throbber
Jennifer Dressman,‘ James Butler, John Hempenstall, and Christos Reppas
`
`
`
`
`
`
`
`
`
`ince the Biopharmaceutics Classification System
`(BCS) was introduced several years ago, it has be»
`come a benchmark in the regulation of bioequiva-
`lence of oral drug products both in the United
`'
`States and abroad (1). The concept behind the BCS is that
`if two drug products yield the same concentration pro—
`file along the gastrointestinal (GI) tract. they will result
`inthe same plasma profile after oral administration. This
`concept can be summarized by the following equation
`
`
`
`
`.
`E
`
`.-
`
`_
`
`.'_
`
`_.
`
`I
`
`I'
`
`'
`"
`
`_
`
`'
`
`'_
`'
`
`'
`
`__
`I
`
`._
`'31 3
`_I
`;
`
`.
`' Eff
`'_
`
`" '
`
`'E"
`
`I
`
`'
`
`"
`
` j: 1DW CW
`
`.
`
`'
`
`-
`
`-
`
`System (805) has proven to be a valuable/ 3
`tool for the regulation ofEchangesm oral/E. .
`drug products during scale-up and after
`product approvalThis article reviewers the
`criteria for classifymg drugs accord'ng.to
`the BCS and discusses further potential
`applications of the BCS, including the
`development of new drugs, the approval
`of generics, and the regulation of
`controlled-release products.
`
`
`
`gastrointestinal tract based on
`physiological as Well as drug and dosage-
`formrelated conSIderatIons She‘5 a
`professor at the Institute Of Pharmaceutical Technology.
`Johann Wolfgang Goethe University, Marie- Curie-str. 9
`Blozentrum 50439 Frankfurt, Germany. tel. 49 69 7982 9680.
`fax 49 59 7932 9594, e-mail dressmangiem,untfrankfmfie,
`Dr. Dressman is a member of the Editorial Advisory Board of
`Pharmaceutical Technology. James Butler '5 principal
`scientist. strategic technologies. at Pharmaceutical Sciences.
`GIaxoSmithKline R&D, Ware, UK. John Hem penstall is
`director of product-line extensions at Pharmaceutical
`Development, GlaxoSmithKilne R&D. Harlow. UK. Christos
`Reppas is an assistant professor at School of Pharmacy.
`The UWVBTSEW 0f Athens: Athens. Greece.
`*To Whom 3" corms’mr'dence Show be addressed‘
`
`68
`
`Pharmaceutical Technology Jun/2501
`
`in which Iis the flux across the gut wall. Pw is the permea—
`bility of the gut wall to the drug, and Cw is the concen-
`tration profile at the gut wall. In terms of bioequivalence,
`it is assumed that highly permeable, highly soluble drugs
`housed in rapidly dissolving drug products will be bio~
`equivalent and that, unless major changes are made to the
`formulation, dissolution data can be used as a surrogate
`for pharmacokinetic data to demonstrate bioequivalence
`of two drug products. The BCS thus enables manufac-
`turers to reduce the costs of approving scale-up and post-
`approval changes (SUPAC) to certain oral drug products
`(rapidly dissolving products of Class I drugs; see Table I)
`without compromising public safety interests.
`After several years of experience with the BCS, several
`issues have arisen: First, is the BCS fail safe? Second, should
`biowaivers be limited to Class 1 drugs. or could we extend
`them to other classes? Third, what about controlled~
`release dosage forms? Fourth, how early in the develop—
`ment process can we apply the BCS principles, and should
`the same cutoff values be applied to developing both new
`drug products and SUPAC applications? Although these
`issues already have been addressed to some extent in the
`.
`literatum we mus‘ contmue to gather data and ex19‘3““
`ence in order to resolve them. In this article we have tried
`to summarize current thinking and to make some sug-
`gestions about where we should head with the BCS in the
`coming years.
`
`?
`_
`'5 the 303 fall safe-
`FDA has set quite restrictive limitations on which drugs
`and drug products would be candidates for biowaivers
`under the BCS. The permeability requirement states that
`MYLAN EXHIBIT 1031
`MYLAN EXHIBIT 1031
`
`

`

`
`
`ample time to be dissolved. As long as
`these drugs meet the permeability crite-
`rion, biowaivers for products that dissolve
`rapidly at pH vaIUes typical of the small
`intestine could be considered.
