solubility estimation in git

Upload: sayuj-nath

Post on 03-Jun-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/11/2019 Solubility Estimation in GIT

    1/12

    Estimating drug solubility in the gastrointestinal tract

    J.B. Dressmana, M. Vertzoni b, K. Goumas c , C. Reppasb ,

    a Department of Pharmaceutical Technology, Johann Wolfgang Goe the University, Frankfurt, Germanyb Department of Pharmaceutical Technology, National & Kapodistrian University of Athens, Greece

    c Department of Gastroenterology, Red Cross Hospital of Athens, Greece

    Received 23 April 2007; accepted 10 May 2007

    Available online 29 May 2007

    Abstract

    Solubilities measured in water are not always indicative of solubilities in the gastrointestinal tract. The use of aqueous solubility to predict oral

    drug absorption can therefore lead to very pronounced underestimates of the oral bioavailability, particularly for drugs which are poorly soluble

    and lipophilic. Mechanisms responsible for enhancing the luminal solubility of such drugs are discussed. Various methods for estimating intra-

    lumenal solubilities are presented, with emphasis on the two most widely implemented methods: determining solubility in fluids aspirated from the

    human gastrointestinal tract, and determining solubility in so-called biorelevant media, composed to simulate these fluids. The ability of the

    biorelevant media to predict solubility in human aspirates and to predict plasma profiles is illustrated with case examples.

    2007 Published by Elsevier B.V.

    Keywords: Solubility; Gastrointestinal tract; Humans; Dogs; Simulated media; Oral absorption

    Contents

    1. Why estimate intra-lumenal solubility of drugs? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 591

    2. Solubilization mechanisms in the gastrointestinal tract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 592

    3. Procedures for aspirating and measuring solubility in gastrointestinal fluids . . . . . . . . . . . . . . . . . . . . . . . . . . . . 594

    4. Estimation of drug solubility in the stomach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 595

    4.1. Fasted state, gastric . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 595

    4.2. Fed state, gastric . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 595

    5. Estimation of drug solubility in the small intestine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 597

    5.1. Fasted state, small intestine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 597

    5.2. Fed state, small intestine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 598

    6. Use of drug dissolution and solubility data in predicting plasma profiles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 599

    7. Conclusions future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 600

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 601

    1. Why estimate intra-lumenal solubility of drugs?

    After oral administration, intra-lumenal drug concentrations

    influence the rate of appearance in plasma and, in certain

    situations they can determine the total amount reaching the

    general circulation. In turn, the solubility under gastrointestinal

    (GI) conditions sets the upper limit to the intra-lumenal

    concentration that can be achieved.

    Advanced Drug Delivery Reviews 59 (2007) 591602

    www.elsevier.com/locate/addr

    This review is part of the Advanced Drug Delivery Reviewstheme issue on

    Drug solubility: How to measure it, how to improve it". Corresponding author. Department of Pharmaceutical Technology, National

    & Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou

    Athens, Greece. Tel.: +30 210 727 4678.

    E-mail address: [email protected](C. Reppas).

    0169-409X/$ - see front matter 2007 Published by Elsevier B.V.doi:10.1016/j.addr.2007.05.009

    mailto:[email protected]://dx.doi.org/10.1016/j.addr.2007.05.009http://dx.doi.org/10.1016/j.addr.2007.05.009mailto:[email protected]
  • 8/11/2019 Solubility Estimation in GIT

    2/12

    The stomach, although not the primary site for drug

    absorption, provides the first site at which an orally administered

    formulation can quantitatively release its drug. For compounds

    highly soluble at gastric pH, complete dissolution can occur in the

    stomach. For such compounds, gastric emptying may well limit

    the subsequent rate of absorption from the small intestine (e.g.

    [1]). For poorly soluble weak acids like ibuprofen [2] littledissolution will occur in the stomach. By contrast, the small

    intestine with its higher pH offers a more favorable environment

    for dissolution of acids. For ibuprofen and similar weak acids,

    emptying from the stomach becomes rate limiting to the onset of

    dissolution and hence absorption. For poorly soluble neutral

    compounds, dissolution will be slow in the gastric region and in

    many cases will not be complete before the drug reaches the first

    absorptive sites in the small intestine. Incomplete dissolution in

    the GI tract of such compounds can severely restrict their oral

    bioavailability. Finally, for poorly soluble weak bases, solubility

    is likely to be higher in the (preprandial) stomach than elsewhere

    in the GI tract. This can result in a supersaturation as the drugmoves out of the stomach into the higher pH small intestine.

    Precipitation in the small intestine may result, though this process

    appears to be hindered by the bile components[3].

    In the fed state, the intra-gastric performance of immediate

    release tablets has to date been studied primarily in dogs. In

    these experiments, food components have been shown to delay

    the dissolution of highly soluble compounds during gastric

    residence [4,5]. Similar observations have been made very

    recently in humans by Brouwers et al. [6]. In this study a food-

    induced delay in the dissolution of fosamprenavir in the fed

    stomach was reflected in changes in the plasma profile of

    amprenavir[6].

    In the small intestine, the drug concentration at the intestinalwall,Cw, is one of the two principal determinants of the rate of

    drug uptake and transport across the cellular membrane of the

    intestinal epithelium, the other being permeability. Depending

    on the mechanism of transport, the drug flux through the

    intestinal mucosa, J, can be described with the following

    equations:

    For passive transport : JCWPW 1

    For carrier mediated transport : J JmaxCW

    CWKM2

    where Pw is the effective membrane permeability coefficient,

    Jmaxis the maximum drug flux through the membrane and KMis the MichaelisMenten constant. Since the small intestine is

    the primary site of absorption for the great majority of

    compounds, the concentration of interest for the calculation of

    flux is the one developed in this region. Intra-intestinal drug

    concentrations after oral administration of drug powder were

    first measured about ten years ago[7]. Recently, intra-intestinal

    drug concentrations have been measured after oral drug

    administration using procedures that do not perturb the natural

    fluid balance and composition in the region substantially and,

    thus, enable administration of marketed dosage forms and/or

    typical meals[6,8,9,10]. These studies have proved very useful

    in answering detailed questions about factors important to

    absorption of several drugs. For example, it appears that in the

    fed state concentrations of danazol in the aqueous phase of the

    intestinal contents may not correlate with blood levels (despite

    the limited aqueous solubility of this compound) [10], that

    amprenavir's permeability from the marketed formulation is not

    influenced by p-gP effects [9], and the delayed absorption offosamprenavir in fed state is not related to intra-intestinal but

    rather to intra-gastric processes[6].

    Drawbacks with direct measurements of intra-gastric or

    intra-intestinal drug concentrations are the specialized proce-

    dures used, the associated costs and ethical issues in terms of

    exposing humans to the procedure and drugs without any direct

    therapeutic benefit to the subject. As a result, data are usually

    collected from just a limited number of subjects and studies

    reported in the literature are few [6,8,9,10]. One way to

    eliminate some of these drawbacks would be to use imaging

    techniques. However, such techniques would also be expensive

    to apply and, at best, they are still in their infancy with regard tothis application[11]. Another way to improve the cost:benefit

    ratio of the experiments is to collect human aspirates without

    prior administration of the drug and use them for measuring

    the parameters of interest. Analogously, intestinal permeability

    in humans is usually performed off-sitein cell cultures rather

    than directly in human subjects. Interestingly, it has recently

    been established that using human aspirates (containing the

    drug and formulation excipients) as the medium for permeabil-

    ity studies can lead to surprisingly different results than when

    permeability is measured from solutions consisting of simple

    buffers[9]. Similarly, it is expected that determining solubilities

    in human aspirates will help us to estimate intra-lumenal

    dissolution kinetics much better than studying solubility insimple buffer solutions (e.g.[12]).

    Another reason that estimation of intestinal permeability and

    intra-lumenal solubility has become of major interest during the

    last decade is that both of these parameters are required for

    application of the Biopharmaceutics Classification Scheme

    principles[13] to drug development.

