tighter as it progresses
`distally, which has been implicated in a reduced
`paracellular absorption in the colon (39).
`The epithelium is supported underneath by lamina
`propria and a layer of smooth muscle called muscularis
`mucosa (3-10 cells thick). These three layers, i.e., the
`epithelium,
`lamina propria, and muscularis mucosa,
`together constitute the intestinal mucosa (40). On the
`apical surface, the epithelium along with lamina propria
`projects to form villi. The cell membranes of epithelial cells
`that comprise the villi contain uniform microvilli, which
`give the cells a fuzzy border, collectively called a brush
`border. These structures, although greatly increase the
`absorptive surface area of the small intestine, provide an
`additional enzymatic barrier since the intestinal digestive
`enzymes are contained in the brush border. In addition, on
`the top of the epithelial layer lies another layer, the UWL, as
`previously described. The metabolic and biochemical
`components of the epithelial barrier will be discussed.
`
`Gastrointestinal transit
`
`Advanced CR dosage forms can provide a precise control
`over release rates or release patterns for most drugs, which
`is attractive from a clinical point of view, particularly
`minimization of peak and trough variations in plasma drug
`concentration, and chronotherapy. For example,
`the GI
`
`

`
`891
`
`Furthermore, the brush border activity is generally
`greater in the proximal small intestine (duodenum ~
`jejunum > ileum >> colon) and involves enzymes
`such as alkaline phosphatase, sucrase, isomaltase, and
`a considerable number of peptidases (7).
`The intracellular metabolism occurs in the cyto-
`plasm of enterocytes and involves the major class
`of phase I metabolizing enzymes (i.e., cytochrome
`P450s,
`in particular CYP3A4),
`several phase II
`conjugating enzymes, and others such as esterases.
`Important examples of intestinal phase II metabolism
`include sulfation of isoproterenol and terbutaline, and
`glucuronidation of morphine and propofol (52). It
`is obvious that
`intestinal epithelium as a site of
`preabsorptive metabolism may significantly contrib-
`ute to the low bioavailability of therapeutic peptides
`and ester type drugs like aspirin, although it could
`serve as a key site for targeted delivery of ester or
`amide prodrugs (52).
`
`3.
`
`First-pass hepatic metabolism. As an absorbed drug
`reaches the liver through the portal circulation, a
`fraction of the administered dose is biotransformed
`
`before it reaches the systemic circulation. This is
`known as first—pass hepatic metabolism. In comparison
`with intestine, the liver dominates the process of first-
`pass metabolism for most drugs by virtue of its large
`mass, multiplicity of enzyme families present, and its
`unique anatomical position (13). However, this is not
`true for some drugs, such as terbutaline, in which case
`the sulfation occurs predominantly in the small
`intestine, as well as for midazolam, in which first-
`pass extraction by intestinal mucosa and liver appears
`to be comparable (44 i 14% versus 43 i 24%) (52). It
`is also important to emphasize here. that efflciency of
`first—pass metabolism (both hepatic and intestinal)
`varies considerably among different animal species
`and human subjects, which should be taken into
`account when deriving the estimates of oral bioavail-
`ability, particularly in the case of poorly soluble drugs.
`
`P-Glycoprotein and other
`efflux systems
`
`it has been recognized that P—Gp can
`In recent years,
`significantly contribute to the barrier function of the
`intestinal mucosa. P—Gp is an integral membrane protein,
`about 170-180 kDa, encoded by the MDRI gene in
`humans and contains 12 putative transmembrane domains
`and two ATP binding sites (53). In the small intestine and
`colon, P—Gp is expressed almost exclusively within the
`brush border membrane of mature enterocytes, where it
`
`Drug Delivery—Oral Route
`
`Food effect
`
`The coadministration of drugs with food is known to result
`in decreased, delayed,
`increased, or accelerated drug
`absorption, which may have pharmacokinetic and
`pharmacodynamic implications
`(46). Most often,
`the
`food effect is nonspecific and manifested as an interplay
`between physiological effects of food (such as delayed
`gastric emptying, stimulated bile secretion, increased liver
`blood flow, and alterations in gastric and duodenal pH),
`physicochemical characteristics of the drug (e.g., water
`solubility), or
`its
`formulation (e.g.,
`size,
`structural
`organization, and dissolution profiles). In general, drugs
`that are most
`influenced are those that are primarily
`absorbed from the upper regions of the GI tract and/or are
`poorly water—soluble. Apparently, food does not have a
`clinically significant effect on the absorption of moder-
`ately soluble drugs having a pH—independent solubility,
`and those that are completely absorbed (e.g., glipizide,
`isosorbide-5-mononitrate, felodipine, and nifedipine) from
`the GI tract. Furthermore, food may indirectly influence
`the drug absorption by affecting drug release from both
`hydrophilic matrix as well as lipid matrix formulations. In
`the former case, the effect has been attributed to increased
`
`hydrodynamic mechanical stress, which is caused by
`increased gastric motility (47). In the latter, the effect is
`due to increased pancreatic and biliary secretions, which in
`turn affect the integrity of matrix (48).
`Other potential factors that affect oral delivery are pH
`of the upper GI tract (49), which varies as a function of
`age, food and certain disease states, circadian rhythm (50,
`51), various diseases of the GI tract (49), and drug—drug
`interactions (7).
`
`
`
`Metabolic and Biochemical Barriers
`
`to Oral Drug Delivery
`
`Presystemic metabolism
`
`to presystemic
`Orally administered drugs are subject
`metabolism, which is comprised of three subtypes of
`mechanisms:
`
`1- Lumenal metabolism. This may be triggered by
`digestive enzymes
`secreted from the pancreas
`(amylase,
`lipases, and peptidases including trypsin
`and or-chymotrypsin), and those derived from the
`bacterial flora of the gut, especially within the lower
`part of the GI tract.
`
`First-pass intestinal metabolism. This includes brush
`border metabolism and intracellular metabolism. The
`
`former occurs at the surface of the enterocytes by the
`enzymes present within the brush border membrane.
`
`