`Another issue is that the requirement
`for “not less than 35% dissolution within
`
`30 min" may be too conservative in some
`dosing circumstances. Although in the
`fasted state it is quite possible that tran»
`sit time through the stomach is short
`(half-emptying times for water as short
`as 8—10 min have been reported in the lit—
`erature), if the dosage form is given with
`a meal, more than likely it will spend at
`least an hour or two in the stomach.
`Under these circumstances even slowly
`dissolving products still may show ab—
`sorption patterns that are controlled by
`gastric emptying. A case example is that
`of certain immediate-release (IR) parace—
`tamol tablets. Galia et al. showed that
`
`Panadol tablets release very slowly in sim-
`ulated fed»state conditions (milk) (8). It
`was subsequently shown by Reppas and
`Nicolaides that gastric emptying contin—
`ues to be rate limiting to absorption of
`paracetamol, even in the fed state (7).
`These results suggest that in cases in
`which the drug is routinely administered
`with meals, it may be possible to relax the
`criteria for dissolution.
`
`Can the 868 be extended to
`rapidly dissolving products of
`Class III substances?
`
`it has been suggested by Blume and Schug
`that because the absorption of Class III
`drugs is essentially controlled by the gut
`wall permeability to the drug and not by
`the drug's solubility, biowaivers for rapidly
`dissolving products of Class HI drugs also
`could be justified (8). Although in terms
`of the BCS theory this concept is clearly
`valid, some physiological issues would
`have to he addressed on a case-by—case
`basis. First, one must establish why the
`
`www.pharrnapertai.com
`
`‘3 Paracetamol formulation. 630 mg sodium bicarbonate
`+ Paracetamol formulation, 400 mg sodium bicarbonate
`A Paracetamol formulation, 375 mg sodium bicarbonate
`X Panadoi tablets
`
`D Panadol soluble, 1342 mg sodium bicarbonate
`
` I.l
`
`l l
`
`Figure 1: Mean paracetamol serum concentrations tollewmg 500 mg oral paracetamol.
`
`
`intrinsic solubility: 0.064 regimi. and pKa: 4.39
`
`Figure 2: The pH—soiubtlity profile of Ibuprofen at 37 C.
`
`(see Figure 1) even
`though the dissolution
`of the products in vitro
`was similar (4). This ex-
`ample shows that even
`though an excipient
`change may seem com-
`pletely innocuous. if the
`new excipient alters the
`GI physiology, then it
`may very well alter the
`plasma profile also.
`Regulatory authorities
`must be very careful
`about defining what
`constitutes a "major
`change" to the formu-
`lation to address the potential physiologi-
`cal issues.
`
`the permeability of the drug is common"
`surate with 290% absorption from a so-
`lution. The solubility requirement is that
`the dose~to-solubiiity ratio (D13) of the
`drug must be $250 mL over a pH range
`of l to 7.5. and the dissolution require-
`ment for the drug product is that disso-
`lution must be >85% complete within
`30 min (3). For products meeting these
`criteria. gastric emptying, rather than the
`release performance of the drug product.
`will be the key factor in determining the
`plasma profile; therefore, variability in the
`plasma profile will be under physiologi-
`cal control and not dictated by the dosage
`form.
`
`Even for rapidly dissolving products of
`Classl drugs, however. it is possible to man-
`ufacture bioinequivalent products if ex-
`cipients that modify gastric emptying are
`added. For example, Grattan et al. showed
`that the addition of sodium bicarbonate to
`the paracetamol (acetaminophen) formu—
`lation produced a faster and higher peak
`concentration of paracetamol in plasma
`
`70
`
`Pharmaceutical Technology JULY 2001
`
`llre the BBS criteria too restrictive?
`On the other hand, some drugs that are
`currently classified as Class II are consis-
`tently and completely absorbed after oral
`administration. These are typically poorly
`soluble weak acids with pKa values of $4.5
`and intrinsic solubilities (solubility of the
`un—ionized form) of 20.01 mg/mL. At pH
`values typical of the fasted state in the je-
`junum (about pH 6.5), these drugs will
`have soiubilities of E1 mg/mL, resulting
`in fast and reliable dissolution of the drug.
`
`Currently, these drugs are classified as
`Class Ii drugs because they are poorly sol-
`uble at gastric pH, in which pH<< pK.
`Figure 2 shows a typical solubility versus
`pH profile for ibuprofen (5)
`Because the small—intestinal transit time
`is more reliable, and in the fasted state.