    This article discusses the procedures necessary to obtain

    reliable results for solubility in human aspirates and additionally

    discusses whether the composition of the GI fluids can be

    simulated with media that can be collected from animals or

    manufactured in the laboratory.

    2. Solubilization mechanisms in the gastrointestinal tract

    Although the underlying driver for solubility in the GI fluids

    is the aqueous solubility of the drug, the solubility in the GI tract

    may additionally be influenced by the pH profile, by

    solubilization via naturally occurring surfactants and food

    components, as well as by complexation with food and native

    components of the GI milieu. Since these additional influences

    can result in orders of magnitude changes in solubility (see

    Table 1for some examples), it is worthwhile addressing them in

    some detail.

    The pH profile in the GI tract is of primary importance for

    drugs that can ionize in this range. Rearranging the Henderson

    592 J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    3/12

    Hasselbalch equation (e.g. [17]) we see that for a monobasic

    compound the concentration of acid required for saturation of

    the medium will be enhanced at pH values where ionizationoccurs:

    Cs Cs;0 110pHpka

    3

    where Cs,0 is the solubility of the non-ionized acid form

    (intrinsic solubility) and Cs is the total solubility (sum of

    intrinsic solubility and the existing concentration of ionized

    form) at the pH of interest. For a monoacidic compound the

    equivalent equation is:

    Cs Cs;0 110pkapH

    4

    Equations for more complex ionization reactions can be foundelsewhere in this issue[18].

    The pH in the stomach in the fasted state has been the subject

    of many studies over the years and the general consensus is that

    in healthy adult humans the fasted pH usually lies in the range

    pH 13. Elevated pH can be observed in a modest percentage of

    elderly subjects due to waning ability to produce gastric acid.

    The effect is particularly pronounced in the Japanese popula-

    tion, although the incidence of achlorhydria there does appear to

    be falling with time[19]. In North Americans, elevated pH in

    the elderly is the exception rather than the rule [20]. On the

    other hand, gastric pH can be elevated by pharmacological

    interventions such as H2-receptor antagonists and proton pumpinhibitors, which are used widely in Western populations. By

    contrast, hyper-secretion of acid is very rare, mostly associated

    with specific diseases such as ZollingerEllison syndrome[21].

    After meal intake, pH in the stomach usually rises due to

    buffering effects of the meal contents, and may initially reach

    values of up to 7, depending on meal composition. With the

    continuous secretion of gastric acid, the pH value then trends

    back down to baseline over a period of several hours[22,23].

    In the small intestine the pH exhibits a profile, with lowest

    pH proximally and somewhat higher pH values in the distal

    regions. pH values in the duodenum in the fasted state tend to lie

    slightly below neutral (pH 66.5) [22,23]. The pH in the

    proximal small intestine is influenced more by meal intake than

    the pH in the distal regions, as might logically be expected from

    the huge swings in pH observed in the stomach between the

    fasted and fed states. After meal intake, pH will be influenced

    by the chyme coming into the small intestine from the stomach.

    Thus, after meal intake the pH values may actually rise initially

    in the proximal small intestine. With time, as the incoming

    chyme becomes ever more acidic the pH will actually drop aslow as 55.5, even in the jejunum [22,23]. Meanwhile, pH

    values in the distal ileum appear to remain stable at around pH

    7.5 [24]. This is consistent with digestion and absorption

    occurring primarily in the proximal part of the small intestine:

    up to one-half of the small intestine can be removed without

    disturbing the ability to sustain nutritional balance[25].

    The pH in the proximal large intestine reverts to more acidic

    values, typically between 5 and 6.5[26], due to fermentation of

    undigested foodstuffs (cellulosics and the like) to short chain fatty

    acids (e.g.butyrate, propionate, acetate) by the colonic bacteria.

    The wide range of pH values encountered by ionizable drug

    substances within the GI tract suggests that large swings insolubility may occur, with implications forCw(Eqs. (1) and (2))

    and hence the efficiency of absorption.

    The second major influence on solubility in the GI tract is

    solubilization. Solubilization mechanisms include micellar

    solubilization by either native or co-ingested surfactants,

    binding to peptides or proteins and solubilization in lipid

    components of the meal.

    In the stomach, the source of surfactants is not so clear,

    although it has been consistently observed that the surface

    tension of gastric fluids is commensurate with a significant level

    of surfactant (e.g. [23]). In some subjects, the reflux of bile

    components into the stomach appears to be the source of

    surfactant behavior, but in others no bile components can bedetected in gastric aspirates [27]. In addition to native

    surfactants, meal intake offers the potential for solubilization

    of drugs during gastric residence. Macheras et al. have

    demonstrated that chlorothiazide and hydrochlorothiazide are

    well solubilized by casein micelles in milk[28]whereas more

    lipophilic compounds, such as indomethacin and diazepam, are

    additionally solubilized into the milk fat [29]. On the other

    hand, meal components can have an adverse effect on solubility

    if an insoluble complex with the drug is formed. The classical

    example here is complexation with calcium, which precipitates

    bisphosphonates and tetracyclines, rendering them insoluble

    and thus unavailable for absorption[30].In the small intestine the primary source of solubilization is

    clear the bile components such as bile salt conjugates,

    phospholipids and cholesterol team up (additionally with

    lipolysis products in the fed state) to create mixed micelles

    that can solubilize lipophilic molecules very well. Indeed,

    correlations have been established for solubilization by mixed

    micelles as a function of logPfor neutral compounds[31]. The

    concentration of mixed micelles is much higher after meal

    intake, as the gall bladder contracts in response to a meal and

    empties its contents into the duodenum at the level of the

    Sphincter of Oddi. In addition,in vitrostudies indicate that the

    solubilization capacity of the micelles is enhanced by the

    incorporation of lipolysis products [32]. It should be noted,

    Table 1

    Mean equilibrium solubility data in g/ml for three drugs, illustrating the large

    differences between solubility in simple aqueous media and biorelevant media/

    aspirates

    Felodipine Ketoconazole Dipyridamole

    (Intrinsic) aqueous solubility 1 a 6.9b 5.0b

    Solubility in FaSSGF 1.4a

    9054a

    11,417a

    Solubility in HGFfasted 0.4a 9025 a 8530 a

    Solubility in FeSSIF 188 c 406540 d 181246 d

    Solubility in HIFfed 412c 476989 d 160254 d

    a From Ref.[15].b Intrinsic solubility (solubility of the non-ionized form) from Ref. [14].c Numbers were extracted from the graphs of Ref.[16].d Data vary with the aspirations times of HIFfed and with composition of

    FeSSIF[14].

    593J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    4/12

    however, that absorption of lipolysis products is generally

    completed in the jejunum and the bile salts are reabsorbed

    actively in the ileum [33], thus solubilization effects are

    restricted primarily to the upper small intestine.

    As there is a paucity of information in the literature about

    surface tension or surfactants in the colonic fluids, it is not

    possible to comment at this time on potential solubilization inthis region.

    Other factors that can influence the capacity of the GI tract to

    dissolve drugs in a pharmacokinetically relevant way include

    the volume of fluids available in the region(s) of interest and the

    passage time of the drug/dosage from up to and through the

    regions where the drug is most efficiently absorbed.

    3. Procedures for aspirating and measuring solubility in

    gastrointestinal fluids

    Given the large number of factors affecting solubility,a priori

    prediction of intra-lumenal solubility is practically impossible.Measurement of drug concentrations in situ requires highly

    specialized expertise and is complicated and costly, with the result

    that few studies of this kind have been performed (see above). A

    third, physiologically relevant and generally more practical

    approach to estimating drug solubility in the GI tract is to aspirate

    fluids from the human GI tract and measure the solubility in these

    fluids ex vivo. With this approach, issues include subject selection,

    conditions under which the fluids are aspirated, maintaining the

    quality of the aspirated sample ex vivo, the methodology for

    measuring the solubility, and associated costs.

