throbber
e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 1 ( 2 0 0 7 ) 249–261
`
`a v a i l a b l e a t w w w . s c i e n c e d i r e c t . c o m
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / e j p s
`
`Conference report
`When poor solubility becomes an issue:
`From early stage to proof of concept夽
`S. Stegemann a, F. Leveiller b, D. Franchi c, H. de Jong d, H. Lind´en e,∗
`
`a Capsugel, Rijksweg 11, 2880 Bornem, Belgium
`b AstraZeneca, 22187 Lund, Sweden
`c GSK, Via A. Fleming 4, 37135 Verona, Italy
`d Servier, 14 rue de Bezons, 92415 Courbevoie, France
`e EUFEPS, Walingatan 26A, 11181 Stockholm, Sweden
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 16 May 2007
`Accepted 16 May 2007
`Published on line 21 May 2007
`
`Keywords:
`Drug solubility
`Bioenhancement
`Formulation
`Poor water solubility
`
`Drug absorption, sufficient and reproducible bioavailability and/or pharmacokinetic profile
`in humans are recognized today as one of the major challenges in oral delivery of new drug
`substances. The issue arose especially when drug discovery and medicinal chemistry moved
`from wet chemistry to combinatorial chemistry and high throughput screening in the mid-
`1990s. Taking into account the drug product development times of 8–12 years, the apparent
`R&D productivity gap as determined by the number of products in late stage clinical devel-
`opment today, is the result of the drug discovery and formulation development in the late
`1990s, which were the early and enthusiastic times of the combinatorial chemistry and high
`throughput screening. In parallel to implementation of these new technologies, tremen-
`dous knowledge has been accumulated on biological factors like transporters, metabolizing
`enzymes and efflux systems as well as on the physicochemical characteristics of the drug
`substances like crystal structures and salt formation impacting oral bioavailability. Research
`tools and technologies have been, are and will be developed to assess the impact of these
`factors on drug absorption for the new chemical entities.
`The conference focused specifically on the impact of compounds with poor solubility on
`analytical evaluation, prediction of oral absorption, substance selection, material and for-
`mulation strategies and development. The existing tools and technologies, their potential
`utilization throughout the drug development process and the directions for further research
`to overcome existing gaps and influence these drug characteristics were discussed in
`detail.
`
`© 2007 Elsevier B.V. All rights reserved.
`
`夽 The publication is the summary of the EUFEPS Meeting prepared by the scientific committee: “When poor solubility becomes an issue:
`from early stage to proof of concept.” which took place in GlaxoSmithKline research facility in Verona (Italy) on 26–27 April 2006.
`∗
`Corresponding author. Tel.: +46 8 723 5086; fax: +46 8 411 3217.
`E-mail address: hans.linden@eufeps.org (H. Lind ´en).
`0928-0987/$ – see front matter © 2007 Elsevier B.V. All rights reserved.
`doi:10.1016/j.ejps.2007.05.110
`
`MYLAN EXHIBIT 1011
`
`

`

`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 1 ( 2 0 0 7 ) 249–261
`
`250
`
`Contents
`
`250
`Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`1.
`251
`Physical/chemical properties of the drug substance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`2.
`252
`Biopharmaceutical evaluation of the drug substance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.
`254
`4. Drug candidate selection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`255
`5.
`Formulation strategies for solubility and bioenhancement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`258
`6.
`Biopharmaceutical evaluation of drug delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`7.
`Future outlook. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 259
`8.
`Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`259
`Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`260
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`260
`
`1.
`
`Introduction
`
`With the introduction of combinatorial chemistry and high
`throughput screening, the properties of new chemical enti-
`ties shifted towards higher molecular weight and increasing
`lipophilicity that results in decreasing aqueous solubility
`(Lipinski et al., 1997; Lipinski, 2000).
`Solubility in different solvents is an intrinsic material char-
`acteristic for a defined molecule. To achieve a pharmacological
`activity, the molecules must in general exhibit certain sol-
`ubility in physiological intestinal fluids to be present in the
`dissolved state at the site of absorption. The aqueous solubil-
`ity is a major indicator for the solubility in the intestinal fluids
`and its potential contribution to bioavailability issues.
