throbber

`
`
`
`Jennifer Dressman,* James Butter, John Hempenstall, and Christos Reppas
`
`
`
`
`
`
`
`
`
`System (BCS)has proven tobea valuablejf
`tool for the regulation ofchangesiin oral’.
`drug products during scale-up and after
`productapproval.Thisarticle reviews the
`criteria for.classifying drugsaccording|to”
`the BCS anddiscusses further potential
`applicationsof theBCS,including the
`developmentof new drugs, the approval
`of generics, and the regulation of
`controlied-release products.
`
`
`
`Jennifer Dressman, PhD,currently is
`‘researching the developmentof a predictive
`-Modelfor the delivery of drugs via the
`gastrointestinal tract, based on
`physiological as weil as drug- and dosage-
`form-related considerations. She is a
`professerat the Institute of Pharmaceutical Technology,
`Johann Wolfgang Goethe University, Marie-Curie-str. 9,
`Biozentrum 60439 Frankfurt, Germany,tel. 49 69 7982 9680,
`fax 49 69 7982 9694, e-mail dressman@em.uni-frankfurt.de.
`Dr. Dressman is a memberof the Editorial Advisory Board of
`Pharmaceutical Technology. James ButlerIs principal
`scientist, strategic technologies, at Pharmaceutical Sciences,
`GlaxoSmithKline R&D, Ware, UK, John Hempenstall is
`director of product-line extensions at Pharmaceutical
`Development, GlaxoSmithKtine R&D, Harlow, UK. Christos
`Reppas js an assistant professor at Schoal of Pharmacy,
`The University of Athens, Athens, Greece.
`“fo whom all correspondence shoud be addressed,
`
`68
`
`Pharmaceutical Technology suty 2001
`
`
`nce the Biopharmaceutics Classification System
`
`_
`(BCS) was introducedseveral years ago,it has be-
`ome a benchmark in the regulation of bioequiva-
`
`lence of oral drug products both in the United
`States and abroad (1). The concept behind the BCSis that
`if two drug products yield the same concentration pro-
`file along the gastrointestinal (GI) tract, they will result
`in the same plasmaprofile after oral administration. This
`concept can be summarized by the following equation
`
`J = Py Cy
`
`in which Jis the flux across the gutwall, P,is the permea-
`bility of the gut wall to the drug, and C,, 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 TableI)
`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 I 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 sarne 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
`literature, we must continue to gather data and experi-
`ence in order to resolve them. In this article we havetried
`to summarize current thinking and to make some sug-
`gestions about where we should head with the BCS in the
`coming years.
`
`Is the BCSfail safe?
`FDAhasset quite restrictive limitations on which drugs
`and drug products would be candidates for biowalvers
`under the BCS, The permeability requirementstates that
`MYLAN EXHIBIT 1031
`MYLAN EXHIBIT 1031
`
`

`

`
`
`ample time to be dissolved. As longas
`these drugs meet the permeability crite-
`rion, biowaivers for products that dissolve
`rapidly at pH values typical of the small
`intestine could be considered.
`Anotherissue 1s that the requirement
`for “not less than 85% dissolution within
`36 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 8-16 min have been reportedin thelit-
`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
`Panadoltablets release very stowly in sim-
`ulated fed-state conditions (milk) (6). 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 BCSbe extended to
`rapidly dissolving products of
`Class III substances?
`It has been suggested by Blume and Schug
`that because the absorption of Class Ii]
`drugs is essentially controlled by the gut
`wall permeability to the drug and not by
`the drug's solubility, biowalvers for rapidly
`dissolving products of Class III drugs also
`could bejustified (8), Although in terms
`of the BCS theory this conceptts clearly
`valid, some physiological issues would
`have to be addressed on a case-by-case
`basis, First, one must establish why the
`
`© Paracetamol formulation, 630 mg sodium bicarbonate
`+ Paracetamol formutation, 400 mg sodium bicarbonate
`4 Paracetamol formufation, 375 mg sodium bicarbonate
`* Panadoltablets
`
`0 Panadol soluble, 1342 mg sodium bicarbonate
`
`
`Intrinsic solubility: 0.664 mg/ml. and pk: 4.39
`
`k :
`
`Figure 1: Mcan paracetamol serum concentrationsfollowing 500 mg oral paracetamol.
`{see Figure 1) even
`though the dissolution
`ofthe praducts 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
`mayvery 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 physiolog!-
`cal issues.
`
`Figure 2: The pH-solubility profile of ibuprofen at 37 °C.
`
`the permeability of the drug is commen-
`surate with 290% absorption from a so-
`lution, The solubility requirementis that
`the dose-to-solubility ratio (D:S) of the
`drug must be =250 mi. over a pH range
`of I 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
`plasmaprofile; therefore,variability in the
`plasmaprofile will be under physiologi-
`cal control and notdictated by the dosage
`form.
`Even for rapidly dissolving products of
`Class I 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
`
`Are the BCScriteria too restrictive?
`On the other hand, some drugs that are
`currently classified as Class IT are consis-
`tently and completely absorbed after oral
`administration. These are typically poorly
`soluble weak acids with pK, values of <4.5
`and intrinsic solubilities (solubility of the
`un-ionized form) of =0.01 mg/mL. At pH
`values typical of the fasted state in the je-
`junum (about pH 6.5), these drugs will
`have solubilities of =1 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 pli<< pX,,
`Figure 2 shows a typical solubility versus
`pHprofile 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 orderof 3 h), drugs with
`these physical characteristics will have
`
`www.pharmaportal.com
`70=Pharmaceutical Technolagy suty 2007
`
`