    First, the composition of aspirated samples can vary

    dramatically with the demographics and medical history of

    the subjects from which the aspirates are collected. Early in thedevelopment process, the drug will typically be administered to

    young, healthy subjects to assess safety. In bridging bioequi-

    valence studies performed later in the clinical development,

    formulations are also typically tested in healthy adults.

    Therefore it seems reasonable to use a set of subjects with

    similar characteristics to obtain aspirates of the GI fluids.

    Second, it is necessary to write a well-defined protocol for

    the aspiration studies. It is very important to stipulate fasting

    requirements prior to the study day, to define water intake when

    performing aspiration in the fasted state, and to exactly define

    meal intake (composition, rate of ingestion) and timing of meal

    intake in relation to timing of aspiration when performingaspiration in the fed state.

    Aspirations can be performed after nasal or oral intubation,

    with the nasal route considered more practical by gastroenter-

    ologists since the tube arrives at the trachealesophageal

    junction at an angle which is more favorable to entry into the

    esophagus.

    In the fasted state, aspirations after administration of 200

    250 ml of water allow for the protocol to be closer to that of a

    standard pharmacokinetic study. In addition, since water flux is

    limited in the fasted stomach, such amounts of water will

    dramatically affect the composition of gastric contents and,

    therefore, the ability of the drug to dissolve during gastric

    residence may be greatly modified. Additionally, for aspiration

    from the fasted upper small intestine, administration of 200 or

    250 ml water before the aspiration procedure will improve the

    chances of aspirating adequate volumes.

    In the fed state, aspirates will vary in composition with the

    size and type of meal and with the time after the meal's

    consumption (e.g.[23]). One way to resolve this problem would

    be to administer the same meal typically administered inbioavailability/bioequivalence studies. However, solid meals

    create problems with sample aspiration due to potential

    clogging of the aspiration port. As a result, alternative liquid

    meals have been suggested [34,35]. The duration of the

    aspiration period after consumption of the meal should be as

    long as the residence time in the region of the GI lumen from

    which samples are aspirated.

    The third issue is the handling of the samples after aspiration.

    Upon collection, certain physicochemical parameters should be

    measured immediately (e.g. pH and buffer capacity) and, to

    maintain the composition of the aspirated sample until the

    solubility experiment is performed, it is necessary to deactivateenzymes immediately using a method that itself only minimally

    (or more preferably, not at all) affects the composition of the

    sample, before storing the sample under deep-freeze conditions

    (20 C or lower).

    Individual or pooled aspirates can be used for solubility

    estimations. Individual samples have the advantage of enabling

    correlations with levels of specific components in each sample

    to be developed and thus to determine the most important

    factors affecting luminal solubility of the compound in question.

    Pooled samples from a large number of volunteers offer greater

    volumes, and results for solubility in the GI fluids can be

    compared across a set of compounds. If samples are to be

    pooled, the volume taken from each individual sample shouldbe held constant, to ensure that the pooled sample is equally

    representative of all subjects.

    The fourth issue is the methodology for measuring solubility.

    To date, all available drug solubility data in human GI fluids

    have been measured by equilibrium solubility methods which

    are generally preferred to kinetic solubility measurements[17].

    When measuring equilibrium solubility, an excess of pure drug

    powder (typically two to three times higher than the expected

    amount to needed to saturate the medium[36]) should be used.

    On the one hand, the time allowed to reach equilibrium should

    be as short as possible to minimize composition changes in the

    aspirate. On the other hand, enough time should be allowed toenable the system to attain equilibrium. In some cases, a

    supersaturation may be generated. This can be avoided in many

    cases by using the most stable crystal form of the drug powder

    at the highest possible level of purity. Another problem that can

    arise during the solubility measurement is conversion of the

    drug to another compound, especially if the conversion is

    enzymatically catalyzed [37]. In such cases, kinetic solubility

    measurements [38] may be more useful than the equilibrium

    solubility approach.

    Up till now, measurements of solubility in fed state human

    aspirates have been made in the total luminal contents, with no

    distinction between concentrations achieved in lipid, micellar and

    aqueous phases. One problem with this approach is that it is

    594 J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    5/12

    difficult in some cases to separate the excess solid drug from the

    rest of the sample prior to analysis without simultaneous phase

    separation. The other is that the concentrations achieved in the

    phase driving drug absorption may be over or underestimated if

    only the total concentration is reported, leading to false estimates

    of the expected impact on bioavailability. Postulating that it is the

    micellar phase concentration which drives absorption, it would bepreferable to report solubilities determined in this phase of the

    postprandial luminal contents to predict food effects.

    The fifth issue relates to resources necessary to perform the

    studies. Since collection of aspirates has to be carefully planned

    and executed by highly trained and experienced staff, and since

    the aspirates themselves require special handling to generate

    reproducible and meaningful results, measurement of solubility

    in human aspirates is a tedious and costly process. Hence, use of

    human aspirates for screening the solubility of a large number of

    compounds would not be cost-effective. Therefore, the

    identification of alternative media for estimating luminal

    solubility, e.g. intestinal aspirates from animals or simulatedGI fluids generated in the laboratory, would be highly desirable.

    4. Estimation of drug solubility in the stomach

    4.1. Fasted state, gastric

    Typically, human gastric aspirates in the fasted state

    (HGFfasted) are collected using simple naso- or orogastric

    tubes from the antrum of healthy adult volunteers after a ten

    hour (overnight) fast [27,39]. To better reflect the dosing

    conditions in a standard pharmacokinetic study, it is preferable

    to collect aspirates in suitably fasted subjects after they have

    been administrated a glass of water (250 ml). Assuming thatin a bioavailability study the disintegrated particles of an

    immediate release dosage form as well as any drug that has

    already dissolved will empty from the stomach together with the

    co-administered water, samples to be used for solubility studies

    should be aspirated approximately 15 min to a half-hour after

    water administration [15,23]. This timing will also facilitate

    collection of enough volume (about 20 ml) to carry out a

    solubility measurement. To avoid changes in aspirate compo-

    sition, samples should be deep frozen immediately until

    solubility studies are performed (typically to 80 C)[23,27].

    Solubility data in HGFfasted samples to date have been

    performed in the absence of enzyme inhibitors[15]. To preventmicrobial growth during the solubility experiments, 6 mM

    NaN3 and 0.01 mM chloramphenicol can be added [39].

    However, the effect of these agents on the solubility data has not

    been elucidated. Immediately after equilibration, HGFfastedsamples can be either filtered through 0.45 m regenerated

    cellulose filters (recommended)[15]or centrifuged (5000 rpm,

    1020 min)[39]. Quantification of the dissolved compound is

    typically performed with HPLC and standard curves are

    constructed in the corresponding medium.

    Theonly animal aspirates that have been studied as alternatives

    toHGFfasted media for estimating intra-gastricsolubility are canine

    gastric aspirates collected in the fasted state (CGFfasted)[15]. In

    order to allow for the faster gastric emptying rates in dogs than in

    humans in the fasted state, administration of a little more than

    250 ml of water prior to aspirations with aspiration approximately

    10 min after the water administration is recommended.

    In contrast to the paucity of alternative animal models,

    various simulated media have been proposed[40]. It should be

    noted that media designed to simulate intra-gastric conditions

    and frequently used in biorelevant dissolution studies tend tooverestimate intra-gastric solubility values if they contain

    synthetic surfactants[15,39].

    Fig. 1compares the solubility values of four compounds in

    HGFfasted, CGFfasted and Fasted State Simulating Gastric Fluid

    (FaSSGF), a biorelevant medium that contains only physiolog-

    ically relevant substances[15,40]. CGFfasted, with its higher pH,

    appears to be less useful than FaSSGF for predicting the intra-

    gastric solubility of drugs. However, it is worth noting that

    solubility data in FaSSGF are not always more predictive than

    simple HCl solutions (Fig. 1, felodipine), so the search for a

    ubiquitously applicable in vitro medium for the accurate

    estimation of intra-gastric solubility continues[15].