`During drug substance development the molecules are
`screened in nanomolar receptor assays. The molecules with
`the best receptor binding are selected for further pre-clinical
`studies, for which more drug substance is synthesized.
`Already at this stage, solubility is of critical importance,
`because solubility estimates for the poorly defined drug sub-
`stance serve its pharmacological and toxicological profiling.
`When going first into humans, sufficient and well charac-
`terized solubility becomes even more critical. From now on
`the solubility or dissolution of the dose ranges in the vari-
`ous biophysiological media to which the drug substance or
`formulated drug substance will be exposed is expected to be
`reproducible and remain unchanged for the final development
`and eventually marketing.
`It is well accepted today throughout the scientific com-
`munity that drug substance solubility and especially aqueous
`drug substance solubility is an issue for the drug discovery as
`well as the early and late stage pharmaceutical development
`
`process and therefore needs to be addressed very early on,
`during compound design and optimization.
`The solubility or dissolution of the drug substance can be
`mainly altered on two levels, through material engineering of
`the drug substance or through formulation approaches. What-
`ever route is taken to enhance or modify the solubility and/or
`dissolution of a lead substance, it needs to be scalable to a
`commercially viable process later on in the development.
`Besides the aqueous solubility of a drug substance, its per-
`meability is a second critical aspect for oral bioavailability.
`The Biopharmaceutical Classification System (BCS) was intro-
`duced in the mid-1990s to classify the drug substances with
`respect to their aqueous solubility and membrane permeabil-
`ity (Amidon et al., 1995). Drug substances, for which solubility
`enhancement can improve the oral bioavailability, are sub-
`stances that are classified in class 2 (poor soluble/permeable)
`and class 4 (poor soluble/poor permeable). Especially for class
`2 substances, solubility enhancement is part of the strategies
`to improve the oral bioavailability.
`The BCS classification takes into account the required dose
`since low dosed drugs will sufficiently dissolve in the intesti-
`nal fluids of the GI tract to be absorbed, while higher doses
`of drugs with similar aqueous solubility will not. To gener-
`ally describe “solubility” the Pharmacopoeia (USP) uses seven
`different solubility expressions as shown in Table 1. The Euro-
`pean Pharmacopoeia uses similar solubility definitions except
`the ‘practically insoluble’ characteristic, which is not specified
`(European Pharmacopoeia 5.0).
`When looking at the product launches between 1995 and
`2002, out of 100 substances 14 were considered class I, 12 were
`classified as class II, 28 were class III and 46 were class IV
`substances (Mehta, 2002). If a drug substance exhibits a poor
`
`Table 1 – Solubility definition in the USP
`Description forms
`Parts of solvent required
`(solubility definition)
`for one part of solute
`
`Solubility range
`(mg/ml)
`
`Solubility assigned
`(mg/ml)
`
`Very soluble (VS)
`Freely soluble (FS)
`Soluble
`Sparingly soluble (SPS)
`Slightly soluble (SS)
`Very slightly soluble (VSS)
`Practically insoluble (PI)
`
`<1
`From 1 to 10
`From 10 to 30
`From 30 to 100
`From 100 to 1000
`From 1000 to 10,000
`>10.000
`
`>1000
`100–1000
`33–100
`10–33
`1–10
`0.1–1
`<0.1
`
`1000
`100
`33
`10
`1
`0.1
`0.01
`
`

`

`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 1 ( 2 0 0 7 ) 249–261
`
`251
`
`aqueous solubility, the product development will have to focus
`on the investigation of various other drug substance character-
`istics like its physicochemical, biopharmaceutical properties
`and the targeted dose to identify the potential impact of the
`solubility on the further product development. Today, about
`35–40% of the lead substances are known to have an aqueous
`solubility of less than 10 ␮M or 5 mg/ml at pH 7 and it is not
`expected that this figure will change in the future.
`
`Physical/chemical properties of the drug
`2.