`

`
`
`
`a With 1132 mg SAPP
`4 Alone
`° All excipiants of effervescent tablet except SAPP
`« Effervescent tablet containing 1132 mg SAPP
`
`
`
`Figure 3: Mean serum ranitidine concentrations following 150 mg oral solution doses of
`ranitidine.
`
`permeability of the gut wall to the drugts
`low. If the permeability is low but uniform
`along the entire GItract (including the
`proximal colon), biowaivers might be con-
`sidered. However,if there is an absorption
`window ora gradientin 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
`bioavallability of the drug.
`Several compounds belonging to the
`H2 receptor antagonist group are classi-
`cai examples of Class III drugs. It was
`shown in the literature some years ago that
`the shapeof the plasmaprofile of cimeti-
`dine is highly dependent uponthe gastric
`pH atthe time of administration, with the
`characteristic double peak eliminated if
`the drug is given underelevated gastric
`pH conditions (9), Further, excipients that
`accelerate transit in the upper GI tract
`such as sodium acid pyrophosphate (10)
`and mannitol {11} have been clearly
`shownto reduce the extent of absorption
`of ranitidine and cimetidine, respectively,
`The results from Koch etal. are shown in
`Figure 3 (10). The 50% reduction in C,...
`illustrates how importantthe influence of
`excipients that can alter the GI motility
`can be to the absorption of Ciass Il drugs.
`
`Can the BCS be applied to
`controlied-release drug products?
`Under the current definition, the BCS Is
`applicable only to immediate-release
`dosage forms because only the perme-
`72
`Pharmaceutical Technology duty 2007
`
`ability in the jejunum is considered, To
`extend the BCS to controlled-release (CR)
`dosage forms, one must assess the per-
`mieability at all points in the GI tract where
`release of the drug is foreseen (12). 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, jet alone
`for biowaivers 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 smail intestine, and the colon dif-
`fer greatly. Important parameters that
`vary with location in the GI tract include
`the volumeof fluid available for dissolu-
`tion, osmolarity of the contents, the hy-
`drodynamic (motility) conditions, and
`the secretion of various enzymes and
`other para-GIsecretions that could po-
`tentially affect the release rate, Similarity
`of the dissolution profiles under ail ap-
`propriate G] 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 ofthe
`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 severalsites
`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 BES 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
`drugis suffictently well absorbed to make
`an oral dosage form feasible. When new
`drug substances are belng 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.
`ManyClass II and Class IIT drugs are avail-
`able on the market, and several that meet
`Class IV criteria are available (see Table I).
`One problem with applying the BCScri-
`terla to new drug substances is that, early
`in preformutation/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 thumbis that
`compounds with aqueous solubilities
`> 100 pg/mL seldom exhibit dissolution
`rate-limited absorption. Alternatively, one
`can estimate the maximum absorbable
`dose on the basis of the usual volumes of
`GI fluids available underthe 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. pharmaportal.com
`
`

`

`
`Drug in sofution
`at uptake sites
`Pt
`f
`
`Decomposition
`Adsorption
`Complexation
`
`---4-¢-<I
`j
`j
`I
`+
`4