    4.2. Fed state, gastric

    The problematic aspiration and the heterogenous composi-

    tion of gastric contents after administration of typical solid

    meals, the dramatically changing composition with time after

    administration, and the presence of various phases (i.e. solid,

    aqueous, micellar, and lipid) make drug solubility in the

    stomach difficult to define and measure.

    To facilitate the aspiration procedure and to reduce the

    degree of heterogeneity of gastric contents, aspirates can be

    collected after administration of a liquid meal (HGFfed).Table 2

    shows the composition of two meals that have been recom-mended by the U.S. FDA, the composition of corresponding

    liquid meals that have been used to facilitate aspiration of

    samples from the fed stomach, and the composition of gastric

    contents after administrations of these liquid meals to healthy

    volunteers after a twelve hour fast.

    When using aspirates as the solubility medium, there is an

    important issue with respect to the changing composition of the

    medium during the solubility experiment. Typically, termina-

    tion of lipase activity can be achieved by transferring the

    aspirate to glass vials containing a methanol solution of lipase

    inhibitors (5% v/v: 100 mM diisopropyl fluorophosphate,

    50 mM acetophenone, 250 mM phenylboronic acid) [42]whereas inhibition of pepsin's proteolytic activity can be

    achieved by titrating the sample to pH 1[23]. Finally, to prevent

    any bacterial growth, 5 l of an aqueous solution of 4% NaN3w/v and 5% w/v chloramphenicol per milliliter of gastric

    aspirate can be added [42]. It goes without saying that all of

    these additions can impact the solubility value measured, and,

    therefore, deep-freezing of the aspirates immediately upon

    collection at80 C may alternatively be considered.

    What about the possibility of simulating the digestion

    processin vitro? Indeed, the changing intra-gastric environment

    with time after the meal's administration can be adequately

    reproducedin vitrowith regard to pH and pepsin levels.Fig. 2

    shows two relevant examples, with cow's milk (heat-treated)

    595J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    6/12

    and Ensure Plus as the starting point for the media, in

    comparison with the actual intra-gastric pH profile after

    administration of 500 ml Ensure Plus or a normal solid/liquidmeal to healthy volunteers. With regard to sample work-up,

    cow's milk is much more practical than using Ensure Plus as

    the starting point for digestion. In addition, the lower nutrient

    content of milk compared to Ensure Plus or to meals usually

    administered in drug absorption studies [34] makes it more

    representative of the intra-gastric conditions, where significant

    amounts of secretions (i.e. significant dilutions of the meal)

    occur[23]. At least partly for this reason, although cow's milk

    deviates significantly from the caloric content of typical FDA

    meals [34], the in vitro pH profile with time lies within theexpected intra-gastric values (Fig. 2).

    Another issue when using intra-gastric fluids (either

    simulated or aspirated) in the fed state is that of total

    composition vs. aqueous phase composition. For example,

    when using cow's milk, the aqueous phase may contain lipids,

    casein micelles, and/or casein molecules and its physical

    composition will depend on the pH [43]. Separation of the

    Table 2

    Composition and volumes of standard U.S. FDA meals and of liquid meals in comparison with the resulting intra-gastric composition after administration the liquid

    meals to healthy fasted adults a

    Meal suggested by

    FDA until 2002[34]

    Meal suggested by

    FDA after 2002[41]

    Glucose/Olive oil/

    Egg meal[42]

    HGFfedcomposition after

    Glucose/Olive oil/Egg meal [42]

    Ensure

    Plus [23]

    HGFfed composition after

    Ensure Plus[23]

    Proteins (g/l) 56.5 1516.6% of total

    calories

    33 N.M. 54.9 23.311.2

    (30210 min)

    Carbohydrates

    (g/l)

    142.3 2527.8% of total

    calories

    177 12010 200 152.149.1

    (14 h) (30210 min)

    Lipids (g/l) 52.6 6055.6% of total

    calories

    175 150 (13 h) 53.3 N.M.

    50 (4 h)

    pH 5.3 N.M. 7.0 4.02.5 6.7 6.42.7

    (14 h) (30210 min)

    Calories

    (kcal)

    648 9001000 960 N.A. 750 N.A.

    Volume (ml) 513 N.M. 400 N.A. 500 N.A.

    a N.A.: not applicable; N.M.: not measured.

    Fig. 1. Solubility of ketoconazole, dipyridamole, miconazole and felodipine in HGFfasted, CGFfastedand media simulating intra-gastric conditions in the fasted state[15].

    pHeq represents the pH value measured in the sample after solubility equilibrium had been attainted.

    596 J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    7/12

    aqueous phase from the precipitated (digested) phase, the lipid

    droplets, and the undissolved drug presents another challenge.

    Usually, immediate centrifugation (up to 11,400 g, 10 C,

    10 min) leads to separation of the aqueous phase from

    precipitated/digested phase, most of lipid droplets, and the

    undissolved drug [44]. If centrifugation does not work, i.e. if

    drug solid particles cannot be separated, one must additionally

    filter the supernatant. In this case the filter efficiency has to

    balance the rate of filtration against the ability of the filter to

    exclude undissolved material, and adsorption of the drug to the

    filter has to be ruled out (Diakidou et al. unpublished data). Thepart of the sample to be analyzed (supernatant or filtrate) is then

    treated analogously to a plasma sample, in that a further protein

    precipitation step may be required, and can usually be analyzed

    by standard HPLC techniques. Quantification of the drug in the

    aqueous phase or in the whole (aspirated) medium is performed

    with standard curves constructed in the corresponding medium.

    This means that estimating solubility at various time points will

    typically require the construction of several standard curves (to

    take into account the changing composition with time after meal

    administration).

    As an alternative to measurement of solubility concomitant

    with simulation of digestion, it is also possible to construct aseries of media to simulate the composition of the gastric

    contents at various stages of the digestive process, so-called

    snapshot media. The snapshot media reflect the composi-

    tion of the gastric contents after administration of 500 ml Ensure

    Plus, as reported by Kalantzi et al.[23]early, in the middle and

    late in the digestive process. Parameters important to drug

    solubility and dissolution such as pH, buffer capacity, surface

    activity and osmolarity are all simulated in these media, which

    use cow's milk as the starting point for preparation (Janen

    et al. unpublished data). Such media have the advantage of

    being completely defined, as opposed to starting with a

    composition like cow's milk and simulating digestion, in

    which case reproducibility may be an issue.

    5. Estimation of drug solubility in the small intestine

    5.1. Fasted state, small intestine

    Human intestinal fluids in the fasted state (HIFfasted) are

    usually collected at or about the beginning of the jejunum of

    healthy fasted volunteers[6,8,9,16,23,27,37,39,45]. This site ispreferred, because it is possible to locate the aspiration tubes in

    this region reproducibly (as opposed to the upper and middle

    duodenum), the aspiration tubes can be placed in this region

    reasonably quickly (as opposed to the lower small intestine), and

    the volumes that can be aspirated are greater than is possible at

    more distal locations in the small intestine. Some studies have

    been conducted without prior administration of any water

    [27,37,39,45]. It is also possible to use a procedure that involves

    administration of 180250 ml water before the aspiration is

    started[6,8,9,23]. This comes closer to simulating conditions in

    a bioavailability study. In one version, the water is administered

    via a tube to the antrum of the volunteer and samples areaspirated from the end of the duodenum as depicted in Fig. 3.