`substance
`
`Solubility is an intrinsic material property that can only be
`influenced by chemical modification of the molecule as such,
`like salt complex or prodrug formation. In contrast to this,
`dissolution is an extrinsic material property that can be influ-
`enced by various chemical, physical or crystallographic means
`like complexation, particle size, surface properties, solid state
`modification or solubilization enhancing formulation strate-
`gies.
`Solubility is one of the key physicochemical parameters of
`a new molecule that needs to be assessed and understood
`very early on in drug discovery and drug candidate selec-
`tion process. Starting with the first amount of the substance
`synthesized, commonly a few milligrams, the first series of
`analytical tests include that of the equilibrium solubility of
`the substance in a given solvent, as well as the apparent or
`dynamic solubility, which reflects the concentration of the
`substance in solution under certain conditions. The value of
`equilibrium solubility is often limited by the test duration,
`which is normally between 4 and 24 h. The solubility and
`apparent solubility of the substance in an aqueous system are
`dependent on several factors (Table 2).
`Rather than single point determinations, a solubility pro-
`file of the substance is required to identify potential issues
`for drug precipitation in vivo. Especially the pH profiling for
`weakly basic salts is of critical importance as their solubility
`will vary in the intestinal pH (typically pH 1–8) and pre-
`cipitation may occur. The pH profiling should be performed
`in different bio-relevant buffer systems to mimic the high
`concentrations of the most common counter ions of phar-
`maceutical salts in GI fluids. This screen allows detection
`of a potential counter ion exchange and formation of more
`stable/less soluble salts of a molecule that will lead to precip-
`itation in vivo. The pH profile additionally provides the basic
`guidance to choose among potential solubilization strategies.
`Conversion of the molecule to other salts or hydrates needs
`to be taken into account and evaluated in the solubility profil-
`
`Table 2 – Factors influencing solubility
`
`Composition of the aqueous media
`Temperature
`pH
`Solid state (amorphous, crystalline)
`Polymorph type
`Counter ions (salt formation)
`Ionic strength
`
`ing. Different salts of the substance can be formed dependent
`on the buffer systems as well as their ionic strength. It is gen-
`erally accepted that there should be a minimum difference
`of three units between the pKa value of the ionizable group
`and of the possible counter ion (Bowker, 2002). (Ampholytes
`display a more complex solubilization process (two step reac-
`tion) including micelle formation at high ionic strength. Buffer
`salts as well as hydrates might have different solubility char-
`acteristics.)
`Therefore different analytical methods have been devel-
`oped to assess the substance in solution or solid state
`(precipitates) (Giron and Grant, 2002). Analytical methods for
`the substance in pH-controlled solutions are LC coupled with
`UV and MS. For the solid state analysis, powder X-ray diffrac-
`tion, energy dispersive X-ray (EDX), Raman spectroscopy, IR,
`microscopy (polarized light microscopy (PLM), environmental
`scanning electron microscopy (ESEM)) and thermal analysis
`are used.
`The solubility profiling should include any other biorele-
`vant dissolution media like simulated gastric fluid (SGF) with
`and without enzymes, fasted state simulated intestinal fluid
`(FaSSIF) and fed state simulated intestinal fluid (FeSSIF) at pH
`5.0 and 6.5 or human gastric or intestinal fluids (HIF’s).
`In parallel to the analytical characterization of the initial
`material of the drug substance, substantial efforts are invested
`into understanding and optimization of the crystalline struc-
`ture and to identify a potential pseudo-thermodynamic stable
`form of the substance. These investigations are looking into
`the polymorphs, solvates and salts formed by the substance
`under various conditions to identify the most suitable material
`for dosage form development, scaling up and later manufac-
`turing.
`Polymorphs appear in a number of different structures as
`non-mixed polymorphs (free base or acid) or as mixed poly-
`morphs like salts, co-crystals (Vishweshwar et al., 2006), guest
`substances, hydrates or solvates (Bernstein, 2002; Brittain,
`1999).