`
`longer than the gastric residence time
`(generally on the order of 3 h), drugs with
`these physical characteristics will have
`
`

`

`
`
`
`A With 1132 mg SAPP
`A Alone
`0 All excipianis of effervescent tablet except SAPP
`o Effervescent tablet containing 1132 mg SAPP
`
`
`
`Figure 3: Mean serum ramtldine concentrations followmg 150 mg oral solution doses of
`ranitidine.
`
`permeability of the gut wall to the drug is
`low. If the permeability is low but uniform
`along the entire GI tract (including the
`proximal colon), biowalvers might be con-
`sidered. Howaver, if there is an absorption
`window or a gradient in the permeability
`of the gut wall to the drug (with decreas-
`ing permeability in distal regions), excip-
`ients that accelerate gut motility could sig~
`nificantly reduce the contact time of the
`drug with the sites at which permeability
`is favorable and therefore lower the
`
`bioavailability of the drug.
`Several compounds belonging to the
`HE receptor antagonist group are c1assi~
`cal examples of Class III drugs. It was
`shown in the literature some years ago that
`the shape of the plasma profile of cimeti-
`dine is highly dependent upon the gastric
`pH at the time of administration, with the
`characteristic double peak eliminated if
`the drug is given under elevated gastric
`pH conditions (9) , Further, excipients that
`accelerate transit in the upper GI tract
`such as sodium acid pyrophosphate (10)
`and mannitol
`(1 l) have been clearly
`shown to reduce the extent of absorption
`of ranitidine and cimetidine, respectively.
`The results from Koch et al. are shown in
`
`Figure 3 (10) . The 50% reduction in CPeak
`illustrates how important the influence of
`excipients that can alter the GI motility
`can be to the absorption of Class llI drugs.
`
`Can the BBS be applied to
`controlled-release drug products?
`Under the current definition, the BCS is
`
`applicable only to immediate—release
`dosage forms because only the perme-
`
`72
`
`Pharmaceutical Technology JULY2001
`
`ability in the Jejunum is considered. To
`extend the BCS to controlled-release (CR)
`dosage forms, one must assess the per-
`meability at all points in the GI tract where
`release of the drug is foreseen {l2}. As
`pointed out by Corrigan, it is unlikely that
`drugs with low permeability in either the
`ileum or colon will prove to be suitable
`candidates for CR dosage forms, let alone
`for blowaivers based on dissolution tests
`
`(5). He has proposed a useful subclassifi—
`cation scheme for CR products that is
`based on the site dependency of both the
`drug solubility and permeability.
`A further consideration is the selection
`
`of appropriate dissolution conditions to
`simulate the release profile of the dosage
`form as it moves through the GI tract.
`Conditions for dissolution in the stom-
`ach, the small intestine, and the colon dif-
`fer greatly. Important parameters that
`vary with location in the GI tract include
`the volume of fluid available for dissolu-
`
`tion, osmolarity of the contents, the hy—
`drodynamic (motility) conditions, and
`the secretion of various enzymes and
`other para—GI secretions that could po-
`tentially affect the release rate. Similarity
`of the dissolution profiles under all ap-
`propriate GI conditions would have to be
`shOWn for the two drug products. Al-
`though our understanding of the com—
`position of lumenal contents as they move
`along the GI tract is far better than it was
`a decade ago, a more complete charac-
`terization is still needed. Still almost to-
`
`tally lacking is an understanding of the
`relationship between the hydrodynamics
`in the gut and those in the currently avail-
`
`able dissolution testers. This thrOWs a de-
`
`gree of uncertainty into the interpreta-
`tion of dissolution results in terms of in
`
`vivo performance. eVen when the com~
`position of the lumenal contents can be
`simulated well in the in vitro tests. Al-
`
`though a problem is posed by the limi—
`tation to establishing in vivo-in vitro cor-
`relations for IR products, the problem is
`compounded for CR dosage forms be-
`cause the hydrodynamics at several sites
`within the GI tract must then be simu-
`lated. As a result, in vitro release profiles
`of CR dosage forms with different release
`mechanisms must be interpreted very
`cautiously,
`
`Application of the 368 to the
`development of new
`drug substances
`Because the BCS was originally developed
`as a basis for determining bioequivalence
`of oral drug products, it assumes that the
`drug is sufficiently well absorbed to make
`an oral dosage form feasible. When new
`drug substances are being developed,
`however, this assumption is not appro-
`priate, and one must consider other fac—
`tors than just the solubility and permea-
`bility to determine whether an oral dosage
`form can be successfully developed. An
`overview of the events in the GI tract fol—
`
`lowing oral drug administration is de~
`picted in Figure 4.