    Alternatively, the water can be swallowed and aspirations can be

    performed from a tube that allows access in the small intestine

    [6,8,9]. In the latter case the tube has two lumens, both of which

    end in the small intestine. One is used for aspirating samples and

    the other for alleviation of any pressure reduction generated in

    the intestine by the aspiration procedure. Regardless of the exact

    methodology, aspirates are collected from the end of duodenum/

    start of the jejunum and are typically stored at20 C (or lower),

    until used [16,23,27,39,45]. Deactivation of trypsin can be

    achieved by adding Phenylmethylsulfonyl fluoride (PMSF) to a

    Fig. 3. Representative X-ray picture showing the position of a two-lumen tube in

    the upper GI lumen of a volunteer who was administered a liquid meal into the

    antrum of the stomach (administration ports) with sample aspiration from the

    end of the duodenum (aspiration ports). Details of the specific tube can be foundelsewhere[23].

    Fig. 2. pH values in the stomach after administration of 500 ml Ensure Plus to

    healthy fasted adults [23] (box plots from 30 to 210 min), pH values in the

    stomach after administration of a solid meal (1000 kcal, pH 5.7) to fasted healthy

    subjects[22](box plots from 0 to 240 min), meanSD pH values in 500 ml

    cow's milk after addition of acidic solutions of pepsin in physiologically

    appropriate quantities [44] (), and meanSD pH values of 500 ml Ensure

    Plus after addition of acidic solutions of pepsin in physiologically appropriate

    quantities following the same procedure with that applied in milk ().

    597J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    8/12

    final concentration of 1 mM[23]. Deactivation of lipase can be

    achieved with one of the following methods: the cocktail

    mentioned earlier for deactivation of lipase in the fed stomach

    [42]at double the concentration[35,46]; using tetrahydrolistatin

    (orlistat) at a final concentration of 1 mg/ml[16,47]; or usingp-

    bromophenylboronic acid at a final concentration of 1 mM[48].

    Regardless of the method used, the total volume of inhibitorsolution should be less than 2% (by volume) of the collected

    luminal contents[35]. Sometimes, to prevent microbial growth

    during the solubility tests, 6 mM NaN3 and 0.01 mM

    chloramphenicol are added[39]. As with gastric aspirates, all

    these treatments may affect the finally estimated solubility value

    since they all impact the composition of the aspirated sample.

    However, there are some data suggesting that orlistat does not

    affect solubility data in HIFfasted [16]. To date, solubility data

    in HIFfasted have been measured in aspirates that have been

    treated several different ways: addition of orlistat[16], addition

    of antimicrobial growth agents [39] or without addition of

    chemicals (aspirates were simply deep frozen immediately uponcollection) [14]. After equilibrium, samples are centrifuged

    (500010,000 rpm or at 10,000 g for about 1020 min)

    [14,16,39], and then quantified with HPLC techniques. Per-

    forming solubility studies in individual aspirates, Pedersen

    et al. [39]were able to demonstrate that the total solubility of

    danazol (logP= 4.53) correlated nicely with the bile salt content

    of the aspirate, whereas for the less lipophilic hydrocortisone

    (logP= 1.66) no correlation was seen. These results are

    consistent with solubilization theory for bile salts[49].

    Canine aspirates and simulated media have been proposed to

    avoid the collection and use of human aspirates. One of the

    disadvantages of using canine aspirates is that canine gall

    bladder shows brief alternating excursions of filling andemptying with the number of emptying events exceeding the

    filling events during phases II of the IMMCs[50]and this may

    lead to highly variable estimations of solubility [14].

    Simulated media contain phosphatidylcholine and bile salts

    (usually trihydroxy bile salts of taurine having various levels of

    purity), but usually contain a non-physiological buffer system

    e.g. phosphates. Porter and Charman have reviewed this topic

    [51]. One of the reasons for not using the physiological buffer

    (bicarbonate) is that bicarbonates are unstable with time,

    seeking to come to equilibrium with carbon dioxide in the

    atmosphere. To maintain a constant pH, bicarbonate-containing

    media must be continuously sparged with carbon dioxide andtitrated with sodium hydroxide[52,53]. This leads to changes in

    buffer capacity, ionic strength and osmolality during the

    experiment, changes which themselves are not physiological.

    Due to these practical difficulties, bicarbonates are usually

    replaced with stable buffer systems in media used for solubility

    measurement. The anion in the buffer system may theoretically

    affect the solubility product of a weakly basic compound with a

    pkahigher than 5, the solubility of extremely highly lipophilic

    compounds due to salting in/out properties (of the anion), and/

    or the stability of the dissolving compound[54]. In addition, the

    anion of the buffer system may be important for the conversion

    of a prodrug to its active form [37]. For these reasons,

    biorelevant media with alternative buffer species have been

    suggested[54]. It should be pointed out, however, that to date

    no data have been brought forward showing that the presence of

    phosphates affects estimates of intra-intestinal solubility. In fact,

    Kalantzi et al. were able to demonstrate that the fasted state

    simulating intestinal fluids, originally proposed in 1998 [55],

    are able to predict solubility in human aspirates well [14].The

    use of a crude mixture of bile salts and maleates might slightlyimprove the prediction of intra-lumenal solubility in some cases

    [14].

    5.2. Fed state, small intestine

    Human duodenal aspirates for solubility studies in the fed

    state (HIFfed) have been collected and characterized after

    administration of various liquid meals to the antrum or the

    jejunum of healthy fasted volunteers (Table 2). Aspirated

    samples are typically transferred quickly to glass vials

    containing lipase and trypsin inhibitors as well as agents

    for preventing microbial growth, as described above for theintestinal aspirates collected in the fasted state. However, since

    these treatments may have an impact on the measured

    solubility values, studies to date have been performed by

    adding orlistat only [16] or no inhibitors [23] in the tested

    aspirates.

    Intestinal aspirates in the fed state are less heterogenous than

    the corresponding gastric aspirates and, although their physi-

    cochemical characteristics change with time, the changes are

    not as pronounced as in gastric aspirates. In fact, the solubility

    of ketoconazole and dipyridamole in pooled human aspirates

    collected after administration of Ensure Plus was not

    significantly affected by the time of aspiration, for times up to

    120 min after administration of Ensure Plus[14].The solubility of compounds in HIFfedcan be several orders

    of magnitude higher than the corresponding values in the fasted

    state [14,16]. Although to date all relevant solubility experi-

    ments have been performed using the entire aspirated sample, it

    should be born in mind that a more detailed characterization of

    solubility could be obtained if the phases are separated prior to

    solubility determination. Complete phase separation is more

    difficult in intestinal than in gastric aspirates because fine

    emulsions may have been formed (seeFig. 4).

    If solubility increases are phase-dependent (lipid, micellar,

    aqueous), successful prediction of effects on absorption will be

    contingent on correctly identifying the increase in solubility inthe absorption-relevant phase. For example, if the increase in

    solubility is primarily due to incorporation of the drug in the

    lipid phase, distribution into the aqueous phase may limit the

    increase inin vivodissolution rate in the aqueous phase. On the

    other hand, if the increase in solubility is due to micellar

    solubilization in the aqueous phase, increases in dissolution and

    subsequently absorption rates should follow NoyesWhitney's

    considerations more directly [56]. Even so, the intra-lumenal

    dissolution rate can be slower than that expected from solubility

    data[16], most likely due to the slower diffusion of the micelle-

    bound drug to the bulk solution[57].

    Canine aspirates and simulated media have also been studied

    as alternatives to HIFfedsamples. Persson et al.[16]found good

    598 J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    9/12

    agreement between canine and human results for drug solubility

    in intestinal fluids when small (200 ml) meals were adminis-

    tered to both species. However, when larger meals were

    administered to both species, the solubilities in canine aspirates

    overestimated solubilities in human aspirates. This is probably

    due to the higher levels of bile salts in the postprandial canine

    small intestine and makes extrapolation from canine to human

    data rather precarious[14].An issue with simulated media is the identity of buffer

    species used[54], although in this case (unlike in the fasted state

    scenario) bicarbonate is only one of many buffers contributing

    to the overall buffer capacity. In fact, data published to date

    show that solubility in FeSSIF adequately predicts or only

    slightly underestimates solubility in HIFfed [14]. Based on

    Table 3however, FeSSIF is being redesigned to have a pH value

    higher than 5 and contain less bile salt. Also, as demonstrated in

    various in vitro setups[32,58], the simulated medium should

    contain a biorelevant amount of lipolysis products in order to

    adequately address the influence of meal digestion products on

    solubility. This point is especially relevant for highly lipophilic

    compounds, and a significant amount of work in this direction

    has already been done to help optimize orally administered lipid

    dosage forms[59].