`While the polymorph screening focused more on the num-
`ber of different polymorphs by crystallization in different
`solvents in the past, it now has shifted towards qualitative
`data on polymorph formation under thermodynamically con-
`trolled conditions to get more accurate data on the potential
`risk for polymorphic changes in the final dosage form under
`the expected storage conditions. These constant and con-
`trolled conditions include pressure, temperature, solvent and
`time.
`Once a polymorph is found and characterized, additional
`polymorph evaluation experiments should be performed to
`provide information on its kinetic stability.
`The evaluation starts with determining, e.g. if the sub-
`stance forms only one anhydrate or several anhydrates. Once
`different polymorphs have been identified it is important to
`characterize whether they are monotropically or enantiotrop-
`ically related. In case of an enantiotropic phase transition,
`the transition temperature as well as the temperature stabil-
`ity range must be very well characterized. Long-term stability
`is tested in slurry experiments for at least 2 months in dif-
`ferent non-solvate forming solvents and/or at temperatures
`in the stability range of the anhydrates. From this series of
`polymorph experiments the most thermodynamically stable
`
`

`

`252
`
`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 1 ( 2 0 0 7 ) 249–261
`
`form is selected and will be further evaluated in competi-
`tive slurry experiments at room temperature with any other
`known anhydrates and solvates. Reversibility between the dif-
`ferent forms will be included in the evaluation in long-term
`slurry experiments and summarized using binary and ternary
`phase diagrams. As an example, formoterol fumarate has been
`found to form seven modifications in the polymorph screen.
`Running the polymorph experiments, the dihydrate form has
`been determined as the most stable solid form due to its favor-
`able capacity of forming efficient hydrogen bonds between the
`formoterol and the fumarate leading to a well packed crystal
`structure (Jarring et al., 2006).
`To improve physicochemical properties of the drug sub-
`stance with regard to manufacturing, isolation and long-term
`storage, as well as to improve solubility and/or dissolution
`properties of the drug substance, salt formation is tradition-
`ally preferred by medicinal chemists for weak bases or weak
`acids. Since only 20–30% of the new molecules form salts eas-
`ily, the 70–80% remain challenging (Serajuddin and Pudipeddi,
`2002).
`Salt formation is a two-step process in which proton trans-
`fer in solution has to take place followed by a crystallization
`step. Identification of a suitable solvent to achieve sufficient
`attractive forces of the substance salt as well as to overcome
`ion and molecule solvation is required. To achieve a ther-
`modynamically stable salt with sufficient aqueous solubility,
`it is important to understand the salt structure formed by
`a specific counter ion. Counter ions act as a template that
`interacts through non-covalent bonds with the molecule by
`their conformation and bonding capabilities, which directly
`influence the three-dimensional structure. Data bases on the
`possible salt structures of functional groups can provide some
`valuable predictions on the number of possible solvates and
`hydrates and their coordination number defined as the num-
`ber of non-covalent bonds formed by the ion (e.g. Cambridge
`Crystallographic Data Center). Selected salts of a molecule will
`be assessed in a salt screening following the same principle
`as polymorph screening to investigate the long term stability
`as well as its conversion to other, more stable salts and its
`precipitation in different aqueous and bio-relevant media.
`With the increasing knowledge about the implications of
`polymorphs and salts in drug discovery, automated tools
`are being developed to standardize and implement these
`experiments as a routine process in drug discovery and lead
`substance selection (York, 1999; Rohl, 2003).