`First, it should be remembered that the
`drug substance does not have to meet the
`Class I criteria of high permeability and
`solubility for the drug to be successfully
`formulated in an oral solid dosage form.
`Many Class II and Class III drugs are avail"
`able on the market, and several that meet
`Class IV criteria are available (see Table ll).
`One problem with applying the BCS cri-
`teria to new drug substances is that, early
`in preformulatlon/formulation, the dose
`is not yet accurately known. So at this
`point, the D:S can only be expressed as a
`likely range. A helpful rule of thumb is that
`compounds with aqueous solubilities
`>100 pg/mL seldom exhibit dissolution
`ratemlimited absorption. Alternatively, one
`can estimate the maximum absorbable
`dose on the basis of the usual volumes of
`GI fluids available under the anticipated
`dosing conditions and the solubility of the
`drug. With regard to the solubility of the
`drug, it may be useful to consider the
`
`www.pharmaporia|.cem
`
`

`

`
`Drug in solution
`at uptake sites
`iii
`
`Decomposition
`Adsorption
`Complexation
`
`Decomposition
`iii
`Adsorption
`Drug in solution
`at uptake sites
`Complexatlon
`
`Figure 4: Steps In drug absorption and sources of incomplete hioava: a lily c owmg or
`administration of a solid dosage form.
`
`
`
`
`
`would lead to a quite different interpreta-
`tion of the chances for absorption in vivo.
`For promising compounds that are both
`ionizable and lipophilic, extensive solubil-
`ity experiments in biorelevant media will
`help characterize the likely solubility be
`havior in viVO. Several publications address
`the composition and applications of these
`media (6,13—16). An alternative approach
`is to use aspirates from human volunteers.
`although volumes aspirated typically are
`small and the choice of experiments and
`apparatus therefore is limited (17).
`Another issue is the use of 250 mL as
`the volume in which a dose must be dis-
`solved. This amount is a conservative es-
`timate of the volume of fluid available in
`
`physicochemical properties of the drug
`when deciding which media to use for the
`solubility determinations. For example,
`measuring solubility at all pH values rec—
`ommended by the BCS is unnecessary for
`neutral compounds in early development.
`Later, when formulations are compared,
`dissolution data for the drug product over
`the entire Gl pll range will be useful in 88*
`tablishing the robustness of release from
`the formulation under Gl conditions.
`
`Lipophilic drugs may be very poorly sol-
`uble in water and in simple buffers, but in
`the GI fluids they can often be solubilized
`by the bile to a significant extent. Increases
`in solubility of one to two orders of mag—
`nitude are possible for compounds with
`log P values of ;4. In some cases this
`
`74
`
`Pharmaceutical Technology worms:
`
`the gut under fasting-state conditions and
`is based on the volume usually ingested
`along with the dosage form in a pharma-
`cokinetic study (the so-called FDA glass of
`Water). The actual volume available is a
`composite of the ingested fluid and the se-
`cretions of the GI tract. Although these
`amounts tend to be modest in the fasted
`state, secretions in the fed state contribute
`substantially to the overall fluid volume.
`which may be as high as 1.5 L in both the
`stomach and upper small intestine. De-
`pending on whether drug administration
`is to be on an empty stomach or with
`meals, it is reasonable to adjust the volume
`used to assess the capacity of the GI fluids
`to dissolve the dose. A useful starting point
`would be to use a volume of 300 mL for
`the fasted stomach. 500 ml. for the fasting
`
`small intestine, and up to I L for the post-
`prandial stomach and small intestine.
`A further consideration is the choice of
`
`model for assessing the permeability. Ale
`though perfusions in humans will pro—
`duce the most reliable results (18) and are
`clearly the “gold standard,” these require
`too much time and money to make them
`practicable for screening new drug sub—
`stances. Many animal- and celluculture
`models have been developed, each with its
`own set of advantages and disadvantages.
`For example. the Caro 2 cells can be used
`with confidence to assess transcelluiar dif—
`fusion and can be standardized to ensure
`
`reproducible results, but they tend to un-
`derestimate paracellular and active mecha—
`nisms. cannot be employed to determine
`regional permeability within the gut, and
`tend to overestimate efflux via the Pa
`
`glycoproteins. In situ perfusions in rats.
`although they are much better in terms of
`forecasting active transport and can be
`used to determine regional permeability,
`take more time and effort to produce a re-
`liable permeability estimate. In any case,
`it is a good idea to have more than one
`permeability screen at the disposal of the
`laboratory in order to build confidence
`and robustness into the screening system.