    6. Use of drug dissolution and solubility data in predicting

    plasma profiles

    Various examples in the literature have demonstrated the

    utility of dissolution and solubility data measured under

    biorelevant conditions in the prediction of drug plasma levelsand/or the fraction of drug absorbed[12,60,61,62].

    For compounds with low dose:solubility ratios and which are

    highly permeable, predictions are indeed quite successful, as is the

    case with danazol in the fed state (using dissolution data collected

    with the flow-through apparatus) (Fig. 5), glibenclamide in the

    fasted state (using dissolution data collected with the rotating

    Fig. 5. Mean observed plasma concentration profiles of danazol in the fasted ()

    and in thefedstates() in comparisonwith predictedprofiles that were based on

    dissolution data collected with the flow-through apparatus (32 ml/min) in a

    mediumwith similar composition to HIFfasted composition (e.g. [39]) (),andat

    8 ml/min in a medium with similar composition to HIFfed(e.g.[16,23]) (- - - - -).(Reproduced with permission from Ref.[60]).

    Table 3

    Average composition of contents in the upper small intestine after administration

    of standard meals to healthy fasted adults a

    Intestinal

    composition after

    400 ml glucose/

    olive oil/egg mealb

    [35,46]

    Intestinal

    composition

    after 500 ml

    Ensure Plusb

    [23,14]

    Intestinal composition

    after NuTRIflex

    administration

    (180 ml over

    90 min) c [16]

    Proteins (g/l) N.M. 10 5.0 0.1

    Carbohydrates

    (g/l)

    N.M. 5060 N.M.

    Total neutral

    lipids

    55100 (g/l)

    (14 h)

    45.058.3 mM 221 mM

    (0.53 h)

    Phospholipids 3.05.8 mM 3 0.3 mM

    (0.53 h)

    Bile salts

    (mM)

    6.713.4 (14 h) 11.25.2 8.0 0.1

    (0.53 h)

    pH 67 6.65.2 6.1

    (0.53.5 h)

    a N.M.: not measured.b Please seeTable 2for the exact composition of this meal.c Unlike the other meals on this Table, NuTRIflex was administered directly

    to the jejunum. NuTRIflex composition: pH: 5.4, proteins: 19 g/l, lipids:

    12 mM, phospholipids: 3 mM [16]. 180 ml contains 138 kcal nitrogen 0.8 g,

    amino acids 5.8 g, glucose 11.5 g, and lipids 7.2 g [16].

    Fig. 4. Upper panel: Schematic representation of the various phases in intestinal

    aspirates collected in the fed state (corresponding to the photograph shown in the

    lower panel), after ultracentrifugation. Lower panel: Set of aspirated samples

    from the end of the duodenum after administration of a glucose/olive oil/egg

    meal [35,42,45] to a healthy volunteer. Numbers correspond to minutes after

    administration of the meal. First row shows the samples immediately after

    collection and second row shows the same samples after ultracentrifugation

    (410,174 g, 37 C, 2 h).

    599J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    10/12

    paddle apparatus)[61] and troglitazone in the fed state (using

    dissolution data collected with the rotating paddle apparatus) [12].

    For compounds with high dose:solubility ratios and which

    are highly permeable, predictions to date are successful in the

    fed state, but in the fasted state the plasma profiles appear to be

    affected significantly by the hydrodynamics.Fig. 6illustrates an

    example with atovaquone tablets (Wellvone). The dose:solubility ratios of atovaquone in FaSSIF and FeSSIF have

    been estimated to be 25 and 80 l, respectively. Dissolution data

    collected with the rotating paddle apparatus and biorelevant

    media in combination with the corresponding solubility data led

    to successful prediction of plasma levels in the fed state. In the

    fasted state, predictions, while much better than using

    dissolution results in compendial media, were not as accurate

    [12]. Another example is danazol in the fasted state. Its dose:

    solubility ratio in HIFfasted is about 20 l [16]. By applying a

    simulation methodology similar to that for atovaquone[12], the

    predicted average plasma profiles for Danatrol capsules using

    solubility data in HIFfasted[16]and dissolution data in FaSSIFwith the rotating paddle apparatus [55], provide much better

    predictions than data collected in USP simulated intestinal fluid.

    However, they fail to accurately predict the average maximum

    concentration [60] (Fig. 7). Substantial improvements can be

    achieved using solubilities close to those obtained in HIFfasted[16] and dissolution data obtained with the flow-through

    apparatus[60](Fig. 5).

    Figs. 57in combination with other relevant published data

    suggest that although estimation of intra-lumenal dissolution

    has greatly facilitated our ability to predict intra-lumenal

    performance of solid dosage forms, hydrodynamics may in

    some cases be crucial for accurate predictions of plasma

    levels.

    7. Conclusions

    future directions

    The ability to predict solubility in the upper gastrointestinal

    tract would clearly be advantageous to discovery and

    development programs in the pharmaceutical industry. Since

    direct measurement of luminal concentrations is cumbersome,

    recent efforts have been directed at determination of solubility

    in human gastrointestinal fluids and in developing media which

    can simulate these appropriately. As is evident from the

    foregoing discussion, the collection of aspirates from the

    human intestinal tract is fraught with technical challenges,

    especially in the fed state. The reproducibility of solubility data

    is highly dependent on the aspiration protocol and the way theaspirates are processed and stored. Careful attention also has to

    be given to the experimental procedure of the solubility

    determination itself. Although fluids from animals seem like a

    reasonable way of addressing some of the challenges, results to

    date with dogs have been disappointing. For all these reasons,

    in vitro surrogate media for predicting drug solubility in the

    upper gastrointestinal tract appear to be the way forward. With

    media already available or being developed for the upper

    gastrointestinal tract, the next logical step is to design media to

    represent conditions in the lower regions. Work is currently

    underway to characterize fluids collected from the proximal

    colon and to use these as a basis for design of the corresponding

    biorelevant media.

    Fig. 7. Mean danazol plasma levels after single dose administration of one

    Danatrol capsule (100 mg danazol per capsule) with 200 ml of water to nine

    healthy fasted volunteers[60]() and predicted profiles that were constructed

    using a simulation procedure identical with that used in Ref.[12]and dissolution

    data collected with the rotating paddle apparatus (100 rpm) in USP simulated

    intestinal fluid (.....) and in FaSSIF ()[55].

    Fig. 6. (A) Median observed plasma data in the fasted state after single

    administration of two Wellvone tablets to healthy subjects () and predicted

    profiles that were based on dissolution data collected with the rotating paddle

    apparatus (100 rpm) in USP simulated intestinal fluid (.....) and in FaSSIF ().

    (B) Median observed plasma data in the fed state after single administration of

    two Wellvone tablets to healthy subjects () and predicted profiles that were

    based on dissolution data collected with the rotating paddle apparatus (100 rpm)

    in USP simulated intestinal fluid (.....) and in FeSSIF (

    ). (Reproduced withpermission from Ref.[12]).

    600 J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    11/12

    References

    [1] J. Dressman, J. Butler, J. Hempenstall, C. Reppas, The BCS: where do we

    go from here? Pharm. Technol. 25 (2001) 6876.

    [2] H. Potthast, J.B. Dressman, H.E.Junginger, K.K. Midha, H. Oeser, V.P. Shah,

    H. Vogelpoel, D.M. Barends, Biowaiver monographs for immediate release

    solid oral dosage forms: ibuprofen, J. Pharm. Sci. 94 (2005) 21212131.