`To overcome poor aqueous solubility or erratic bioavail-
`ability, chemical modification leading to a prodrug has
`successfully been used for several substances. The most com-
`mon used prodrug approach is the incorporation of a polar
`or ionizable moiety into the molecule. The incorporation of
`N-acyloxyalkyl moieties of different chain length leads to
`a reduced crystal lattice interaction and decreasing melting
`point with increasing number of methylene groups (Stella
`et al., 1998). In vivo studies in dogs with the N-acyloxyalkyl
`derivatives of phenytoin confirmed a higher bioavailability in
`the fed state that did not correlate with decreasing water sol-
`ubility (Stella et al., 1999). Prodrugs also might reduce the
`pre-systemic metabolism of the substance in the GI tract
`or the release of the drug substance itself by enzymatic
`cleavage of the prodrug moiety close to the site of drug
`
`absorption. Fosphenytoin is a phosphate prodrug of pheny-
`toin that is metabolized by phosphatases to release its active
`moiety phenytoin. In vivo studies in dogs and humans have
`demonstrated a better bioavailability after oral, intramuscular
`and intravenous administration. Additionally fosphenytoin
`has shown a better safety profile compared to its original
`form phenytoin sodium (Stella, 1996). Examples for success-
`ful phosphate prodrugs are fosphenytoin (CerebyxTM) and
`amprenavir phosphate (VX-175/GW 908). Another successful
`prodrug development is bortezomib, a boronic acid pro-
`drug (VelcadeTM) (Sanchez-Serrano, 2006). Current research
`is focusing on the development of new prodrug moieties
`like N-glycines, sulfenamides and cysteamines to improve
`bioavailability by better solubility or metabolic stability.
`Once the most favorable thermodynamically stable form
`of a lead substance is determined and its solubility profile is
`characterized, investigations into its biopharmaceutical char-
`acteristics using in vitro, in silico or in vivo trials in animals
`will provide the necessary information on the substances
`predicted in vivo performance and for the design of a drug
`delivery system.
`
`Biopharmaceutical evaluation of the
`3.
`drug substance
`
`The biopharmaceutical evaluation and prediction is another
`crucial part of the lead drug candidate selection process. It
`is well established today that drug substances that do not
`meet at least certain biopharmaceutical criteria are returned
`to the medicinal chemist for lead optimization (Clark and
`Grootenhuis, 2003).
`One of the most critical aspects of in vitro screening assays
`is the determination of the true concentration of the free drug
`in the assay. The concentration of free drug in the assay is
`normally calculated rather than measured. Unexpected low
`solubility and precipitation of the drug in the media due to
`the dilution sequence (e.g. dilution from an aqueous buffer
`solution or from DMSO solution) has been reported. When
`the calculated drug concentration in the assay is not reached,
`wrong conclusions are most likely to be drawn regarding effi-
`cacy, toxicity or permeability (Di and Kerns, 2004).
`The main in vitro tools used for the assessment of drug
`absorption and permeability are listed in Table 3.
`The CaCo-2 cell monolayers are well-established in vitro
`systems that are used in various stages of the drug develop-
`ment process to assess drug absorption and the underlying
`processes (Shah et al., 2006). CaCo-2 cell lines provide valuable
`information about the permeability and absorption potential
`
`Table 3 – In vitro systems to identify permeability and
`absorption
`
`CaCo-2 cell lines
`Madin-Darby canine kidney (MDCK) cell lines
`Parallel artificial membrane permeability assay (PAMPA)
`Immobilized artificial membrane chromatography (IAM)
`Advanced compartmental absorption and transit (ACAT)
`Lipophilicity (log D and log P)
`
`

`

`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 1 ( 2 0 0 7 ) 249–261
`
`253
`
`of a drug substance. They can further be used in later devel-
`opment to provide information about the potential impact of
`metabolic or efflux systems on the drug substance and to clas-
`sify the drug according to the BCS.
`When using CaCo-2 cell monolayers to measure perme-
`ability of poorly soluble substances attention must be drawn
`to possible drug accumulation within the cells, binding to
`proteins or adhesion to plastic surfaces. To avoid variability
`beyond ±10% in the permeability data obtained with CaCo-2
`cell tests, standardization and calibration are critical.
`The parallel artificial membrane permeability assay
`(PAMPA) is an artificial, intrinsic permeability measurement
`that is used for drug lead substance selection and for pre-
`clinical studies in a high throughput manner. The PAMPA
`system is based on synthetic phospholipids and fatty acids
`that mimic physiological conditions. PAMPA is used as a low
`cost alternative to cell based systems for early ADME screening
`(Avdeef, 2005). High confidence in the predictions using PAMPA
`has been reported for drugs with good effective permeability
`−6 cm/s) and the poor effective permeability (<2.5
`−6 cm/s),
`(>10
`while the results for drugs in between are difficult to interpret.