`If the drug is poorly soluble but highly
`permeable, formulation efforts will con—
`centrate on improving the dissolution
`profile. For example, the combined effects
`of formulating the drug as amorphous
`solid dispersion and administering it in
`the fed state are shown for troglitazone
`
`in Figure 5. Combined, these two ap-
`proaches shift the solubility—dissolution
`characteristics from those of a very poorly
`soluble drug (D28 >10,000 mL) to those
`of a drug product with a D13 within the
`range of values encountered in the gut
`after meals.
`
`Figure 6 summarizes some further pos-
`sibilities for improving the absorption of
`drugs with less than optimal permeabil~
`ity and solubility characteristics. If perv
`meability rather than solubility is the
`main problem. formulation approaches
`are less numerous and less reliable. In ex-
`treme cases, it may be appropriate to con—
`sider developing another analog with
`more appropriate blopharmaceutical
`characteristics.
`Even when allowance is made for the
`differences in solubility and permeability
`
`www.pharmaportai.com
`
`
`
`
`
`
`
`
`
`
`
`

`

` scale-up and postal:-
`exciplents? required to optimize the
`
`giggling: :18 rfugugf,
`P P 0 a 3’
`Will be used increasingly
`in the early develop~
`merit of new drugs, in—
`eluding for analog se—
`lection as well as for
`initial formulation 3p“
`proaches.As our knowl»
`edge of GI physiology
`becomes more sophia
`ticated ll'lVitI‘O dissolu—
`'
`’
`no“ teas Win be able to
`better Simulate the con—
`ditioris in the GI tract;
`-
`-
`Th“ m tum Win lead to
`more powerful predic-
`tions of in vivo perfor-
`mance and ultimately to
`a significant reduction
`in the number of ani-
`
`mal and human studies
`
`formulation. Together
`with screens for other
`limitations to oral ab—
`
`
`
`E. Gaiia at 3.1.. "Evaluation of Various Dissow
`lotion Media for Predicting in Vivo Perfor-
`mance of Class} and H Compounds," Pharm.
`Res. 15 (5). 698—705 (2998).
`7. C. Reppas and E. Nicolaides, "Analysis of
`Drug Dissolution Data" in Methods for As-
`sesslng Oral DrugAbsoypilon. J. Dressman
`and H. Lennernas, Eds. Marcel Dekker, New
`York, 2000), pp. 229—254.
`8. H. Biume and B. Schug, ”The Biopharma—
`ceutics Classification System (BCS): Class II
`Drugs a“ Better Candidates for BA/BE
`Waiver?" Eur. J. Pharm. Sci. 9 (2), 117—121
`(199%
`..
`9. V. Mummaneni and }. Dressman, Gastric
`PH Influences on the Appearance of Double
`Peaks in the Plasma Cflncemratmmflm
`Profiles of Cimetidlne after Oral AdminESv
`tration in Dogs," Pharm. Res.12 (5). 780—786
`(l 995).
`10. KM. Koch et al.. "Effect of Sodium Acid ry—
`rophosphate on Raniticline Bioavaiiahllity
`and Gastrointestinal Transit Time," Pharm.
`Res. 10 (7), 1027-1030 (1993).
`D.A. Adkin. et al., "The Effect of Mannitol
`on the Oral Bioavailability of Clmetidine." J.
`Pharm. Sci. 8402114054409 (1995).
`. I. Wilding, "Evolution of the Biopharma-
`ceutics Classification System (BCS) to Mad
`ified Release (MR) Formulations: What Do
`We Need to Consider?" Eur. J. Pharm. Sci. 8
`(3), 151L159 (1999).
`i3. J, Dressman et al.. “Dissolution Testing as a
`
`5 1-
`
`12
`
`Prognostic Tool for Oral Drug Absorption:
`Immediate Release Dosage Forms." Pharm.
`Res. 15 (1), 1142 (1998}.
`14. E. Nicolaides at it” "Firecagting the IrBVivo
`formance of our ow- olubiiity
`rugs
`H T
`.
`B
`gggfimhgirsl?ggr5 gigglgfltégg (1egggiour
`t5. R. Lobenberg et a!” "Dissolution Testing as
`2133Prognostic "£001 for Oral DGrug Abslorption:
`1
`l
`"
`Phisrjhuhg:178231393; $030: am do,
`'
`'
`'
`..