    [3] E.S. Kostewicz, M. Wunderlich,U. Brauns, R. Becker, T. Bock, J.B.Dressman,Predicting the precipitation of poorly soluble weak bases upon entry in the

    small intestine, J. Pharm. Pharmacol. 56 (2004) 4351.

    [4] B. Abrahamsson, T. Albery, A. Eriksson, I. Gustafsson, M. Sjoeberg, Food

    effects on tablet disintegration, Eur. J. Pharm. Sci. 22 (2004) 165172.

    [5] L. Kalantzi, B. Polentarutti, T. Albery, D. Laitmer, B. Abrahamsson,

    J. Dressman, C. Reppas, The delayed dissolution of paracetamol products in

    thecanine fed stomach can bepredictedin vitrobut it does notaffect theonset

    of plasma levels. Int. J. Pharm. 296 (2005) 8793.

    [6] J. Brouwers, J. Tack, P. Augustijns, Parallel monitoring of plasma and

    intraluminal drug concentrations in man after oral administration of

    fosamprenavir in the fasted and fed state, Pharm. Res. 19 (April 2007)

    online.

    [7] L. Bonlokke, F.N. Christensen, L. Knutson, H.G. Kristensen, H. Lennernas,

    A newapproach fordirect in vivo dissolution studies of poorly soluble drugs,

    Pharm. Res. 14 (1997) 14901492.[8] J. Brouwers, F. Ingels, J. Tack, P. Augustijns, Determination

    of intraluminal theophylline concentrations after oral intake of an

    immediate- and a slow-release dosage form, J. Pharm. Pharmacol. 57

    (2005) 987996.

    [9] J. Brouwers, J. Tack, F. Lammert, P. Augustijns, Intraluminal drug and

    formulation behavior and integration in in vitro permeability estimation: a

    case study with amprenavir, J. Pharm. Sci. 95 (2006) 372383.

    [10] M. Vertzoni, K. Goumas, V. Kalioras, M. Symillides, H. Archontaki,

    C. Reppas, Danazol levels in the various phases of intraduodenal contents

    versus plasma concentrations after single oraladministrations in the fed state,

    Preliminary Data, AAPS Annual Meeting, Poster # T2197, San Antonio,

    2006.

    [11] W. Weitschies, R.S. Wedemeyer, O. Kosch, K. Fach, S. Nagel, E. Sderlin,

    L. Trahms, B. Abrahamsson, H. Mnnikes, Impact of the intragastric

    location of extended release tablets on food interactions, J. Control.Release 108 (2005) 375385.

    [12] E. Nicolaides, M. Symillides, J.B. Dressman, C. Reppas, Biorelevant

    dissolution testing to predict the plasma profile of lipophilic drugs after

    oral administration, Pharm. Res. 18 (2001) 380388.

    [13] G.L. Amidon, H. Lennernas, V.P. Shah, J.R. Crison, A theoretical basis for

    a biopharmaceutics drug classification: the correlation of in vitro drug

    product dissolution and in vivo bioavailability, Pharm. Res. 12 (1995)

    413420.

    [14] L. Kalantzi, E. Persson, B. Polentarutti, B. Abrahamsson, K. Goumas,

    J. Dressman, C. Reppas, Canine intestinal contents vs simulated media

    for the assessment of solubility of two weak bases in the human small

    intestinal contents, Pharm. Res. 23 (2006) 13731381.

    [15] M. Vertzoni, E. Pastelli, D. Psachoulias, L. Kalantzi, C. Reppas,

    Estimation of intragastric solubility of drugs: In what medium? Pharm.

    Res. 24 (2007) 909917.[16] E. Persson, A.S. Gustafsson, A. Carlsson, R. Nilsson, L. Knuston, P. Forsell,

    G. Hanish, H. Lennerns, B. Abrahamsson, The effects of food on the

    dissolution of poorly soluble drugs in human and in model small intestine

    fluids, Pharm. Res. 22 (2005) 21412151.

    [17] S.N. Bhattachar, L.A. Deschenes, J.A. Wesley, Solubility: it's not just for

    physical chemists, Drug Discov. Today 11 (2006) 10121018.

    [18] A. Avdeef, Solubility of sparingly-soluble ionizable drugs, Adv. Drug Del.

    Rev. 59 (2007) 568590 (this issue).

    [19] M. Morihara, N. Aoyagi, N. Kaniwa, S. Kojima, H. Ogata, Assessment of

    gastric acidity of Japanese subjects over the last 15 years, Biol. Pharm.

    Bull. 24 (2001) 313315.

    [20] T.L. Russell, J.L. Barnett, R.R. Berardi, L.C. Dermentzoglou, K.M. Jarvenpaa,

    S.S. Schmaltz, J.B. Dressman, Upper gastrointestinal pH in seventy-nine

    healthy North American men and women N65 years, Pharm. Res. 10 (1993)

    187196.

    [21] J. del Valle, T. Yamada, ZollingerEllison syndrome, in: T. Yamada, et al.,

    (Eds.), Textbook of Gastroenterology, 62, J.B. Lippincott Co., USA, 1991,

    pp. 13401452.

    [22] J.B. Dressman, R.R. Berardi, L.C. Dermentzoglou, T.L. Russell, S.P.

    Schmaltz, J.L. Barnett, K.M. Jarvenpaa, Upper gastrointestinal (GI) pH in

    young healthy men and women, Pharm. Res. 7 (1990) 756761.

    [23] L. Kalantzi, K. Goumas, V. Kalioras, B. Abrahamsson, J. Dressman, C. Reppas,

    Characterization of the human upper gastrointestinal contents underconditions simulating bioavailability/bioequivalence studies, Pharm. Res.

    23 (2006) 165176.

    [24] D.F. Evans, G. Pye, R. Bramley, A.G. Clark, T.J. Dyson, J.D. Hardcastle,

    Measurement of gastrointestinal pH profiles in normal ambulant human

    subjects, Gut 29 (1988) 10351041.

    [25] T.A. Brasitus, M.D. Stirn, Short bowel syndrome, Chapter 74, pp. 15411554,

    Textbook of Gastroenterology, Chief Editor T. Yamada, JB Lippincott Co.,

    New York, 1991.

    [26] S.G. Nugent, D. Kumar, D.S. Rampton, D.F. Evans, Intestinal luminal pH

    in inflammatory bowel disease: possible determinants and implications for

    therapy with aminosalisylates and other drugs, Gut 48 (2001) 571577.

    [27] A. Lindahl, A.L. Ungell, L. Knuston, H. Lennernas, Characterization of

    fluids from the stomach and proximal jejunum in men and women, Pharm.

    Res. 14 (1997) 497502.

    [28] P.E. Macheras, M.A. Koupparis, S.G. Antimisiaris, Effect of temperatureand fat content on the solubility of hydrochlorothiazide and chlorothiazide

    in milk, J. Pharm. Sci. 78 (1989) 933936.

    [29] P.E. Macheras, M.A. Koupparis, S.G. Antimisiaris, Drug binding and

    solubility in milk, Pharm. Res. 7 (1990) 537541.

    [30] Alendronic Acid, Martindale: The Complete Drug Reference, 34th edition,

    Pharmaceutical Press, London, 2005, pp. 765766.

    [31] S.D. Mithani, V. Bakatselou, C.N. TenHoor, J.B. Dressman, Prediction of

    increase in solubility of drugs as a function of bile salt concentration,

    Pharm Res 13 (1996) 163167.

    [32] G. Kossena, B. Boyd, C. Porter, W. Charman, Separationand characterization

    of the colloidal phases produced on digestion of common formulation lipids

    and assessment of their impact on the apparent solubility of selected poorly

    water-soluble drugs, J. Pharm. Sci. 92 (2003) 634648.

    [33] H. Davenport, Physiology of the Digestive Tract, 5th edition, Yearbook

    medical Publishers, Chicago, 1982, pp. 155173.[34] S. Klein, J. Butler, J. Hempenstall, C. Reppas, J.B. Dressman, Media to

    simulate postprandial stomach I. Matching the physicochemical character-

    istics of standard breakfasts, J. Pharm. Pharmacol. 56 (2004) 605610.