`For efficient and rapid investigation into mechanisms of
`drug absorption, CaCo-2 cell monolayers are combined with
`the PAMPA assay (Kerns et al., 2004). Drugs that correlate in
`the comparison are mainly absorbed through passive diffu-
`sion. Substances, which show higher permeability in PAMPA
`than in a CaCo-2 model, are substrates for efflux or for reduced
`passive diffusion of acids under CaCo-2 pH gradient set-
`up. Substances with a higher CaCo-2 cell permeability when
`compared to the PAMPA system have underlying absorptive
`mechanisms like paracellular, active transport or increased
`passive diffusion in a CaCo-2 cell pH gradient.
`Immobilized artificial membranes (IAM) are solid phase
`models of liposome membranes for partitioning studies of
`drugs into membranes. IAM chromatography is a simple
`experimental tool that allows for large number screening
`tests in the drug discovery phase to identify the potential
`of a drug substance of being absorbed, independently from
`other factors involved in poor bioavailability like pre-systemic
`metabolism, efflux systems or transporters (Pidgeon et al.,
`1995).
`Poor prediction still persists for the drug absorption in the
`colon. Due to the reduced paracellular or carrier-mediated
`absorption, efflux mechanisms, the reduced surface area, the
`colonic solubility environment (pH 6–7), the reduced volume
`and mixing and the unknown chemical metabolism of the sub-
`stance along the GI tract, the contribution to drug absorption
`still remains unclear. However, poorly soluble substances and
`drugs from extended release formulations will reach the colon
`at a considerable concentration and stay there for a long time.
`Even if absorbed slowly, this may contribute substantially to
`absolute bioavailability.
`One of the first prediction models used a mathemati-
`cal approach to estimate the fraction dose absorbed (Oh
`et al., 1993). This mathematical calculation uses only four
`parameters to estimate the dose that can be absorbed: initial
`saturation, absorption number, dose number and dissolution
`number.
`For the predictions of human drug absorption today the
`specific absorption rate (SAR) or human absorbable dose (Dabs)
`
`model is frequently used. The SAR plots the absorption num-
`ber versus the dose number (Collins and Rose, 2004). The Dabs
`model is based on the permeability of a substance, its sol-
`ubility in simulated intestinal fluids, the surface area of the
`human GI tract defined as 800 cm2 and a GI transit time of
`3.3 h (Yu, 1999). Plotting the Dabs versus the expected thera-
`peutic dose already gives some hint whether drug absorption
`will be the rate-limiting step for a conventional dosage form.
`For example, pioglitazone, a weak base with a good solubility
`at low pH will not require an enabling formulation due to its
`solubilization in the stomach; neither would tadalafil, a non-
`ionizable substance over the physiological pH range and a low
`solubility in all relevant media, because of its low dose (20 mg).
`Another model proposed is using the maximum absorbable
`dose (MAD) concept taking into account a small intestinal
`transit time of 4.5 h, a small intestinal fluid volume of 250 ml,
`the substance solubility and an absorption constant for the
`drug substance (Johnson and Swindell, 1996). The MAD val-
`ues provide good predictions if the projected human dose can
`be absorbed. Evaluating a series of substances it can serve
`as a guiding tool for rank ordering of the potential lead drug
`candidates in a specific series of substances (Curatolo, 1998).
`Using the MAD model as a starting point, computational
`systems have been developed to simulate drug absorption in
`vivo (Johnson, 2003). Additional input parameters like water
`absorption from the GI tract and changing gastrointestinal
`permeability have been introduced to improve the simula-
`tions.