`Decomposition
`Adsorption
`Drug in solution
`Complexation
`
`at uptake sites
`
`--Ne—o
`
`omeo
`Transitee ee
`
`
`
`
`small intestine, and up to 1 L for the post-
`prandial stomach and small intestine.
`A further consideration is the choice of
`model for assessing the permeability. Al-
`though perfusions in humans will pro-
`duce the mostreliable 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 cell-culture
`models have been developed, each with its
`ownset of advantages and disadvantages.
`For example, the Caco 2 cells can be used
`with confidenceto assess transcellular 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 P-
`glycoproteins. In situ perfusions in rats,
`although they are muchbetter in termsof
`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 pooriy 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 shownfor troglitazone
`in Figure 5, Combined, these two ap-
`proachesshift the solubility-dissolution
`characteristics from those of a very poorly
`soluble drug (D:S >10,000 mL) to those
`of a drug product with a D:S 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 per-
`meability rather than solubility is the
`main problem, formulation approaches
`are less numerous andless reliable, In ex-
`treme cases, It may be appropriate to con-
`sider developing another analog with
`more appropriate biopharmaceutical
`characteristics.
`Even when allowance is made for the
`differences in solubility and permeability
`
`
`
`
`
`
`
`
`
`
`
`Figure 4: Steps in drug absorption and sourcesof incomplete bioavailability following 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 Hpophilic, extensive solubil-
`ity experiments in biorelevant media will
`help characterize thelikely solubility be-
`haviorin 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
`apparatusthereforeis limited (17).
`Anotherissue 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 offluid available in
`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 ts 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 smail 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 GIfluids
`to dissolve the dose, A useful starting point
`would be to use a volume of 300 mL for
`the fasted stomach, 5€0 mL. for the fasting
`
`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 BCSis unnecessary for
`neutral compoundsin early development,
`Later, when formulations are compared,
`dissolution data for the drug product over
`the entire GI pH range will be useful in es-
`tablishing the robustness of release from
`the formulation under GI 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 passible for compounds with
`log P values of 24. In some cases this
`www, pharmaportai.com
`74~—Pharmaceutical Technology vury 2007
`
`