    [35] O. Hernell, J. Staggers, M. Carey, Physicalchemical behaviour of dietary

    and biliary lipids during intestinal digestion and absorption. 2. Phase

    analysis and aggregation states of luminal lipids during duodenal fat

    digestion in healthy adult human beings, Biochemistry 29 (1990)

    20412056.

    [36] K. Kawakami, K. Miyoshi, Y. Ida, Impact of the amount of excess solids

    on appararent solubility, Pharm. Res. 22 (2005) 15371543.

    [37] J. Brouwers, J. Tack, F. Lammert, P. Augustijns, In vitro behavior of a

    phosphate ester prodrug of amprenavir in human intestinal fluids and in the

    Caco-2 system: illustration of intraluminal supersaturation, Int. J. Pharm.

    95 (2006) 372383.

    [38] J. Alsenz, M. Kansy, High throughput solubility measurement in drugdiscovery and development, Adv. Drug Del. Rev. 59 (2007) 546567

    (this issue).

    [39] B.L. Pedersen, A. Mllertz, H. Brndsted, H.G. Kristensen, A comparison

    of the solubility of danazol in human and simulated gastrointestinal fluids,

    Pharm. Res. 17 (2000) 891894.

    [40] M. Vertzoni, J. Dressman, J. Butler, J. Hempenstall, C. Reppas, Simulation

    of fasting gastric conditions and its importance for the in vivo dissolution

    of lipophilic compounds, Eur. J. Pharm. Biopharm. 60 (2005) 413417.

    [41] Guidance for Industry, Food Effect, Bioavailability and Fed Bioequiva-

    lence Studies, U.S. Department of Health and Human Services, FDA,

    CDER, December 2002.

    [42] M. Armand, P. Borel, B. Pasquier, C. Dubois, M. Senft, M. Andre, J. Peyrot,

    J. Salducci, D. Lairon, Physicochemical characteristics of emulsions during

    fat digestion in human stomach and duodenum, Am. J. Phys. 271 (1996)

    G172G183.

    601J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602

  • 8/11/2019 Solubility Estimation in GIT

    12/12

    [43] P. Walstra, R. Jennes, H.T. Badings, Dairy Chemistry and Physics, John

    Wiley and Sons, New York, 1984, pp. 89270.

    [44] N. Fotaki, M. Symillides, C. Reppas, Canine versus in vitro data for

    predicting input profiles of L-sulpiride after oral administration, Eur. J.

    Pharm. Sci. 26 (2005) 324333.

    [45] M. Perez de la Cruz Moreno, M. Oth, S. Deferme, F. Lammert, J. Tack,

    J. Dressman, P. Augustijns, Characterization of fasted-state human

    intestinal fluids collected from duodenum and jejunum, J. Pharm.Pharmacol. 58 (2006) 10791089.

    [46] M. Armand, P. Borel, C. Dubois, M. Senft, J. Peyrot, J. Salducci, H. Lafont,

    D. Lairon, Characterization of emulsions and lipolysis of dietary lipids in the

    human stomach, Am. J. Phys. 266 (1994) G372G381.

    [47] F. Carriere,C. Renou, S. Ransac, V. Lopez,J. De Caro, F. Ferrato, A. De Caro,

    A. Fleury, P. Sanwald-Ducray, H. Lengsfeld, C. Beglinger, P. Hadvary,

    R. Verger, R. Laugier, Inhibition of gastrointestinal lipolysis by Orlistat

    during digestion of test meals in healthy volunteers, Am. J. Physiol.

    Gastrointest. Liver Physiol. 281 (2001) G16G28.

    [48] D.R. Fine, P.L. Zentler-Munro, T.C. Northfield, Three different methods of

    inhibiting lipolysis in human chyme in vitro: efficiency and effect on phase

    distribution of lipids, Clin. Sci. 79 (1990) 349355.

    [49] A. Glomme, J. Mrz, J.B. Dressman, in: B. Testa, S. Krmer, H. Wunderli-

    Allensprach, G. Folkers (Eds.), Predicting the Intestinal Solubility of

    Poorly Soluble Drugs, Pharmacokinetic Profiling in Drug Research,Wiley-VCH, (Zurich), 2006, pp. 259280, ISBN 3-906390-35-7.

    [50] H. Abiru, S.K. Sarna, R.E. Condon, Contractile mechanisms of gallbladder

    filling and emptying in dogs, Gastroenterology 106 (1994) 16521661.

    [51] C. Porter, W. Charman, In vitro assessment of oral lipid based

    formulations, Adv. Drug Deliv. Rev. 50 (2001) S127S147.

    [52] D. McNamara, K. Whitney, S. Goss, Use of a physiologic bicarbonate

    buffer system for dissolution characterization of ionisable drugs, Pharm.

    Res. 20 (2003) 16411646.

    [53] H.M. Fadda, A.W. Basit, Dissolution of pH responsive formulations in

    media resembling intestinal fluids: bicarbonate versus phosphate buffers,

    J. Drug Del. Sci. Tech. 15 (2005) 273279.

    [54] M. Vertzoni, N. Fotaki, E. Kostewicz, E. Stippler, C. Leuner, E.

    Nicolaides, J. Dressman, C. Reppas, Dissolution media simulating the

    intralumenal composition of the small intestine: physiological issues and

    practical aspects, J. Pharm. Pharmacol. 56 (2004) 453462.

    [55] E. Galia, E. Nicolaides, D. Horter, R. Lobenberg, C. Reppas, J.B. Dressman,

    Evaluation of variousdissolution media forpredicting in vivoperformance of

    class I and II drugs, Pharm. Res. (1998) 698705.

    [56] J.B. Dressman, K. Schamp, K. Beltz, J. Alsenz, Characterizing releasefrom lipid based formulations, in: D. Hauss (Ed.), Lipid-Based Formula-

    tions for Oral Drug Delivery: Enhancing the Bioavailability of Poorly

    Water-Soluble Drugs, in press, Chapter 10, 241255.

    [57] P. Macheras, C. Reppas, Dissolution and in vitro penetration behaviours of

    dicoumarol, nitrofurantoin and sulfamethizole in the presence of protein,

    Int. J. Pharm. 37 (1987) 103112.

    [58] J.. Christensen, K. Schultz, B. Mollgaard, H.G. Kristensen, A. Mullertz,

    Solubilisation of poorly water-soluble drugs during in vitro lipolysis of

    medium- and long-chain triacylglycerols, Eur. J. Pharm. Sci. 23 (2004)

    287296.

    [59] N.H. Zangenberg, A. Mllertz, H.G. Kristensen, L. Hovgaard, A dynamic

    in vitro lipolysis model II: evaluation of the model, Eur. J. Pharm. Sci. 14

    (2001) 237244.

    [60] V.H. Sunesen, B.L. Pedersen, H.G. Kristensen, A. Mllertz, In vivo in vitro

    correlations for a poorly soluble drug danazol using the flow-throughdissolution method with biorelevant dissolution media, Eur. J. Pharm. Sci.

    24 (2005) 305313.

    [61] R. Lbenberg, J. Krmer, V. Shah, G.L. Amidon, J.B. Dressman, Dissolution

    as a prognostic tool for oral drug absorption: dissolution behaviour of

    glibenclamide a case II compound, Pharm. Res. 17 (2000) 439444.

    [62] R. Takano, K. Sugano, A. Higashida, Y. Hayashi, M. Machida, Y. Aso,

    S. Yamashita, Oral absorption of poorly water-soluble drugs: computer

    simulation of fraction absorbed in humans from a miniscale dissolution

    test, Pharm. Res. 23 (2006) 11441156.

    602 J.B. Dressman et al. / Advanced Drug Delivery Reviews 59 (2007) 591602