`
`
`
`Controlled-Release Preparations
`
`The currently employed CR technologies for ora1_
`delivery are diffusion-controlled systems. solven
`vated systems, and chemically controlled system 2.
`Diffusion—controlled systems
`include monolithic}:
`reservoir devices in which diffusion of the drug-1
`rate—limiting step, respectively, through a polymer H
`or a polymeric membrane. Solvent-activated systems" .
`be either osmotically controlled or controlled by po _.,._
`swelling. Chemically controlled systems release drug
`polymeric degradation (surface or bulk matrix erosi
`cleavage of drug from a polymer chain (63).
`-._..
`examples of commercial products that have
`developed based on these CR principles are provid
`Table 3. It is worth mentioning here that the so—c ll
`programmed—release (“tailored—_release”) profile of a ii--
`CR product is rarely the outcome of a single pharma --
`tical principle. Depending on the specific physicoche u -
`properties of the drug in question and desired therap
`objectives, different formulation and CR principles
`-13:
`proportionally combined within the same dosage
`This task appears to be simpler when realized in tar as
`appropriate selection of polymers and excipients -~-~
`incorporate desired principles.
`
`Targeted-Release Preparations
`Site-specific oral drug delivery requires spatial Place
`of a drug delivery device at a desired site within th_
`tract. Although it is virtually possible to localize 3 d '.
`within each part of GI tract, the attainment of site-SP‘
`delivery in the oral cavity and the rectum is fem
`I,
`easier than in the stomach and the small and
`"E
`intestines. The latter
`requires consideration of
`longitudinal and transverse aspects of G1 cons“
`(66). Some of the potential CR and site-specific DDSS A
`be described.
`
`'
`
`‘M
`
`892
`
`Drug Delive
`
`ry§0 '
`
`fast disintegrating tablets and granules th
`at use 4 I
`cent mixtures, such as sodium carbonate
`._
`bicarbonate) and citric acid (or
`tartaric (or 5.
`superdisintegrants,
`such as
`sodium Starch aci
`croscarmellose sodium, and crospovidon El
`.
`technologies in fast-dispersing dosage fore.
`u‘:
`modified tableting systems, floss or Shearfofls
`ogy, which employs application of centrifugainf in
`controlled temperature, and freeze drying 6
`these technologies are briefly described in (fig)-leS
`
`acts as an energy—dependent drug efflux pump. Although
`P-gp appears to be distributed throughout the GI tract,
`its
`levels
`are
`higher
`in more
`distal
`regions
`(stomach < jejunum < colon) (54). Moreover, several
`studies have shown that P—Gp and CYP3A4 have similar
`substrate specificity [reviewed in Ref.
`(55)]. The
`colocalization of P—Gp and CYP3A4 in the mature
`enterocytes and their overlapping substrate specificity
`reasonably suggest that the function of these two pro-
`teins may be synergistic and appear to be coordinately
`regulated (56). Consequently, a greater proportion of the
`drug will be metabolized since the repetitive two-way
`kinetics (drug exsorption from the enterocyte into the
`lumen via P—Gp and reabsorption back into the
`enterocyte) will simply prolong the drug exposure to
`CYP3A4 (57). This mechanism not only limits the
`absorption of a wide variety of drugs, including peptides,
`but also poses a threat for potential drug interactions
`when attempts are made to inhibit CYP3A4 or P—Gp (58).
`Other mechanisms of drug efflux process may involve
`organic cation and anion transporters, which have been
`described elsewhere (32).
`
`CURRENT TECHNOLOGIES IN
`ORAL DRUG DELIVERY
`
`Over the last 3 decades, many novel oral drug therapeutic
`systems have been invented along with the appreciable
`development of drug delivery technology. Although these
`advanced DDS are manufactured or
`fabricated in
`
`traditional pharmaceutical formulations, such as tablets,
`capsules, sachets, suspensions, emulsions, and solutions,
`they are superior to the conventional oral dosage forms in
`terms of their
`therapeutic efficacies,
`toxicities, and
`stabilities (59). Based on the desired therapeutic
`objectives, oral DDS may be assorted into three
`categories:
`immediate—release preparations, controlled-
`release preparations, and targeted—release preparations.
`
`Immediate-Release Preparations
`
`These preparations are primarily intended to achi

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