`

`ty
`Amorphous!
`{fed}
`
`fy
`
`Crystalline
`(fasted)
`
`proval, In the future,
`BCSconcepts probably
`will be used increasingly
`in the early develop-
`ment of new drugs, In-
`cluding for analog se-
`lection as well as for
`
`
`Figure 5: Troglitazone 200 mg: the effect of food and form on the
`potential for solubility limited bioavailability.
`
`Amorphous (fasted) scale-up and postap-
`
`
`. E, Gatia et al., “Evaluation of Various Disso-
`lution Media for Predicting In Vivo Perfor-
`mance of Class and H Compounds,” Pharm.
`Res. 15 (5), 698-705 (1998).
`. C, Reppas and E, Nicolaides, “Analysis of
`Drug Dissolution Data” in Methods for As-
`sessing Oral Drug Absorption, J, Dressman
`and H. Lennernas, Eds. (Marcel Dekker, New
`York, 2000}, pp. 228-254.
`. H. Blume andB. Schug, “The Biopharma-
`initial formulation ap-
`ceutles Classification System (BCS): ClassII
`Drugs —- Better Candidates for BA/BE
`proaches. As our knowl-
`Watver?” Bur. J. Pharm, Sci. 9 (2), 117-121
`edge of GI physiology
`(1999}.
`becomes more sophis-
`. ¥, Mummanert and J}. Dressman, “Gastric
`ticated, in vitro dissolu-
`pH Influences on the Appearance of Double
`tion tests will be able to
`Peaks in the Plasma Concentration-Time
`Profiles of Cimetidine after Oral Adminis-
`better simulate the con-
`tration in Dogs,” Pharm. Res, 12 (6), 780-786
`ditions in the GI tract.
`(995).
`This in turn will lead to
`10,
`K.M.Kochet al., “Effect of Sodium Acid Py-
`more powerful predic-
`<——_
`rophosphate on Ranitidine Bioavailability
`=
`k
`and Gastrointestinal Transit Time,” Pharm.
`tions of In vivo perfor-
`Nanoparticles
`FSelebt more soluble salt/polymorph
`Res, 10 (7), 1027-1030 (1993).
`mance and ultimately to
`. DA. Adkin, et al., “The Effect of Mannitol
`|
`“Si Liquid-filed capsutes (ofy
`a significant reduction
`on the Oral Bioavailability of Cimetidine,” j,
`i
`1
`& self-amulsifying vehicias)
`in the numberof ani-
`Solic dispersions|
`Pharm. Sel, 84(12),1405-1409 (1995).
`TAddltion of surfactaatto
`mal and human studies
`I. Wilding, “Evolution of the Biopharma-
`solld dosage form
`required to optimize the
`ceutics Classification System (BCS) to Mad-
`Liquid-filled
`ified Release (MR) Formulations: What Do
`formulation, Together
`capsules with
`absorption-
`We Need to Consider?” Eur. f. Pharm. Sci, 8
`with screens for other
`enhancing
`(3), 157-159 (1999).
`excipients?
`limitations to oral ab-
`J, Dressmanetal., “Dissolution Testing as a
`sorption,the BCS paves
`Prognostic Tool for Oral Drug Absorption:
`the way for (r)evolution
`Immediate Release Dosage Forms,’ Pharm.
`Res, 15 (1), 11-22 (1998}.
`in the drug develop-
`E, Nicolaideset al,, “Forecasting the In Vivo
`ment process,
`Performance of Four Low-Solubility Drugs
`from Their In Vitro Dissolution Behaviour,”
`Pharm, Res, 16 (12}, 1876-1882 (1999).
`R, Lébenberget al., “Dissolution Testing as
`a Prognostic Tool for Oral Drug Absorption:
`Dissolution Behaviour of Glibenclamide,”
`Pharm. Res, 17 {4}, 439-444 (2000),
`J, Dressman and C., Reppas, “In Vitro In Vivo
`Correlations for Lipophitic, Poorly Water-
`Soluble Deugs,” Bur. J. Pharm, Sei, 14 Suppl.
`2,573-S80 (2001).
`B, Pedersonetal., “A Comparisonofthe Sot-
`ubility of Danazol in Human and Simulated
`Gastrointestinal Fluids,” Pharm. Res. 17 (1),
`891-894 (2000).
`H. Lennernas, “Human Perfusion Studies,’
`in Methods in Oral Drug Absorption, J. Dress-
`man and H. Lennernas, Eds. (Marcel Dekker,
`New York, 2000}, pp. 99-17. PT
`
`
`
`12.
`
`13.
`
`14,
`
`£5,
`
`16.
`
`17.
`
`18,
`
`www. pharmaportal.cam
`
`3—
`
`Mucoadhesion,
`absorption-
`1 erchancing HE
`excipients,
`effiux
`inhibitors
`
`WV
`
`Figure 6: Possible effects of various formulations on developabilily.
`
`requirements for oral drug product de-
`velopmentvis-a-vis biowaivercriteria ac-
`cording to the BCS,further factorsstill
`must be considered for new drugs. These
`include the possibility of decomposition
`under GI conditions and the assessment
`of first-pass metabolism both in the gut
`wail and theliver. Appraising decompo-
`sition in the gutis relatively simple using
`biorelevant media and exposure times
`based on longest anticipated exposure
`times. For sensitive compounds, appro-
`priate enzymes(e.g., pepsin and gastric li-
`pases for the stornach, pancreatic enzymes
`for the jejunum, and bacterial enzymes
`for the colon) must be added to the
`medium in relevant concentrations. As far
`as first-pass metabolism in the gut wailis
`concerned, it may be possible to screen for
`metabolites in the permeability model de-
`pending on how the modelis 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 suvy 2007
`
`References
`1. G. Amidon etal., “Theoretical Basls for a Bio-
`pharmaceutical Drug Classification: The Cor-
`relation of In Vitro Drug Product Dissclu-
`tion and In Vivo Bioavailability,” Pharm, Res.
`12 (3), 413-420 (1995),
`2, Center for Drug Evaluation, FDA,“Guidance
`for Industry on Dissolution Testing of Im-
`mediate Release Solid Oral Dosage Forms”
`{August 1997).
`3. Center for Drug Evaluation, FDA,“Guidance
`for Industry on the Waiver of In Vivo
`Bioavailability and Bioequivalence for Im-
`mediate Release Solid Oral Dosage Forms
`Based on a Biopharmaceuties Classification
`System” (August 2.000).
`4, T. Grattan et al., "A Flve-way Crossover
`Human Volunteer Study to Compare the
`Pharmacokinetics of Paracetamol Following
`Oral Administration of Two Commerciaily
`Available Paracetamol Tablets and Three De-
`velopment Tablets Containing Paracetamol
`in Combination with Sodium Bicarbonate
`or Calctum Carbonate,” Bur. J. Pharm. Bio-
`pharm. 49 (3), 225-229 (2000).
`5, OJ. Corrigan. “The Biopharmaceutics Drug
`Classification and Drugs Administered In Ex-
`tended Release (ER) Formulations,” Adv. Exp.
`Med. Biol. 423, 11-128 (1997).
`
`

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