`‘
`16' 3' Dressman and 0' Reppas' In Vim) In Vivo
`Correlations for Lipcphiiic. Poorly Waterw
`Soluble Drugs,” Eon]. Pharm. Sci. 11 Suppl.
`2, $73680 (2001).
`.
`d
`.,“A
`l
`fth
`i
`.
`‘7 3 PB “50591581
`Eifmpar 501;:
`BSOd
`ability of
`anazol in
`urnan an
`imulate
`Gastrointestinal Fluids .. Pharm Res 17 (7)
`‘
`891—894 (2090)
`’
`'
`'
`18. H. Lennemas, "Human Perfusion Studies,"
`in Methods in Oral Drug Absorption, J. Dress—
`man and H. Lennerniis, Eds. (Marcel Dekker,
`New York. 2000), pp. QQul l7. PI'
`
`II
`
`4.:fil-norpnous (faSted)
`H
`Cast...”ne
`(fasted)
`
`
`_
`.
`Figure 5: Troglltazone 200 mg: the effect of food and form on the
`potential for solubility limited bioavaiiability.
`
`3
`
`I
`
`Nanopaniclas
`E Selebt more soluble saltlpoiymorph
`' 4.3—...—
`l
`. fl Liquid-filled capsules (oily
`j
`1
`it self-emulsifying vehicles)
`:
`I
`Solid dispersions
`T‘Addlllon of surfactant to
`solid dosage form
`Liquid-filled
`capsules with
`absorption-
`enhancing
`
`w
`
`Mucoadneslon,
`absorption-
`l enhancing iii
`exclpieats,
`effiux
`inhibitors
`
`Sfirptior; 3123;3031133‘183
`E 9 way or r 3V0 ution
`in the drug develop-
`men: process.
`
`.
`.
`.
`.
`.
`.
`.
`Figure 6. Possmle effects of various formulations on developablllly.
`
`requirements for oral drug product de-
`velopment vis—a-vis biowaiver criteria ac- References
`cording to the BCS. further factors still
`I. G. Arnidon etalb‘ThegreticalBasts fgr agio-
`must be considered for new drugs. These
`EgglfiicififlwtEZgDrIElEEEEZt 31:50::
`.
`.
`.
`include the POSSiblhty 0f decomposuion
`tion and InVivo Bloavailability."Pharm. Res.
`under GI conditions and the assessment
`12 (3). 4134120 (1995),
`f first- ass metabolism both in the ut
`2 C t
`f Dr
`E l
`tion FDA “(3 id n
`0
`p
`g
`an or or
`ug va ua
`'
`'
`'
`u a ce
`wall and the “VET- Appraislng decompo-
`for Induséry on gissoitclltiolnlgl'estingFof 1m;
`sition in the gut is relatively simple using
`231:“: 15;?“ did m “age 0““
`-
`“S
`‘
`biorelevant media and exposure times
`3. Center for Drug Evaluation,FDA,"Guldance
`based on ion est
`l’itici ated ex osure
`g
`a
`P
`p
`for industry on the Waiver of In Vivo
`times. For sensitive compounds. appro—
`Bioavailability and Bioequivalence for Im-
`priate enzymes (e.g.. P393” and 335515 ll'
`mediate Release Solid Oral Dosage Forms
`Based on a Biopharmaceutics Classification
`pesos for the stomach, pancreatic enzymes
`System" (August 2000).
`for the jejunum, and bacterial enzymes
`4. T. Grattan et al.. "A Five—way Crossover
`Human Volunteer Study to Compare the
`for the colon) must be added to the
`Pharmacokinetics of Paracetamol Following
`medium in relevant concentrations. As far
`Oral Administration of Two Commercially
`Available Paracetamol Tablets and Three De-
`velopment Tablets Containing Paracetamol
`in Combination with Sodium Bicarbonate
`or Calcium Carbonate," Eur. J. Pharm. Blo—
`pharm. 49 (3), 225—229 (2000).
`5. 0.1. Corrigan. "The Biopharmaceutics Drug
`Classification and Drugs Administered in Ex-
`tended Release OER) Formulations," Adv. Exp.
`Med. Biol. 423.1[1—128{1997).
`
`as first—pass metabolism in the gut wall is
`concerned. it may be possible to screen for
`metabolites in the permeability model de-
`pending on how the model is set up.
`
`Summary
`in summary, the BCS has proven to be an
`extremely useful tool for the regulation of
`bioequivalence of drug products during
`
`76
`
`Pharmaceutical Technology JULY 2001
`
`www.pharmaportal.com
`
`

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