`Another computational system for the simulation of drug
`absorption is based on the advanced compartmental absorp-
`tion and transit (ACAT) model (Agoram et al., 2001). The ACAT
`model is an extended version of the compartmental absorp-
`tion and transit (CAT) model that uses seven small intestinal
`compartments for prediction. The ACAT takes into account
`the colon as another compartment for drug absorption, which
`cannot be neglected for poorly soluble and poorly permeable
`substances as well as for sustained release formulations. The
`selection of the dissolution media for each compartment is
`an important factor that affects the accuracy of the prediction
`in the ACAT model. Using FaSSIF media for example, provides
`better correlations than simple buffer systems at pH 6.5 for
`soluble substances, however, for poorly soluble substances
`under-predictions are more likely for pH 6.5 buffer, while over-
`predictions are the case for the FaSSIF media. The output of
`the ACAT model has been improved by including other param-
`eters on the drug substance like particle size/radius, density,
`diffusion coefficient, log D, pKa and molecular weight.
`The simulations for both systems are relatively good, but
`tend to be less accurate for the poorly soluble substances due
`to the difficulty in modeling the colonic drug absorption.
`The major limitations seen today in the in silico models
`concern the potential drug precipitation in the GI tract (e.g.
`weak bases, salts of poorly soluble substances), the impact
`of colonic drug absorption, especially of poorly soluble sub-
`stances when at high dose and the effect of the drug particle
`size on absorption. While further validation work is ongoing,
`the existing in silico tools already provide important informa-
`tion for lead candidate selection and/or optimization and can
`be developed further to improve the accuracy of the simulation
`(Kuentz et al., 2006).
`
`

`

`254
`
`e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 1 ( 2 0 0 7 ) 249–261
`
`Following the biopharmaceutical evaluation of the sub-
`stance’s solubility/dissolution behavior in aqueous or bio-
`relevant media, permeability, chemical and metabolic stability
`are assessed, in vitro. Thereafter, the absorption, food effect,
`first pass metabolism, PK profiles are finally quantified in
`different animal species (rats, dogs, monkeys) in a dose-
`escalating manner. These studies are designed to establish
`first in vivo oral exposure rate and dose linearity. For these
`studies the drug is administered as a solid, suspension or
`solution. These studies should also provide information on
`any dependency of absorption on particle size as well as on
`the upper limit of exposure, the clearance and potential food
`effects of a standard formulation.
`Later on, in particular when potential for extended release
`formulation development is considered, more in depth inves-
`tigations into the mechanism of drug absorption can be
`performed using, for instance, a regional perfusion model that
`can be applied to animals (e.g. dogs) or humans (Lennern ¨as
`et al., 1992). The regional perfusion model is also used to
`establish a correlation between the human permeability and
`the CaCo-2 cell permeability (Lennern ¨as et al., 1997) and
`provides important information for the design of the drug
`delivery system. To facilitate and improve the investigation
`into regional drug absorption and its underlying mechanisms,
`special capsules have been developed that permit a time- and
`position-controlled release of the drug substance or formula-
`tion in defined areas of the GI tract (Wilding et al., 2000)
`To get at least some data on a substance performance in
`humans before lead substance selection or entering into the
`clinical development, the European Medicines Agency (EMEA)
`recently accepted the concept of a single micro-dosing study
`in humans (EMEA, 2003). Micro-dosing studies allow one single
`dose of less than 1/100th of the calculated pharmacologi-
`cal dose and not more than 100 ␮g in total dose for small
`molecules. Even so there is only very limited experience of
`micro-dosing studies and their value in the drug development
`process so far. Micro-dosing is expected to become a useful
`tool in identifying substances with critical biopharmaceutical
`properties as well as improving significantly in silico predic-
`tions of drug absorption and deposition (Wilding and Bell,
`2005).
`While some tools provide highly valuable data for lead sub-
`stance selection for one substance, they might not for others.
`Therefore the series of different in vitro, in vivo and predictive
`methods and tools provide a good base for putting together a
`meaningful evaluation program that needs to be designed for
`each individual substance based on the information gathered
`during drug discovery and substance characterization.
`
`4.
`
`Drug candidate selection
`
`Multidisciplinary teams are becoming an essential part of
`the lead candidate selection process in the pharmaceutical
`industry. Their objective is to address early on the various
`characteristics of the drug substances from the chemical as
`well as the pharmacological, toxicological and biopharma-
`ceutical point of

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